Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Biochem ; 2024 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-38341833

RESUMEN

BACKGROUND: WD repeat domain 12 (WDR12) plays a crucial role in the ribosome biogenesis pathway. However, its biological function in colorectal cancer (CRC) remains poorly understood. Therefore, this study aims to investigate the roles of WDR12 in the occurrence and progression of CRC, as well as its underlying mechanisms. METHODS: The expression of WDR12 was assessed through The Cancer Genome Atlas (TCGA) and the Human Protein Atlas (HPA) database. Functional experiments including Celigo assay, MTT assay, and Caspase-3/7 assay were conducted to validate the role of WDR12 in the malignant progression of CRC. Additionally, mRNA chip-sequencing and ingenuity pathway analysis (IPA) were performed to identify the molecular mechanism. RESULTS: WDR12 expression was significantly upregulated in CRC tissues compared to normal colorectal tissues. Knockdown of WDR12 reduced proliferation and promoted apoptosis of CRC cell lines in vitro and in vivo experiments. Furthermore, WDR12 expression had a significantly inverse association with diseases and functions, including cancer, cell cycle, DNA replication, recombination, cellular growth, proliferation and repair, as revealed by IPA analysis of mRNA chip-sequencing data. Moreover, the activation of cell cycle checkpoint kinases proteins in the cell cycle checkpoint control signaling pathway was enriched in the WDR12 knockdown CRC cell lines. Additionally, downregulation of rac family small GTPase 1 (RAC1) occurred upon WDR12 knockdown, thereby facilitating the proliferation and anti-apoptosis of CRC cells. CONCLUSION: Our study demonstrates that the WDR12/RAC1 axis promotes tumor progression in CRC. Therefore, WDR12 may serve as a novel oncogene and a potential target for individualized therapy in CRC. These findings provide an experimental foundation for the clinical development of drugs targeting the WDR12/RAC1 axis.

2.
Environ Res ; 218: 115063, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36528045

RESUMEN

Bacteria have evolved several mechanisms to resist Cd toxicity, which are crucial for Cd detoxication and have the potential to be used for bioremediation of Cd. Geobacter species are widely found in anaerobic environments and play important roles in natural biogeochemical cycles. However, the transcriptomic response of Geobacter sulfurreducens under Cd stress have not been fully elucidated. Through integrated analysis of transcriptomic and protein-protein interaction (PPI) data, we uncovered a global view of mRNA changes in Cd-induced cellular processes in this study. We identified 182 differentially expressed genes (|log2(fold change)| > 1, adjusted P < 0.05) in G. sulfurreducens exposed to 0.1 mM CdCl2 using RNA sequencing (RNA-seq). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that CdCl2 significantly affected sulfur compound metabolic processes. In addition, through PPI network analysis, hub genes related to molecular chaperones were identified to play important role in Cd stress response. We also identified a Cd-responsive transcriptional regulator ArsR2 (coded by GSU2149) and verified the function of ArsR2-ParsR2 regulatory circuit in Escherichia coli. This study provides new insight into Cd stress response in G. sulfurreducens, and identified a potential sensor element for Cd detection.


Asunto(s)
Geobacter , Transcriptoma , Cadmio/toxicidad , Geobacter/genética , Perfilación de la Expresión Génica
3.
Acta Derm Venereol ; 103: adv9400, 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37787418

RESUMEN

Type 2 diabetes is associated with an increased risk of herpes zoster and postherpetic neuralgia. However, the association of type 1 diabetes with herpes zoster or postherpetic neuralgia remains unclear. This retrospective cohort study using Taiwan's Health Insurance Research Database included 199,566 patients with type 1 diabetes and 1,458,331 with type 2 diabetes, identified during the period 2000 to 2012. Patients with type 1 diabetes had a significantly higher risk of developing herpes zoster than those with type 2 diabetes (p < 0.001). Across all age groups, the impact of diabetes on herpes zoster was greater in type 1 than in type 2 diabetes. Patients with both type 1 and type 2 diabetes had a 1.45-fold higher risk of post-herpetic neuralgia than those without diabetes (hazard ratio 1.45, 95% confidence interval 1.28-1.65; hazard ratio 1.45, 95% confidence interval 1.37-1.52, respectively), and there was no difference between the 2 types of diabetes (hazard ratio 1.06; 95% confidence interval 0.93-1.21). The results recommend consideration of herpes zoster vaccination at an earlier age in patients with type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Herpes Zóster , Neuralgia Posherpética , Humanos , Neuralgia Posherpética/diagnóstico , Neuralgia Posherpética/epidemiología , Neuralgia Posherpética/complicaciones , Estudios de Cohortes , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/epidemiología , Estudios Retrospectivos , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/epidemiología , Herpes Zóster/epidemiología , Herpesvirus Humano 3
4.
J Cell Physiol ; 236(5): 3896-3905, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33283880

RESUMEN

Lung cancer is a leading cause of cancer-related death worldwide. In this study, we used lung adenocarcinoma cells as a model, as lung adenocarcinoma has the highest mortality rate among all lung cancers. For the past few years, medical treatments or lung cancer have been limited because of chemotherapy resistance. Therefore, understanding the pathogenesis of the development of drug resistance in lung cancer is urgent. Gemcitabine is widely prescribed in the chemotherapeutic treatment of lung cancers. In this study, we developed gemcitabine-resistant lung adenocarcinoma cells (A549-GR) from the A549 cell line. The results showed that apoptotic protein expression and reactive oxygen species (ROS) generation were reduced in A549-GR cells compared to A549 cells. Interestingly, we found that signal transducer and activator of transcription 3 (STAT3) translocated to the nucleus and mitochondria to affect the apoptotic pathway and ROS generation, respectively. Furthermore, treatment with STAT3 small interfering RNA diminished the increase in ROS production, proliferation and antiapoptotic proteins in A549-GR cells. Taken together, the study demonstrated that STAT3 acts as an essential regulator and moderates apoptosis through two major mechanisms to induce gemcitabine resistance in cells; and these findings provide a potential target for the treatment of gemcitabine-resistant lung cancer.


Asunto(s)
Apoptosis , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Neoplasias Pulmonares/metabolismo , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Apoptosis/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citosol/metabolismo , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/efectos de los fármacos , Modelos Biológicos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Gemcitabina
5.
Environ Toxicol ; 36(4): 654-664, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33314651

RESUMEN

Skin pigmentation is resulted from several processes, such as melanin synthesis transportation and abnormal melanin accumulation in keratinocytes. Various studies have suggested that seven traditional Chinese herbal extracts from Atractylodes macrocephala, Paeonia lactiflora, Bletilla striata, Poria cocos, Dictamnus dasycarpus, Ampelopsis japonica and Tribulus terrestris (which we collectively named ChiBai), show several protective effects toward skin-related diseases. Lactobacillus rhamnosus, a lactic acid bacterium, has been reported to treat skin inflammation and atopic dermatitis. In this study, the broth produced by the cofermentation of ChiBai with Lactobacillus rhamnosus was studied for its effects on skin pigmentation through in vitro and in vitro experiments. In the in vitro experiments, we found that the fermented broth of ChiBai (FB-ChiBai) suppressed alpha-melanocyte stimulating hormone (α-MSH)-induced melanogenesis in B16F0 murine melanoma cells without any cytotoxicity at a concentration of 0.5%. FB-ChiBai significantly attenuated melanin production, tyrosinase activities and melanogenesis-related signaling pathways. Treatment with FB-ChiBai also reduced the nuclear translocation and promoter binding activities of MITF. In the in vivo experiments, FB-ChiBai was topically applied to the dorsal skin of C57BL/6J nude mice and concurrently irradiated with UVB, three times a week for 8 weeks. The results indicated that FB-ChiBai alleviated UVB-induced hyperpigmentation by reducing epidermal hyperplasia and inhibiting the CREB/MITF/tyrosinase pathway. In conclusion, our data indicated that the anti-melanogenic effects of FB-ChiBai are mediated by the inhibition of CREB/MITF/tyrosinase signaling pathway. The findings suggest that FB-ChiBai can protect against UV-B irradiation and that it might be used as an agent in cosmetic products to protect against UVB-induced hyperpigmentation.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Lacticaseibacillus rhamnosus/metabolismo , Monofenol Monooxigenasa/metabolismo , Pigmentación de la Piel/efectos de los fármacos , Rayos Ultravioleta , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Medicamentos Herbarios Chinos/metabolismo , Fermentación , Humanos , Melaninas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Factor de Transcripción Asociado a Microftalmía/metabolismo , Transducción de Señal , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación , Pigmentación de la Piel/efectos de la radiación , alfa-MSH/antagonistas & inhibidores
6.
Environ Toxicol ; 36(4): 607-619, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33270331

RESUMEN

Ultraviolet (UV) irradiation is a crucial factor that leads to skin photoaging and results in increased DNA damage, oxidative stress, and collagen degradation. Jasmine flowers have been utilized as a traditional medicine in Asia to treat various diseases, including dermatitis, diarrhea, and fever. Furthermore, the fermented broth of Lactobacillus rhamnosus has been reported to exert protective effects on the skin. In the present study, jasmine flower extract was fermented with L. rhamnosus. We investigated the antioxidant and collagen-promoting effects on UVB/H2 O2 -induced HS68 dermal fibroblast cell damage. The results indicated that treatment with the fermented flower extracts of Jasminum sambac (F-FEJS) could enhance the viability of HS68 cells. Furthermore, the UVB/H2 O2 -induced excessive production of reactive oxygen species, degradation of collagen, activation of MAPKs, including P38, ERK, and JNK, and premature senescence were remarkably attenuated by F-FEJS in dermal fibroblast cells. The nuclear accumulation of p-c-jun, which is downstream of MAPK, and the inactivation of p-smad2/3, which is one of the crucial transcription factors that enhance collagen synthesis, were reversed in response to F-FEJS treatment in UVB/H2 O2 -exposed cells. Notably, the expression of antioxidant genes, such as HO-1, and the nuclear translocation of Nrf2 were further enhanced by F-FEJS in UVB/H2 O2 -treated cells. Interestingly, the F-FEJS-induced increase in ARE luciferase activity indicated the activation of Nrf2/ARE signaling. In conclusion, our findings demonstrated that F-FEJS can effectively ameliorate UVB/H2 O2 -induced dermal cell aging and may be considered a promising ingredient in skin aging therapy.


Asunto(s)
Antioxidantes/farmacología , Senescencia Celular , Fibroblastos/efectos de los fármacos , Jasminum/química , Lacticaseibacillus rhamnosus/metabolismo , Extractos Vegetales/farmacología , Antioxidantes/aislamiento & purificación , Antioxidantes/metabolismo , Línea Celular , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Fermentación , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Flores/química , Humanos , Peróxido de Hidrógeno/toxicidad , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Envejecimiento de la Piel/efectos de los fármacos , Envejecimiento de la Piel/efectos de la radiación , Rayos Ultravioleta
7.
Environ Toxicol ; 35(2): 115-123, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31566298

RESUMEN

Galangin, a natural flavonol, has anti-inflammatory and antioxidative potential. However, the cytoprotective effects of galangin against oxidative-induced aging in human fibroblasts have not been well studied. IGF-1 signaling pathway is associated with the control of aging and longevity in human. The goal of this study was to investigate the effects of galangin on human skin fibroblast HS68 cells under H2 O2 exposure to induce aging. In this study, we demonstrate that galangin could decrease the levels of pro-inflammatory proteins and enhanced collagen formation through promoting the IGF-1R pathway. Furthermore, aging markers such as senescence-associated ß-galactosidase p53, p21Cip1/WAF1 , and p16INK4A were upregulated under H2 O2 exposure and galangin could reverse its effects. Taken together, these data indicated that anti-inflammatory and antiaging activities of galangin may be mediated through the IGF-1R signaling pathway. These findings may provide the evidence for galangin to develop as an antiwrinkle product on human skin.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antiinflamatorios/farmacología , Fibroblastos/efectos de los fármacos , Flavonoides/farmacología , Peróxido de Hidrógeno/toxicidad , Factor I del Crecimiento Similar a la Insulina/metabolismo , Piel/efectos de los fármacos , Envejecimiento/metabolismo , Biomarcadores/metabolismo , Línea Celular , Colágeno/biosíntesis , Humanos , Oxidación-Reducción , Transducción de Señal , Piel/metabolismo
8.
J Cell Physiol ; 234(8): 13557-13570, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30659610

RESUMEN

The prevalence of chronic hyperglycemia and its complications, imposing a critical burden on the worldwide economy and the global healthcare system, is a pressing issue. Mounting evidence indicates that oxidative stress and hypoxia, two noticeable features of hyperglycemia, play a joint crucial role in mediating cellular apoptosis. However, the underlying detailed molecular mechanism remains elusive. Triggered by the observation that insulin-like growth factor (IGF1)-binding protein 3 (IGFBP3) can mediate, in renal cells, high-glucose-induced apoptosis by elevating oxidative stress, we wish to, in this study, know whether or not the similar scenario holds in cardiac cells and, if so, to find its relevant molecular key players, thereby dissecting the underlying molecular pathway. Specifically, we used a combination of three different cellular sources (H9c2 cells, diabetic rats, and neonatal rat ventricular cardiomyocytes) as our model systems of study. We made use of Co-IP assay and western blot analysis in conjunction with loss-of-function reasoning, gain-of-function logic, and inhibitor treatment as our main analytical tools. As a result, briefly, our main findings are that hyperglycemia can induce cardiac IGFBP3 overexpression and secretion, that high levels of IGFBP3 can sequester IGF1 from IGF1 survival pathway, leading to apoptosis, and that IGFBP3 gene upregulation is hypoxia-inducible factor (HIF)1α-dependent and reactive oxygen species dependent. Piecing these findings together allows us to propose the improved molecular regulatory mechanism. In conclusion, we have established the molecular roles of IGFBP3, HIF1, and prolyl hydroxylase domain in connecting oxidative stress with hypoxia and in cellular apoptosis under hyperglycemia.


Asunto(s)
Hiperglucemia/metabolismo , Hiperglucemia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Animales , Apoptosis/fisiología , Línea Celular , Supervivencia Celular/fisiología , Hiperglucemia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Regulación hacia Arriba
9.
Cancer Sci ; 110(9): 2822-2833, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31278883

RESUMEN

Kinesin family member C1 (KIFC1) is implicated in the clustering of multiple centrosomes to maintain tumor survival and is thought to be an oncogene in several kinds of cancers. In our experiments, we first performed bioinformatics analysis to investigate the expression levels of KIFC1 in bladder cancer (BC) specimens and normal bladder epitheliums and then, using our samples, verified findings by quantitative real-time PCR and western blotting assays. All data showed that KIFC1 was significantly upregulated in BC specimens at both the mRNA and protein levels. Immunohistochemical studies in a cohort of 152 paraffin-embedded BC tissues displayed that upregulated expression of KIFC1 clearly correlated with pT status (P = .014) and recurrent status (P = .002). Kaplan-Meier survival analysis and log-rank test indicated that patients with BC with high KIFC1 expression had both shorter cancer-specific survival (P < .001) and recurrence-free survival time (P < .001) than those with low KIFC1 expression. Furthermore, ectopic downregulation of KIFC1 weakened BC cell proliferation and migration both in vitro and in vivo, whereas upregulation of KIFC1 enhanced this in vitro. Overexpression of KIFC1 phosphorylated GSK3ß and promoted Snail through activating AKT (protein kinase B0) to induce proliferation and epithelial-mesenchymal transition (EMT) and, therefore, substantially promoted BC migration and metastasis. Our study revealed an oncogenic role for KIFC1 to promote BC cell proliferation and EMT via Akt/GSK3ß signaling; KIFC1 might be a promising prognostic biomarker as well as a therapeutic target for BC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Transición Epitelial-Mesenquimal , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Cinesinas/metabolismo , Recurrencia Local de Neoplasia/diagnóstico , Neoplasias de la Vejiga Urinaria/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Fosforilación , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Regulación hacia Arriba , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/mortalidad , Urotelio/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Cell Biochem ; 119(4): 3363-3372, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29130531

RESUMEN

Metabolic syndrome is a risk factor for the development of cardiovascular diseases. Myocardial cell damage leads to an imbalance of energy metabolism, and many studies have indicated that short-term hypoxia during myocardial cell injury has a protective effect. In our previous animal studies, we found that short-term hypoxia in the heart has a protective effect, but long-term hypoxia increases myocardial cell injury. Palmitic acid (PA)-treated H9c2 cardiomyoblasts and neonatal rat ventricle cardiomyocytes were used to simulate hyperlipidemia model, which suppress cluster of differentiation 36 (CD36) and activate glucose transporter type 4 (GLUT4). We exposed the cells to short- and long-term hypoxia and investigated the protective effects of hypoxic preconditioning on PA-induced lipotoxicity in H9c2 cardiomyoblasts and neonatal rat cardiomyocytes. Preconditioning with short-term hypoxia enhanced both CD36 and GLUT4 metabolism pathway protein levels. Expression levels of phospho-PI3K, phospho-Akt, phospho-AMPK, SIRT1, PGC1α, PPARα, CD36, and CPT1ß induced by PA was reversed by short-term hypoxia in a time-dependent manner. PA-induced increased GLUT4 membrane protein level was reduced in the cells exposed to short-term hypoxia and si-PKCζ. Short-term hypoxia, resveratrol and si-PKCζ rescue H9c2 cells from apoptosis induced by PA and switch the metabolic pathway from GLUT4 dependent to CD36 dependent. We demonstrate short-term hypoxic preconditioning as a more efficient way as resveratrol in maintaining the energy metabolism of hearts during hyperlipidemia and can be used as a therapeutic strategy.


Asunto(s)
Antígenos CD36/metabolismo , Proteínas de Unión al ADN/metabolismo , Ácidos Grasos/metabolismo , Hiperlipidemias/metabolismo , Miocitos Cardíacos/citología , Ácido Palmítico/efectos adversos , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Línea Celular , Glucosa/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Redes y Vías Metabólicas , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ácido Palmítico/farmacología , Ratas
11.
Environ Toxicol ; 33(1): 93-103, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29087013

RESUMEN

BACKGROUND: Doxorubicin (Dox) is an effective anticancer agent. However, its effectiveness is limited by its cardiotoxic effects. It has also been reported that the mitogen-activated protein kinase family and NF-κB can be activated by Dox treatment. DATS has been shown to be a potent antioxidant with cardioprotective effects. We investigate whether Dox induces cardiac apoptosis through JNK- and ERK-dependent NF-κB upregulation that can be reduced by DATS treatment. METHODS AND MATERIAL: H9c2 cells were treated with 0.5-1.5 µM Dox for 24 hours. Dox promoted apoptosis and ROS generation and inhibited viability in a dose-dependent manner. Then, the phosphorylation levels of JNK, ERK, and NF-κB evaluated by western blot were elevated. We used inhibitors of JNK, ERK, and NF-κB to determine which of these proteins were involved in Dox-induced apoptosis. Furthermore, Dox-exposed cells were treated with DATS at doses of 1, 5, and 10 µM, and the data demonstrated that ROS generation and apoptotic proteins were decreased and that ERK and NF-κB were downregulated in a dose-dependent manner. Additionally, six-week-old rats were divided into three groups (n = 6 per group) designed as an eight-week study. Normal, Dox (at dose 3.75 mg/kg by ip) administered with or without DATS (at dose 40 mg/kg by gavage) treatment groups. The results indicate that cardiac dysfunction, apoptosis, and JNK, ERK, and NF-κB activation by Dox were reversed by treatment with DATS. CONCLUSION: DATS appears to suppress Dox-induced cardiomyocyte apoptosis by inhibiting NADPH oxidase-related ROS production and the downstream JNK/ERK/NF-κB signaling pathway; DATS may possess clinical therapeutic potential by blocking Dox-induced cardiotoxicity.


Asunto(s)
Compuestos Alílicos/farmacología , Apoptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfuros/farmacología , Acetilcisteína/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Doxorrubicina/toxicidad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Corazón/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar
12.
Environ Toxicol ; 33(2): 234-247, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29134746

RESUMEN

The physiological regulation of Oestrogen receptor α (ERα) and peroxisome proliferator-activated receptor alpha (PPARα) in Hepatocellular carcinoma (HCC) remains unknown. The present study we first treat the cells with fenofibrate and further investigated the possible mechanisms of 17ß-estradiol (E2 ) and/or ERα on regulating PPARα expression. We also found higher PPARα expression in the tumor area than adjacent areas and subsequently compared PPARα expression in four different hepatic cancer cell lines. Hep3B cells were found to express more PPARα than the other cell lines. Using the PPARα agonist fenofibrate, we found that fenofibrate increased Hep3B cell proliferation efficiency by increasing cell cycle proteins, such as cyclin D1 and PCNA, and inhibiting p27 and caspase 3 expressions. Next, we performed transient transfections and immuno-precipitation studies using the pTRE2/ERα plasmid to evaluate the interaction between ERα and PPARα. ERα interacted directly with PPARα and negatively regulated its function. Moreover, in Tet-on ERα over-expressed Hep3B cells, E2 treatment inhibited PPARα, its downstream gene acyl-CoA oxidase (ACO), cyclin D1 and PCNA expression and further increased p27 and caspase 3 expressions. However, over-expressed ERα plus 17-ß-estradiol (10-8 M) reversed the fenofibrate effect and induced apoptosis, which was blocked in ICI/melatonin/fenofibrate-treated cells. This study illustrates that PPARα expression and function were negatively regulated by ERα expression in Hep3B cells.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Fenofibrato/toxicidad , Hipolipemiantes/toxicidad , PPAR alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , PPAR alfa/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , ARN Mensajero/metabolismo
13.
J Cell Physiol ; 232(11): 3020-3029, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28500736

RESUMEN

In our previous study palmitic acid (PA) induced lipotoxicity and switches energy metabolism from CD36 to GLUT4 in H9c2 cells. Low level of high density lipoprotein (HDL) is an independent risk factor for cardiac hypertrophy. Therefore, we in the present study investigated whether HDL can reverse PA induced lipotoxicity in H9c2 cardiomyoblast cells. In this study, we treated H9c2 cells with PA to create a hyperlipidemia model in vitro and analyzed for CD36 and GLUT4 metabolic pathway proteins. CD36 metabolic pathway proteins (phospho-AMPK, SIRT1, PGC1α, PPARα, CPT1ß, and CD36) were decreased by high PA (150 and 200 µg/µl) concentration. Interestingly, expression of GLUT4 metabolic pathway proteins (p-PI3K and pAKT) were increased at low concentration (50 µg/µl) and decreased at high PA concentration. Whereas, phospho-PKCζ, GLUT4 and PDH proteins expression was increased in a dose dependent manner. PA treated H9c2 cells were treated with HDL and analyzed for cell viability. Results showed that HDL treatment induced cell proliferation efficiency in PA treated cells. In addition, HDL reversed the metabolic effects of PA: CD36 translocation was increased and reduced GLUT4 translocation, but HDL treatment significantly increased CD36 metabolic pathway proteins and reduced GLUT4 pathway proteins. Rat neonatal cardiomyocytes showed similar results. In conclusion, HDL reversed palmatic acid-induced lipotoxicity and energy metabolism imbalance in H9c2 cardiomyoblast cells and in neonatal rat cardiomyocyte cells.


Asunto(s)
Antígenos CD36/metabolismo , Metabolismo Energético/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Lipoproteínas HDL/farmacología , Miocitos Cardíacos/efectos de los fármacos , Ácido Palmítico/toxicidad , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Animales Recién Nacidos , Cardiotoxicidad , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Citoprotección , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , PPAR alfa/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Transporte de Proteínas , Proteolisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Sirtuina 1/metabolismo , Factores de Tiempo
14.
J Cell Biochem ; 118(7): 1659-1669, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27859591

RESUMEN

Hyperglycemia leads to excess reactive oxygen species (ROS) generation, which causes many diabetic complications, such as cardiomyopathy. Nuclear factor erythroid 2-related factor 2 (Nrf2), a redox-sensing transcription factor, can up-regulate its downstream antioxidant gene expressions in response to oxidative stress. However, the regulatory signal pathway in which high glucose (HG) induces Nrf2 activation is still unclear. Our results demonstrated that HG (33 mM) can indeed stimulate Nrf2 protein expression and translocation into the nucleus in cardiomyocytes, enhancing the downstream antioxidant protein levels. Using siRNAs, p38, JNK, PKCα, and PKCδ, as well as ROS scavengers, it was observed that the dependence of PKCα/PKCδ on ROS production to enhance JNK and p38 phosphorylation mediated HG-induced cardiac Nrf2 expression and activation. Knockdown of Nrf2 by siRNA transfection increased cleaved-caspase3, reduced Bcl2 in the cellular protein level and further exacerbated HG-induced apoptosis. In addition, all of these proteins induced by HG in vitro were also increased in STZ-induced diabetic rat ventricles in vivo. Our study demonstrated that HG-induced cardiac Nrf2 activation occurs through PKCα/PKCδ-ROS-JNK/p38 signaling. These findings may provide a therapeutic target to counteract the oxidative stress associated with diabetic cardiomyopathy. J. Cell. Biochem. 118: 1659-1669, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Glucosa/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Miocitos Cardíacos/citología , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Cell Biochem ; 118(11): 3785-3795, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28374891

RESUMEN

High levels of circulating low-density lipoproteins (LDL, plasma proteins that carry cholesterol and triglycerides) are associated with type 2 diabetes, arteriosclerosis, obesity, and hyperlipidemia. In the heart, the accumulation of oxidized low-density lipoprotein (Ox-LDL) has been proposed to play a role in the development of cardiovascular disease. We obtain cholesterol from animals and animal-derived foods such as milk, eggs, and cheese. In previous studies, the ratio of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) was shown to be important for our health. High levels of LDL cholesterol lead to atherosclerosis, increasing the risk of heart attack and ischemic stroke. In this study, we utilized Ox-LDL-treated H9c2 cardiomyoblast cells as a simulated hyperlipidemia model. CD36 metabolism pathway proteins (phospho-Akt, SIRT1, PGC1α, PPARα, CPT1ß, and CD36) increased at low doses of Ox-LDL. However, high doses (150 and 200 mg/dL) of Ox-LDL reduced the levels of these proteins. Interestingly, expression of GLUT4 metabolism pathway proteins (phospho-PKCζ) were reduced at low doses, while the expression of phospho-AMPK, phospho-PI3K, phospho-PKCζ, GLUT4, and PDH proteins increased at high doses. Ox-LDL acute treatment induces apoptosis in cardiomyocytes as evidenced by apoptotic nuclei apparition, caspase-3 activation, and cytochrome c release from mitochondria. In our results, Ox-LDL induced lipotoxicity in cardiomyocytes, and subsequent exposure to short-term hypoxia or reversed the Ox-LDL-induced metabolic imbalance. The same result was obtained with the pharmacological activation of SIRT1 by resveratrol and si-PKCζ. The mechanism of metabolic switching during Ox-LDL lipotoxicity seems to be mediated by SIRT1 and PKC ζ. J. Cell. Biochem. 118: 3785-3795, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Antígenos CD36/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Hiperlipidemias/metabolismo , Lipoproteínas LDL/metabolismo , Mioblastos Cardíacos/metabolismo , Animales , Hipoxia de la Célula , Línea Celular , Hiperlipidemias/patología , Mioblastos Cardíacos/patología , Ratas
16.
Cancer Sci ; 108(11): 2166-2175, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28837252

RESUMEN

Chromobox homolog 8 (CBX8), also known as human polycomb 8, is a repressor that maintains the transcriptionally repressive state in various cellular genes, and has been reported to promote tumorigenesis. In the present study, we examined CBX8 expression in eight pairs of muscle invasive bladder cancer tissues and adjacent non-tumor tissues, and found that CBX8 was frequently upregulated in muscle invasive bladder cancer tissues when compared to adjacent non-tumor tissues. Analysis showed that high expression of CBX8 in 152 muscle invasive bladder cancer specimens was associated with progression of the T, N, and M stages (P = 0.004, 0.005, <0.001, respectively). Furthermore, Kaplan-Meier survival analysis and log-rank test showed that muscle invasive bladder cancer patients with high CBX8 expression had a poor rate of overall survival (P < 0.001) and 5-year recurrence-free survival (P < 0.001) compared to patients with low CBX8 expression. High CBX8 expression predicted poor overall survival and 5-year recurrence-free survival in T and N stages of muscle invasive bladder cancer patients. Moreover, knockdown of CBX8 inhibited cell proliferation of urothelial carcinoma of the bladder both in vitro and in vivo. In addition, CBX8 depletion resulted in cell cycle delay of urothelial carcinoma cells of the bladder at the G2/M phase by the p53 pathway. The data suggest that high expression of CBX8 plays a critical oncogenic role in aggressiveness of urothelial carcinoma cells of the bladder through promoting cancer cell proliferation by repressing the p53 pathway, and CBX8 could be used as a novel predictor for muscle invasive bladder cancer patients.


Asunto(s)
Neoplasias de los Músculos/genética , Complejo Represivo Polycomb 1/genética , Proteína p53 Supresora de Tumor/genética , Neoplasias de la Vejiga Urinaria/genética , Anciano , Animales , Apoptosis/genética , Carcinogénesis/genética , Proliferación Celular/genética , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Persona de Mediana Edad , Neoplasias de los Músculos/patología , Neoplasias de los Músculos/secundario , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Environ Toxicol ; 32(4): 1390-1398, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27566995

RESUMEN

The Warburg effect plays a critical role in tumorigenesis, suggesting that specific agents targeting Warburg effect key proteins may be a promising strategy for cancer therapy. Previous studies have shown that diallyl trisulfide (DATS) inhibits proliferation of breast cancer cells by inducing apoptosis in vitro and in vivo. However, whether the Warburg effect is involved with the apoptosis-promoting action of DATS is unclear. Here, we show that the action of DATS is associated with downregulation of lactate dehydrogenase A (LDHA), an essential protein of the Warburg effect whose upregulation is closely related to tumorigenesis. Interestingly, inhibition of the Warburg effect by DATS in breast cancer cells did not greatly affect normal cells. Furthermore, DATS inhibited growth of breast cancer cells, particularly in MDA-MB-231, a triple-negative breast cancer (TNBC) cell, and reduced proliferation and migration; invasion was reversed by over-expression of LDHA. These data suggest that DATS inhibits breast cancer growth and aggressiveness through a novel pathway targeting the key enzyme of the Warburg effect. Our study shows that LDHA downregulation is involved in the apoptotic effect of DATS on TNBC. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1390-1398, 2017.


Asunto(s)
Compuestos Alílicos/farmacología , Antineoplásicos/farmacología , L-Lactato Deshidrogenasa/genética , Sulfuros/farmacología , Neoplasias de la Mama Triple Negativas/enzimología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Metabolismo de los Hidratos de Carbono , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Regulación hacia Abajo , Ensayos de Selección de Medicamentos Antitumorales , Represión Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Metástasis de la Neoplasia , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología
18.
Environ Toxicol ; 32(12): 2419-2427, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28834114

RESUMEN

Human skin aging is a progressive process that includes intrinsic aging and extrinsic photodamage, both of which can cause an accumulation of reactive oxygen species (ROS), resulting in dermal fibrosis dysfunction and wrinkle formation. Galangin is a flavonoid that exhibits anti-inflammatory and antioxidative potential. Previous studies have reported that galangin has antioxidative activity against ROS-mediated stress. The aim of the present study is to determine the antiaging effects of galangin on dermal fibroblasts exposed to H2 O2 . In this study, we established a hydrogen peroxide-induced inflammation and aging model using human HS68 dermal fibroblasts. Stimulation of fibroblasts with H2 O2 is associated with skin aging and increased expression of inflammation-related proteins, along with downregulation of collagen I/III formation and expression of antioxidative proteins. Galangin effectively reduced NF-κB activation, the expression of inflammation-related proteins and cell aging. Galangin also reversed H2 O2 -activated cell senescence in HS68 cells. Our results reveal that galangin protects human dermal fibroblasts by inhibiting NF-κB activation, decreases the expression of inflammatory factors and upregulates IGF1R/Akt-related proteins, indicating that galangin may be a potential candidate for developing natural antiaging products that protect skin from damage caused by ROS.


Asunto(s)
Antioxidantes/farmacología , Senescencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Flavonoides/farmacología , Línea Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , FN-kappa B/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Piel/citología
19.
Int J Mol Sci ; 18(4)2017 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-28441761

RESUMEN

Secretion of multifunctional estrogen and its receptor has been widely considered as the reason for markedly higher frequency of heart disease in men than in women. 17ß-Estradiol (E2), for instance, has been reported to prevent development of cardiac apoptosis via activation of estrogen receptors (ERs). In addition, protein phosphatase such as protein phosphatase 1 (PP1) and calcineurin (PP2B) are also involved in cardiac hypertrophy and cell apoptosis signaling. However, the mechanism by which E2/ERß suppresses apoptosis is not fully understood, and the role of protein phosphatase in E2/ERß action also needs further investigation. In this study, we observed that E2/ERß inhibited isoproterenol (ISO)-induced myocardial cell apoptosis, cytochrome c release and downstream apoptotic markers. Moreover, we found that E2/ERß blocks ISO-induced apoptosis in H9c2 cells through the enhancement of calcineurin protein degradation through PI3K/Akt/MDM2 signaling pathway. Our results suggest that supplementation with estrogen and/or overexpression of estrogen receptor ß gene may prove to be effective means to treat stress-induced myocardial damage.


Asunto(s)
Apoptosis/efectos de los fármacos , Calcineurina/metabolismo , Estradiol/farmacología , Receptor beta de Estrógeno/antagonistas & inhibidores , Isoproterenol/toxicidad , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Cicloheximida/farmacología , Citocromos c/metabolismo , Receptor beta de Estrógeno/metabolismo , Leupeptinas/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Ratas
20.
Cell Biochem Funct ; 34(8): 606-612, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27859413

RESUMEN

ZAK (sterile alpha motif and leucine zipper containing kinase AZK), a serine/threonine kinase with multiple biochemical functions, has been associated with various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy. In our previous reports, we found that the activation of ZAKα signaling was critical for cardiac hypertrophy. In this study, we show that the expression of ZAKα activated apoptosis through both a FAS-dependent pathway and a mitochondria-dependent pathway by subsequently inducing caspase-3. ZAKß, an isoform of ZAKα, is dramatically expressed during cardiac hypertrophy and apoptosis. The interaction between ZAKα and ZAKß was demonstrated here using immunoprecipitation. The results show that ZAKß has the ability to diminish the expression level of ZAKα. These findings reveal an inherent regulatory role of ZAKß to antagonize ZAKα and to subsequently downregulate the cardiac hypertrophy and apoptosis induced by ZAKα.


Asunto(s)
Apoptosis , Cardiomegalia/metabolismo , Cardiomegalia/patología , Proteínas Quinasas/metabolismo , Animales , Supervivencia Celular , Espacio Intracelular/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Unión Proteica , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA