Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 26(9): 5087-5096, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33483691

RESUMO

The fine-tuning of neuroinflammation is crucial for brain homeostasis as well as its immune response. The transcription factor, nuclear factor-κ-B (NFκB) is a key inflammatory player that is antagonized via anti-inflammatory actions exerted by the glucocorticoid receptor (GR). However, technical limitations have restricted our understanding of how GR is involved in the dynamics of NFκB in vivo. In this study, we used an improved lentiviral-based reporter to elucidate the time course of NFκB and GR activities during behavioral changes from sickness to depression induced by a systemic lipopolysaccharide challenge. The trajectory of NFκB activity established a behavioral basis for the NFκB signal transition involved in three phases, sickness-early-phase, normal-middle-phase, and depressive-like-late-phase. The temporal shift in brain GR activity was differentially involved in the transition of NFκB signals during the normal and depressive-like phases. The middle-phase GR effectively inhibited NFκB in a glucocorticoid-dependent manner, but the late-phase GR had no inhibitory action. Furthermore, we revealed the cryptic role of basal GR activity in the early NFκB signal transition, as evidenced by the fact that blocking GR activity with RU486 led to early depressive-like episodes through the emergence of the brain NFκB activity. These results highlight the inhibitory action of GR on NFκB by the basal and activated hypothalamic-pituitary-adrenal (HPA)-axis during body-to-brain inflammatory spread, providing clues about molecular mechanisms underlying systemic inflammation caused by such as COVID-19 infection, leading to depression.


Assuntos
Depressão/metabolismo , NF-kappa B , Receptores de Glucocorticoides , Animais , Encéfalo/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Camundongos , NF-kappa B/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Glucocorticoides/metabolismo
2.
Biochem Pharmacol ; 182: 114290, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33075311

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are the most commonly prescribed medications for alleviating pain and inflammation but may cause gastrointestinal tract damage. Proton pump inhibitors (PPI) prevent NSAID-induced gastric damage but may aggravate intestinal damage via dysbiosis and intestinal permeability alteration. Currently, there is growing interest regarding the influence of potassium competitive acid blockers (PCAB) on NSAID-induced enteropathy. Here, we investigated the relative changes in indomethacin-induced enteropathy by combining indomethacin with pantoprazole (as PPI) or revaprazan (as PCAB). We examined intestinal permeability-related molecular changes in in vitro Caco-2 cell models and in an in vivo indomethacin-induced enteropathy rat model. Indomethacin alone or in combination with pantoprazole significantly increased relative lucifer yellow dye flux and decreased relative trans-epithelial electrical resistance and tight junction protein (TJP) expression compare to normal cells. In contrast, indomethacin combined with revaprazan significantly preserved TJPs compare to indomethacin-treated cells. MLC phosphorylation, Rho activation, and ERK activation responsible for TJP were significantly increased by indomethacin alone or a combination of indomethacin and pantoprazole but not by a combination of indomethacin and revaprazan. Intestinal damage scores significantly increased with indomethacin and pantoprazole combination but not with indomethacin and revaprazan combination. Indomethacin and pantoprazole combination significantly activated Rho-GTPase, p-MLC, and p-ERK but significantly decreased TJP expression. However, indomethacin and revaprazan combination significantly preserved TJPs and inactivated Rho-GTPase, MLC, and ERK. Hence, revaprazan rather than PPIs should be co-administered with NSAIDs to mitigate NSAID-induced intestinal damage.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Indometacina/toxicidade , Intestino Delgado/efeitos dos fármacos , Inibidores da Bomba de Prótons/farmacologia , Pirimidinonas/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , Junções Íntimas/efeitos dos fármacos , Animais , Células CACO-2 , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Junções Íntimas/metabolismo , Junções Íntimas/patologia
3.
Neurobiol Aging ; 90: 93-98, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32169355

RESUMO

Partitioning-defective 1 (PAR-1), a conserved cell polarity regulator, plays an important role in synaptic development, and its mutation affects the formation of synaptic boutons and localization of postsynaptic density protein Discs large (Dlg) at the neuromuscular junction (NMJ) in Drosophila. Drosophila PAR-1 and its human homolog, Microtubule affinity-regulating kinases (MARK), are also known to be implicated in Alzheimer's disease (AD) by controlling tau-mediated Aß toxicity. However, the molecular mechanisms of PAR-1 function remain incompletely understood. Here we identified Pod-1, an actin-microtubule crosslinker, which functionally and physically interacts with PAR-1 in Drosophila. Pod-1 prominently co-localizes with PAR-1 in the postsynaptic region and regulates PAR-1 activity at the NMJ. Synaptic defects, including the reduction of boutons and delocalization of Dlg caused by PAR-1 overexpression, were rescued by Pod-1 knockdown. Conversely, the reduction of synaptic boutons in PAR-1 overexpressed NMJ was synergistically enhanced by the overexpression of Pod-1. Furthermore, Pod-1 increases the PAR-1 dependent S262 phosphorylation of tau, which is known to contribute to tau-mediated Aß toxicity. In line with the change of tau phosphorylation, Pod-1 knockdown rescued tau-mediated synaptic toxicity at the NMJ. Our results suggest that Pod-1 may act as a modulator of PAR-1 in synaptic development and tau-mediated toxicity.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Quinase 3 da Glicogênio Sintase/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Sinapses/fisiologia , Proteínas tau/metabolismo , Animais , Drosophila , Proteínas de Drosophila/genética , Epistasia Genética , Quinase 3 da Glicogênio Sintase/genética , Junção Neuromuscular/metabolismo , Fosforilação/genética
4.
Biol Open ; 8(1)2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30341106

RESUMO

Bioluminescence imaging has proven to be a highly sensitive technique for assessing in vitro transcriptional activity toward understanding gene regulation patterns; however, application of this technique is limited for brain research. In particular, the poor spatiotemporal resolution is a major hurdle for monitoring the dynamic changes of transcriptional activity in specific regions of the brain during longitudinal analysis of living animals. To overcome this limitation, in this study, we modified a lentivirus-based luciferase glucocorticoid receptor (GR) reporter by inserting destabilizing sequence genes, and then the reporter was stereotaxically injected in the mouse infralimbic prefrontal cortex (IL-PFC). Using this strategy, we could successfully pin-point and monitor the dynamic changes in GR activity in IL-PFC during normal stress adaptation. The modified reporter showed a 1.5-fold increase in temporal resolution for monitoring GR activity compared to the control, with respect to the intra-individual coefficients of variation. This novel in vivo method has broad applications, as it is readily adaptable to different types of transcription factor arrays as well spanning wide target regions of the brain to other organs and tissues.

5.
Cell Death Dis ; 9(11): 1125, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420654

RESUMO

Leucine-rich repeat kinase 2 (LRRK2), originally identified as a causative genetic factor in Parkinson's disease, is now associated with a number of pathologies. Here, we show that brain injury induces a robust expression of endogenous LRRK2 and suggest a role of LRRK2 after injury. We found that various in vitro and in vivo models of traumatic brain injury (TBI) markedly enhanced LRRK2 expression in neurons and also increased the level of hypoxia-inducible factor (HIF)-1α. Luciferase reporter assay and chromatin immunoprecipitation revealed direct binding of HIF-1α in LRRK2 proximal promoter. We also found that HIF-1α-dependent transcriptional induction of LRRK2 exacerbated neuronal cell death following injury. Furthermore, application of G1023, a specific, brain-permeable inhibitor of LRRK2, substantially prevented brain tissue damage, cell death, and inflammatory response and alleviated motor and cognitive defects induced by controlled cortical impact injury. Together, these results suggest HIF-1α-LRRK2 axis as a potential therapeutic target for brain injury.


Assuntos
Lesões Encefálicas Traumáticas/genética , Córtex Cerebral/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Transcrição Gênica , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Sequência de Bases , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/prevenção & controle , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Cultura Primária de Células , Regiões Promotoras Genéticas , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Desempenho Psicomotor/efeitos dos fármacos , Transdução de Sinais
6.
J Diet Suppl ; 15(1): 11-23, 2018 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-28485986

RESUMO

Polyphenolic compounds have been suggested to be involved in the preservation of neural function via the production of neurotrophic factors in the brain. The nonedible joint part of lotus root (a rhizome of Nelumbo nucifera) has been reported to contain large amounts of polyphenolic compounds and, therefore, is expected to improve neural function by stimulating the production of brain-derived neurotrophic factor (BDNF) in glial cells. The effect of the aqueous extract prepared from the joint part of lotus root on BDNF gene expression was examined in C6 glioma cells as an in vitro model. This extract was shown to increase BDNF messenger ribonucleic acid (mRNA) levels to the elevation of HO-1 mRNA levels in the glioma cells, but failed to cause the elevation of BDNF mRNA levels in the cells pretreated with a HO-1 antisense oligodeoxynucleotide (ODN) or an HO-1 enzyme inhibitor zinc protoporphyrin (ZnPP). These findings strongly suggest that the aqueous extract prepared from the nonedible joint part of lotus root might be able to stimulate BDNF gene expression by enhancing HO-1 activity in the glioma cells, proposing the possibility that the joint part of lotus root might potentially improve neural function through the stimulation of BDNF production in glial cells.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Nelumbo/química , Extratos Vegetais/farmacologia , Rizoma/química , Animais , Linhagem Celular Tumoral , Glioma/genética , Neuroglia/metabolismo , RNA Mensageiro/efeitos dos fármacos , Ratos
7.
Int J Mol Sci ; 17(11)2016 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-27869673

RESUMO

Environmental contamination by trinitrotoluene is of global concern due to its widespread use in military ordnance and commercial explosives. Despite known long-term persistence in groundwater and soil, the toxicological profile of trinitrotoluene and other explosive wastes have not been systematically measured using in vivo biological assays. Zebrafish embryos are ideal model vertebrates for high-throughput toxicity screening and live in vivo imaging due to their small size and transparency during embryogenesis. Here, we used Single Plane Illumination Microscopy (SPIM)/light sheet microscopy to assess the developmental toxicity of explosive-contaminated water in zebrafish embryos and report 2,4,6-trinitrotoluene-associated developmental abnormalities, including defects in heart formation and circulation, in 3D. Levels of apoptotic cell death were higher in the actively developing tissues of trinitrotoluene-treated embryos than controls. Live 3D imaging of heart tube development at cellular resolution by light-sheet microscopy revealed trinitrotoluene-associated cardiac toxicity, including hypoplastic heart chamber formation and cardiac looping defects, while the real time PCR (polymerase chain reaction) quantitatively measured the molecular changes in the heart and blood development supporting the developmental defects at the molecular level. Identification of cellular toxicity in zebrafish using the state-of-the-art 3D imaging system could form the basis of a sensitive biosensor for environmental contaminants and be further valued by combining it with molecular analysis.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Substâncias Explosivas/toxicidade , Trinitrotolueno/toxicidade , Poluentes Químicos da Água/toxicidade , Animais , Apoptose/efeitos dos fármacos , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/patologia , Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/embriologia , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/patologia , Microscopia Intravital , Melanócitos/efeitos dos fármacos , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
8.
Toxicol Sci ; 154(2): 430-441, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27587462

RESUMO

Hydroquinone (HQ), a major benzene metabolite, occurs naturally in various plants and is manufactured for commercial use. Although HQ displays various biological effects, its neuroprotective effects following ischemic insults have not been investigated. In this study, we first examined neuroprotective effects of HQ in a rat model of transient focal cerebral ischemia. Animals were subjected to transient middle cerebral artery occlusion for 120 min. HQ (50 or 100 mg/kg) or vehicle was intraperitoneally administered once at 30 min after ischemia-reperfusion. Neuroprotection by treatment with 100 mg/kg of HQ was shown using evaluation of neurological deficits, positron-emission tomography (PET) and 2,3,5-triphenyltetrazoliumchloride (TTC) staining. In addition, HQ treatment significantly attenuated ischemia-induced Evans blue dye extravasation from blood vessels and significantly increased immunoreactivities of SMI-71 (an endothelial BBB marker) and glucose transporter-1 (GLUT-1, an endothelial cell marker) in ischemic cortex compared to the vehicle-treated ischemia-operated group. Confocal microscopy and western blot analysis also showed that HQ treatment maintained expressions of tight junction proteins (zonula occludens-1 and occludin) in the ischemic cortex. Post-treatment with HQ protected neurons from transient focal cerebral ischemic injury and the neuroprotective effect of HQ might be closely associated with prevention of BBB disruption via maintaining SMI-71 and GLUT-1 expressions as well as prevention of the degradation of zonula occludens-1 and occludin proteins.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Hidroquinonas/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/ultraestrutura , Citoproteção , Modelos Animais de Doenças , Transportador de Glucose Tipo 1/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Atividade Motora/efeitos dos fármacos , Ocludina/metabolismo , Tomografia por Emissão de Pósitrons , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Proteína da Zônula de Oclusão-1/metabolismo
9.
Molecules ; 21(3): 277, 2016 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-26927055

RESUMO

Lindera obtusiloba extracts are commonly used as an alternative medicine due to its numerous health benefits in Korea. However, the antidepressant-like effects of L. obtusiloba extracts have not been fully elucidated. In this study, we aimed to determine whether L. obtusiloba extracts exhibited antidepressant-like activity in rats subjected to forced swim test (FST)-induced depression. Acute treatment of rats with L. obtusiloba extracts (200 mg/kg, p.o.) significantly reduced immobility time and increased swimming time without any significant change in climbing. Rats treated with L. obtusiloba extracts also exhibited a decrease in the limbic hypothalamic-pituitary-adrenal (HPA) axis response to the FST, as indicated by attenuation of the corticosterone response and decreased c-Fos immunoreactivity in the hippocampus CA3 region. In addition, L. obtusiloba extracts, at concentrations that were not affected by cell viability, significantly decreased luciferase activity in response to cortisol in a concentration-dependent manner by the glucocorticoid binding assay in HeLa cells. Our findings suggested that the antidepressant-like effects of L. obtusiloba extracts were likely mediated via the glucocorticoid receptor (GR). Further studies are needed to evaluate the potential of L. obtusiloba extracts as an alternative therapeutic approach for the treatment of depression.


Assuntos
Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Lindera/química , Extratos Vegetais/farmacologia , Animais , Antidepressivos/química , Antidepressivos/uso terapêutico , Depressão/etiologia , Depressão/fisiopatologia , Modelos Animais de Doenças , Células HeLa , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Ratos , Receptores de Glucocorticoides , Natação
10.
Neurochem Int ; 90: 204-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26364578

RESUMO

Zinc oxide nanoparticles (ZnO NPs) are known to induce oxidative stress and modulate an inflammatory process in various cell types. Although the cytotoxic effects of ZnO NPs in various cell types have been evaluated, few neurotoxic surveys on ZnO NPs as well as rescue studies have been reported. This study was designed to examine the neurotoxic ZnO NP concentration according to exposure time and dose, and the mechanisms that underlie ZnO NP-induced neurotoxicity in the SH-SY5Y human neuroblastoma cell line. A significant reduction in neuronal viability as well as distinct morphological findings resulted from application of 15 µM ZnO NPs. Apoptotic injury-as measured by annexin V and caspase 3/7 activities-was significantly elevated at 12 h and 24 h, but not 6 h, after ZnO NP exposure. However, electron microscopy revealed typical necrotic characteristics, such as swelling or loss of cell organelles and rupture of the cytosolic or nuclear membrane at 12 h and 24 h after ZnO NP exposure. In rescue studies, the lipoxygenase (LOX) inhibitor esculetin attenuated ZnO NP-induced neuronal injury. The elevation of PI3 kinase (PI3K) and p-Akt/Akt activities induced by ZnO NP was significantly decreased by esculetin or LY294002. Allopurinol, N-acetyl-l-cysteine and α-tocopherol protected ZnO NP-induced cytotoxicity. Sodium nitroprusside (SNP)-induced neurotoxicity and ZnO NP-mediated NO overproduction were ameliorated by esculetin. Esculetin reduced the production of reactive oxygen species (ROS) and the depletion of antioxidant enzymes induced by ZnO NPs. The concentration of zinc from the dissolution of ZnO NPs increased in proportion to increases in the ZnO NPs concentration. These results suggest that ZnO NPs induce apoptosis via the PI3K/Akt/caspase-3/7 pathway and necrosis by LOX-mediated ROS production elevation.


Assuntos
Apoptose/efeitos dos fármacos , Lipoxigenase/metabolismo , Nanopartículas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Óxido de Zinco/farmacologia , Antioxidantes/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Necrose/induzido quimicamente , Fosfatidilinositol 3-Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo
11.
J Nutr Biochem ; 26(11): 1368-78, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26297476

RESUMO

Piceatannol, a polyphenol which exhibits anticancer activities, is found in grapes, red wine and berries. It has been shown to inhibit several transcription factor pathways. The present study was conducted to determine whether oral administration of piceatannol inhibits mammary tumor progression. 4T1 mammary carcinoma cells were injected into the mammary fat pad of syngeneic female BALB/c mice. Starting 1 day later, piceatannol (10- or 20-mg/kg body weight/day) was administered by oral gavage for 30 days. Piceatannol treatment reduced tumor growth. In tumor tissues, piceatannol treatment reduced the expression of transcription factors P-NFκB p65, P-STAT3 and HIF-1α and multiple proteins involved in regulation of cell cycle progression (Ki67, cyclin D1, cyclin A, CDK2, CDK4), angiogenesis (VEGF-A, VEGFR-2, VE-cadherin, CD31) and lymphangiogenesis (VEGF-C, LYVE-1), as well as macrophage infiltration. Piceatannol significantly increased apoptotic cells and expression of both Bax and cleaved caspase-3 but reduced Bcl-2 expression in tumor tissues. In addition, piceatannol reduced the number and volume of pulmonary tumor nodules and expression of MMP-9 in both lung and tumor. It also reduced tissue levels of cytokines/chemokines, including M-CSF and MCP-1. In vitro results revealed that piceatannol inhibited migration of 4T1 cells and monocytes, as well as secretion of MCP-1 and M-CSF by 4T1 cells. 4T1 cell-conditioned medium stimulated monocyte migration, which was suppressed by a CCR2 antibody. These results indicate that alteration in tumor microenvironment (macrophages, transcription factors, etc.) is an important mechanism by which piceatannol inhibits tumor proliferation, angiogenesis and lymphangiogenesis, leading to suppression of mammary tumor growth and metastasis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Macrófagos/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Estilbenos/administração & dosagem , Estilbenos/farmacologia , Administração Oral , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Feminino , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Macrófagos/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos Endogâmicos BALB C , Neovascularização Patológica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição RelA/metabolismo
12.
Autophagy ; 11(5): 796-811, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25946189

RESUMO

The EWSR1 (EWS RNA-binding protein 1/Ewing Sarcoma Break Point Region 1) gene encodes a RNA/DNA binding protein that is ubiquitously expressed and involved in various cellular processes. EWSR1 deficiency leads to impairment of development and accelerated senescence but the mechanism is not known. Herein, we found that EWSR1 modulates the Uvrag (UV radiation resistance associated) gene at the post-transcription level. Interestingly, EWSR1 deficiency led to the activation of the DROSHA-mediated microprocessor complex and increased the level of Mir125a and Mir351, which directly target Uvrag. Moreover, the Mir125a- and Mir351-mediated reduction of Uvrag was associated with the inhibition of autophagy that was confirmed in ewsr1 knockout (KO) MEFs and ewsr1 KO mice. Taken together, our data indicate that EWSR1 is involved in the post-transcriptional regulation of Uvrag via a miRNA-dependent pathway, resulting in the deregulation of autophagy inhibition. The mechanism of Uvrag and autophagy regulation by EWSR1 provides new insights into the role of EWSR1 deficiency-related cellular dysfunction.


Assuntos
Autofagia , Proteínas de Ligação a Calmodulina/deficiência , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Autofagia/genética , Sequência de Bases , Proteínas de Ligação a Calmodulina/metabolismo , Regulação para Baixo/genética , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Células NIH 3T3 , Proteína EWS de Ligação a RNA , Proteínas de Ligação a RNA , Transcrição Gênica
13.
Carcinogenesis ; 36(9): 1028-39, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26025912

RESUMO

We reported previously that high-fat diet (HFD) feeding stimulated solid tumor growth and lymph node (LN) metastasis in C57BL/6N mice injected with B16F10 melanoma cells. ß-caryophyllene (BCP) is a natural bicyclic sesquiterpene found in many essential oils and has been shown to exert anti-inflammatory activities. To examine whether BCP inhibits HFD-induced melanoma progression, 4-weeks old, male C57BL/6N mice were fed a control diet (CD, 10 kcal% fat) or HFD (60 kcal% fat + 0, 0.15 or 0.3% BCP) for the entire experimental period. After 16 weeks of feeding, B16F10s were subcutaneously injected into mice. Three weeks later, tumors were resected, and mice were killed 2 weeks post-resection. Although HFD feeding increased body weight gain, fasting blood glucose levels, solid tumor growth, LN metastasis, tumor cell proliferation, angiogenesis and lymphangiogenesis, it decreased apoptotic cells, all of which were suppressed by dietary BCP. HFD feeding increased the number of lipid vacuoles and F4/80+ macrophage (MΦ) and macrophage mannose receptor (MMR)+ M2-MΦs in tumor tissues and adipose tissues surrounding the LN, which was suppressed by BCP. HFD feeding increased the levels of CCL19 and CCL21 in the LN and the expression of CCR7 in the tumor; these changes were blocked by dietary BCP. In vitro culture results revealed that BCP inhibited lipid accumulation in 3T3-L1 preadipocytes; monocyte migration and monocyte chemoattractant protein-1 secretion by B16F10s, adipocytes and M2-MΦs; angiogenesis and lymphangiogenesis. The suppression of adipocyte and M2-cell accumulation and the inhibition of CCL19/21-CCR7 axis may be a part of mechanisms for the BCP suppression of HFD-stimulated melanoma progression.


Assuntos
Antineoplásicos/farmacologia , Dieta Hiperlipídica/efeitos adversos , Melanoma Experimental/tratamento farmacológico , Sesquiterpenos/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Células 3T3 , Adipócitos/metabolismo , Animais , Peso Corporal , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL19/antagonistas & inibidores , Quimiocina CCL19/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL21/antagonistas & inibidores , Quimiocina CCL21/metabolismo , Gorduras na Dieta , Lectinas Tipo C/metabolismo , Linfonodos/patologia , Metástase Linfática , Macrófagos/citologia , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Obesidade/patologia , Sesquiterpenos Policíclicos , Distribuição Aleatória , Receptores CCR7/antagonistas & inibidores , Receptores CCR7/biossíntese , Receptores de Superfície Celular/metabolismo , Neoplasias Cutâneas/patologia , Gordura Subcutânea/citologia , Gordura Subcutânea/patologia , Vacúolos/patologia , Aumento de Peso/efeitos dos fármacos
14.
Artigo em Inglês | MEDLINE | ID: mdl-25058912

RESUMO

Recently, lecithin-derived phosphatidylserine (PS), which originates from marine life, has received much attention as a viable alternative to bovine cerebral cortex PS. In this study, the use of squid phosphatidylcholine-transphosphatidylated PS (SQ-PS) was evaluated through examination of its ameliorating effects on age-associated learning and memory deficits in rats. Aged rats were orally administered SQ-PS (10, 20, or 50 mg/kg per day) once a day for seven days 30 min prior to behavioral assessment in a Morris water maze. SQ-PS administration produced significant dose-dependent improvements in escape latency for finding the platform in the Morris water maze in the aged rats even though Soy-PS administration also exhibited comparable improvements with SQ-PS. Biochemical alterations in the hippocampal cholinergic system, including changes in choline acetyltransferase and acetylcholinesterase immunoreactivity, were consistent with the behavioral results. In addition, SQ-PS treatment significantly restored age-associated decreases of choline transporter and muscarinic acetylcholine receptor type 1 mRNA expression in the hippocampus. These results demonstrate that orally administered SQ-PS dose-dependently aids in the improvement of memory deficits that occur during normal aging in rats. This suggests that SQ-PS may be a useful therapeutic agent in the treatment of diminished memory function in elderly people.


Assuntos
Decapodiformes/química , Transtornos da Memória/dietoterapia , Fosfatidilserinas/administração & dosagem , Acetilcolinesterase/metabolismo , Administração Oral , Fatores Etários , Animais , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Transtornos da Memória/fisiopatologia , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Retenção Psicológica/efeitos dos fármacos , Glycine max/química
15.
Int J Cancer ; 136(2): 258-70, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24844408

RESUMO

To examine the effects of high-fat diet (HFD) on melanoma progression, HFD-fed C57BL/6N mice were subcutaneously injected with syngeneic B16F10 melanoma cells. At 3 weeks post-injection, the tumors were resected; the mice were then sacrificed at 2 weeks post-resection. HFD stimulated melanoma growth and lymph node (LN) metastasis as well as tumor and LN lymphangiogenesis. Lipid vacuoles in the tumor and M2-macrophage (MΦ)s in the adipose and tumor tissues were increased in HFD-fed mice. CCL19 and CCL21 contents were higher in LNs than in tumors. HFD increased both CCL19 and CCL21 levels in LNs and CCR7 in tumors. Adipose tissue-conditioned media (CM) from HFD-fed mice enhanced lymphangiogenesis, and mature adipocyte (MA)/M2-MΦ co-culture CM markedly stimulated the tube formation of lymphatic endothelial cell (LEC)s and B16F10 migration. Monocyte migration was moderately stimulated by B16F10 or MA CM, but tremendously stimulated by B16F10/M2-MΦ co-culture CM, which was enhanced by MA/B16F10/M2-MΦ co-culture CM. The co-culture results revealed that MAs increased CCL2, M-CSF and CCR7 mRNAs in B16F10s; vascular endothelial growth factor (VEGF)-D mRNA in M2-MΦs; and CCL19, CCL21 and VEGF receptor (VEGFR)3 mRNA in LECs. M2-MΦs increased CCL2, M-CSF and VEGF-A mRNAs in B16F10s, whereas B16F10s increased VEGF-C mRNAs in M2-MΦs and VEGFR3 mRNA in LECs. These results indicate that in HFD-fed mice, MA-induced CCL2 and M-CSF in tumor cells increase M2-MΦs in tumor; the crosstalk between tumor cells and M2-MΦs further increases cytokines and angiogenic and lymphangiogenic factors. Additionally, MA-stimulated CCL19, CCL21/CCR7 axis contributes to increased LN metastasis in HFD-fed mice.


Assuntos
Adipócitos/patologia , Dieta Hiperlipídica/efeitos adversos , Linfangiogênese , Macrófagos/patologia , Melanoma Experimental/patologia , Obesidade/etiologia , Adipócitos/metabolismo , Aloenxertos , Animais , Apoptose , Western Blotting , Proliferação de Células , Quimiocinas/metabolismo , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Leptina/fisiologia , Metástase Linfática , Macrófagos/metabolismo , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica , Obesidade/fisiopatologia , Análise Serial de Proteínas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 4 de Melanocortina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Microambiente Tumoral
16.
Mol Carcinog ; 54(1): 72-82, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24729546

RESUMO

We previously reported that a high-fat diet (HFD) and M2-macrophages induce changes in tumor microenvironments and stimulate tumor growth and metastasis of 4T1 mammary cancer cells in BALB/c mice. In this study, we attempted to determine whether benzyl isothiocyanate (BITC) inhibits HFD-induced changes in tumor progression and in tumor microenvironments. Four groups of female BALB/c mice (4-week-old) were fed on a control diet (CD, 10 kcal% fat) and HFD (60 kcal% fat) containing BITC (0, 25, or 100 mg/kg diet) for 20 weeks. Following 16 weeks of feeding, 4T1 cells (5×10(4) cells) were injected into the mammary fat pads, and animals were killed 30 d after the injection. HFD feeding increased solid tumor growth and the number of tumor nodules in the lung and liver, as compared to the CD group, and these increases were inhibited by BITC supplementation. The number of lipid vacuoles, CD45+ leukocytes and CD206+ M2-macrophages, expression of Ki67, levels of cytokines/chemokines, including macrophage-colony stimulating factor (M-CSF) and monocyte chemoattractant protein-1, and mRNA levels of F4/80, CD86, Ym1, CD163, CCR2, and M-CSF receptor were increased in the tumor tissues of HFD-fed mice, and these increases were inhibited by BITC supplementation. In vitro culture results demonstrated that BITC inhibited macrophage migration as well as lipid droplet accumulation in 3T3-L1 cells. These results suggest that suppression of lipid accumulation and macrophage infiltration in tumor tissues may be one of the mechanisms by which BITC suppresses tumor progression in HFD-fed mice.


Assuntos
Gorduras na Dieta/efeitos adversos , Isotiocianatos/administração & dosagem , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Microambiente Tumoral/efeitos dos fármacos , Células 3T3-L1 , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Isotiocianatos/farmacologia , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Obesidade/metabolismo
17.
Appl Biochem Biotechnol ; 175(2): 892-901, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25349086

RESUMO

Blue-green algae are known to contain biologically active proteins and non-protein substances and considered as useful materials for manufacturing the nutritional supplements. Particularly, Spirulina has been reported to contain a variety of antioxidants, such as flavonoids, carotenoids, and vitamin C, thereby exerting their protective effects against the oxidative damage to the cells. In addition to their antioxidant actions, polyphenolic compounds have been speculated to cause the protection of neuronal cells and the recovery of neurologic function in the brain through the production of brain-derived neurotrophic factor (BDNF) in glial cells. Then, the protein-deprived extract was prepared by removing the most part of protein components from aqueous extract of Spirulina platensis, and the effect of this extract on BDNF gene transcription was examined in C6 glioma cells. Consequently, the protein-deprived extract was shown to cause the elevation of BDNF mRNA levels following the expression of heme oxygenase-1 (HO-1) in the glioma cells. Therefore, the non-protein components of S. platensis are considered to stimulate BDNF gene transcription through the HO-1 induction in glial cells, thus proposing a potential ability of the algae to indirectly modulate the brain function through the glial cell activity.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Glioma/patologia , Heme Oxigenase-1/metabolismo , Spirulina/química , Transcrição Gênica/efeitos dos fármacos , Animais , Proteínas de Bactérias/isolamento & purificação , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ratos , Água/química
18.
Artigo em Inglês | MEDLINE | ID: mdl-25477995

RESUMO

Caffeic acid phenethyl ester (CAPE) is an active component of propolis that has a variety of potential pharmacological effects. Although we previously demonstrated that propolis has antidepressant-like activity, the effect of CAPE on this activity remains unknown. The present study assessed whether treatment with CAPE (5, 10, and 20 µmol/kg for 21 days) has an antidepressant-like effect in mice subjected to chronic unpredictable stress via tail suspension (TST) and forced swim (FST) tests. CAPE administration induced behaviors consistent with an antidepressant effect, evidenced by decreased immobility in the TST and FST independent of any effect on serum corticosterone secretion. Western blots, conducted subsequent to behavioral assessment, revealed that CAPE significantly decreased glucocorticoid receptor phosphorylation at S234 (pGR(S234)), resulting in an increased pGR(S220/S234) ratio. We also observed negative correlations between pGR(S220)/(S234) and p38 mitogen-activated protein kinase (p38MAPK) phosphorylation, which was decreased by CAPE treatment. These findings suggest that CAPE treatment exerts an antidepressant-like effect via downregulation of p38MAPK phosphorylation, thereby contributing to enhanced GR function.

19.
Cell Biol Int ; 38(10): 1132-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24800957

RESUMO

Polyamines are widely distributed in living organisms, and considered to play a potential role in various cellular processes. The effects of polyamines on gene expression as well as cell proliferation have been suggested to be closely associated with the physiological and pathological functions. However, it seems necessary to investigate their potential roles in the regulation of cellular metabolism and functions. Previously, glial cells have been suggested to be involved in the protection and preservation of neuronal functions, probably through the production of neurotrophic factors in the brain. On the other hand, neuroactive 5α-reduced steroids promote glial cell differentiation, resulting in enhancement of their ability to produce brain-derived neurotrophic factor (BDNF). Based on these findings, polyamines are assumed to stimulate the expression of the gene encoding steroid 5α-reductase (5α-R), which can induce the production of neuroactive 5α-reduced steroids in glial cells. The effects of polyamines on 5α-R mRNA levels in C6 glioma cells were examined as a model experiment. In consequence, spermine (SPM) and spermidine (SPD), but not putrescine (PUT), have been shown to elevate 5α-R mRNA levels without activating the 5α-R promoter. Furthermore, SPM increased 5α-R mRNA levels under the conditions in which the mRNA biosynthesis was inhibited. Therefore, it can be speculated that polyamines increase 5α-R mRNA levels as a consequence of suppressing the degradation of mRNA.


Assuntos
3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Poliaminas/farmacologia , Estabilidade de RNA/efeitos dos fármacos , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linhagem Celular Tumoral , Glioma/metabolismo , Glioma/patologia , Oligonucleotídeos Antissenso/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Fator de Transcrição Sp1/antagonistas & inibidores , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
J Neurosci Res ; 92(6): 795-807, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24481585

RESUMO

Risperidone, an atypical antipsychotic drug, has been discovered to have some beneficial effects beyond its original effectiveness. The present study examines the neuroprotective effects of risperidone against ischemic damage in the rat and gerbil induced by transient focal and global cerebral ischemia, respectively. The results showed that pre- and posttreatment with 4 mg/kg risperidone significantly protected against neuronal death from ischemic injury. Many NeuN-immunoreactive neurons and a few F-J B-positive cells were found in the rat cerebral cortex and gerbil hippocampal CA1 region (CA1) in the risperidone-treated ischemia groups compared with those in the vehicle-treated ischemia group. In addition, treatment with risperidone markedly attenuated the activation of microglia in the gerbil CA1. On the other hand, we found that treatment with risperidone significantly maintained the antioxidants levels in the ischemic gerbil CA1. Immunoreactivities of superoxide dismutases 1 and 2, catalase, and glutathione peroxidase were maintained in the stratum pyramidale of the CA1; the antioxidants were very different from those in the vehicle-treated ischemia groups. In brief, our present findings indicate that posttreatment as well as pretreatment with risperidone can protect neurons in the rat cerebral cortex and gerbils CA1 from transient cerebral ischemic injury and that the neuroprotective effect of risperidone may be related to attenuation of microglial activation as well as maintenance of antioxidants.


Assuntos
Antioxidantes/metabolismo , Fármacos Neuroprotetores/farmacologia , Risperidona/farmacologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Western Blotting , Isquemia Encefálica/complicações , Modelos Animais de Doenças , Gerbillinae , Imuno-Histoquímica , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...