Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Eur J Med Chem ; 210: 112952, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33139114

RESUMO

ALS is a rare type of progressive neurological disease with unknown etiology. It results in the gradual degeneration and death of motor neurons responsible for controlling the voluntary muscles. Identification of mutations in the superoxide dismutase (SOD) 1 gene has been the most significant finding in ALS research. SOD1 abnormalities have been associated with both familial as well as sporadic ALS cases. SOD2 is a highly inducible SOD that performs in concurrence with SOD1 to detoxify ROS. Induction of SOD2 can be obtained through activation of NF-Ò¡Bs. We previously reported that SRI-22819 increases NF-Ò¡B expression and activation in vitro, but it has poor ADME properties in general and has no oral bioavailability. Our initial studies were focused on direct modifications of SRI-22819. There were active compounds identified but no improvement in microsomal stability was observed. In this context, we focused on making more significant structural changes in the core of the molecule. Ataluren, an oxadiazole compound that promotes read-through and expression of dystrophin in patients with Duchenne muscular dystrophy, bears some structural similarity to SRI-22819. Thus, we synthesized a series of SRI-22819 and Ataluren (PTC124) hybrid compounds. Several compounds from this series exhibited improved activity, microsomal stability and lower calculated polar surface area (PSA). This manuscript describes the synthesis and biological evaluation of SRI-22819 analogs and its hybrid combination with Ataluren.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , NF-kappa B/agonistas , Esclerose Lateral Amiotrófica/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Simulação de Acoplamento Molecular , NF-kappa B/metabolismo , Oxidiazóis/química , Oxidiazóis/farmacocinética , Oxidiazóis/farmacologia , Relação Estrutura-Atividade , Superóxido Dismutase/metabolismo
2.
Brain Res Bull ; 164: 339-349, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32795490

RESUMO

BACKGROUND AND PURPOSE: DTNBP1 gene variation and lower dysbindin-1 protein are associated with schizophrenia. Previous evidence suggests that downregulated dysbindin-1 expression results in lower expression of copper transporters ATP7A (intracellular copper transporter) and SLC31A1 (CTR1; extracellular copper transporter), which are required for copper transport across the blood brain barrier. However, whether antipsychotic medications used for schizophrenia treatment may modulate these systems is unclear. EXPERIMENTAL APPROACH: The current study measured behavioral indices of neurological function in dysbindin-1 functional knockout (KO) mice and their wild-type (WT) littermates with or without quetiapine treatment. We assessed serum and brain copper levels, ATP7A and CTR1 mRNA, and copper transporter-expressing cellular population transcripts: TTR (transthyretin; choroid plexus epithelial cells), MBP (myelin basic protein; oligodendrocytes), and GJA1 (gap-junction protein alpha-1; astrocytes) in cortex and hippocampus. KEY RESULTS: Regardless of genotype, quetiapine significantly reduced TTR, MBP, CTR1 mRNA, and serum copper levels. Neurological function of untreated KO mice was abnormal, and ledge instability was rescued with quetiapine. KO mice were hyperactive after 10 min in the open-field assay, which was not affected by treatment. CONCLUSIONS AND IMPLICATIONS: Dysbindin-1 KO results in hyperactivity, altered serum copper, and neurological impairment, the last of which is selectively rescued with quetiapine. Antipsychotic treatment modulates specific cellular populations, affecting myelin, the choroid plexus, and copper transport across the blood brain barrier. Together these results indicate the widespread impact of antipsychotic treatment, and that alteration of dysbindin-1 may be sufficient, but not necessary, for specific schizophrenia pathology.


Assuntos
Encéfalo/metabolismo , Cobre/metabolismo , Disbindina/genética , Esquizofrenia/genética , Animais , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Encéfalo/efeitos dos fármacos , Transportador de Cobre 1/genética , Transportador de Cobre 1/metabolismo , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Camundongos , Camundongos Knockout , Fumarato de Quetiapina/farmacologia , Fumarato de Quetiapina/uso terapêutico , Fatores de Risco , Esquizofrenia/tratamento farmacológico , Esquizofrenia/metabolismo
3.
Scand J Pain ; 17: 316-324, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28927908

RESUMO

BACKGROUND AND AIMS: Obesity is a significant health concern in the Western world and the presence of comorbid conditions suggests an interaction. The overlapping distributions of chronic pain populations and obesity suggests that an interaction may exist. Poor quality diet (high carbohydrates, saturated fats, omega-6 polyunsaturated fatty acids) can lead to increased adiposity which can activate immune cells independent of the activating effect of the diet components themselves. This dual action can contribute to chronic inflammation that may alter susceptibility to chronic pain and prolong recovery from injury. However, traditional examinations of diet focus on high-fat diets that often contain a single source of fat, that is not reflective of an American diet. Thus, we examined the impact of a novel human-relevant (high-carbohydrate) American diet on measures of pain and inflammation in rats, as well as the effect on recovery and immune cell activation. METHODS: We developed a novel, human-relevant Standard American Diet (SAD) to better model the kilocalorie levels and nutrient sources in an American population. Male and female rats were fed the SAD over the course of 20 weeks prior to persistent inflammatory pain induction with Complete Freund's Adjuvant (CFA). Mechanical and thermal sensitivity were measured weekly. Spontaneous pain, open field locomotion and blood glucose levels were measured during diet consumption. Body composition was assessed at 20 weeks. Following full recovery from CFA-induced hypersensitivity, blood was analyzed for inflammatory mediators and spinal cords were immunohistochemically processed for microglial markers. RESULTS: Chronic consumption of the SAD increased fat mass, decreased lean mass and reduce bone mineral density. SAD-fed rats had increased leptin levels and pro-inflammatory cytokines in peripheral blood serum. Following CFA administration, mechanical sensitivity was assessed and recovery was delayed significantly in SAD-fed animals. Sex differences in the impact of the SAD were also observed. The SAD increased body weight and common T-cell related inflammatory mediators in female, but not male, animals. In males, the SAD had a greater effect on bone mineral density and body composition. Long-term consumption of the SAD resulted in elevated microglial staining in the dorsal horn of the spinal cord, but no sex differences were observed. CONCLUSIONS: We demonstrate the negative effects of an American diet on physiology, behavior and recovery from injury. SAD consumption elevated pro-inflammatory mediators and increased microglial activation in the spinal cord. While there were sex differences in weight gain and inflammation, both sexes showed prolonged recovery from injury. IMPLICATIONS: These data suggest that poor quality diet may increase susceptibility to chronic pain due to persistent peripheral and central immune system activation. Furthermore, consumption of a diet that is high in carbohydrates and omega-6 polyunsaturated fatty acid is likely to lead to protracted recovery following trauma or surgical procedures. These data suggest that recovery of a number of patients eating a poor quality diet may be expedited with a change in diet to one that is healthier.


Assuntos
Tecido Adiposo , Comportamento Animal/fisiologia , Glicemia , Densidade Óssea , Dieta Ocidental/efeitos adversos , Inflamação , Microglia/imunologia , Dor/imunologia , Medula Espinal/imunologia , Animais , Citocinas/sangue , Feminino , Inflamação/sangue , Inflamação/complicações , Inflamação/etiologia , Inflamação/imunologia , Leptina/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
4.
J Neurosci ; 34(43): 14375-87, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339750

RESUMO

Accumulating evidence strongly implicates the transcriptional coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in the pathophysiology of multiple neurological disorders, but the downstream gene targets of PGC-1α in the brain have remained enigmatic. Previous data demonstrate that PGC-1α is primarily concentrated in inhibitory neurons and that PGC-1α is required for the expression of the interneuron-specific Ca(2+)-binding protein parvalbumin (PV) throughout the cortex. To identify other possible transcriptional targets of PGC-1α in neural tissue, we conducted a microarray on neuroblastoma cells overexpressing PGC-1α, mined results for genes with physiological relevance to interneurons, and measured cortical gene and protein expression of these genes in mice with underexpression and overexpression of PGC-1α. We observed bidirectional regulation of novel PGC-1α-dependent transcripts spanning synaptic [synaptotagmin 2 (Syt2) and complexin 1 (Cplx1)], structural [neurofilament heavy chain (Nefh)], and metabolic [neutral cholesterol ester hydrolase 1 (Nceh1), adenylate kinase 1 (Ak1), inositol polyphosphate 5-phosphatase J (Inpp5j), ATP synthase mitochondrial F1 complex O subunit (Atp5o), phytanol-CoA-2hydroxylase (Phyh), and ATP synthase mitrochondrial F1 complex α subunit 1 (Atp5a1)] functions. The neuron-specific genes Syt2, Cplx1, and Nefh were developmentally upregulated in an expression pattern consistent with that of PGC-1α and were expressed in cortical interneurons. Conditional deletion of PGC-1α in PV-positive neurons significantly decreased cortical transcript expression of these genes, promoted asynchronous GABA release, and impaired long-term memory. Collectively, these data demonstrate that PGC-1α is required for normal PV-positive interneuron function and that loss of PGC-1α in this interneuron subpopulation could contribute to cortical dysfunction in disease states.


Assuntos
Interneurônios/metabolismo , Parvalbuminas/biossíntese , Fatores de Transcrição/biossíntese , Transcrição Gênica/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo
5.
Front Cell Neurosci ; 8: 441, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25610371

RESUMO

Alterations in the expression and activity of the transcriptional coactivator peroxisome proliferator-activated receptor γ coactivator-1α (ppargc1a or PGC-1α) have been reported in multiple movement disorders, yet it is unclear how a lack of PGC-1α impacts transcription and function of the cerebellum, a region with high PGC-1α expression. We show here that mice lacking PGC-1α exhibit ataxia in addition to the previously described deficits in motor coordination. Using q-RT-PCR in cerebellar homogenates from PGC-1α(-/-) mice, we measured expression of 37 microarray-identified transcripts upregulated by PGC-1α in SH-SY5Y neuroblastoma cells with neuroanatomical overlap with PGC-1α or parvalbumin (PV), a calcium buffer highly expressed by Purkinje cells. We found significant reductions in transcripts with synaptic (complexin1, Cplx1; Pacsin2), structural (neurofilament heavy chain, Nefh), and metabolic (isocitrate dehydrogenase 3a, Idh3a; neutral cholesterol ester hydrolase 1, Nceh1; pyruvate dehydrogenase alpha 1, Pdha1; phytanoyl-CoA hydroxylase, Phyh; ubiquinol-cytochrome c reductase, Rieske iron-sulfur polypeptide 1, Uqcrfs1) functions. Using conditional deletion of PGC-1α in PV-positive neurons, we determined that 50% of PGC-1α expression and a reduction in a subset of these transcripts could be explained by its concentration in PV-positive neuronal populations in the cerbellum. To determine whether there were functional consequences associated with these changes, we conducted stereological counts and spike rate analysis in Purkinje cells, a cell type rich in PV, from PGC-1α(-/-) mice. We observed a significant loss of Purkinje cells by 6 weeks of age, and the remaining Purkinje cells exhibited a 50% reduction in spike rate. Together, these data highlight the complexity of PGC-1α's actions in the central nervous system and suggest that dysfunction in multiple cell types contribute to motor deficits in the context of PGC-1α deficiency.

6.
Hum Mol Genet ; 22(24): 4988-5000, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23886663

RESUMO

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset Parkinson's disease (PD). Emerging evidence suggests a role for LRRK2 in the endocytic pathway. Here, we show that LRRK2 is released in extracellular microvesicles (i.e. exosomes) from cells that natively express LRRK2. LRRK2 localizes to collecting duct epithelial cells in the kidney that actively secrete exosomes into urine. Purified urinary exosomes contain LRRK2 protein that is both dimerized and phosphorylated. We provide a quantitative proteomic profile of 1673 proteins in urinary exosomes and find that known LRRK2 interactors including 14-3-3 are some of the most abundant exosome proteins. Disruption of the 14-3-3 LRRK2 interaction with a 14-3-3 inhibitor or through acute LRRK2 kinase inhibition potently blocks LRRK2 release in exosomes, but familial mutations in LRRK2 had no effect on secretion. LRRK2 levels were overall comparable but highly variable in urinary exosomes derived from PD cases and age-matched controls, although very high LRRK2 levels were detected in some PD affected cases. We further characterized LRRK2 exosome release in neurons and macrophages in culture, and found that LRRK2-positive exosomes circulate in cerebral spinal fluid (CSF). Together, these results define a pathway for LRRK2 extracellular release, clarify one function of the LRRK2 14-3-3 interaction and provide a foundation for utilization of LRRK2 as a biomarker in clinical trials.


Assuntos
Proteínas 14-3-3/metabolismo , Exossomos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Células Epiteliais/metabolismo , Humanos , Túbulos Renais Coletores/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Mutação , Neurônios/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/líquido cefalorraquidiano , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Ratos , Ratos Transgênicos
7.
Biochem Biophys Res Commun ; 379(2): 578-82, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19118529

RESUMO

Recent evidence suggests that the transcriptional coactivator peroxisome proliferator activated receptor gamma coactivator 1alpha (PGC-1alpha) is involved in the pathology of Huntington's Disease (HD). While animals lacking PGC-1alpha express lower levels of genes involved in antioxidant defense and oxidative phosphorylation in the brain, little is known about other targets for PGC-1alpha in neuronal cells and whether there are ways to pharmacologically target PGC-1alpha in neurons. Here, PGC-1alpha overexpression in SH-SY5Y neuroblastoma cells upregulated expression of genes involved in mitochondrial function, glucose transport, fatty acid metabolism, and synaptic function. Overexpression also decreased vulnerability to hydrogen peroxide-induced cell death and caspase 3 activation. Treatment of cells with the histone deacetylase inhibitors (HDACi's) trichostatin A and valproic acid upregulated PGC-1alpha and glucose transporter 4 (GLUT4). These results suggest that PGC-1alpha regulates multiple pathways in neurons and that HDACi's may be good candidates to target PGC-1alpha and GLUT4 in HD and other neurological disorders.


Assuntos
Regulação da Expressão Gênica , Proteínas de Choque Térmico/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Apoptose/genética , Transporte Biológico/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Ácidos Graxos/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 4/genética , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Doença de Huntington/enzimologia , Doença de Huntington/genética , Doença de Huntington/metabolismo , Peróxido de Hidrogênio/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neuroblastoma , Neurônios/enzimologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ácido Valproico/farmacologia
8.
Neurosci Lett ; 439(3): 269-74, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18538475

RESUMO

Recent evidence indicates that mitochondrial homeostasis is critical for myelination and maintenance of peripheral nerve function. Mice lacking the metabolic transcriptional coactivator peroxisome proliferator activated receptor gamma coactivator 1alpha (PGC-1alpha) show reductions in expression of myelin-related proteins and exhibit myelin-associated lesions, so we identified PGC-1alpha target genes in Schwann cells (SCs) in vitro to determine potential roles for PGC-1alpha in glia and tested whether PGC-1alpha was sufficient for SC differentiation and myelination. Forskolin-induced differentiation was associated with an upregulation of PGC-1alpha mRNA and protein, and while overexpression of PGC-1alpha upregulated genes such as manganese superoxide dismutase and estrogen-related receptor alpha, it was not sufficient for induction of differentiation. Both PGC-1alpha overexpression and forskolin exposure caused an increase in the mitochondrial fusion-related protein mitofusin 1. These studies suggest that PGC-1alpha might be a potential target to promote mitochondrial stability during differentiation and myelination.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colforsina/farmacologia , Proteínas de Ligação a RNA/metabolismo , Receptores de Estrogênio/metabolismo , Células de Schwann/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Indóis , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , RNA Mensageiro/metabolismo , Ratos , Receptores de Estrogênio/genética , Nervo Isquiático/citologia , Superóxido Dismutase/genética , Transfecção , Receptor ERRalfa Relacionado ao Estrogênio
9.
J Neurosci ; 23(28): 9459-68, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14561876

RESUMO

Conflicting data have emerged regarding the role of complement activation in the pathophysiology of cerebral ischemia. On the basis of considerable evidence implicating inflammatory mediators in the progression of neonatal brain injury, we evaluated the contribution of complement activation to cerebral hypoxic-ischemic (HI) injury in the neonatal rat. To elicit unilateral forebrain HI injury, 7-d-old rats underwent right carotid ligation followed by 1.5-2 hr of exposure to 8% oxygen. Using immunoprecipitation and Western blot assays, we determined that HI induces local complement cascade activation as early as 8 hr post-HI; there was an eightfold increase in the activation fragment inactivated C3b at 16 hr. With immunofluorescence assays and confocal microscopy, both C3 and C9 were localized to injured neurons 16 and 24 hr post-HI. To investigate the contribution of systemic complement to brain injury, we administered the complement-depleting agent cobra venom factor (CVF) 24 hr before HI lesioning and evaluated both acute HI-induced complement deposition and the extent of resulting tissue injury 5 d after lesioning. CVF depleted both systemic and brain C3 by the time of surgery and reduced infarct size. Analysis of lesioned, CVF-treated animals demonstrated minimal neuronal C3 deposition but no reduction in C9 deposition. C3-immunoreactive microglia were identified in injured areas. These results indicate that complement activation contributes to HI injury in neonatal rat brain, systemic administration of CVF does not eliminate complement deposition within injured brain, and microglia may represent an important local source of C3 after acute brain injury.


Assuntos
Encéfalo/fisiopatologia , Ativação do Complemento , Hipóxia-Isquemia Encefálica/etiologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Complemento C3b/metabolismo , Complemento C9/metabolismo , Proteínas Inativadoras do Complemento/farmacologia , Modelos Animais de Doenças , Venenos Elapídicos/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Microglia/metabolismo , Microglia/patologia , Ratos , Ratos Sprague-Dawley
10.
Stroke ; 33(3): 795-801, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11872906

RESUMO

BACKGROUND AND PURPOSE: Macrophage inflammatory protein (MIP)-1alpha is a well-characterized monocyte chemoattractant; its role in regulating monocyte and microglial recruitment and activation in the injured neonatal brain is unknown. We evaluated the impact of acute hypoxic-ischemic (HI) brain injury on the expression of MIP-1alpha in neonatal rat brain. METHODS: To elicit forebrain ischemic injury, 7-day-old (P7) rats underwent right carotid ligation, followed by 2.5 hours of 8% oxygen exposure. We used an enzyme-linked immunosorbent assay and immunohistochemistry to detect MIP-1alpha; double-labeling immunofluorescence assays were analyzed with confocal microscopy to identify cellular sources of MIP-1alpha. Immunocytochemistry assays were also used to detect 2 MIP-1alpha receptors, CCR1 and CCR5. RESULTS: We found marked increases in tissue concentrations of MIP-1alpha in the HI cerebral hemisphere, peaking from 8 to 72 hours after lesioning. Immunocytochemistry assays revealed that MIP-1alpha was constitutively expressed in physiologically activated microglia; from 8 to 120 hours after lesioning, MIP-1alpha immunoreactive monocytes and microglia accumulated in the lesion territory. In immunoreactive cells, MIP-1alpha was diffusely distributed throughout the cytoplasm at early post-HI time intervals; by 72 hours, MIP-1alpha immunoreactivity was typically concentrated adjacent to the nucleus, a pattern indicative of active MIP-1alpha production. In P7 to P12 brain, many cells expressed MIP-1alpha receptors; both CCR1 and CCR5 immunoreactivity were localized to endothelium and ependyma; CCR1-immunoreactive astrocytes and neurons and CCR5-immunoreactive microglia were also identified. CONCLUSIONS: These data implicate MIP-1alpha as a mediator of the complex and sustained inflammatory response initiated by perinatal HI braininjury.


Assuntos
Encéfalo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Proteínas Inflamatórias de Macrófagos/biossíntese , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação , Encéfalo/patologia , Quimiocina CCL3 , Quimiocina CCL4 , Modelos Animais de Doenças , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Receptores CCR1 , Receptores CCR5/metabolismo , Receptores de Quimiocinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA