Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 362
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34462350

RESUMO

Intraventricular hemorrhage (IVH) results in periventricular inflammation, hypomyelination of the white matter, and hydrocephalus in premature infants. No effective therapy exists to prevent these disorders. Peroxisome proliferator activated receptor-γ (PPAR-γ) agonists reduce inflammation, alleviate free radical generation, and enhance microglial phagocytosis, promoting clearance of debris and red blood cells. We hypothesized that activation of PPAR-γ would enhance myelination, reduce hydrocephalus, and promote neurological recovery in newborns with IVH. These hypotheses were tested in a preterm rabbit model of IVH; autopsy brain samples from premature infants with and without IVH were analyzed. We found that IVH augmented PPAR-γ expression in microglia of both preterm human infants and rabbit kits. The treatment with PPAR-γ agonist or PPAR-γ overexpression by adenovirus delivery further elevated PPAR-γ levels in microglia, reduced proinflammatory cytokines, increased microglial phagocytosis, and improved oligodendrocyte progenitor cell (OPC) maturation in kits with IVH. Transcriptomic analyses of OPCs identified previously unrecognized PPAR-γ-induced genes for purinergic signaling, cyclic adenosine monophosphate generation, and antioxidant production, which would reprogram these progenitors toward promoting myelination. RNA-sequencing analyses of microglia revealed PPAR-γ-triggered down-regulation of several proinflammatory genes and transcripts having roles in Parkinson's disease and amyotrophic lateral sclerosis, contributing to neurological recovery in kits with IVH. Accordingly, PPAR-γ activation enhanced myelination and neurological function in kits with IVH. This also enhanced microglial phagocytosis of red blood cells but did not reduce hydrocephalus. Treatment with PPAR-γ agonist might enhance myelination and neurological recovery in premature infants with IVH.


Assuntos
Hemorragia Cerebral Intraventricular/metabolismo , Proteínas da Mielina/biossíntese , PPAR gama/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/deficiência , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Animais Recém-Nascidos , Antiporters/deficiência , Antiporters/metabolismo , Hemorragia Cerebral Intraventricular/patologia , Modelos Animais de Doenças , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/metabolismo , Humanos , Recém-Nascido Prematuro , Microglia/metabolismo , Doenças Mitocondriais/metabolismo , Oligodendroglia/patologia , PPAR gama/agonistas , Transtornos Psicomotores/metabolismo , Coelhos , Rosiglitazona/farmacologia , Análise de Sequência de RNA/métodos
2.
Int J Mol Sci ; 22(11)2021 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-34071138

RESUMO

Schwann cells play an important role in peripheral nerve function, and their dysfunction has been implicated in the pathogenesis of diabetic neuropathy and other demyelinating diseases. The physiological functions of insulin in Schwann cells remain unclear and therefore define the aim of this study. By using immortalized adult Fischer rat Schwann cells (IFRS1), we investigated the mechanism of the stimulating effects of insulin on the cell proliferation and expression of myelin proteins (myelin protein zero (MPZ) and myelin basic protein (MBP). The application of insulin to IFRS1 cells increased the proliferative activity and induced phosphorylation of Akt and ERK, but not P38-MAPK. The proliferative potential of insulin-stimulated IFRS1 was significantly suppressed by the addition of LY294002, a PI3 kinase inhibitor. The insulin-stimulated increase in MPZ expression was significantly suppressed by the addition of PD98059, a MEK inhibitor. Furthermore, insulin-increased MBP expression was significantly suppressed by the addition of LY294002. These findings suggest that both PI3-K/Akt and ERK/MEK pathways are involved in insulin-induced cell growth and upregulation of MPZ and MBP in IFRS1 Schwann cells.


Assuntos
Insulina/farmacologia , Células de Schwann/efeitos dos fármacos , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Cromonas/farmacologia , Neuropatias Diabéticas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Morfolinas/farmacologia , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptor de Insulina/biossíntese , Receptor de Insulina/genética , Transdução de Sinais/efeitos dos fármacos
3.
Hepatol Commun ; 5(2): 334-348, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33553979

RESUMO

CKLF-like MARVEL transmembrane domain containing 6 (CMTM6) was identified as a regulator of programmed death ligand 1 (PD-L1), which induces antitumor immunity in several cancers. This study aimed to clarify the relationship between CMTM6 and PD-L1 expression and clinical outcomes in patients with hepatocellular carcinoma (HCC). In total, 259 patients with HCC who had undergone hepatic resection were enrolled. Immunohistochemical staining for CMTM6 and PD-L1 was performed. The relationships between CMTM6 expression and the clinicopathological characteristics and outcomes were analyzed. Additionally, the stabilization of PD-L1 expression and regulation of malignant activities by CMTM6 were examined in vitro. Our patients were divided into high (n = 65, 25.1%) and low (n = 194, 74.9%) CMTM6 expression groups. High CMTM6 expression was significantly associated with malignant aggregates, including poor differentiation (P < 0.0001), microscopic intrahepatic metastasis (P = 0.0369), and multiple intrahepatic recurrences (P = 0.0211). CMTM6 expression was significantly correlated with PD-L1 expression in HCC tissues (P < 0.0001). The patients were classified into three groups: high CMTM6/PD-L1 positive (n = 21), high CMTM6/ PD-L1 negative (n = 44), and low CMTM6 (n = 194) expression pattern groups. Overall survival was significantly different among the three groups (P < 0.0001). Additionally, immunohistochemical double staining revealed that CMTM6 and PD-L1 were co-expressed on HCC cells. In vitro, PD-L1 expression was enhanced at late time points in the presence of CMTM6 expression. CMTM6 also regulated epithelial-to-mesenchymal transition and stemness phenotypes in HCC cells. Conclusion: Our large cohort study found that CMTM6 co-expressed with PD-L1 was strongly associated with the clinical outcome in patients with HCC. The evaluation of CMTM6 combined with PD-L1 in HCC might be useful for patient selection in immune checkpoint therapy.


Assuntos
Antígeno B7-H1/biossíntese , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas com Domínio MARVEL/biossíntese , Proteínas da Mielina/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Estudos de Coortes , Transição Epitelial-Mesenquimal , Feminino , Humanos , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico
5.
J Neuroimmune Pharmacol ; 16(1): 169-180, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-31776836

RESUMO

Despite the introduction of antiretroviral (ARV) therapy (ART), approximately 30-50% of people living with human immunodeficiency virus-1 (HIV-1) will develop a spectrum of measurable neurocognitive dysfunction, collectively called HIV-associated neurocognitive disorder (HAND). While the clinical manifestations of HAND have changed with the advent of ART, certain pathological features have endured, including white matter alterations and dysfunction. The persistence of white matter alterations in the post-ART era suggests that ARV drugs themselves may contribute to HAND pathology. Our group has previously demonstrated that two ARV compounds from the protease inhibitor (PI) class, ritonavir and lopinavir, inhibit oligodendrocyte maturation and myelin protein production. We hypothesized that other members of the PI class, saquinavir and darunavir, could also negatively impact oligodendrocyte differentiation. Here we demonstrate that treating primary rat oligodendrocyte precursor cells with therapeutically relevant concentrations of either ARV drug results in a concentration-dependent inhibition of oligodendrocyte maturation in vitro. Furthermore, we show that acidifying endolysosomal pH via a mucolipin transient receptor potential channel 1 (TRPML1) agonist provides protection against saquinavir- and darunavir-induced inhibition of oligodendrocyte maturation. Moreover, our findings suggest, for the first time, an imperative role of proper endolysosomal pH in regulating OL differentation, and that therapeutic targeting of endolysosomes may provide protection against ARV-induced oligodendrocyte dysregulation. Graphical Abstract Treatment of primary rat oligodendrocyte precursor cells with therapeutically relevant concentrations of either antiretroviral compound of the protease inhibitor class, darunavir or saquinavir, results in a concentration-dependent inhibition of oligodendrocyte maturation in vitro. Additionally, in darunavir or saquinavir-treated cultures we observed a concentration-dependent decrease in the number of acidic lysosomes, via immunostaining with LysoTracker Red, compared with vehicle-treated cultures. Finally, we showed that acidifying endolysosomal pH via a mucolipin transient receptor potential channel 1 (TRPML1) agonist provides protection against saquinavir- or darunavir-induced inhibition of oligodendrocyte maturation. Our findings suggest, for the first time, a critical role of proper endolysosomal pH in regulating OL differentation, and that therapeutic targeting of endolysosomes may provide protection against antiretroviral-induced oligodendrocyte dysregulation.


Assuntos
Darunavir/farmacologia , Endossomos/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , Lisossomos/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Saquinavir/farmacologia , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Darunavir/toxicidade , Depressão Química , Relação Dose-Resposta a Droga , Endossomos/química , Inibidores da Protease de HIV/toxicidade , Concentração de Íons de Hidrogênio , Lisossomos/química , Proteínas da Mielina/biossíntese , Estresse Oxidativo , Ftalimidas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Saquinavir/toxicidade , Canais de Potencial de Receptor Transitório/agonistas
6.
Exp Neurol ; 335: 113486, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32991932

RESUMO

Myelination is extremely important in achieving neural function. Hypomyelination causes a variety of neurological diseases. However, little is known about how hypomyelination occurs. Here we investigated the effect of dendritic cell factor 1(Dcf1) on myelination, using in vitro and in vivo models and found that Dcf1 is essential for normal myelination, motor coordination and balance. Lack of Dcf1 downregulated myelin-associated proteins, such as myelin basic protein (MBP), myelin associated glycoprotein (MAG), and 2'3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) in the hippocampus and corpus callosum of Dcf1-null mice, as a result, the myelin sheath of these mice became thinner. Transmission electron microscopy revealed hypomyelination in Dcf1-deficient mice. Motor coordination and balance tests confirmed impaired neurological function in Dcf1-null mice. Gain-of-function analysis via In utero electroporation showed that hypomyelination could be rescued by re-expression of Dcf1 in Dcf1-null mouse brain. Dcf1-null mice exhibited a phenotype similar to that of cuprizone-induced demyelinated mice, thereby supporting the finding of hypomyelination caused by Dcf1 knockout. Mechanistically, we further revealed that insufficient Dcf1 leads to hyperactivation of the Wnt/ß-catenin signaling pathway. Our work describes the role of Dcf1 in maintaining normal myelination, and this could help improve the current understanding of hypomyelination and its pathogenesis.


Assuntos
Doenças Desmielinizantes/genética , Proteínas de Membrana/genética , Bainha de Mielina/genética , Proteínas do Tecido Nervoso/genética , Via de Sinalização Wnt/genética , Animais , Corpo Caloso/metabolismo , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/psicologia , Regulação para Baixo/genética , Eletroporação , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas da Mielina/biossíntese , Equilíbrio Postural , Desempenho Psicomotor
7.
Cancer Immunol Immunother ; 70(2): 417-429, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32770259

RESUMO

The "macrotrabecular-massive" (MTM) pattern of hepatocellular carcinoma (HCC) has been suggested to represent a distinct HCC subtype and is associated with specific molecular features. Since the immune microenvironment is heterogenous in HCC, it is important to evaluate the immune microenvironment of this novel variant. CMTM6, a key regulator of PD-L1, is an important immunocheckpoint inhibitor. This study aimed to evaluate the prognostic effect of CMTM6/PD-L1 coexpression and its relationship with inflammatory cells in HCC. We analyzed 619 HCC patients and tumors were classified into MTM and non-MTM HCC subtypes. The expression levels of CMTM6 and PD-L1 in tumor and inflammatory cells were evaluated by immunohistochemistry. The density of inflammatory cells in the cancer cell nest was calculated. Tumoral PD-L1 expression and inflammatory cell density were higher in the MTM type than in the non-MTM type. CMTM6-high expression was significantly associated with shorter OS and DFS than CMTM6-low expression in the whole HCC patient population and the MTM HCC patient population. Moreover, MTM HCC patients with CMTM6/PD-L1 coexpression experienced a higher risk of HCC progression and death. In addition, CMTM6/PD-L1 coexpression was shown to be related to a high density of inflammatory cells. Notably, a new immune classification, based on CMTM6/PD-L1 coexpression and inflammatory cells, successfully stratified OS and DFS in MTM HCC. CMTM6/PD-L1 coexpression has an adverse effect on the prognosis of HCC patients, especially MTM HCC patients. Our study provides evidence for the combination of immune status assessment with anti-CMTM6 and anti-PD-L1 therapy in MTM HCC patients.


Assuntos
Antígeno B7-H1/biossíntese , Carcinoma Hepatocelular/imunologia , Neoplasias Hepáticas/imunologia , Proteínas com Domínio MARVEL/imunologia , Proteínas da Mielina/imunologia , Adolescente , Adulto , Idoso , Antígeno B7-H1/imunologia , Carcinoma Hepatocelular/patologia , Feminino , Humanos , Imunofenotipagem , Proteínas com Domínio MARVEL/biossíntese , Masculino , Pessoa de Meia-Idade , Proteínas da Mielina/biossíntese , Prognóstico , Estudos Retrospectivos , Análise de Sobrevida , Adulto Jovem
8.
J Biol Chem ; 295(34): 11963-11970, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32647009

RESUMO

Charcot-Marie-Tooth disease (CMT) is a neuropathy of the peripheral nervous system that afflicts ∼1:2500 people. The most common form of this disease (CMT1A, 1:4000) is associated with duplication of chromosome fragment 17p11.2-12, which results in a third WT PMP22 allele. In rodent models overexpressing the PMP22 (peripheral myelin protein 22) protein and in dermal fibroblasts from patients with CMT1A, PMP22 aggregates have been observed. This suggests that overexpression of PMP22 under CMT1A conditions overwhelms the endoplasmic reticulum quality control system, leading to formation of cytotoxic aggregates. In this work, we used a single-cell flow-cytometry trafficking assay to quantitatively examine the relationship between PMP22 expression and trafficking efficiency in individual cells. We observed that as expression of WT or disease variants of PMP22 is increased, the amount of intracellular PMP22 increases to a greater extent than the amount of surface-trafficked protein. This was true for both transiently transfected cells and PMP22 stable expressing cells. Our results support the notion that overexpression of PMP22 in CMT1A leads to a disproportionate increase in misfolding and mistrafficking of PMP22, which is likely a contributor to disease pathology and progression.


Assuntos
Doença de Charcot-Marie-Tooth/metabolismo , Regulação da Expressão Gênica , Proteínas da Mielina/biossíntese , Agregação Patológica de Proteínas/metabolismo , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Células HEK293 , Humanos , Proteínas da Mielina/genética , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Transporte Proteico
9.
Neuromolecular Med ; 22(3): 420-436, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32638208

RESUMO

Neonatal sepsis is associated with cognitive deficit in the later life. Axonal myelination plays a pivotal role in neurotransmission and formation of learning and memory. This study aimed to explore if systemic lipopolysaccharide (LPS) injection would induce hypomyelination in the prefrontal cortex and hippocampus in developing septic neonatal rats. Sprague-Dawley rats (1-day old) were injected with LPS (1 mg/kg) intraperitoneally. By electron microscopy, axonal hypomyelination was evident in the subcortical white matter and hippocampus. The expression of myelin proteins including CNPase, MBP, PLP and MAG was downregulated in both areas of the brain at 7, 14 and 28 days after LPS injection. The frequency of MBP and PLP-positive oligodendrocyte was significantly reduced using in situ hybridization in the cerebral cortex and hippocampus at the corresponding time points after LPS injection, whereas the expression of NG2 and PDGFRα was noticeably increased. In tandem with this was reduction of Olig1 and Olig2 expressions which are involved in differentiation/maturation of OPCs. Expression of NFL, NFM, and NFH was significantly downregulated, indicating that axon development was disrupted after LPS injection. Morris Water Maze behavioral test, Open field test, Rotarod test, and Pole test were used to evaluate neurological behaviors of 28 days rats. The rats in the LPS group showed the impairment of motor coordination, balance, memory, and learning ability and represented bradykinesia and anxiety-like behavior. The present results suggest that following systemic LPS injection, differentiation/maturation of OPCs was affected which may be attributed to the inhibition of transcription factors Olig1 and Olig2 expression resulting in impairment to axonal development. It is suggested that this would ultimately lead to axonal hypomyelination in the prefrontal cortex and hippocampus, which may be associated with neurological deficits in later life.


Assuntos
Hipocampo/patologia , Proteínas da Mielina/deficiência , Transtornos do Neurodesenvolvimento/etiologia , Córtex Pré-Frontal/patologia , Sepse/patologia , Substância Branca/patologia , Animais , Animais Recém-Nascidos , Ansiedade/etiologia , Axônios/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Hipocampo/metabolismo , Hipocinesia/etiologia , Lipopolissacarídeos/toxicidade , Masculino , Transtornos da Memória/etiologia , Microscopia Eletrônica , Teste do Labirinto Aquático de Morris , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia , Fator de Transcrição 2 de Oligodendrócitos/biossíntese , Fator de Transcrição 2 de Oligodendrócitos/genética , Oligodendroglia/patologia , Teste de Campo Aberto , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Teste de Desempenho do Rota-Rod , Sepse/complicações , Sepse/metabolismo
10.
Sci Rep ; 9(1): 7728, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31118452

RESUMO

Pregnant women with MS experience fewer relapses, especially during the third trimester. In this study, we explore the cellular and molecular events that bring about the protective effect of late pregnancy on the course of de/remyelination in rats. Using cellular, molecular, and ultrastructural methods, we explored remyelination in response to a focal demyelination in the corpus callosum of late pregnant, virgin, and postpartum rats. We further explored the role of GABAA receptor (GABAAR) in the promyelinating effect observed during late pregnancy. Remyelination in response to a gliotoxin-induced demyelination in the corpus callosum was enhanced in late pregnant rats when compared to that seen in virgin and postpartum rats. This pregnancy-associated promyelinating effect was lost when either the GABAAR was blocked or when 5α-reductase, the rate limiting enzyme for the endogenous GABAAR activator allopregnanolone, was inhibited. Taken together, these data suggest that the pregnancy-associated pro-myelination operates, at least in part, through a GABAergic activated system.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Proteínas do Tecido Nervoso/fisiologia , Complicações na Gravidez/fisiopatologia , Receptores de GABA-A/fisiologia , Remielinização , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/fisiologia , Inibidores de 5-alfa Redutase/farmacologia , Animais , Bicuculina/farmacologia , Corpo Caloso/patologia , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Feminino , Finasterida/farmacologia , Antagonistas GABAérgicos/farmacologia , Lisofosfatidilcolinas/toxicidade , Masculino , Microglia/fisiologia , Esclerose Múltipla , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Bainha de Mielina/patologia , Proteínas do Tecido Nervoso/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/patologia , Oligodendroglia/patologia , Gravidez , Pregnanolona/farmacologia , Transtornos Puerperais/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos
11.
Ann Neurol ; 85(6): 887-898, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30945774

RESUMO

OBJECTIVE: Charcot-Marie-Tooth (CMT) disease is most commonly caused by duplication of a chromosomal segment surrounding Peripheral Myelin Protein 22, or PMP22 gene, which is classified as CMT1A. Several candidate therapies reduce Pmp22 mRNA levels in CMT1A rodent models, but development of biomarkers for clinical trials in CMT1A is a challenge given its slow progression and difficulty in obtaining nerve samples. Quantitative PCR measurements of PMP22 mRNA in dermal nerves were performed using skin biopsies in human clinical trials for CMT1A, but this approach did not show increased PMP22 mRNA in CMT1A patients compared to controls. One complicating factor is the variable amounts of Schwann cells (SCs) in skin. The objective of the study was to develop a novel method for precise evaluation of PMP22 levels in skin biopsies that can discriminate CMT1A patients from controls. METHODS: We have developed methods to normalize PMP22 transcript levels to SC-specific genes that are not altered by CMT1A status. Several CMT1A-associated genes were assembled into a custom Nanostring panel to enable precise transcript measurements that can be normalized to variable SC content. RESULTS: The digital expression data from Nanostring analysis showed reproducible elevation of PMP22 levels in CMT1A versus control skin biopsies, particularly after normalization to SC-specific genes. INTERPRETATION: This platform should be useful in clinical trials for CMT1A as a biomarker of target engagement that can be used to optimize dosing, and the same normalization framework is applicable to other types of CMT. ANN NEUROL 2019;85:887-898.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/patologia , Proteínas da Mielina/genética , Células de Schwann/patologia , Pele/patologia , Adolescente , Adulto , Idoso , Animais , Biomarcadores/metabolismo , Biópsia , Doença de Charcot-Marie-Tooth/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas da Mielina/biossíntese , Células de Schwann/metabolismo , Pele/metabolismo , Adulto Jovem
12.
J Neurol Sci ; 398: 79-90, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30685714

RESUMO

BACKGROUND: Charcot-Marie-Tooth diseases (CMT) are due to abnormalities of many genes, the most frequent being linked to PMP22 (Peripheral Myelin Protein 22). In the past, only spontaneous genetic anomalies occurring in mouse mutants such as Trembler (Tr) mice were available; more recently, several rodent models have been generated for exploration of the pathophysiological mechanisms underlying these neuropathies. METHODS: Based on the personal experience of our team, we describe here the pathological hallmarks of most of these animal models and compare them to the pathological features observed in some CMT patient nerves (CMT types 1A and E; hereditary neuropathy with liability to pressure palsies, HNPP). RESULTS: We describe clinical data and detailed pathological analysis mainly by electron microscopy of the sciatic nerves of these animal models conducted in our laboratory; lesions of PMP22 deficient animals (KO and mutated PMP22) and PMP22 overexpressed models are described and compared to ultrastructural anomalies of nerve biopsies from CMT patients due to PMP22 gene anomalies. It is of note that while there are some similarities, there are also significant differences between the lesions in animal models and human cases. Such observations highlight the complex roles played by PMP22 in nerve development. CONCLUSION: It should be borne in mind that we require additional correlations between animal models of hereditary neuropathies and CMT patients to rationalize the development of efficient drugs.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Modelos Animais de Doenças , Neuropatia Hereditária Motora e Sensorial/genética , Proteínas da Mielina/genética , Animais , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Expressão Gênica , Neuropatia Hereditária Motora e Sensorial/metabolismo , Neuropatia Hereditária Motora e Sensorial/patologia , Humanos , Camundongos , Proteínas da Mielina/biossíntese , Mutação Puntual/genética , Ratos , Roedores
13.
Histochem Cell Biol ; 151(5): 385-394, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30357511

RESUMO

A membrane skeletal molecular complex, protein 4.1G-membrane palmitoylated protein 6 (MPP6)-Lin7-cell adhesion molecule 4 (CADM4), is incorporated in Schwann cells, especially in Schmidt-Lanterman incisures (SLIs), in the mouse peripheral nervous system (PNS). MPP6, Lin7, and CADM4 are transported to SLIs by 4.1G. In this study, we created MPP6-deficient mice and evaluated myelin structure and MPP6 protein complexes. In SLIs in MPP6-deficient nerves, Lin7 was rarely detected by immunohistochemistry and western blotting, but the localization and amount of CADM4 and 4.1G were not altered. Motor activity was not significantly impaired in a tail-suspension test, but the sciatic nerves of MPP6-deficient mice had thicker myelin in internodes by electron microscopy compared to that of wild-type mice. These results indicate that the MPP6-Lin7 complex regulates myelin formation.


Assuntos
Guanilato Quinases/metabolismo , Proteínas Ligadas a Lipídeos/metabolismo , Proteínas da Mielina/biossíntese , Sistema Nervoso Periférico/metabolismo , Animais , Western Blotting , Genótipo , Guanilato Quinases/deficiência , Guanilato Quinases/genética , Imuno-Histoquímica , Proteínas Ligadas a Lipídeos/deficiência , Proteínas Ligadas a Lipídeos/genética , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Mutação , Proteínas da Mielina/química , Sistema Nervoso Periférico/citologia
14.
J Clin Invest ; 128(1): 359-368, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29202483

RESUMO

Charcot-Marie-Tooth disease type 1A (CMT1A) is caused by duplication of peripheral myelin protein 22 (PMP22) and is the most common hereditary peripheral neuropathy. CMT1A is characterized by demyelination and axonal loss, which underlie slowed motor nerve conduction velocity (MNCV) and reduced compound muscle action potentials (CMAP) in patients. There is currently no known treatment for this disease. Here, we show that antisense oligonucleotides (ASOs) effectively suppress PMP22 mRNA in affected nerves in 2 murine CMT1A models. Notably, initiation of ASO treatment after disease onset restored myelination, MNCV, and CMAP almost to levels seen in WT animals. In addition to disease-associated gene expression networks that were restored with ASO treatment, we also identified potential disease biomarkers through transcriptomic profiling. Furthermore, we demonstrated that reduction of PMP22 mRNA in skin biopsies from ASO-treated rats is a suitable biomarker for evaluating target engagement in response to ASO therapy. These results support the use of ASOs as a potential treatment for CMT1A and elucidate potential disease and target engagement biomarkers for use in future clinical trials.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Neurônios Motores/metabolismo , Proteínas da Mielina/antagonistas & inibidores , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Pele/metabolismo , Potenciais de Ação/genética , Animais , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/patologia , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Pele/patologia
15.
J Neurosci ; 37(50): 12297-12313, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29109239

RESUMO

Schwann cell c-Jun is implicated in adaptive and maladaptive functions in peripheral nerves. In injured nerves, this transcription factor promotes the repair Schwann cell phenotype and regeneration and promotes Schwann-cell-mediated neurotrophic support in models of peripheral neuropathies. However, c-Jun is associated with tumor formation in some systems, potentially suppresses myelin genes, and has been implicated in demyelinating neuropathies. To clarify these issues and to determine how c-Jun levels determine its function, we have generated c-Jun OE/+ and c-Jun OE/OE mice with graded expression of c-Jun in Schwann cells and examined these lines during development, in adulthood, and after injury using RNA sequencing analysis, quantitative electron microscopic morphometry, Western blotting, and functional tests. Schwann cells are remarkably tolerant of elevated c-Jun because the nerves of c-Jun OE/+ mice, in which c-Jun is elevated ∼6-fold, are normal with the exception of modestly reduced myelin thickness. The stronger elevation of c-Jun in c-Jun OE/OE mice is, however, sufficient to induce significant hypomyelination pathology, implicating c-Jun as a potential player in demyelinating neuropathies. The tumor suppressor P19ARF is strongly activated in the nerves of these mice and, even in aged c-Jun OE/OE mice, there is no evidence of tumors. This is consistent with the fact that tumors do not form in injured nerves, although they contain proliferating Schwann cells with strikingly elevated c-Jun. Furthermore, in crushed nerves of c-Jun OE/+ mice, where c-Jun levels are overexpressed sufficiently to accelerate axonal regeneration, myelination and function are restored after injury.SIGNIFICANCE STATEMENT In injured and diseased nerves, the transcription factor c-Jun in Schwann cells is elevated and variously implicated in controlling beneficial or adverse functions, including trophic Schwann cell support for neurons, promotion of regeneration, tumorigenesis, and suppression of myelination. To analyze the functions of c-Jun, we have used transgenic mice with graded elevation of Schwann cell c-Jun. We show that high c-Jun elevation is a potential pathogenic mechanism because it inhibits myelination. Conversely, we did not find a link between c-Jun elevation and tumorigenesis. Modest c-Jun elevation, which is beneficial for regeneration, is well tolerated during Schwann cell development and in the adult and is compatible with restoration of myelination and nerve function after injury.


Assuntos
Dosagem de Genes , Bainha de Mielina/fisiologia , Regeneração Nervosa/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Células de Schwann/metabolismo , Animais , Axônios/patologia , Núcleo Celular/metabolismo , Transformação Celular Neoplásica , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Bainha de Mielina/ultraestrutura , Compressão Nervosa , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas c-jun/biossíntese , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/biossíntese , Recuperação de Função Fisiológica , Nervo Isquiático/lesões , Nervo Isquiático/patologia
16.
Nat Commun ; 8: 14241, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28117328

RESUMO

Multiple Sclerosis (MS) is an inflammatory demyelinating disorder in which remyelination failure contributes to persistent disability. Cholesterol is rate-limiting for myelin biogenesis in the developing CNS; however, whether cholesterol insufficiency contributes to remyelination failure in MS, is unclear. Here, we show the relationship between cholesterol, myelination and neurological parameters in mouse models of demyelination and remyelination. In the cuprizone model, acute disease reduces serum cholesterol levels that can be restored by dietary cholesterol. Concomitant with blood-brain barrier impairment, supplemented cholesterol directly supports oligodendrocyte precursor proliferation and differentiation, and restores the balance of growth factors, creating a permissive environment for repair. This leads to attenuated axon damage, enhanced remyelination and improved motor learning. Remarkably, in experimental autoimmune encephalomyelitis, cholesterol supplementation does not exacerbate disease expression. These findings emphasize the safety of dietary cholesterol in inflammatory diseases and point to a previously unrecognized role of cholesterol in promoting repair after demyelinating episodes.


Assuntos
Colesterol na Dieta/administração & dosagem , Colesterol/sangue , Esclerose Múltipla/terapia , Proteínas da Mielina/biossíntese , Animais , Axônios/patologia , Biomarcadores/sangue , Encéfalo/citologia , Encéfalo/patologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Colesterol/metabolismo , Colesterol na Dieta/efeitos adversos , Cuprizona/toxicidade , Suplementos Nutricionais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/sangue , Esclerose Múltipla/induzido quimicamente , Oligodendroglia/citologia , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Cultura Primária de Células , Células-Tronco/fisiologia
17.
Mol Neurobiol ; 54(1): 227-237, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26738853

RESUMO

Stroke induces new myelinating oligodendrocytes that are involved in ischemic brain repair. Molecular mechanisms that regulate oligodendrogenesis have not been fully investigated. MicroRNAs (miRNAs) are small non-coding RNA molecules that post-transcriptionally regulate gene expression. MiR-146a has been reported to regulate immune response, but the role of miR-146a in oligodendrocyte progenitor cells (OPCs) remains unknown. Adult Wistar rats were subjected to the right middle cerebral artery occlusion (MCAo). In situ hybridization analysis with LNA probes against miR-146a revealed that stroke considerably increased miR-146a density in the corpus callosum and subventricular zone (SVZ) of the lateral ventricle of the ischemic hemisphere. In vitro, overexpression of miR-146a in neural progenitor cells (NPCs) significantly increased their differentiation into O4+ OPCs. Overexpression of miR-146a in primary OPCs increased their expression of myelin proteins, whereas attenuation of endogenous miR-146a suppressed generation of myelin proteins. MiR-146a also inversely regulated its target gene-IRAK1 expression in OPCs. Attenuation of IRAK1 in OPCs substantially increased myelin proteins and decreased OPC apoptosis. Collectively, our data suggest that miR-146a may mediate stroke-induced oligodendrogenesis.


Assuntos
MicroRNAs/biossíntese , Oligodendroglia/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Masculino , Proteínas da Mielina/biossíntese , Oligodendroglia/patologia , Ratos , Ratos Wistar , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/prevenção & controle
18.
J Neurosci ; 36(42): 10853-10869, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798140

RESUMO

To determine whether L-type voltage-operated Ca2+ channels (L-VOCCs) are required for oligodendrocyte progenitor cell (OPC) development, we generated an inducible conditional knock-out mouse in which the L-VOCC isoform Cav1.2 was postnatally deleted in NG2-positive OPCs. A significant hypomyelination was found in the brains of the Cav1.2 conditional knock-out (Cav1.2KO) mice specifically when the Cav1.2 deletion was induced in OPCs during the first 2 postnatal weeks. A decrease in myelin proteins expression was visible in several brain structures, including the corpus callosum, cortex, and striatum, and the corpus callosum of Cav1.2KO animals showed an important decrease in the percentage of myelinated axons and a substantial increase in the mean g-ratio of myelinated axons. The reduced myelination was accompanied by an important decline in the number of myelinating oligodendrocytes and in the rate of OPC proliferation. Furthermore, using a triple transgenic mouse in which all of the Cav1.2KO OPCs were tracked by a Cre reporter, we found that Cav1.2KO OPCs produce less mature oligodendrocytes than control cells. Finally, live-cell imaging in early postnatal brain slices revealed that the migration and proliferation of subventricular zone OPCs is decreased in the Cav1.2KO mice. These results indicate that the L-VOCC isoform Cav1.2 modulates oligodendrocyte development and suggest that Ca2+ influx mediated by L-VOCCs in OPCs is necessary for normal myelination. SIGNIFICANCE STATEMENT: Overall, it is clear that cells in the oligodendrocyte lineage exhibit remarkable plasticity with regard to the expression of Ca2+ channels and that perturbation of Ca2+ homeostasis likely plays an important role in the pathogenesis underlying demyelinating diseases. To determine whether voltage-gated Ca2+ entry is involved in oligodendrocyte maturation and myelination, we used a conditional knock-out mouse for voltage-operated Ca2+ channels in oligodendrocyte progenitor cells. Our results indicate that voltage-operated Ca2+ channels can modulate oligodendrocyte development in the postnatal brain and suggest that voltage-gated Ca2+ influx in oligodendroglial cells is critical for normal myelination. These findings could lead to novel approaches to intervene in neurodegenerative diseases in which myelin is lost or damaged.


Assuntos
Canais de Cálcio Tipo L/genética , Bainha de Mielina/fisiologia , Células-Tronco Neurais/fisiologia , Oligodendroglia/fisiologia , Animais , Animais Recém-Nascidos , Proliferação de Células , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Knockout , Proteínas da Mielina/biossíntese , Cultura Primária de Células
19.
J Neurosci Res ; 94(12): 1520-1530, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27426866

RESUMO

Krabbe's disease is a leukodystrophy resulting from deficiency of galactosylceramidase and the accumulation of galactosylsphingosine (psychosine) in the nervous system. Psychosine is believed to cause central demyelination by killing oligodendrocytes. Quantitative analysis of this process is lacking. To address this, we generated a new transgenic reporter twitcher line in which myelinating oligodendrocytes are genetically marked by the expression of LacZ under control of the myelin basic protein (MBP) promoter. MBP-LacZ-twitcher transgenic mice were used for unbiased stereological quantification of ß-galactosidase+ oligodendrocytes in the spinal cord. As expected, we found decreased numbers of these cells in mutant cords, paralleling the severity of clinical disease. The decrease of oligodendrocytes does not correlate well with the increase of psychosine. The new MBP-LacZ-twitcher line will be a useful genetic tool for measuring changes in oligodendrocyte numbers in different regions of the mutant CNS and in preclinical trials of therapies to prevent demyelination. © 2016 Wiley Periodicals, Inc.


Assuntos
Óperon Lac/genética , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/patologia , Oligodendroglia/patologia , Envelhecimento , Animais , Células Cultivadas , Genes Reporter/genética , Camundongos , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Mutação , Proteínas da Mielina/biossíntese , Proteínas da Mielina/genética , Neuroglia/metabolismo , Psicosina/metabolismo , beta-Galactosidase/metabolismo
20.
Folia Neuropathol ; 53(2): 147-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26216117

RESUMO

Multiple sclerosis (MS) lesions are characterized by inflammatory demyelination and reactive gliosis, and although remyelination occurs in some lesions it is limited and incomplete. Leukemia inhibitory factor (LIF) is an important cytokine that stimulates oligodendrocyte proliferation and survival in vitro. Opalin is a unique molecular marker for mature oligodendrocytes. The aim of this study was to demonstrate the role of LIF on Opalin and myelin oligodendrocyte glycoprotein (MOG) expression in the cerebral cortex of cuprizone-induced MS mice. The mice were treated with cuprizone for five weeks in order to induce MS. The mice were then divided into 3 groups. The first group was injected intraperitoneally (IP) with LIF for six weeks in the amount of 30 µg/kg bw per day. The second group (SHAM) was injected IP with normal saline and the third group was left without injection as a control. After six weeks the mice were killed, the cerebral cortex was harvested, and the expression of MOG and Opalin was studied. Using western blotting we found that LIF increases Opalin and MOG expression in the cerebral cortex extracts as compared to SHAM and control groups. However, no significant difference in the Opalin and MOG expression was seen between SHAM and control groups. It is concluded that LIF may have an important role in the process of remyelination by increasing Opalin expression and MOG expression.


Assuntos
Córtex Cerebral/metabolismo , Doenças Desmielinizantes/metabolismo , Fator Inibidor de Leucemia/metabolismo , Proteínas da Mielina/biossíntese , Glicoproteína Mielina-Oligodendrócito/biossíntese , Animais , Western Blotting , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Quelantes/toxicidade , Cuprizona/toxicidade , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Fator Inibidor de Leucemia/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...