Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 50(6): 908-18, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23806336

RESUMEN

Fanconi anemia (FA) is a rare genetic disorder characterized by an increased susceptibility to squamous cell cancers. Fifteen FA genes are known, and the encoded proteins cooperate in a common DNA repair pathway. A critical step is the monoubiquitination of the FANCD2 protein, and cells from most FA patients are deficient in this step. How monoubiquitinated FANCD2 suppresses squamous cell cancers is unknown. Here we show that Fancd2-deficient mice are prone to Ras-oncogene-driven skin carcinogenesis, while Usp1-deficient mice, expressing elevated cellular levels of Fancd2-Ub, are resistant to skin tumors. Moreover, Fancd2-Ub activates the transcription of the tumor suppressor TAp63, thereby promoting cellular senescence and blocking skin tumorigenesis. For FA patients, the reduction of FANCD2-Ub and TAp63 protein levels may account for their susceptibility to squamous cell neoplasia. Taken together, Usp1 inhibition may be a useful strategy for upregulating TAp63 and preventing or treating squamous cell cancers in the general non-FA population.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Genes Supresores de Tumor , Fosfoproteínas/genética , Transactivadores/genética , Activación Transcripcional , Animales , Proteínas de Arabidopsis , Proliferación Celular , Células Cultivadas , Senescencia Celular , Daño del ADN , Resistencia a la Enfermedad/genética , Endopeptidasas/deficiencia , Endopeptidasas/genética , Anemia de Fanconi/genética , Femenino , Genes ras , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias de Células Escamosas/inducido químicamente , Neoplasias de Células Escamosas/genética , Neoplasias de Células Escamosas/patología , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Transactivadores/metabolismo , Proteasas Ubiquitina-Específicas , Ubiquitinación
2.
Int J Mol Sci ; 22(6)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33802169

RESUMEN

In order to treat Coronavirus Disease 2019 (COVID-19), we predicted and implemented a drug delivery system (DDS) that can provide stable drug delivery through a computational approach including a clustering algorithm and the Schrödinger software. Six carrier candidates were derived by the proposed method that could find molecules meeting the predefined conditions using the molecular structure and its functional group positional information. Then, just one compound named glycyrrhizin was selected as a candidate for drug delivery through the Schrödinger software. Using glycyrrhizin, nafamostat mesilate (NM), which is known for its efficacy, was converted into micelle nanoparticles (NPs) to improve drug stability and to effectively treat COVID-19. The spherical particle morphology was confirmed by transmission electron microscopy (TEM), and the particle size and stability of 300-400 nm were evaluated by measuring DLSand the zeta potential. The loading of NM was confirmed to be more than 90% efficient using the UV spectrum.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Biología Computacional/métodos , Sistemas de Liberación de Medicamentos/métodos , Células A549 , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Benzamidinas/química , Benzamidinas/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Análisis por Conglomerados , Simulación por Computador , Bases de Datos Farmacéuticas , Portadores de Fármacos/química , Reposicionamiento de Medicamentos , Estabilidad de Medicamentos , Ácido Glicirrínico/química , Ácido Glicirrínico/uso terapéutico , Guanidinas/química , Guanidinas/uso terapéutico , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Micelas , Microscopía Electrónica de Transmisión , Estructura Molecular , Nanopartículas/química , Tamaño de la Partícula
3.
Korean J Physiol Pharmacol ; 25(6): 575-583, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34697268

RESUMEN

Composition of the gut microbiota changes with aging and plays an important role in age-associated disease such as metabolic syndrome, cancer, and neurodegeneration. The gut microbiota composition oscillates through the day, and the disruption of their diurnal rhythm results in gut dysbiosis leading to metabolic and immune dysfunctions. It is well documented that circadian rhythm changes with age in several biological functions such as sleep, body temperature, and hormone secretion. However, it is not defined whether the diurnal pattern of gut microbial composition is affected by aging. To evaluate aging effects on the diurnal pattern of the gut microbiome, we evaluated the taxa profiles of cecal contents obtained from young and aged mice of both sexes at daytime and nighttime points by 16S rRNA gene sequencing. At the phylum level, the ratio of Firmicutes to Bacteroidetes and the relative abundances of Verrucomicrobia and Cyanobacteria were increased in aged male mice at night compared with that of young male mice. Meanwhile, the relative abundances of Sutterellaceae, Alloprevotella, Lachnospiraceae UCG-001, and Parasutterella increased in aged female mice at night compared with that of young female mice. The Lachnospiraceae NK4A136 group relative abundance increased in aged mice of both sexes but at opposite time points. These results showed the changes in diurnal patterns of gut microbial composition with aging, which varied depending on the sex of the host. We suggest that disturbed diurnal patterns of the gut microbiome can be a factor for the underlying mechanism of age-associated gut dysbiosis.

4.
Emerg Infect Dis ; 26(10): 2465-2468, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32673193

RESUMEN

We analyzed reports for 59,073 contacts of 5,706 coronavirus disease (COVID-19) index patients reported in South Korea during January 20-March 27, 2020. Of 10,592 household contacts, 11.8% had COVID-19. Of 48,481 nonhousehold contacts, 1.9% had COVID-19. Use of personal protective measures and social distancing reduces the likelihood of transmission.


Asunto(s)
Trazado de Contacto/estadística & datos numéricos , Infecciones por Coronavirus/epidemiología , Brotes de Enfermedades , Neumonía Viral/epidemiología , Adolescente , Adulto , Distribución por Edad , Factores de Edad , Anciano , Anciano de 80 o más Años , Betacoronavirus , COVID-19 , Niño , Preescolar , Infecciones por Coronavirus/transmisión , Composición Familiar , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Pandemias , Neumonía Viral/transmisión , República de Corea/epidemiología , SARS-CoV-2 , Adulto Joven
5.
Korean J Physiol Pharmacol ; 24(3): 203-212, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32392911

RESUMEN

Promyelocytic leukemia (PML) gene, through alternative splicing of its C-terminal region, generates several PML isoforms that interact with specific partners and perform distinct functions. The PML protein is a tumor suppressor that plays an important role by interacting with various proteins. Herein, we investigated the effect of the PML isoforms on oncostatin M (OSM)-induced signal transducer and activator of transcription-3 (STAT-3) transcriptional activity. PML influenced OSM-induced STAT-3 activity in a cell type-specific manner, which was dependent on the p53 status of the cells but regardless of PML isoform. Interestingly, overexpression of PML exerted opposite effects on OSM-induced STAT-3 activity in p53 wild-type and mutant cells. Specifically, overexpression of PML in the cell lines bearing wild-type p53 (NIH3T3 and U87-MG cells) decreased OSM-induced STAT-3 transcriptional activity, whereas overexpression of PML increased OSM-induced STAT-3 transcriptional activity in mutant p53-bearing cell lines (HEK293T and U251-MG cells). When wild-type p53 cells were co-transfected with PML-IV and R273H-p53 mutant, OSM-mediated STAT-3 transcriptional activity was significantly enhanced, compared to that of cells which were transfected with PML-IV alone; however, when cells bearing mutant p53 were co-transfected with PML-IV and wild-type p53, OSM-induced STAT-3 transcriptional activity was significantly decreased, compared to that of transfected cells with PML-IV alone. In conclusion, PML acts together with wild-type or mutant p53 and influences OSM-mediated STAT-3 activity in a negative or positive manner, resulting in the aberrant activation of STAT-3 in cancer cells bearing mutant p53 probably might occur through the interaction of mutant p53 with PML.

6.
J Neuroinflammation ; 16(1): 246, 2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31791357

RESUMEN

BACKGROUND: Neuroinflammation plays a pivotal role in the pathogenesis of Parkinson's disease (PD). Thus, the development of agents that can control neuroinflammation has been suggested as a promising therapeutic strategy for PD. In the present study, we investigated whether the phosphodiesterase (PDE) 10 inhibitor has anti-inflammatory and neuroprotective effects in neuroinflammation and PD mouse models. METHODS: Papaverine (PAP) was utilized as a selective inhibitor of PDE10. The effects of PAP on the expression of pro-inflammatory molecules were examined in lipopolysaccharide (LPS)-stimulated BV2 microglial cells by ELISA, RT-PCR, and Western blot analysis. The effects of PAP on transcription factors were analyzed by the electrophoretic mobility shift assay, the reporter gene assay, and Western blot analysis. Microglial activation and the expression of proinflammatory molecules were measured in the LPS- or MPTP-injected mouse brains by immunohistochemistry and RT-PCR analysis. The effect of PAP on dopaminergic neuronal cell death and neurotrophic factors were determined by immunohistochemistry and Western blot analysis. To assess mouse locomotor activity, rotarod and pole tests were performed in MPTP-injected mice. RESULTS: PAP inhibited the production of nitric oxide and proinflammatory cytokines in LPS-stimulated microglia by modulating various inflammatory signals. In addition, PAP elevated intracellular cAMP levels and CREB phosphorylation. Treatment with H89, a PKA inhibitor, reversed the anti-inflammatory effects of PAP, suggesting the critical role of PKA signaling in the anti-inflammatory effects of PAP. We verified the anti-inflammatory effects of PAP in the brains of mice with LPS-induced systemic inflammation. PAP suppressed microglial activation and proinflammatory gene expression in the brains of these mice, and these effects were reversed by H89 treatment. We further examined the effects of PAP on MPTP-injected PD model mice. MPTP-induced dopaminergic neuronal cell death and impaired locomotor activity were recovered by PAP. In addition, PAP suppressed microglial activation and proinflammatory mediators in the brains of MPTP-injected mice. CONCLUSIONS: PAP has strong anti-inflammatory and neuroprotective effects and thus may be a potential candidate for treating neuroinflammatory disorders such as PD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Papaverina/uso terapéutico , Trastornos Parkinsonianos/prevención & control , Inhibidores de Fosfodiesterasa/uso terapéutico , Animales , Antiinflamatorios/farmacología , Línea Celular Transformada , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Fármacos Neuroprotectores/farmacología , Papaverina/farmacología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/enzimología , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
7.
Cell Physiol Biochem ; 45(5): 1863-1877, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29510393

RESUMEN

BACKGROUND/AIMS: The signal transducer and activator of transcription 6 (STAT6) transcription factor mediates PPARγ-regulated gene expression in macrophages. However, it remains largely unknown how proximal membrane signaling events initiated by apoptotic cell recognition upregulate PPARγ expression and activate the lung homeostatic program. METHODS: The STAT6 inhibitor AS1517499 was used to determine the role of STAT6 in mediating PPARγ activity, anti-inflammatory effects, and anti-fibrotic effects induced by apoptotic cell instillation after bleomycin treatment into C57BL/6 mice. Bronchoalveolar lavage fluid, alveolar macrophages and lungs were harvested at days 2, 7, and 14 and then analyzed by real-time PCR, immunoblotting, ELISA, immunocytochemistry and immunohistochemistry assays. RESULTS: Our data demonstrate that apoptotic cell instillation after bleomycin results in prolonged enhancement of STAT6 phosphorylation in alveolar macrophages and lung. Co-administration of the STAT6 inhibitor, AS1517499, reversed the enhanced PPARγ expression and activity induced by apoptotic cell instillation after bleomycin treatment. By reducing the expression of PPARγ target genes, including CD36, macrophage mannose receptor, and arginase 1, AS1517499 inhibited efferocytosis and restored pro-inflammatory cytokine expression, neutrophil recruitment, protein levels, hydroxyproline content, and expression of fibrosis markers, including type 1 collagen α2, fibronectin, and α-smooth muscle actin. STAT6 inhibition reversed the expression profile of hepatocyte growth factor and interleukin-10. CONCLUSION: These results indicate that prolonged STAT6 activation following one-time apoptotic cell instillation facilitates continuous PPARγ activation, resulting in the resolution of bleomycin-induced lung inflammation and fibrosis.


Asunto(s)
Apoptosis/efectos de los fármacos , PPAR gamma/metabolismo , Fibrosis Pulmonar/patología , Pirimidinas/farmacología , Factor de Transcripción STAT6/antagonistas & inhibidores , Animales , Arginasa/metabolismo , Bleomicina/toxicidad , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Antígenos CD36/metabolismo , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Interleucina-10/metabolismo , Células Jurkat , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Factor de Transcripción STAT6/metabolismo
8.
Neurochem Res ; 42(12): 3504-3514, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29019035

RESUMEN

The lectin, concanavalin A (Con A), is the most extensively investigated member of the lectin family of plant proteins, but its effects on cortical neurons and astrocytes are poorly understood. In cultured cortical neurons and astrocytes, Con A exhibited dose-dependent neurotoxicity, but this was not observed in astrocytes. Similarly, in the cortical areas of rat brains, intracranial administration of Con A caused neuronal but no astrocyte damage. Methyl-α-D-mannopyranoside, a competitor of Con A, blocked Con A-induced cell death, whereas AMPA/KA receptor antagonists showed partial blocking effects. Furthermore, the mRNA levels of TNF-α, IL-1ß, and IL-6 were elevated in astrocytes and cortical neurons treated with Con A. Intracellular reactive oxygen species (ROS) levels were increased in Con A-treated cortical neurons, and N-acetyl-cysteine (NAC, an antioxidant) and diphenyleneiodonium (DPI, a NADPH oxidase inhibitor) reduced intracellular ROS accumulation. Likewise, AG556 (a TNF-α inhibitor) and AG82 (a tyrosine kinase inhibitor) both reduced Con A-induced intracellular ROS accumulation. Furthermore, Con A-induced tyrosine phosphorylation was decreased by NAC and by AG556. Taken together, Con A-induced apoptosis in cortical neurons occurred as a sequel to Con A binding to neuronal glycoproteins and intracellular ROS accumulation. Interestingly, Con A-induced cellular damage was observed in cortical neurons but not in astrocytes or microglia.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Concanavalina A/farmacología , Neuronas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Astrocitos/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Masculino , Microglía/metabolismo , NADPH Oxidasas/metabolismo , Neuronas/metabolismo , Compuestos Onio/farmacología , Fosforilación , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
9.
J Immunol ; 193(5): 2384-93, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25049354

RESUMEN

Matrix metalloproteinases (MMPs) play important roles in normal brain development and synaptic plasticity, although aberrant expression of MMPs leads to brain damage, including blood-brain barrier disruption, inflammation, demyelination, and neuronal cell death. In this article, we report that MMP-8 is upregulated in LPS-stimulated BV2 microglial cells and primary cultured microglia, and treatment of MMP-8 inhibitor (M8I) or MMP-8 short hairpin RNA suppresses proinflammatory molecules, particularly TNF-α secretion. Subsequent experiments showed that MMP-8 exhibits TNF-α-converting enzyme (TACE) activity by cleaving the prodomain of TNF-α (A(74)/Q(75), A(76)/V(77) residues) and, furthermore, that M8I inhibits TACE activity more efficiently than TAPI-0, a general TACE inhibitor. Biochemical analysis of the underlying anti-inflammatory mechanisms of M8I revealed that it inhibits MAPK phosphorylation, NF-κB/AP-1 activity, and reactive oxygen species production. Further support for the proinflammatory role of microglial MMP-8 was obtained from an in vivo animal model of neuroinflammatory disorder. MMP-8 is upregulated in septic conditions, particularly in microglia. Administration of M8I or MMP-8 short hairpin RNA significantly inhibits microglial activation and expression/secretion of TNF-α in brain tissue, serum, and cerebrospinal fluid of LPS-induced septic mice. These results demonstrate that MMP-8 critically mediates microglial activation by modulating TNF-α activity, which may explain neuroinflammation in septic mouse brain.


Asunto(s)
Encefalopatías/inmunología , Encéfalo/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Metaloproteinasa 8 de la Matriz/inmunología , Proteínas del Tejido Nervioso/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Proteínas ADAM/inmunología , Proteína ADAM17 , Animales , Encéfalo/patología , Encefalopatías/inducido químicamente , Encefalopatías/patología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Lipopolisacáridos/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Endogámicos ICR , Microglía/inmunología , Microglía/patología , FN-kappa B/inmunología , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Sepsis/inducido químicamente , Sepsis/inmunología , Sepsis/patología , Factor de Transcripción AP-1/inmunología
10.
Biochem Biophys Res Commun ; 458(3): 520-524, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25677620

RESUMEN

Homologous-recombination (HR)-dependent repair defective cells are hypersensitive to poly (ADP-ribose) polymerase (PARP) inhibitors. Combinations of defective HR pathway and PARP inhibitors have been an effective chemotherapeutic modality. We previously showed that knockdown of the WD40-repeat containing protein, Uaf1, causes an HR repair defect in mouse embryo fibroblast cells and therefore, increases sensitivity to PARP inhibitor, ABT-888. Similarly, here, we show that ferulic acid reduces HR repair, inhibits RAD 51 foci formation, and accumulates γ-H2AX in breast cancer cells. Moreover, ferulic acid, when combined with ABT-888, renders breast cancer cells become hypersensitive to ABT-888. Our study indicates that ferulic acid in combination with ABT-888 treatment may serve as an effective combination chemotherapeutic agent as a natural bioactive compound.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Bencimidazoles/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ácidos Cumáricos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Antioxidantes/administración & dosificación , Bencimidazoles/administración & dosificación , Mama/efectos de los fármacos , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ácidos Cumáricos/administración & dosificación , Daño del ADN/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Femenino , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación/efectos de los fármacos
11.
J Neuroinflammation ; 12: 133, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26173397

RESUMEN

BACKGROUND: ß-Lapachone (ß-LAP) is a natural naphthoquinone compound isolated from the lapacho tree (Tabebuia sp.), and it has been used for treatment of rheumatoid arthritis, infection, and cancer. In the present study, we investigated whether ß-LAP has anti-inflammatory effects under in vitro and in vivo neuroinflammatory conditions. METHODS: The effects of ß-LAP on the expression of inducible nitric oxide synthase (iNOS), cytokines, and matrix metalloproteinases (MMPs) were examined in lipopolysaccharide (LPS)-stimulated BV2 microglial cells and rat primary microglia by ELISA, reverse transcription polymerase chain reaction (RT-PCR), and Western blot analysis. Microglial activation and the expression levels of proinflammatory molecules were measured in the LPS-injected mouse brain by immunohistochemistry and RT-PCR analysis. The detailed molecular mechanism underlying the anti-inflammatory effects of ß-LAP was analyzed by electrophoretic mobility shift assay, reporter gene assay, Western blot, and RT-PCR analysis. RESULTS: ß-LAP inhibited the expression of iNOS, proinflammatory cytokines, and MMPs (MMP-3, MMP-8, MMP-9) at mRNA and protein levels in LPS-stimulated microglia. On the other hand, ß-LAP upregulated the expressions of anti-inflammatory molecules such as IL-10, heme oxygenase-1 (HO-1), and the tissue inhibitor of metalloproteinase-2 (TIMP-2). The anti-inflammatory effect of ß-LAP was confirmed in an LPS-induced systemic inflammation mouse model. Thus, ß-LAP inhibited microglial activation and the expressions of iNOS, proinflammatory cytokines, and MMPs in the LPS-injected mouse brain. Further mechanistic studies revealed that ß-LAP exerts anti-inflammatory effects by inhibiting MAPKs, PI3K/AKT, and NF-κB/AP-1 signaling pathways in LPS-stimulated microglia. ß-LAP also inhibited reactive oxygen species (ROS) production by suppressing the expression and/or phosphorylation of NADPH oxidase subunit proteins, such as p47(phox) and gp91(phox). The anti-oxidant effects of ß-LAP appeared to be related with the increase of HO-1 and NQO1 via the Nrf2/anti-oxidant response element (ARE) pathway and/or the PKA pathway. CONCLUSIONS: The strong anti-inflammatory/anti-oxidant effects of ß-LAP may provide preventive therapeutic potential for various neuroinflammatory disorders.


Asunto(s)
Antiinflamatorios/farmacología , Citocinas/metabolismo , Encefalitis/metabolismo , Encefalitis/prevención & control , Metaloproteinasas de la Matriz/metabolismo , Microglía/efectos de los fármacos , Naftoquinonas/farmacología , Animales , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Hemo-Oxigenasa 1/metabolismo , Técnicas In Vitro , Interleucina-10/metabolismo , Lipopolisacáridos/efectos adversos , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
12.
Brain Behav Immun ; 50: 221-231, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26184082

RESUMEN

Visceral adipose tissue is accumulated with aging. An increase in visceral fat accompanied by low-grade inflammation is associated with several adult-onset diseases. However, the effects of visceral adipose tissue inflammation on the normal and ischemic brains of aged are not clearly defined. To examine the role of visceral adipose tissue inflammation, we evaluated inflammatory cytokines in the serum, visceral adipose tissue, and brain as well as blood-brain barrier (BBB) permeability in aged male mice (20 months) underwent sham or visceral fat removal surgery compared with the young mice (2.5 months). Additionally, ischemic brain injury was compared in young and aged mice with sham and visceral fat removal surgery. Interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α levels in examined organs were increased in aged mice compared with the young mice, and these levels were reduced in the mice with visceral fat removal. Increased BBB permeability with reduced expression of tight junction proteins in aged sham mice were also decreased in mice with visceral fat removal. After focal ischemic injury, aged mice with visceral fat removal showed a reduction in infarct volumes, BBB permeability, and levels of proinflammatory cytokines in the ischemic brain compared with sham mice, although the neurological outcomes were not significantly improved. In addition, further upregulated visceral adipose tissue inflammation in response to ischemic brain injury was attenuated in mice with visceral fat removal. These results suggest that visceral adipose tissue inflammation is associated with age-related changes in the brain and contributes to the ischemic brain damage in the aged mice. We suggest that visceral adiposity should be considered as a factor affecting brain health and ischemic brain damage in the aged population.


Asunto(s)
Envejecimiento , Isquemia Encefálica/metabolismo , Inflamación/metabolismo , Grasa Intraabdominal/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/complicaciones , Citocinas/metabolismo , Inflamación/complicaciones , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
13.
Biochim Biophys Acta ; 1831(6): 1016-26, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23384839

RESUMEN

Ceramide is a major molecule among the sphingolipid metabolites which are produced in the brain and other organs and act as intracellular second messengers. Although a variety of physiological roles of ceramide have been reported in the periphery and central nervous systems, the role of ceramide in microglial activation has not been clearly demonstrated. In the present study, we examined the effects of exogenous cell permeable short chain ceramides on microglial activation in vitro and in vivo. We found that C2, C6, and C8 ceramide and C8 ceramide-1-phosphate inhibited iNOS and proinflammatory cytokines in lipopolysaccharide (LPS)-stimulated BV2 microglial cells and rat primary microglia. In addition, the administration of C2 ceramide suppressed microglial activation in the brains of LPS-exposed mice. By HPLC and LC/MS/MS analyses, we found that C2 ceramide on its own, rather than its modified form (i.e. ceramide-1-phosphate or long chain ceramides), mainly work by penetrating into microglial cells. Further mechanistic studies by using the most effective C2 ceramide among the short chain ceramides tested, revealed that C2 ceramide exerts anti-inflammatory effects via inhibition of the ROS, MAPKs, PI3K/Akt, and Jak/STAT pathways with upregulation of PKA and hemeoxygenase-1 expressions. Interestingly, we found that C2 ceramide inhibits TLR4 signaling by interfering with LPS and TLR4 interactions. Therefore, our data collectively suggests the therapeutic potential of short chain ceramides such as C2 for neuroinflammatory disorders such as Alzheimer's disease and Parkinson's disease.


Asunto(s)
Antiinflamatorios/farmacología , Encéfalo/efectos de los fármacos , Ceramidas/farmacología , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Sepsis/prevención & control , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Encéfalo/citología , Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Luciferasas/metabolismo , Ratones , Microglía/citología , Microglía/metabolismo , Nitritos/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sepsis/inmunología , Sepsis/metabolismo
14.
J Pharmacol Exp Ther ; 350(3): 563-77, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24939420

RESUMEN

Growth arrest-specific protein 6 (Gas6)/Mer receptor tyrosine kinase (Mer) signaling modulates cytokine secretion and helps to regulate the immune response and apoptotic cell clearance. Signaling pathways that activate an epithelial growth program in macrophages are still poorly defined. We report that Gas6/Mer/RhoA signaling can induce the production of epithelial growth factor hepatic growth factor (HGF) in macrophages, which ultimately promotes epithelial cell proliferation and wound repair. The RhoA/protein kinase B (Akt)/mitogen-activated protein (MAP) kinases, including p38 MAP kinase, extracellular signal-regulated protein kinase, and Jun NH2-terminal kinase axis in RAW 264.7 cells, was identified as Gas6/Mer downstream signaling pathway for the upregulation of HGF mRNA and protein. Conditioned medium from RAW 264.7 cells that had been exposed to Gas6 or apoptotic cells enhanced epithelial cell proliferation of the epithelial cell line LA-4 and wound closure. Cotreatment with an HGF receptor-blocking antibody or c-Met antagonist downregulated this enhancement. Inhibition of Mer with small interfering RNA (siRNA) or the RhoA/Rho kinase pathway by RhoA siRNA or Rho kinase pharmacologic inhibitor suppressed Gas6-induced HGF mRNA and protein expression in macrophages and blocked epithelial cell proliferation and wound closure induced by the conditioned medium. Our data provide evidence that macrophages can be reprogrammed by Gas6 to promote epithelial proliferation and wound repair via HGF, which is induced by the Mer/RhoA/Akt/MAP kinase pathway. Thus, defects in Gas6/Mer/RhoA signaling in macrophages may delay tissue repair after injury to the alveolar epithelium.


Asunto(s)
Factor de Crecimiento de Hepatocito/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Fosfatidilinositol 3-Quinasa/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteína de Unión al GTP rhoA/biosíntesis , Amidas/farmacología , Animales , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Células Jurkat , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Piridinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Tirosina Quinasa c-Mer , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
15.
Chem Res Toxicol ; 27(5): 919-30, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24725253

RESUMEN

DNA polymerase (pol) κ, one of the Y-family polymerases, has been shown to function in error-free translesion DNA synthesis (TLS) opposite the bulky N(2)-guanyl DNA lesions induced by many carcinogens such as polycyclic aromatic hydrocarbons. We analyzed the biochemical properties of eight reported human pol κ variants positioned in the polymerase core domain, using the recombinant pol κ (residues 1-526) protein and the DNA template containing an N(2)-CH2(9-anthracenyl)G (N(2)-AnthG). The truncation R219X was devoid of polymerase activity, and the E419G and Y432S variants showed much lower polymerase activity than wild-type pol κ. In steady-state kinetic analyses, E419G and Y432S displayed 20- to 34-fold decreases in kcat/Km for dCTP insertion opposite G and N(2)-AnthG compared to that of wild-type pol κ. The L21F, I39T, and D189G variants, as well as E419G and Y432S, displayed 6- to 22-fold decreases in kcat/Km for next-base extension from C paired with N(2)-AnthG, compared to that of wild-type pol κ. The defective Y432S variant had 4- to 5-fold lower DNA-binding affinity than wild-type, while a slightly more efficient S423R variant possessed 2- to 3-fold higher DNA-binding affinity. These results suggest that R219X abolishes and the E419G, Y432S, L21F, I39T, and D189G variations substantially impair the TLS ability of pol κ opposite bulky N(2)-G lesions in the insertion step opposite the lesion and/or the subsequent extension step, raising the possibility that certain nonsynonymous pol κ genetic variations translate into individual differences in susceptibility to genotoxic carcinogens.


Asunto(s)
Aductos de ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Aductos de ADN/química , ADN Polimerasa Dirigida por ADN/química , Variación Genética , Humanos , Modelos Moleculares , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
Brain Behav Immun ; 40: 143-54, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24704569

RESUMEN

We previously reported that bone morphogenetic proteins (BMPs) and their endogenous antagonist noggin are expressed in the brain weeks after an ischemic insult. Here, to define their roles in ischemic brain tissue repair and remodeling, we infused recombinant BMP7 or noggin into the ipsilateral ventricle of mice for 2weeks starting 2weeks after transient middle cerebral artery occlusion (MCAO). Four weeks after MCAO, we measured ischemic brain volume, functional recovery, and molecules related to neurogenesis and angiogenesis such as synaptophysin, GAP-43, and VEGF. Noggin-treated mice but not BMP7-treated mice showed preserved ipsilateral brain volume and reduced neurological deficits compared with artificial cerebrospinal fluids (aCSF)-treated mice. Noggin treatment also decreased glial scar thickness, increased levels of GAP-43 and VEGF protein, and increased the number of Iba1-positive activated microglia in the ipsilateral brain. Furthermore, noggin treatment decreased M1 markers (IL-1ß, TNF-α, IL-12, CCL2 and CD86) and increased M2 markers (IL-1ra, IL-10, arginase 1, CD206 and Ym1) of activated microglia, suggesting a shift from M1 to M2 phenotypes. These results suggest that noggin improves functional recovery from ischemic stroke and enhances alternatively activated microglia, thereby promoting tissue repair and remodeling.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Proteínas Portadoras/uso terapéutico , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Animales , Proteína Morfogenética Ósea 7/uso terapéutico , Encéfalo/metabolismo , Isquemia Encefálica/etiología , Proteínas Portadoras/administración & dosificación , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Proteínas Recombinantes/uso terapéutico
17.
J Immunol ; 189(2): 1062-71, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22696440

RESUMEN

Retinoic acid is the active vitamin A derivative and is well-known to have diverse immunomodulatory actions. In this study, we investigated the impact of all-trans retinoic acid (ATRA), a biologic key metabolite of vitamin A, on the development of arthritis and the pathophysiologic mechanisms by which ATRA might have antiarthritic effects in animal model of rheumatoid arthritis (RA; collagen-induced arthritis [CIA] in DBA/1J mice). We showed that treatment with ATRA markedly suppressed the clinical and histologic signs of arthritis in the CIA mice. It reduced the expression of IL-17 in the arthritic joints. Interestingly, Foxp3(+) regulatory T cells were markedly increased and IL-17-producing CD4(+) T cells (Th17 cells) were decreased in the spleens of ATRA-treated mice. In vitro treatment with ATRA induced the expression of Foxp3 and repressed the IL-17 expression in the CD4(+) T cells in mice. ATRA suppressed the production of total IgG and IgG2a in splenocytes that were stimulated by LPS. It also reduced serum levels of total IgG and IgG2 anti-collagen Abs and germinal center formation in CIA mice. In addition, the ATRA-treated mice showed decreased osteoclast formation in arthritic joints. Moreover, ATRA downregulated the expression of receptor activator of NF-κB ligand, the leading player of osteoclastogenesis, in the CD4(+) T cells and fibroblast-like synoviocytes from patients with RA. Furthermore, ATRA prevented both human monocytes and mice bone marrow-derived monocytes/macrophage cells from differentiating into osteoclasts. These data suggest ATRA might be an effective treatment modality for RA patients.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Mediadores de Inflamación/uso terapéutico , Tretinoina/uso terapéutico , Animales , Artritis Reumatoide/patología , Bovinos , Colágeno Tipo II/toxicidad , Modelos Animales de Enfermedad , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/administración & dosificación , Masculino , Ratones , Ratones Endogámicos DBA , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Osteoclastos/patología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/patología , Tretinoina/administración & dosificación
18.
Indian J Exp Biol ; 52(7): 683-91, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25059036

RESUMEN

Ursolic acid (UA) is a pentacyclic triterpenoid compound that naturally occurs in fruits, leaves and flowers of medicinal herbs. This study investigated the dose-response efficacy of UA (0.01 and 0.05%) on glucose metabolism, the polyol pathway and dyslipidemia in streptozotocin/nicotinamide-induced diabetic mice. Supplement with both UA doses reduced fasting blood glucose and plasma triglyceride levels in non-obese type 2 diabetic mice. High-dose UA significantly lowered plasma free fatty acid, total cholesterol and VLDL-cholesterol levels compared with the diabetic control mice, while LDL-cholesterol levels were reduced with both doses. UA supplement effectively decreased hepatic glucose-6-phosphatase activity and increased glucokinase activity, the glucokinase/glucose-6-phosphatase ratio, GLUT2 mRNA levels and glycogen content compared with the diabetic control mice. UA supplement attenuated hyperglycemia-induced renal hypertrophy and histological changes. Renal aldose reductase activity was higher, whereas sorbitol dehydrogenase activity was lower in the diabetic control group than in the non-diabetic group. However, UA supplement reversed the biochemical changes in polyol pathway to normal values. These results demonstrated that low-dose UA had preventive potency for diabetic renal complications, which could be mediated by changes in hepatic glucose metabolism and the renal polyol pathway. High-dose UA was more effective anti-dyslipidemia therapy in non-obese type 2 diabetic mice.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dislipidemias/tratamiento farmacológico , Glucosa/metabolismo , Polímeros/metabolismo , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Complicaciones de la Diabetes/etiología , Complicaciones de la Diabetes/patología , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/etiología , Dislipidemias/patología , Glucoquinasa/metabolismo , Transportador de Glucosa de Tipo 2/genética , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/metabolismo , Hiperglucemia/complicaciones , Enfermedades Renales/etiología , Enfermedades Renales/patología , Enfermedades Renales/prevención & control , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Endogámicos NOD , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácido Ursólico
19.
J Cell Biochem ; 114(2): 303-13, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22930410

RESUMEN

In our previous study, we screened and isolated genes that were up-regulated after partial pancreatectomy using transcriptomic analysis and glutamate-ammonia ligase (GLUL) was selected for further study based on its effect on differentiation and proliferation. In the immunohistochemical analysis, GLUL was highly up-regulated in the acinar cells and the ductal cells in the pancreas damaged through partial pancreatectomy. Overexpression of GLUL enhanced the proliferation of PANC-1 cells and INS-1 cells. GLUL overexpression shifted the major population of PANC-1 cells from the G0/G1 phase to G2/M phase. In the double thymidine blocking analysis, similar cycle duration was observed between mock cells and GLUL-overexpressing cells while GLUL-overexpressing cells were partially resistant to thymidine blocking. In the FACS analysis of cells stained with Pyronin Y and Hoechst 33342, GLUL-overexpressing cells showed lower population of cells in the G0-quiescent phase than mock cells (5-12%). In addition, GLUL-overexpressing cells had high activation levels of AKT, ERK1/2, JNK, PCNA, c-FOS, and P70S6K in PANC-1 cells. Taken together, these results suggest that GLUL contributes to pancreatic regeneration.


Asunto(s)
Ciclo Celular/genética , Glutamato-Amoníaco Ligasa , Páncreas , Regeneración , Células Acinares/citología , Células Acinares/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/metabolismo , Células HEK293 , Humanos , Páncreas/citología , Páncreas/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-akt/genética , Timidina/farmacología
20.
Pharmacogenet Genomics ; 23(7): 365-73, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23652408

RESUMEN

OBJECTIVE: Human multidrug and toxin extrusion member 2 (MATE2-K, SLC47A2) plays an important role in the renal elimination of various clinical drugs including the antidiabetic drug metformin. The goal of this study was to characterize genetic variants of MATE2-K and determine their association with the pharmacokinetics of metformin. METHODS: We screened DNA samples from 48 healthy Koreans for variants in the promoter and coding regions of MATE2-K and examined the function of common haplotypes in the promoter region using in-vitro luciferase assays. Then, the metformin pharmacokinetic study was carried out to determine the association between MATE2-K promoter haplotypes and metformin pharmacokinetics. RESULTS: Nine variants in the promoter region of MATE2-K and one nonsynonymous variant, p.G211V, were identified. The MATE2-K promoter haplotype 1 containing a known functional polymorphism, g.-130G>A and haplotype 2 containing two polymorphisms, g.-609G>A and g.-396G>A showed a significant increase in reporter activity. Among the 45 individuals who participated in the metformin pharmacokinetic study, 12 healthy Koreans who were homozygous for haplotype 1 or 2 showed a significant increase in renal clearance [539 ± 76 (reference group) vs. 633 ± 102 (variant group) ml/min; P=0.006] and secretion clearance [439 ± 81 (reference group) vs. 531 ± 102 (variant group) ml/min; P=0.007] of metformin compared with that shown by the reference group. CONCLUSION: Our study suggests that common promoter haplotypes of MATE2-K are associated with the pharmacokinetics of metformin.


Asunto(s)
Variación Genética , Hipoglucemiantes/farmacocinética , Metformina/farmacocinética , Proteínas de Transporte de Catión Orgánico/genética , Pueblo Asiatico , Haplotipos , Homocigoto , Humanos , Polimorfismo Genético , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA