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











Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 208: 299-308, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37625657

RESUMO

Progressive death of dopaminergic (DA) neurons is the main cause of Parkinson's disease (PD). The discovery of drug candidates to prevent DA neuronal death is required to address the pathological aspects and alter the process of PD. Azoramide is a new small molecule compound targeting ER stress, which was originally developed for the treatment of diabetes. In this study, pre-treatment with Azoramide was found to suppress mitochondria-targeting neurotoxin MPP+-induced DA neuronal death and locomotor defects in zebrafish larvae. Further study showed that pre-treatment with Azoramide significantly attenuated MPP+-induced SH-SY5Y cell death by reducing aberrant changes in nuclear morphology, mitochondrial membrane potential, intracellular reactive oxygen species, and apoptotic biomarkers. The mechanistic study revealed that Azoramide was able to up-regulate the expression of ER chaperone BiP and thereby prevented MPP+-induced BiP decrease. Furthermore, pre-treatment with Azoramide failed to suppress MPP+-induced cytotoxicity in the presence of the BiP inhibitor HA15. Taken together, these results suggested that Azoramide is a potential neuroprotectant with pro-survival effects against MPP+-induced cell death through up-regulating BiP expression.


Assuntos
1-Metil-4-fenilpiridínio , Neurônios Dopaminérgicos , Chaperona BiP do Retículo Endoplasmático , Neuroblastoma , Animais , Humanos , 1-Metil-4-fenilpiridínio/toxicidade , Apoptose , Morte Celular , Linhagem Celular Tumoral , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neuroblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Peixe-Zebra/metabolismo , Chaperona BiP do Retículo Endoplasmático/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático/metabolismo
2.
Molecules ; 27(12)2022 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-35744822

RESUMO

Eucommia ulmoides Oliver (E. ulmoides) is a popular medicinal herb and health supplement in China, Japan, and Korea, and has a variety of pharmaceutical properties. The neuroendocrine-immune (NEI) network is crucial in maintaining homeostasis and physical or psychological functions at a holistic level, consistent with the regulatory theory of natural medicine. This review aims to systematically summarize the chemical compositions, biological roles, and pharmacological properties of E. ulmoides to build a bridge between it and NEI-associated diseases and to provide a perspective for the development of its new clinical applications. After a review of the literature, we found that E. ulmoides has effects on NEI-related diseases including cancer, neurodegenerative disease, hyperlipidemia, osteoporosis, insomnia, hypertension, diabetes mellitus, and obesity. However, clinical studies on E. ulmoides were scarce. In addition, E. ulmoides derivatives are diverse in China, and they are mainly used to enhance immunity, improve hepatic damage, strengthen bones, and lower blood pressure. Through network pharmacological analysis, we uncovered the possibility that E. ulmoides is involved in functional interactions with cancer development, insulin resistance, NAFLD, and various inflammatory pathways associated with NEI diseases. Overall, this review suggests that E. ulmoides has a wide range of applications for NEI-related diseases and provides a direction for its future research and development.


Assuntos
Eucommiaceae , Hipertensão , Doenças Neurodegenerativas , China , Suplementos Nutricionais , Eucommiaceae/química , Humanos
3.
Oncotarget ; 9(62): 31958-31970, 2018 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-30174789

RESUMO

Angiogenesis is a hallmark for cancer development because it is essential for cancer growth and provides the route for cancer cell migration (metastasis). Understanding the mechanism of angiogenesis and developing drugs that target the process has therefore been a major focus for research on cancer therapy. In this study, we screened 114 FDA-approved anti-cancer drugs for their effects on angiogenesis in the zebrafish. Among those with positive effects, we chose to focus on Ponatinib (AP24534; Iclusig®) for further investigation. Ponatinib is an inhibitor of the tyrosine kinase BCR-ABL in chronic myeloid leukemia (CML), and its clinical trial has been approved by FDA for the treatment of the disease. In recent clinical trials, however, some side effects have been reported for Ponatinib, mostly on blood vessel disorders, raising the possibility that this drug may influence angiogenesis. In this study, we demonstrated that Ponatinib was able to suppress the formation of intersegmental vessels (ISV) and subintestinal vessels (SIV) in the zebrafish larvae. The anti-angiogenic effect of Ponatinib was further validated by other bioassays in human umbilical vein endothelial cells (HUVECs), including cell proliferation and migration, tube formation, and wound healing. Further experiments showed that Ponatinib inhibited VEGF-induced VEGFR2 phosphorylation and its downstream signaling pathways including Akt/eNOS/NO pathway and MAPK pathways (ERK and p38MAPK). Taken together, these results suggest that inhibition of VEGF signaling at its receptor level and downstream pathways may likely be responsible for the antiangiogenic activity of Ponatinib.

4.
Redox Biol ; 9: 50-56, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27372058

RESUMO

The pathological increase in the levels of reactive oxygen species (ROS) in the retinal pigment epithelium (RPE), is implicated in the development of age-related macular degeneration (AMD). The discovery of drug candidates to effectively protect RPE cells from oxidative damage is required to resolve the pathological aspects and modify the process of AMD. In this study, a FDA-approved anti-malaria drug, Artemisinin was found to suppress hydrogen peroxide (H2O2)-induced cell death in human RPE cell-D407 cells. Further study showed that Artemisinin significantly suppressed H2O2- induced D407 cell death by restoring abnormal changes in nuclear morphology, intracellular ROS, mitochondrial membrane potential and apoptotic biomarkers. Western blotting analysis showed that Artemisinin was able to activate extracellular regulated ERK/CREB survival signaling. Furthermore, Artemisinin failed to suppress H2O2-induced cytotoxicity and the increase of caspase 3/7 activity in the presence of the ERK inhibitor PD98059. Taken together, these results suggest that Artemisinin is a potential protectant with the pro-survival effects against H2O2 insult through activation of the ERK/CREB pathway.


Assuntos
Artemisininas/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Free Radic Biol Med ; 97: 158-167, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27242266

RESUMO

The production of nitric oxide (NO) is one of the primary mediators of ischemic damage, glutamate neurotoxicity and neurodegeneration and therefore inhibition of NO-induced neurotoxicity may be considered a therapeutic target for reducing neuronal cell death (neuroprotection). In this study, artemisinin, a well-known anti-malaria drug was found to suppress sodium nitroprusside (SNP, a nitric oxide donor)-induced cell death in the PC12 cells and brain primary cortical neuronal cultures. Pretreatment of PC12 cells with artemisinin significantly suppressed SNP-induced cell death by decreasing the extent of oxidation, preventing the decline of mitochondrial membrane potential, restoring abnormal changes in nuclear morphology and reducing lactate dehydrogenase release and inhibiting caspase 3/7 activities. Western blotting analysis revealed that artemisinin was able to activate extracellular regulated protein kinases (ERK) pathway. Furthermore, the ERK inhibitor PD98059 blocked the neuroprotective effect of artemisinin whereas the PI3K inhibitor LY294002 had no effect. Cumulatively these findings support the notion that artemisinin confers neuroprotection from SNP-induce neuronal cell death insult, a phenomenon coincidentally related to activation of ERK phosphorylation. This SNP-induced oxidative insult in PC12 cell culture model may be useful to investigate molecular mechanisms of NO-induced neurotoxicity and drug-induced neuroprotection, and to generate novel therapeutic concepts for ischemic disease treatment.


Assuntos
Artemisininas/administração & dosagem , Degeneração Neural/tratamento farmacológico , Neurônios/efeitos dos fármacos , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromonas/administração & dosagem , Flavonoides/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Morfolinas/administração & dosagem , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/patologia , Neuroproteção/efeitos dos fármacos , Doadores de Óxido Nítrico/metabolismo , Nitroprussiato/toxicidade , Células PC12 , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Transdução de Sinais/efeitos dos fármacos
6.
Mol Biosyst ; 12(9): 2713-21, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27354305

RESUMO

Rho-associated protein kinase (ROCK) mediated the reorganization of the actin cytoskeleton and has been implicated in the spread and metastatic process of cancer. In this study, structure-based high-throughput virtual screening was used to identify candidate compounds targeting ROCK2 from a chemical library. Moreover, high-content screening based on neurite outgrowth of SH-SY5Y cells (a human neuroblastoma cell line) was used for accelerating the identification of compounds with characteristics of ROCK2 inhibitors. The effects of bioactive ROCK2 inhibitor candidates were further validated using other bioassays including cell migration and wound healing in SH-SY5Y cells. Through the combined virtual and high-content drug screening, the compound 1,3-benzodioxol-5-yl[1-(5-isoquinolinylmethyl)-3-piperidinyl]-methanone (BIPM) was identified as a novel and potent ROCK2 inhibitor. Exposure of SH-SY5Y cells to BIPM led to significant changes in neurite length, cell migration and actin stress fibers. Further experiments demonstrated that BIPM was able to significantly inhibit phosphorylation of cofilin, a regulatory protein of actin cytoskeleton. These results suggest that BIPM could be considered as a promising scaffold for the further development of ROCK2 inhibitors for anti-cancer metastasis.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteínas Quinases/química , Quinases Associadas a rho/química , Sítios de Ligação , Biomarcadores , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Simulação por Computador , Descoberta de Drogas , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Conformação Molecular , Estrutura Molecular , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas , Fluxo de Trabalho , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
7.
Free Radic Biol Med ; 89: 1057-66, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26415025

RESUMO

Progressive degeneration and death of neurons are main causes of neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. Although some current medicines may temporarily improve their symptoms, no treatments can slow or halt the progression of neuronal death. In this study, a pyrimidine derivative, benzyl 7-(4-hydroxy-3-methoxyphenyl)-5-methyl-4,7-dihydrotetrazolo[1,5-a]pyrimidine-6-carboxylate (BHDPC), was found to attenuate dramatically the MPTP-induced death of dopaminergic neurons and improve behavior movement deficiency in zebrafish, supporting its potential neuroprotective activity in vivo. Further study in rat organotypic cerebellar cultures indicated that BHDPC was able to suppress MPP(+)-induced cell death of brain tissue slices ex vivo. The protective effect of BHDPC against MPP(+) toxicity was also effective in human neuroblastoma SH-SY5Y cells through restoring abnormal changes in mitochondrial membrane potential and numerous apoptotic regulators. Western blotting analysis indicated that BHDPC was able to activate PKA/CREB survival signaling and further up-regulate Bcl2 expression. However, BHDPC failed to suppress MPP(+)-induced cytotoxicity and the increase of caspase 3 activity in the presence of the PKA inhibitor H89. Taken together, these results suggest that BHDPC is a potential neuroprotectant with prosurvival effects in multiple models of neurodegenerative disease in vitro, ex vivo, and in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Intoxicação por MPTP/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/toxicidade , Pirimidinas/farmacologia , Tetrazóis/farmacologia , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Western Blotting , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Herbicidas/toxicidade , Humanos , Locomoção/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Teóricos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
8.
Sci Rep ; 5: 10353, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-26037049

RESUMO

Protein disulfide isomerase (PDI) family members including PDI and ERp57 emerge as novel targets for anti-thrombotic treatments, but chemical agents with selectivity remain to be explored. We previously reported a novel derivative of danshensu (DSS), known as ADTM, displayed strong cardioprotective effects against oxidative stress-induced cellular injury in vitro and acute myocardial infarct in vivo. Herein, using chemical proteomics approach, we identified ERp57 as a major target of ADTM. ADTM displayed potent inhibitory effects on the redox activity of ERp57, inhibited the adenosine diphosphate (ADP)-induced expressions of P-selectin and αIIbß3 integrin, and disrupted the interaction between ERp57 and αIIbß3. In addition, ADTM inhibited both arachidonic acid (AA)-induced and ADP-induced platelet aggregation in vitro. Furthermore, ADTM significantly inhibited rat platelet aggregation and thrombus formation in vivo. Taken together, ADTM represents a promising candidate for anti-thrombotic therapy targeting ERp57.


Assuntos
Fibrinolíticos/farmacologia , Isomerases de Dissulfetos de Proteínas/metabolismo , Difosfato de Adenosina/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Moléculas de Adesão Celular/metabolismo , Cloretos/efeitos adversos , Modelos Animais de Doenças , Ativação Enzimática , Compostos Férricos/efeitos adversos , Fibrinolíticos/química , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Humanos , Lactatos/química , Lactatos/farmacologia , Proteínas dos Microfilamentos/metabolismo , Modelos Biológicos , Selectina-P/genética , Selectina-P/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Ativação Plaquetária , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Proteômica , Ratos , Trombose/tratamento farmacológico , Trombose/etiologia , Trombose Venosa/etiologia
9.
J Ethnopharmacol ; 170: 8-15, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25934514

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: The fruit of Schisandra chinensis (Turcz.) Baill, has been traditionally used in management of liver diseases and ageing associated neurodegeneration. The bioactive compound from this medicinal plant would be valuable for its potential use in prevention and treatment of Parkinson׳s disease. AIM OF THE STUDY: The overall objective of the present study was to understand the neuroprotective effect of schisantherin A, a dibenzocyclooctadiene lignan from the fruit of S. chinensis (Turcz.) Baill, and to elucidate its underlying mechanism of action. MATERIAL AND METHODS: This study investigated the protective effect of schisantherin A against selective dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA)-induced neural damage in human neuroblastoma SH-SY5Y cells and zebrafish models. Oxidative stress and related signaling pathways underlying the neuroprotective effect were determined by multiple biochemical assays and Western blot. RESULTS: Pretreatment with schisantherin A offered neuroprotection against 6-OHDA-induced SH-SY5Y cytotoxicity. Moreover, schisantherin A could prevent 6-OHDA-stimulated dopaminergic neuron loss in zebrafish. Our mechanistic study showed that schisantherin A can regulate intracellular ROS accumulation, and inhibit NO overproduction by down-regulating the over-expression of iNOS in 6-OHDA treated SH-SY5Y cells. Schisantherin A also protects against 6-OHDA-mediated activation of MAPKs, PI3K/Akt and GSK3ß. CONCLUSION: These findings demonstrate that schisantherin A may have potential therapeutic value for neurodegenerative diseases associated with abnormal oxidative stress such as Parkinson׳s disease.


Assuntos
Ciclo-Octanos/farmacologia , Dioxóis/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Lignanas/farmacologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antiparkinsonianos/isolamento & purificação , Antiparkinsonianos/farmacologia , Linhagem Celular Tumoral , Ciclo-Octanos/isolamento & purificação , Dioxóis/isolamento & purificação , Neurônios Dopaminérgicos/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Lignanas/isolamento & purificação , Neuroblastoma/metabolismo , Fármacos Neuroprotetores/isolamento & purificação , Óxido Nítrico/metabolismo , Oxidopamina/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Schisandra/química , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra
10.
Biochim Biophys Acta ; 1850(6): 1253-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25731980

RESUMO

BACKGROUND: Animal venoms contain a diverse array of proteins and enzymes that are toxic toward various physiological systems. However, there are also some practical medicinal uses for these toxins including use as anti-bacterial and anti-tumor agents. METHODS: In this study, we identified a nine-residue cryptic oligopeptide, KRFKKFFKK (EVP50) that is repeatedly encoded in tandem within vipericidin sequences. RESULTS: EVP50 displayed in vivo potent lethal toxicity to zebrafish larvae (LD50=6 µM) when the peptide's N-terminus was chemically conjugated to rhodamine B (RhoB). In vitro, RhoB-conjugated EVP50 (RhoB-EVP50) exhibited a concentration-dependent cytotoxic effect toward MCF-7 and MDA-MB-231 breast cancer cells. In MCF-7 cells, the RhoB-EVP50 nonapeptide accumulated inside the cells within minutes. In the cytoplasm, the RhoB-EVP50 induced extracellular calcium influx and intracellular calcium release. Membrane budding was also observed after incubation with micromolar concentrations of the fluorescent EVP50 conjugate. CONCLUSIONS: The conjugate's interference with calcium homeostasis, its intracellular accumulation and its induced membrane dysfunction (budding and vacuolization) seem to act in concert to disrupt the cell circuitry. Contrastively, unconjugated EVP50 peptide did not display neither toxic nor cytotoxic activities in our in vivo and in vitro models. GENERAL SIGNIFICANCE: The synergic mechanism of toxicity was restricted to the structurally modified encrypted vipericidin nonapeptide.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Catelicidinas/farmacologia , Oligopeptídeos/farmacologia , Rodaminas/farmacologia , Venenos de Víboras/química , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos , Antineoplásicos/isolamento & purificação , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Neoplasias da Mama/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Catelicidinas/isolamento & purificação , Catelicidinas/metabolismo , Catelicidinas/toxicidade , Membrana Celular/efeitos dos fármacos , Membrana Celular/patologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Humanos , Larva/efeitos dos fármacos , Dose Letal Mediana , Células MCF-7 , Dados de Sequência Molecular , Oligopeptídeos/isolamento & purificação , Oligopeptídeos/metabolismo , Oligopeptídeos/toxicidade , Rodaminas/metabolismo , Rodaminas/toxicidade , Fatores de Tempo
11.
Free Radic Biol Med ; 84: 331-343, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25769424

RESUMO

Polypharmacology-based strategies using drug combinations with different mechanisms of action are gaining increasing attention as a novel methodology to discover potentially innovative medicines for neurodegenerative disorders. We used this approach to examine the combined neuroprotective effects of two polyphenols, protocatechuic acid (PCA) and chrysin, identified from the fruits of Alpinia oxyphylla. Our results demonstrated synergistic neuroprotective effects, with chrysin enhancing the protective effects of PCA, resulting in greater cell viability and decreased lactate dehydrogenase release from 6-hydroxydopamine-treated PC12 cells. Their combination also significantly attenuated chemically induced dopaminergic neuron loss in both zebrafish and mice. We examined the molecular mechanisms underlying these collective cytoprotective effects through proteomic analysis of treated PC12 cells, resulting in the identification of 12 regulated proteins. Two were further characterized, leading to the determination that pretreatment with PCA and chrysin resulted in (i) increased nuclear factor-erythroid 2-related factor 2 protein expression and transcriptional activity; (ii) modulation of cellular redox status with the upregulated expression of hallmark antioxidant enzymes, including heme oxygenase-1, superoxide dismutase, and catalase; and (iii) decreased levels of malondialdehyde, a known lipid peroxidation product. Treatment with PCA and chrysin also inhibited activation of nuclear factor-κB and expression of inducible nitric oxide synthase. Our findings suggest that natural products, when used in combination, can be effective potential therapeutic agents for treating diseases such as Parkinson disease. A therapy involving both PCA and chrysin exhibits its enhanced neuroprotective effects through a combination of cellular mechanisms: antioxidant cytoprotection and anti-inflammation.


Assuntos
Antiparkinsonianos/farmacologia , Flavonoides/farmacologia , Hidroxibenzoatos/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Animais , Antiparkinsonianos/uso terapêutico , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/fisiologia , Avaliação Pré-Clínica de Medicamentos , Flavonoides/uso terapêutico , Heme Oxigenase-1/metabolismo , Hidroxibenzoatos/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo , Células PC12 , Proteoma/metabolismo , Proteômica , Ratos , Fator de Transcrição RelA/metabolismo , Peixe-Zebra
12.
Neurosci Lett ; 593: 7-12, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25770828

RESUMO

Dibenzocyclooctadiene lignans represent a unique group of natural chemical structures, are considered as protectants against neuronal cell death and cognitive impairment in neurological disorders. Among the family of dibenzocyclooctadiene lignan analogs from the fruit of Schisandra chinensis (Turcz.) Baill, neuroprotective potential of schisantherin A (StA) has not yet been characterized. In this study, 1-methyl-4-phenylpyridinium ion (MPP(+))-incubated SH-SY5Y cells and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice were used to study the neuroprotection of StA. Pretreatment with StA significantly inhibited MPP(+)-induced cytotoxicity in SH-SY5Y cells. Moreover, StA conferred significant protection against MPTP-induced loss of TH-positive dopaminergic neurons in a Parkinson's disease (PD) mice model. Structure activity relationship analysis suggested that methylenedioxy, benzoyloxy and methoxyl groups, in the dibenzocyclooctadiene lignan of StA, were probably functionally important to its neuroprotective activity. In addition, Western blotting analysis demonstrated that StA exhibited neuroprotection against MPP(+) through the regulation of two distinct pathways including increasing CREB-mediated Bcl-2 expression and activating PI3K/Akt survival signaling suggesting that StA might be a promising neuroprotective agent for the prevention of PD.


Assuntos
Antiparkinsonianos/farmacologia , Ciclo-Octanos/farmacologia , Dioxóis/farmacologia , Lignanas/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , 1-Metil-4-fenilpiridínio , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Doença de Parkinson/etiologia , Doença de Parkinson/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais
13.
Cell Signal ; 27(1): 26-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25289861

RESUMO

Hepatoma upregulated protein (HURP) is originally isolated during the search for the genes associated with hepatoma. HURP is upregulated in many human cancers. Culture cells exhibit transformed and invasive phenotype when ectopic HURP is introduced, revealing HURP as an oncogene candidate. Our previous studies demonstrated that Aurora-A regulated the cell transforming activities of HURP by phosphorylating HURP at four serines. To unravel how the Aurora-A/HURP cascade contributes to cell transformation, we firstly noticed that HURP shuttled between cytoplasm and nucleus. The nuclear localization activity of HURP was promoted or abolished by overexpression or knockdown of Aurora-A. Similarly, the HURP phosphorylation mimicking mutant 4E had higher nuclear targeting activity than the phosphorylation deficient mutant 4A. The HURP 4E accelerated G1 progression and upregulated cyclin E1, and the cyclin E1 upregulating and cell transforming activities of HURP were diminished when the nuclear localization signal (NLS) was removed from HURP. Furthermore, HURP employed p38/nuclear factor-κB (NF-κB) cascade to stimulate cell growth. Interestingly, NF-κB trapped HURP in nucleus by interacting with HURP 4E. At last, the HURP/NF-κB complex activated the cyclin E1 promoter. Collectively, Aurora-A/HURP relays cell transforming signal to NF-κB, and the HURP/NF-κB complex is engaged in the regulation of cyclin E1 expression.


Assuntos
Carcinoma Hepatocelular/metabolismo , Núcleo Celular/metabolismo , Ciclina E/metabolismo , Neoplasias Hepáticas/metabolismo , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Oncogênicas/metabolismo , Regulação para Cima , Aurora Quinase A/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Fase G1 , Humanos , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases , Fosforilação , Transporte Proteico
14.
Free Radic Biol Med ; 74: 283-93, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24973649

RESUMO

Parkinson disease (PD) is a neurodegenerative disease with multifactorial etiopathogenesis. The discovery of drug candidates that act on new targets of PD is required to address the varied pathological aspects and modify the disease process. In this study, a small compound, 2-(5-methyl-1-benzofuran-3-yl)-N-(5-propylsulfanyl-1,3,4-thiadiazol-2-yl) acetamide (MBPTA) was identified as a novel Rho-associated protein kinase inhibitor with significant protective effects against 1-methyl-4-phenylpyridinium ion (MPP(+))-induced damage in SH-SY5Y neuroblastoma cells. Further investigation showed that pretreatment of SH-SY5Y cells with MBPTA significantly suppressed MPP(+)-induced cell death by restoring abnormal changes in nuclear morphology, mitochondrial membrane potential, and numerous apoptotic regulators. MBPTA was able to inhibit MPP(+)-induced reactive oxygen species (ROS)/NO generation, overexpression of inducible NO synthase, and activation of NF-κB, indicating the critical role of MBPTA in regulating ROS/NO-mediated cell death. Furthermore, MBPTA was shown to activate PI3K/Akt survival signaling, and its cytoprotective effect was abolished by PI3K and Akt inhibitors. The structural comparison of a series of MBPTA analogs revealed that the benzofuran moiety probably plays a crucial role in the anti-oxidative stress action. Taken together, these results suggest that MBPTA protects against MPP(+)-induced apoptosis in a neuronal cell line through inhibition of ROS/NO generation and activation of PI3K/Akt signaling.


Assuntos
Antioxidantes/farmacologia , Benzofuranos/farmacologia , Doença de Parkinson/tratamento farmacológico , Tiadiazóis/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Acetamidas/química , Ácidos Alcanossulfônicos/química , Antioxidantes/química , Benzofuranos/química , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Descoberta de Drogas , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Terapia de Alvo Molecular , Proteína Oncogênica v-akt/metabolismo , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Compostos de Piridínio/toxicidade , Transdução de Sinais/efeitos dos fármacos , Tiadiazóis/química
15.
PLoS One ; 9(4): e92905, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24690920

RESUMO

In this study, we applied structure-based virtual screening techniques to identify natural product or natural product-like inhibitors of iNOS. The iNOS inhibitory activity of the hit compounds was characterized using cellular assays and an in vivo zebrafish larvae model. The natural product-like compound 1 inhibited NO production in LPS-stimulated Raw264.7 macrophages, without exerting cytotoxic effects on the cells. Significantly, compound 1 was able to reverse MPTP-induced locomotion deficiency and neurotoxicity in an in vivo zebrafish larval model. Hence, compound 1 could be considered as a scaffold for the further development of iNOS inhibitors for potential anti-inflammatory or anti-neurodegenerative applications.


Assuntos
Produtos Biológicos/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Simulação de Acoplamento Molecular , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Produtos Biológicos/análise , Produtos Biológicos/química , Morte Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/química , Ensaios de Triagem em Larga Escala , Isoenzimas/metabolismo , Larva/efeitos dos fármacos , Larva/fisiologia , Ligantes , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Conformação Molecular , Atividade Motora/efeitos dos fármacos , Fármacos Neuroprotetores/análise , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Termodinâmica , Peixe-Zebra
16.
PLoS One ; 9(1): e87556, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498135

RESUMO

BACKGROUND: Dinoponera quadriceps is a predatory giant ant that inhabits the Neotropical region and subdues its prey (insects) with stings that deliver a toxic cocktail of molecules. Human accidents occasionally occur and cause local pain and systemic symptoms. A comprehensive study of the D. quadriceps venom gland transcriptome is required to advance our knowledge about the toxin repertoire of the giant ant venom and to understand the physiopathological basis of Hymenoptera envenomation. RESULTS: We conducted a transcriptome analysis of a cDNA library from the D. quadriceps venom gland with Sanger sequencing in combination with whole-transcriptome shotgun deep sequencing. From the cDNA library, a total of 420 independent clones were analyzed. Although the proportion of dinoponeratoxin isoform precursors was high, the first giant ant venom inhibitor cysteine-knot (ICK) toxin was found. The deep next generation sequencing yielded a total of 2,514,767 raw reads that were assembled into 18,546 contigs. A BLAST search of the assembled contigs against non-redundant and Swiss-Prot databases showed that 6,463 contigs corresponded to BLASTx hits and indicated an interesting diversity of transcripts related to venom gene expression. The majority of these venom-related sequences code for a major polypeptide core, which comprises venom allergens, lethal-like proteins and esterases, and a minor peptide framework composed of inter-specific structurally conserved cysteine-rich toxins. Both the cDNA library and deep sequencing yielded large proportions of contigs that showed no similarities with known sequences. CONCLUSIONS: To our knowledge, this is the first report of the venom gland transcriptome of the New World giant ant D. quadriceps. The glandular venom system was dissected, and the toxin arsenal was revealed; this process brought to light novel sequences that included an ICK-folded toxins, allergen proteins, esterases (phospholipases and carboxylesterases), and lethal-like toxins. These findings contribute to the understanding of the ecology, behavior and venomics of hymenopterans.


Assuntos
Venenos de Formiga/biossíntese , Formigas/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Insetos/biossíntese , Transcriptoma/fisiologia , Animais , Venenos de Formiga/genética , Formigas/genética , Perfilação da Expressão Gênica/métodos , Humanos , Proteínas de Insetos/genética
17.
Mol Biosyst ; 10(2): 206-14, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24264059

RESUMO

Minor variants have significant implications in quasispecies evolution, early cancer detection and non-invasive fetal genotyping but their accurate detection by next-generation sequencing (NGS) is hampered by sequencing errors. We generated sequencing data from mixtures at predetermined ratios in order to provide insight into sequencing errors and variations that can arise for which simulation cannot be performed. The information also enables better parameterization in depth of coverage, read quality and heterogeneity, library preparation techniques, technical repeatability for mathematical modeling, theory development and simulation experimental design. We devised minor variant authentication rules that achieved 100% accuracy in both testing and validation experiments. The rules are free from tedious inspection of alignment accuracy, sequencing read quality or errors introduced by homopolymers. The authentication processes only require minor variants to: (1) have minimum depth of coverage larger than 30; (2) be reported by (a) four or more variant callers, or (b) DiBayes or LoFreq, plus SNVer (or BWA when no results are returned by SNVer), and with the interassay coefficient of variation (CV) no larger than 0.1. Quantification accuracy undermined by sequencing errors could neither be overcome by ultra-deep sequencing, nor recruiting more variant callers to reach a consensus, such that consistent underestimation and overestimation (i.e. low CV) were observed. To accommodate stochastic error and adjust the observed ratio within a specified accuracy, we presented a proof of concept for the use of a double calibration curve for quantification, which provides an important reference towards potential industrial-scale fabrication of calibrants for NGS.


Assuntos
Variação Genética , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Proteínas não Estruturais Virais/genética , Algoritmos , Genoma Bacteriano , Mutação , Plasmídeos , Sensibilidade e Especificidade , Software
18.
Neurosci Lett ; 543: 121-5, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23562886

RESUMO

The overproduction of reactive oxygen species (ROS) has been implicated in the development of neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease (AD). Previous studies have indicated that danshensu (beta-3,4-dihydroxyphenyl-lactic acid), a main hydrophilic component of the Chinese materia medica Radix Salviae Miltiorrhizae (Danshen, Pharmacopoeia of PR China), has ROS scavenging and antioxidant activities, however its mechanism of action was not clear. In this study, we investigated whether the protective effects of danshensu against neurotoxin 6-hydroxydopamine (6-OHDA)-induced oxidative stress involved the Nrf2/HO-1 pathways. Pretreatment with danshensu in PC12 cells significantly attenuated 6-OHDA-induced cytotoxicity and the production of ROS. Danshensu activated the nuclear translocation of Nrf2 to increase heme oxygenase-1 (HO-1), conferring protection against ROS. Danshensu induced the phosphorylation of Akt, and its cytoprotective effect was abolished by PI3K, Akt and HO-1 inhibitors. These results confirmed the crucial role of PI3K/Akt and HO-1 signaling pathways as the underlying mechanistic action of danshensu. Taken together, the results suggest that danshensu enhances HO-1 expression to suppress 6-OHDA-induced oxidative damage via PI3K/Akt/Nrf2 signaling pathways. Moreover, 6-OHDA-induced dopaminergic neuronal loss in zebrafish could be reduced by danshensu, further supporting the neuroprotective potential of danshensu.


Assuntos
Lactatos/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Oxidopamina , Doença de Parkinson/patologia , Animais , Elementos de Resposta Antioxidante , Morte Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Medicamentos de Ervas Chinesas/farmacologia , Heme Oxigenase-1/metabolismo , Lactatos/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/etiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Peixe-Zebra
19.
Chembiochem ; 12(15): 2306-9, 2011 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-21887840

RESUMO

Cellular redox conditions affect Gsp amidase activity in Escherichia coli. Guided by the structure and catalytic mechanism of the amidase, we designed and synthesized an acyloxymethyl ketone-based activity probe containing a biotin handle. This probe was used to monitor Gsp amidase activity in E. coli lysates that had been subjected to oxidative or methylglyoxal-induced stress.


Assuntos
Amidoidrolases/metabolismo , Ensaios Enzimáticos/métodos , Escherichia coli/enzimologia , Glutationa/análogos & derivados , Cetonas/metabolismo , Espermidina/análogos & derivados , Sítios de Ligação , Glutationa/metabolismo , Cetonas/química , Modelos Moleculares , Oxirredução , Espermidina/metabolismo
20.
EMBO J ; 25(24): 5970-82, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17124497

RESUMO

Most organisms use glutathione to regulate intracellular thiol redox balance and protect against oxidative stress; protozoa, however, utilize trypanothione for this purpose. Trypanothione biosynthesis requires ATP-dependent conjugation of glutathione (GSH) to the two terminal amino groups of spermidine by glutathionylspermidine synthetase (GspS) and trypanothione synthetase (TryS), which are considered as drug targets. GspS catalyzes the penultimate step of the biosynthesis-amide bond formation between spermidine and the glycine carboxylate of GSH. We report herein five crystal structures of Escherichia coli GspS in complex with substrate, product or inhibitor. The C-terminal of GspS belongs to the ATP-grasp superfamily with a similar fold to the human glutathione synthetase. GSH is likely phosphorylated at one of two GSH-binding sites to form an acylphosphate intermediate that then translocates to the other site for subsequent nucleophilic addition of spermidine. We also identify essential amino acids involved in the catalysis. Our results constitute the first structural information on the biochemical features of parasite homologs (including TryS) that underlie their broad specificity for polyamines.


Assuntos
Amida Sintases/metabolismo , Escherichia coli/enzimologia , Trifosfato de Adenosina/metabolismo , Amida Sintases/antagonistas & inibidores , Amida Sintases/química , Sequência de Aminoácidos , Sítios de Ligação , Catálise , Cristalografia por Raios X , Dimerização , Inibidores Enzimáticos/farmacologia , Glutationa/metabolismo , Humanos , Cinética , Magnésio/metabolismo , Dados de Sequência Molecular , Fosfatos/metabolismo , Fosforilação , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico , Homologia de Sequência , Espermidina/metabolismo , Eletricidade Estática , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA