Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 37(12): 3127-3137, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28193690

RESUMO

Differentiation and maturation of oligodendrocyte progenitor cells (OPCs) involve the assembly and disassembly of actin microfilaments. However, how actin dynamics are regulated during this process remains poorly understood. Leucine-rich repeat and Ig-like domain-containing Nogo receptor interacting protein 1 (LINGO-1) is a negative regulator of OPC differentiation. We discovered that anti-LINGO-1 antibody-promoted OPC differentiation was accompanied by upregulation of cytoplasmic gelsolin (cGSN), an abundant actin-severing protein involved in the depolymerization of actin filaments. Treating rat OPCs with cGSN siRNA reduced OPC differentiation, whereas overexpression of cGSN promoted OPC differentiation in vitro and remyelination in vivo Furthermore, coexpression of cGSN and LINGO-1 blocked the inhibitory effect of LINGO-1. Our study demonstrates that cGSN works downstream of LINGO-1 signaling pathway, which enhances actin dynamics and is essential for OPC morphogenesis and differentiation. This finding may lead to novel therapeutic approaches for the treatment of demyelinating diseases such as multiple sclerosis (MS).SIGNIFICANCE STATEMENT Myelin loss and subsequent axon degeneration contributes to a variety of neurological diseases, such as multiple sclerosis (MS). Understanding the regulation of myelination by oligodendrocytes is therefore critical for developing therapies for the treatment of MS. We previously demonstrated that leucine-rich repeat and Ig-like domain-containing Nogo receptor interacting protein 1 (LINGO-1) is a negative regulator of oligodendrocyte differentiation and that anti-LINGO-1 promotes remyelination in preclinical animal models for MS and in a phase II acute optic neuritis clinical trial (RENEW). The mechanism by which LINGO-1 regulates oligodendrocyte differentiation is unknown. Here, we demonstrate that LINGO-1 regulates oligodendrocyte differentiation and maturation through the cytoplasmic gelsolin signaling pathway, providing new drug targets for the treatment of demyelination diseases.


Assuntos
Actinas/metabolismo , Diferenciação Celular/fisiologia , Gelsolina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Animais , Células Cultivadas , Citoplasma/metabolismo , Feminino , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
2.
Mol Cell Neurosci ; 60: 36-42, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24583087

RESUMO

Oligodendrocyte differentiation is negatively regulated by LINGO-1 and positively regulated by the ErbB2 receptor tyrosine kinase. In wild-type oligodendrocytes, inhibition of ErbB2 blocks differentiation, whereas activation of ErbB2 promotes differentiation. In LINGO-1(-/-) oligodendrocytes, inhibition of ErbB2 blocks oligodendrocyte differentiation; whereas activation of ErbB2 does not enhance differentiation. Biological and biochemical evidence showing that LINGO-1 can directly bind to ErbB2, block ErbB2 translocation into lipid rafts, and inhibit its phosphorylation for activation. The study demonstrates a novel regulatory mechanism of ErbB2 function whereby LINGO-1 suppresses oligodendrocyte differentiation by inhibiting ErbB2 translocation and activation in lipid rafts.


Assuntos
Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Oligodendroglia/metabolismo , Receptor ErbB-2/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Camundongos , Oligodendroglia/citologia , Fosforilação , Ligação Proteica , Transporte Proteico
3.
Proc Natl Acad Sci U S A ; 109(7): 2521-6, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308476

RESUMO

The boronic acid dipeptide bortezomib inhibits the chymotrypsin-like activity of the 26S proteasome and shows significant therapeutic efficacy in multiple myeloma. However, recent studies suggest that bortezomib may have more complex mechanisms of action in treating cancer. We report here that the endocytosis and lysosomal degradation of the receptor tyrosine kinase C-KIT are required for bortezomib- but not tyrosine kinase inhibitor imatinib-caused apoptosis of t(8;21) leukemia and gastrointestinal stromal tumor cells, suggesting that C-KIT may recruit an apoptosis initiator. We show that C-KIT binds and phosphorylates heat shock protein 90ß (Hsp90ß), which sequestrates apoptotic protease activating factor 1 (Apaf-1). Bortezomib dephosphorylates pHsp90ß and releases Apaf-1. Although the activated caspase-3 is not sufficient to cause marked apoptosis, it cleaves the t(8;21) generated acute myeloid leukemia 1-eight twenty one (AML1-ETO) and AML1-ETO9a fusion proteins, with production of cleavage fragments that perturb the functions of the parental oncoproteins and further contribute to apoptosis. Notably, bortezomib exerts potent therapeutic efficacy in mice bearing AML1-ETO9a-driven leukemia. These data show that C-KIT-pHsp90ß-Apaf-1 cascade is critical for some malignant cells to evade apoptosis, and the clinical therapeutic potentials of bortezomib in C-KIT-driven neoplasms should be further explored.


Assuntos
Ácidos Borônicos/farmacologia , Cromossomos Humanos Par 21 , Cromossomos Humanos Par 8 , Leucemia/patologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirazinas/farmacologia , Translocação Genética , Apoptose , Bortezomib , Humanos , Leucemia/genética , Fosforilação , Ligação Proteica
4.
J Biol Chem ; 287(3): 2237-46, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22123816

RESUMO

It is indicated that there are important molecules interacting with brain nervous systems to regulate feeding and energy balance by influencing the signaling pathways of these systems, but relatively few of the critical players have been identified. In the present study, we provide the evidence for the role of Abelson helper integration site 1 (Ahi1) protein as a mediator of feeding behavior through interaction with serotonin receptor 2C (5-HT(2C)R), known for its critical role in feeding and appetite control. First, we demonstrated the co-localization and interaction between hypothalamic Ahi1 and 5-HT(2C)R. Ahi1 promoted the degradation of 5-HT(2C)R through the lysosomal pathway. Then, we investigated the effects of fasting on the expression of hypothalamic Ahi1 and 5-HT(2C)R. Fasting resulted in an increased Ahi1 expression and a concomitant decreased expression of 5-HT(2C)R. Knockdown of hypothalamic Ahi1 led to a concomitant increased expression of 5-HT(2C)R and a decrease of food intake and body weight. Last, we found that Ahi1 could regulate the expression of neuropeptide Y and proopiomelanocortin. Taken together, our results indicate that Ahi1 mediates feeding behavior by interacting with 5-HT(2C)R to modulate the serotonin signaling pathway.


Assuntos
Regulação do Apetite/fisiologia , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteólise , Proteínas Proto-Oncogênicas/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Humanos , Masculino , Camundongos , Neuropeptídeo Y/biossíntese , Serotonina/metabolismo , Transdução de Sinais/fisiologia
5.
Nat Med ; 12(5): 526-33, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16604089

RESUMO

The hypothalamus responds to circulating leptin and insulin in the control of food intake and body weight. A number of neurotransmitters in the hypothalamus, including gamma-aminobutyric acid (GABA), also have key roles in feeding. Huntingtin-associated protein 1 (Hap1) is expressed more abundantly in the hypothalamus than in other brain regions, and lack of Hap1 in mice leads to early postnatal death. Hap1 is also involved in intracellular trafficking of the GABA(A) receptor. Here, we report that fasting upregulates the expression of Hap1 in the rodent hypothalamus, whereas intracerebroventricular administration of insulin downregulates Hap1 by increasing its degradation through ubiquitination. Decreasing the expression of mouse hypothalamic Hap1 by siRNA reduces the level and activity of hypothalamic GABA(A) receptors and causes a decrease in food intake and body weight. These findings provide evidence linking hypothalamic Hap1 to GABA in the stimulation of feeding and suggest that this mechanism is involved in the feeding-inhibitory actions of insulin in the brain.


Assuntos
Ingestão de Alimentos , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de GABA-A/metabolismo , Animais , Peso Corporal , Eletrofisiologia , Jejum , Humanos , Hipotálamo/citologia , Insulina/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Ubiquitina/metabolismo
6.
Sci Rep ; 13(1): 19529, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945646

RESUMO

Multiple sclerosis (MS), a chronic neurodegenerative disease driven by damage to the protective myelin sheath, is currently incurable. Today, all clinically available treatments modulate the immune-mediated symptoms of the disease but they fail to stop neurodegeneration in many patients. Remyelination, the regenerative process of myelin repair by oligodendrocytes, which is considered a necessary step to protect demyelinated axons and stop neuronal death, is impaired in MS patients. One of the major obstacles to finding effective remyelinating drugs is the lack of biomimetic drug screening platforms that enable quantification of compounds' potential to stimulate 3D myelination in the physiologically relevant axon-like environment. To address this need, we built a unique myelination drug discovery platform, by expanding our previously developed technology, artificial axons (AAs), which enables 3D-printing of synthetic axon mimics with the geometry and mechanical properties closely resembling those of biological axons. This platform allows for high-throughput phenotypic myelination assay based on quantification of 3D wrapping of myelin membrane around axons in response to compounds. Here, we demonstrate quantification of 3D myelin wrapping by rat oligodendrocytes around the axon mimics in response to a small library of known pro-myelinating compounds. This assay shows pro-myelinating activity for all tested compounds consistent with the published in vitro and in vivo data, demonstrating predictive power of AA platform. We find that stimulation of myelin wrapping by these compounds is dose-dependent, providing a facile means to quantify the compounds' potency and efficacy in promoting myelin wrapping. Further, the ranking of relative efficacy among these compounds differs in this 3D axon-like environment as compared to a traditional oligodendrocyte 2D differentiation assay quantifying area of deposited myelin membrane. Together, we demonstrate that the artificial axons platform and associated phenotypic myelin wrapping assay afford direct evaluation of myelin wrapping by oligodendrocytes in response to soluble compounds in an axon-like environment, providing a predictive tool for the discovery of remyelinating therapies.


Assuntos
Esclerose Múltipla , Doenças Neurodegenerativas , Humanos , Ratos , Animais , Biomimética , Axônios/fisiologia , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Esclerose Múltipla/tratamento farmacológico
7.
iScience ; 26(3): 106156, 2023 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-36852281

RESUMO

Promoting myelination capacity of endogenous oligodendrocyte precursor cells (OPCs) is a promising therapeutic approach for CNS demyelinating disorders such as Multiple Sclerosis (MS). To aid in the discovery of myelination-promoting compounds, we generated a genome-engineered human pluripotent stem cell (hPSC) line that consists of three reporters: identification-and-purification tag, GFP, and secreted-NanoLuc, driven by the endogenous PDGFRA, PLP1, and MBP genes, respectively. Using this cell line, we established a high-throughput drug screening platform and performed a small-molecule screen, which identified at least two myelination-promoting small-molecule (Ro1138452 and SR2211) that target prostacyclin (IP) receptor and retinoic acid receptor-related orphan receptor γ (RORγ), respectively. Single-cell-transcriptomic analysis of differentiating OPCs treated with these molecules further confirmed that they promote oligodendrocyte differentiation and revealed several pathways that are potentially modulated by them. The molecules and their target pathways provide promising targets for the possible development of remyelination-based therapy for MS and other demyelinating disorders.

8.
Mol Biol Rep ; 39(2): 789-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21611753

RESUMO

Retinoic acid (RA) plays multiple roles in the nervous system, including induction of neural differentiation, axon outgrowth and neural patterning. Previously, RA for neural differentiation of embryonic stem (ES) cells always relies on embryoid bodies (EBs) formation. Here we report an in vitro adherent monoculture system to induce mouse ES cells into neural cells accompanied with RA. RA (1 µM) treatment, during initial 2 days of differentiation, can enhance the expression of neural markers, such as Nestin, Tuj1 and MAP2, and result in an earlier neural differentiation of ES cells. Furthermore, RA promotes a significant increase in neurite elongation of ES-derived neurons. Our study also implies that RA induced to express Wnt antagonist Dickkopf-1 (Dkk-1) for neural differentiation. However, the mechanisms of RA triggering neural induction remain to be determined. Our simple and efficient strategy is proposed to provide a basis for studying RA signaling pathways in neural differentiation in vitro.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Tretinoína/farmacologia , Animais , Western Blotting , Células-Tronco Embrionárias/efeitos dos fármacos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tubulina (Proteína)/metabolismo
9.
Mol Biol Rep ; 39(10): 9697-705, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22740139

RESUMO

Growing evidence suggests that the brain, in particular the hypothalamus, directly senses hormones and nutrients to initiate feeding behavior and metabolic responses in the control of energy homeostasis. However, the molecular mechanisms underlying this important process have remained largely unknown. Our study provides the evidence for the role of Abelson helper integration site 1 (Ahi1) protein as a sensor of insulin signaling in the hypothalamus. We found that fasting increased the expression of hypothalamic Ahi1 which was accompanied by lower levels of circulating insulin compared with satiated mice, while re-feeding decreased the expression of hypothalamic Ahi1 which was accompanied by higher levels of circulating insulin. We also found the up-regulated expression of hypothalamic Ahi1 in high-fat induced obese mice, db/db mice, and streptozotocin induced diabetic mice. In addition, we demonstrated that insulin could decrease the expression of Ahi1 in neuroblastoma cell line N18TG2. Taken together, our results indicate that hypothalamic Ahi1 functions as a sensor of insulin signaling.


Assuntos
Expressão Gênica , Hipotálamo/metabolismo , Insulina/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Linhagem Celular , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/metabolismo , Dieta Hiperlipídica/efeitos adversos , Privação de Alimentos , Regulação da Expressão Gênica , Hipotálamo/fisiologia , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/metabolismo , Especificidade de Órgãos , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Estreptozocina
10.
J Biol Chem ; 285(14): 10653-61, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20145253

RESUMO

Huntington disease (HD) is caused by an expansion of the polyglutamine (polyQ) repeat (>37Q) in huntingtin (htt), and age of onset is inversely correlated with the length of the polyQ repeat. Mutant htt with expanded polyQ is ubiquitously expressed in various types of cells, including glia, but causes selective neurodegeneration. Our recent study demonstrated that expression of the N-terminal mutant htt with a large polyQ repeat (160Q) in astrocytes is sufficient to induce neurological symptoms in mice (Bradford, J., Shin, J. Y., Roberts, M., Wang, C. E., Li, X.-J., and Li, S. H. (2009) Proc. Natl. Acad. Sci. U.S.A. 106, 22480-22485). Because glia-neuron interactions are critical for maintaining the normal function and survival of neurons in the brain and because mutant htt is more abundant in neurons than in glial cells, it is important to investigate whether glial htt can still contribute to HD pathology when mutant htt is abundantly expressed in neuronal cells. We generated transgenic mice that express mutant htt with 98Q in astrocytes. Unlike our recently generated htt-160Q transgenic mice, htt-98Q mice do not show obvious neurological phenotypes, suggesting that the length of the polyQ repeat determines the severity of glial dysfunction. However, htt-98Q mice show increased susceptibility to glutamate-induced seizure. Mice expressing mutant htt in astrocytes were mated with N171-82Q mice that express mutant htt primarily in neuronal cells. Double transgenic mice expressing mutant htt in both neuronal and glial cells display more severe neurological symptoms and earlier death than N171-82Q mice. These findings indicate a role of glial mutant htt in exacerbating HD neuropathology and underscore the importance of improving glial function in treating HD.


Assuntos
Modelos Animais de Doenças , Doença de Huntington/patologia , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Proteínas Nucleares/metabolismo , Convulsões/patologia , Animais , Comportamento Animal , Western Blotting , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Feminino , Ácido Glutâmico/toxicidade , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neuroglia/citologia , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Convulsões/induzido quimicamente , Convulsões/metabolismo , Expansão das Repetições de Trinucleotídeos/fisiologia
11.
J Clin Invest ; 118(8): 2785-95, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18636121

RESUMO

Joubert syndrome is an autosomal recessive disorder characterized by congenital malformation of the cerebellum and brainstem, with abnormal decussation in the brain. Mutations in the Abelson helper integration site 1 gene, which encodes the protein AHI1, have been shown to cause Joubert syndrome. In this study, we found that mouse Ahi1 formed a stable complex with huntingtin-associated protein 1 (Hap1), which is critical for neonatal development and involved in intracellular trafficking. Hap1-knockout mice showed significantly reduced Ahi1 levels, defective cerebellar development, and abnormal axonal decussation. Suppression of Ahi1 also decreased the level of Hap1; and truncated Ahi1, which corresponds to the mutations in Joubert syndrome, inhibited neurite outgrowth in neuronal culture. Reducing Hap1 expression suppressed the level and internalization of TrkB, a neurotrophic factor receptor that mediates neurogenesis and neuronal differentiation, which led to decreased TrkB signaling. These findings provide insight into the pathogenesis of Joubert syndrome and demonstrate the critical role of the Ahi1-Hap1 complex in early brain development.


Assuntos
Tronco Encefálico/anormalidades , Cerebelo/anormalidades , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Linhagem Celular , Células Cultivadas , Humanos , Imuno-Histoquímica , Rim/citologia , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Transfecção
12.
Artigo em Inglês | MEDLINE | ID: mdl-19342780

RESUMO

The SH3 domain of human AHI1 was cloned and expressed in Escherichia coli. The protein was purified by affinity and size-exclusion chromatography and was crystallized using the sitting-drop vapour-diffusion method at 293 K. A complete data set was collected to 2.5 A resolution at 110 K. The crystal belonged to space group P4(1)2(1)2, with unit-cell parameters a = 67.377, b = 67.377, c = 98.549 A.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/isolamento & purificação , Domínios de Homologia de src , Proteínas Adaptadoras de Transporte Vesicular , Sequência de Aminoácidos , Cromatografia em Gel , Cristalização , Cristalografia por Raios X , Humanos , Luz , Dados de Sequência Molecular , Proteínas Recombinantes/química , Espalhamento de Radiação , Alinhamento de Sequência
13.
J Colloid Interface Sci ; 522: 264-271, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29604445

RESUMO

Monodisperse hollow mesoporous PdCo alloy nanospheres are prepared via a simple galvanic replacement reaction. The as-prepared PdCo hollow nanospheres have small diameter, such as Pd78Co22 nanospheres of diameter about 25 nm and mesoporous shells about 4-5 nm. The Pd78Co22 hollow mesoporous nanospheres possess the largest electrochemical active surface areas (ECSA, 53.91 m2 g-1), mass activity (1488 mA mg-1) and specific activity (2.76 mA cm-2) towards to methanol oxidation relative to the Pd68Co32, Pd92Co8 hollow mesoporous nanospheres and commercial Pd/C catalysts. Moreover, the activity of Pd78Co22 after long-term stability tests is still the best and even better than those of fresh Pd68Co32 and commercial Pd/C catalysts. The PdCo catalysts not only effectively reduce the Pd usage by forming hollow structure, but also fully realize the Pd-Co alloying effects for enhancing the methanol oxidation catalytic performance.

14.
ACS Appl Mater Interfaces ; 10(26): 22248-22256, 2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29900739

RESUMO

Flowerlike NiCo2S4 hollow sub-microspheres are synthesized through Cu2O templates to support Pd nanoparticles as high-efficiency catalysts for the hydrogen evolution reaction (HER). The diameter and shell size of NiCo2S4 hollow sub-microspheres are about 400 and 16 nm, respectively. In addition, the surface of the shells is constructed by petallike nanosheets. About 3 nm Pd particles uniformly incorporate with the flowerlike NiCo2S4 hollow sub-microsphere to form the NiCo2S4/Pd heterostructure. The NiCo2S4/Pd catalysts exhibit significantly lower overpotential of only 87 and 83 mV at 10 mA/cm2 for the HER in both acidic and alkaline conditions, respectively, relative to NiCo2S4 (247, 226 mV) and Pd (175, 385 mV) catalysts. Besides, the NiCo2S4/Pd catalysts also exhibit excellent stability of HER in these two conditions. The superior HER performance of NiCo2S4/Pd might be resulted from the unique architecture of metal nanoparticles anchored on the bimetallic sulfide flowerlike hollow sub-microspheres, which could provide high surface area, lots of active sites, strong synergetic effect, and stable structure.

15.
J Neurosci ; 26(22): 6019-30, 2006 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-16738245

RESUMO

Mutant huntingtin can affect vesicular and receptor trafficking via its abnormal protein interactions, suggesting that impairment of intracellular trafficking may contribute to Huntington's disease. There is growing evidence that huntingtin-associated protein-1 (HAP1) also interacts with microtubule-dependent transporters and is involved in intracellular trafficking. However, it remains unclear how the trafficking of HAP1 is regulated and contributes to neuronal function. Here we report that phosphorylation of HAP1 decreases its association with microtubule-dependent transport proteins dynactin p150 and kinesin light chain and reduces its localization in neurite tips. Suppressing HAP1 expression by RNA interference reduces neurite outgrowth and the level of tropomyosin-related kinase A receptor tyrosine kinase (TrkA), a nerve growth factor receptor whose internalization and trafficking are required for neurite outgrowth. HAP1 maintains the normal level of membrane TrkA by preventing the degradation of internalized TrkA. Mutant huntingtin also reduces the association of HAP1 with dynactin p150 and kinesin light chain and thereby decreases the intracellular level of TrkA. These findings suggest that HAP1 trafficking is critical for the stability of TrkA and neurite function, both of which can be attenuated by mutant huntingtin.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neuritos/fisiologia , Neurônios/fisiologia , Receptor trkA/metabolismo , Animais , Sequência de Bases , Primers do DNA , Genes Reporter , Doença de Huntington , Hipotálamo , Microscopia Confocal , Neuritos/ultraestrutura , Neurônios/citologia , Células PC12 , Feocromocitoma , RNA Interferente Pequeno/genética , Ratos , Gânglio Cervical Superior/fisiologia
16.
Mult Scler J Exp Transl Clin ; 2: 2055217316641704, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28607723

RESUMO

BACKGROUND: Two ongoing phase II clinical trials (RENEW and SYNERGY) have been developed to test the efficacy of anti-LINGO-1 antibodies in acute optic neuritis and relapsing forms of multiple sclerosis, respectively. Across a range of experimental models, LINGO-1 has been found to inhibit neuron and oligodendrocyte survival, axon regeneration, and (re)myelination. The therapeutic effects of anti-LINGO-1 antibodies on optic nerve axonal loss and regeneration have not yet been investigated. OBJECTIVE: In this series of studies we investigate if LINGO-1 antibodies can prevent acute inflammatory axonal loss, and promote axonal regeneration after injury in rodent optic nerves. METHODS: The effects of anti-LINGO-1 antibody on optic nerve axonal damage were assessed using rodent myelin oligodendrocyte glycoprotein experimental autoimmune encephalomyelitis (EAE), and its effects on axonal regeneration were assessed in optic nerve crush injury models. RESULTS: In the optic nerve, anti-LINGO-1 antibody therapy was associated with improved optic nerve parallel diffusivity measures on MRI in mice with EAE and reduced axonal loss in rat EAE. Both anti-LINGO-1 antibody therapy and the genetic deletion of LINGO-1 reduced nerve crush-induced axonal degeneration and enhanced axonal regeneration. CONCLUSION: These data demonstrate that LINGO-1 blockade is associated with axonal protection and regeneration in the injured optic nerve.

17.
ACS Chem Neurosci ; 7(11): 1499-1507, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27504670

RESUMO

Currently, anti-AD drug discovery using target-based approaches is extremely challenging due to unclear etiology of AD and absence of validated therapeutic protein targets. Neuronal death, regardless of causes, plays a key role in AD progression, and it is directly linked to neuroinflammation. Meanwhile, phenotypic screening is making a resurgence in drug discovery process as an alternative to target-focused approaches. Herein, we employed microglia-based phenotypic screenings to search for small molecules that modulate the release of detrimental proinflammatory cytokines. The identified novel pharmacological inhibitor of neuroinflammation (named GIBH-130) was validated to alter phenotypes of neuroinflammation in AD brains. Notably, this molecule exhibited comparable in vivo efficacy of cognitive impairment relief to donepezil and memantine respectively in both ß amyloid-induced and APP/PS1 double transgenic Alzheimer's murine models at a substantially lower dose (0.25 mg/kg). Therefore, GIBH-130 constitutes a unique chemical probe for pathogenesis research and drug development of AD, and it also suggests microglia-based phenotypic screenings that target neuroinflammation as an effective and feasible strategy to identify novel anti-AD agents.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/imunologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Fármacos Neuroprotetores/farmacologia , Piperazinas/farmacologia , Piridazinas/farmacologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Modelos Animais de Doenças , Donepezila , Relação Dose-Resposta a Droga , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Feminino , Indanos/farmacologia , Masculino , Memantina/farmacologia , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/fisiologia , Fragmentos de Peptídeos , Fenótipo , Piperidinas/farmacologia , Distribuição Aleatória , Ratos Sprague-Dawley
18.
Environ Int ; 31(1): 33-42, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15607777

RESUMO

For development of an indirect competitive enzyme-linked immunosorbent assay (ELISA) for the nitrile herbicide bromoxynil, the polyclonal antibodies raised against 2,6-dibromo-4-cyano-phenoxyacetic acid (hapten) conjugated to bovine serum albumin (BSA) by the N-hydroxysuccinimide-activated ester method. Antiserum with a sufficiently high titer to provide the determinations of targeted compounds was obtained only 77 days after the primary immunization. Antiserum A2 was applied to the residual analysis of some water samples, under optimized ELISA condition, the quantitative working range was from 10 to 500 ppb with a limit of detection of 5 ppb. Cross-reactivity to structurally similar agrochemicals and related chemicals was determined. The antiserum showed little cross-reactivity with 2,6-dibromophenol and bromoxynil octanoate ester which have a dibromophenol group as common structure, but showed no cross-reactivity with other herbicides. Each water sample (river water, tap water, purified water, and bottled water) had a matrix effect and was investigated by adding Tween20 in the assay buffer. These four kinds of water samples were fortified with bromoxynil at several concentration levels and were directly analyzed with only dilution with an equal volume of antiserum solution, the mean recovery was 102.3%, and the mean coefficient of variation was 5.96%. The proposed ELISA turned out to be a powerful tool for monitoring of residual bromoxynil in water samples at trace level.


Assuntos
Bromobenzenos/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Herbicidas/análise , Soros Imunes/imunologia , Nitrilas/análise , Água/análise , Animais , Bromobenzenos/síntese química , Reações Cruzadas , Monitoramento Ambiental , Feminino , Haptenos/imunologia , Herbicidas/imunologia , Concentração de Íons de Hidrogênio , Soros Imunes/biossíntese , Imunização , Masculino , Nitrilas/imunologia , Polissorbatos , Coelhos , Reprodutibilidade dos Testes , Rios/química
19.
Eur J Pharmacol ; 451(2): 119-24, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12231380

RESUMO

The neuroprotective effects of verbascoside, one of phenylpropanoid glucoside isolated from the Chinese herbal medicine Buddleja officinalis Maxim, on 1-methyl-4-phenylpyridinium ion (MPP(+)) induced apoptosis and oxidative stress in PC12 neuronal cells were investigated. Treatment of PC12 cells with MPP(+) for 48 h induced apoptotic death as determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry, the activation of caspase-3 measured by the caspase-3 activity assay kit, the reduction in mitochondrial membrane potential with laser scanning confocal microscopy and the increase in the extracellular hydrogen peroxide level. Simultaneous treatment with verbascoside markedly attenuated MPP(+)-induced apoptotic death, increased extracellular hydrogen peroxide level, the activation of caspase-3 and the collapse of mitochondrial membrane potential. These results strongly indicate that verbascoside may provide a useful therapeutic strategy for the treatment of oxidative stress-induced neurodegenerative disease such as Parkinson's disease.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Glucosídeos/farmacologia , Fenóis/farmacologia , Animais , Buddleja/química , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Glucosídeos/química , Peróxido de Hidrogênio/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Células PC12 , Fenóis/química , Ratos
20.
Eur J Endocrinol ; 166(4): 727-34, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22285701

RESUMO

OBJECTIVE: The prevalence of obesity has increased dramatically over the past decade. Gene copy number variants (CNVs) have been recognized as a hereditable source of susceptibility in human complex diseases including obesity. Recent studies have shown that Abelson helper integration site 1 (Ahi1) gene has a significant contribution in the homeostasis regulation in mouse models of obesity. A study was therefore carried out to investigate whether CNVs in AHI1 gene contribute to human obesity. SUBJECTS AND METHODS: We analyzed samples from 70 Chinese overweight adults and 74 healthy controls for DNA copy number change using the Affymetrix single-nucleotide polymorphism (SNP) 6.0 array. Validation of CNVs of AHI1 was achieved by real-time PCR using the ΔΔC(t) method. RESULTS: Copy number gain analysis revealed significant gains (P=0.0017) of AHI1 gene copy number in 17 of 70 (24.3%) samples but only four of 74 (5.4%) controls overall. Then we studied the frequency distribution of CNVs in AHI1 gene according to body mass index (BMI) grade. Five out of 28 (18.5%) at-risk obese, six out of 26 (26.9%) moderate obese, and six out of 17 (29.4%) severe obese subjects studied showed increased AHI1 gene copy number. CONCLUSIONS: The result suggested that there was a significant linear trend for increasing AHI1 gene copy number frequencies with increasing BMI.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Povo Asiático/genética , Variações do Número de Cópias de DNA , Obesidade/genética , Proteínas Adaptadoras de Transporte Vesicular , Adulto , Índice de Massa Corporal , Estudos de Casos e Controles , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/etnologia , População , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA