Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
BMB Rep ; 56(12): 663-668, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37817437

RESUMEN

C-reactive protein (CRP) is an inflammatory marker and risk factor for atherosclerosis and cardiovascular diseases. However, the mechanism through which CRP induces myocardial damage remains unclear. This study aimed to determine how CRP damages cardiomyocytes via the change of mitochondrial dynamics and whether survivin, an anti-apoptotic protein, exerts a cardioprotective effect in this process. We treated H9c2 cardiomyocytes with CRP and found increased intracellular ROS production and shortened mitochondrial length. CRP treatment phosphorylated ERK1/2 and promoted increased expression, phosphorylation, and translocation of DRP1, a mitochondrial fission-related protein, from the cytoplasm to the mitochondria. The expression of mitophagy proteins PINK1 and PARK2 was also increased by CRP. YAP, a transcriptional regulator of PINK1 and PARK2, was also increased by CRP. Knockdown of YAP prevented CRP-induced increases in DRP1, PINK1, and PARK2. Furthermore, CRP-induced changes in the expression of DRP1 and increases in YAP, PINK1, and PARK2 were inhibited by ERK1/2 inhibition, suggesting that ERK1/2 signaling is involved in CRP-induced mitochondrial fission. We treated H9c2 cardiomyocytes with a recombinant TAT-survivin protein before CRP treatment, which reduced CRP-induced ROS accumulation and reduced mitochondrial fission. CRP-induced activation of ERK1/2 and increases in the expression and activity of YAP and its downstream mitochondrial proteins were inhibited by TAT-survivin. This study shows that mitochondrial fission occurs during CRPinduced cardiomyocyte damage and that the ERK1/2-YAP axis is involved in this process, and identifies that survivin alters these mechanisms to prevent CRP-induced mitochondrial damage. [BMB Reports 2023; 56(12): 663-668].


Asunto(s)
Dinaminas , Miocitos Cardíacos , Dinaminas/metabolismo , Survivin/metabolismo , Survivin/farmacología , Dinámicas Mitocondriales/fisiología , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/farmacología , Sistema de Señalización de MAP Quinasas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas/metabolismo
2.
J Med Food ; 26(9): 672-682, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37498372

RESUMEN

This study researched the mineral composition of Korean washed-dehydrated solar salt (WDS) without bittern. It also evaluated the anticancer effects of doenjang (WDSD) prepared using WDS on azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon cancer in C57BL/6 mice. The mineral composition of WDS showed lower Mg (11.71 ± 1.89 g/kg) and S (9.77 ± 2.88 g/kg) contents, and it was confirmed that mice in the WDSD group (AOM/DSS+WDSD) displayed significantly lower weight loss, colon length reduction, and tumor formation compared with the control (Con) group. In addition, pathologically, it was confirmed that the extent of epithelial cell damage and inflammation in the colon tissue of the WDSD group was restored to a state similar to that of the Nor group. Besides, WDSD regulated the protein expression of apoptosis (Bcl-2-associated X protein [Bax], B cell lymphoma-2 [Bcl-2], B cell lymphoma-extra large [Bcl-xL], and caspase 9, caspase 3), and p53, p21, and proinflammatory cytokines (interleukin [IL]-6, tumor necrosis factor [TNF]-α), thereby inducing the apoptosis and cell cycle arrest of cancer cells and suppressing inflammation. In addition, the intestinal microbiota of the mice treated with WDSD were more diverse, with an abundance of Bifidobacterium, a lactic acid bacterium beneficial to colon health, was also a greater presence of Faecalibaculum, which showed antitumor effects. These results indicate that solar salts and their different processing methods affect their functional health-promoting properties. In addition, the inhibitory effect on colon cancer was further enhanced when doenjang was prepared with WDS with low Mg and S content.


Asunto(s)
Colitis , Neoplasias del Colon , Animales , Ratones , Ratones Endogámicos C57BL , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Colon , Citocinas/metabolismo , Interleucina-6/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Cloruro de Sodio Dietético/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Azoximetano/efectos adversos , Sulfato de Dextran/efectos adversos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico
3.
J Med Food ; 26(8): 560-569, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37405755

RESUMEN

The antiobesity effects of kimchi with catechin and lactic acid bacteria as starters were studied in C57BL/6 mice with high-fat diet (HFD)-induced obesity. We prepared four types of kimchi: commercial kimchi, standard kimchi, green tea functional kimchi, and catechin functional kimchi (CFK). Body weight and weight of adipose tissue were significantly lower in the kimchi-treated groups than in the HFD and Salt (HFD +1.5% NaCl) groups. In addition, in the CFK group, the serum levels of triglycerides, total cholesterol, and low-density lipoprotein cholesterol were significantly lower and those of high-density lipoprotein cholesterol were markedly higher than the corresponding levels in the HFD and Salt groups. Moreover, CFK reduced fat cells and crown-like structures in the liver and epididymal fat tissues. The protein expression of adipo/lipogenesis-related genes in the liver and epididymal fat tissues was significantly lower (1.90-7.48-fold) in the CFK group than in the HFD and Salt groups, concurrent with upregulation of lipolysis-related genes (1.71-3.38-fold) and downregulation of inflammation-related genes (3.17-5.06-fold) in epididymal fat tissues. In addition, CFK modulated the gut microbiomes of obese mice by increasing the abundance of Bacteroidetes (7.61%), while in contrast, Firmicutes (82.21%) decreased. In addition, the presence of the Erysipelotrichaceae (8.37%) family in the CFK group decreased, while the number of beneficial bacteria of the families, Akkermansiaceae (6.74%), Lachnospiraceae (14.95%), and Lactobacillaceae (38.41%), increased. Thus, CFK exhibited an antiobesity effect through its modulation of lipid metabolism and the microbiome.


Asunto(s)
Fármacos Antiobesidad , Catequina , Alimentos Fermentados , Lactobacillales , Animales , Ratones , Catequina/farmacología , Catequina/metabolismo , Lactobacillales/metabolismo , Ratones Endogámicos C57BL , Fármacos Antiobesidad/farmacología , Hígado/metabolismo , Dieta Alta en Grasa/efectos adversos , Colesterol
5.
Sci Rep ; 12(1): 3874, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35264673

RESUMEN

Mitochondrial dysfunction is a key element in the progression of Parkinson's disease (PD). The inefficient operation of the electron transport chain (ETC) impairs energy production and enhances the generation of oxidative stress contributing to the loss of dopaminergic cells in the brain. ATPase inhibitory factor 1 (IF1) is a regulator of mitochondrial energy metabolism. IF1 binds directly to the F1Fo ATP synthase and prevents ATP wasting during compromised energy metabolism. In this study, we found treatment with IF1 protects mitochondria against PD-like insult in vitro. SH-SY5Y cells treated with IF1 were resistant to loss of ATP and mitochondrial inner membrane potential during challenge with rotenone, an inhibitor of complex I in the ETC. We further demonstrated that treatment with IF1 reversed rotenone-induced superoxide production in mitochondria and peroxide accumulation in whole cells. Ultimately, IF1 decreased protein levels of pro-apoptotic Bax, cleaved caspase-3, and cleaved PARP, rescuing SH-SY5Y cells from rotenone-mediated apoptotic death. Administration of IF1 significantly improved the results of pole and hanging tests performed by PD mice expressing human α-synuclein. This indicates that IF1 mitigates PD-associated motor deficit. Together, these findings suggest that IF1 exhibits a neuroprotective effect preventing mitochondrial dysfunction in PD pathology.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Ratones , Mitocondrias/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Rotenona/metabolismo , Rotenona/farmacología
6.
FASEB J ; 35(8): e21794, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34314059

RESUMEN

While biglycan (BGN) is suggested to direct diverse signaling cascades, the effects of soluble BGN as a ligand on metabolic traits have not been studied. Herein, we tested the effects of BGN on obesity in high-fat diet (HFD)-induced obese animals and glucose metabolism, with the underlying mechanism responsible for observed effects in vitro. Our results showed that BGN administration (1 mg/kg body weight, intraperitoneally) significantly prevented HFD-induced obesity, and this was mainly attributed to reduced food intake. Also, intracerebroventricular injection of BGN reduced food intake and body weight. The underlying mechanism includes modulation of neuropeptides gene expression involved in appetite in the hypothalamus in vitro and in vivo. In addition, BGN regulates glucose metabolism as shown by improved glucose tolerance in mice as well as AMPK/AKT dual pathway-driven enhanced glucose uptake and GLUT4 translocation in L6 myoblast cells. In conclusion, our results suggest BGN as a potential therapeutic target to treat risk factors for metabolic diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Biglicano/administración & dosificación , Glucosa/metabolismo , Músculo Esquelético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Conducta Alimentaria , Ratones , Ratones Endogámicos ICR , Ratas
7.
Analyst ; 146(15): 4748-4755, 2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34231563

RESUMEN

Since oxidative stress has been recognized as a major factor contributing to the progression of several neurodegenerative disorders, reactive oxygen species (ROS) including superoxide have received great attention as a representative molecular marker for the diagnosis of Alzheimer's disease (AD). Here, superoxide-sensitive fluorogenic molecular probes, benzenesulfonylated resorufin derivatives (BSRs), were newly devised for optical bioimaging of oxidative events in neurodegenerative processes. BSRs, fluorescence-quenched benzenesulfonylated derivatives of resorufin, were designed to recover their fluorescence upon exposure to superoxide through a selective nucleophilic uncaging reaction of the benzenesulfonyl cage. Among BSRs, BSR6 presented the best sensitivity and selectivity to superoxide likely due to the optimal reactivity matching between the nucleophilicity of superoxide and its electrophilicity ascribed to the highly electron-withdrawing pentafluoro-substitution on the benzenesulfonyl cage. Fluorescence imaging of inflammatory cells and animal models presented the potential of BSR6 for optical sensing of superoxide in vitro and in vivo. Furthermore, microglial cell (Bv2) imaging with BSR6 enabled the optical monitoring of intracellular oxidative events upon treatment with an oxidative stimulus (amyloid beta, Aß) or the byproduct of oxidative stress (4-hydroxynonenal, HNE).


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Animales , Sondas Moleculares , Estrés Oxidativo , Especies Reactivas de Oxígeno , Superóxidos
8.
FASEB J ; 34(10): 13445-13460, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32816366

RESUMEN

We investigated the effect of chitinase-3-like protein 1 (CHI3L1) on glucose metabolism and its underlying mechanisms in skeletal muscle cells, and evaluated whether the observed effects are relevant in humans. CHI3L1 was associated with increased glucose uptake in skeletal muscles in an AMP-activated protein kinase (AMPK)-dependent manner, and with increased intracellular calcium levels via PAR2. The improvement in glucose metabolism observed in an intraperitoneal glucose tolerance test on male C57BL/6J mice supported this association. Inhibition of the CaMKK was associated with suppression of CHI3L1-mediated glucose uptake. Additionally, CHI3L1 was found to influence glucose uptake through the PI3K/AKT pathway. Results suggested that CHI3L1 stimulated the phosphorylation of AS160 and p38 MAPK downstream of AMPK and AKT, and the resultant GLUT4 translocation. In primary myoblast cells, stimulation of AMPK and AKT was observed in response to CHI3L1, underscoring the biological relevance of CHI3L1. CHI3L1 levels were elevated in cells under conditions that mimic exercise in vitro and in exercised mice in vivo, indicating that CHI3L1 is secreted during muscle contraction. Finally, similar associations between CHI3L1 and metabolic parameters were observed in humans alongside genotype associations between CHI3L1 and diabetes at the population level. CHI3L1 may be a potential therapeutic target for the treatment of diabetes.


Asunto(s)
Proteína 1 Similar a Quitinasa-3 , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Músculo Esquelético , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular , Proteína 1 Similar a Quitinasa-3/sangre , Proteína 1 Similar a Quitinasa-3/fisiología , Estudios de Asociación Genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
9.
BMC Mol Cell Biol ; 21(1): 42, 2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32522165

RESUMEN

BACKGROUND: Stress is an important cause of skin disease, including hair loss. The hormonal response to stress is due to the HPA axis, which comprises hormones such as corticotropin releasing factor (CRF), adrenocorticotropic hormone (ACTH), and cortisol. Many reports have shown that CRF, a crucial stress hormone, inhibits hair growth and induces hair loss. However, the underlying mechanisms are still unclear. The aim of this study was to examine the effect of CRF on human dermal papilla cells (DPCs) as well as hair follicles and to investigate whether the HPA axis was established in cultured human DPCs. RESULTS: CRF inhibited hair shaft elongation and induced early catagen transition in human hair follicles. Hair follicle cells, both human DPCs and human ORSCs, expressed CRF and its receptors and responded to CRF. CRF inhibited the proliferation of human DPCs through cell cycle arrest at G2/M phase and induced the accumulation of reactive oxygen species (ROS). Anagen-related cytokine levels were downregulated in CRF-treated human DPCs. Interestingly, increases in proopiomelanocortin (POMC), ACTH, and cortisol were induced by CRF in human DPCs, and antagonists for the CRF receptor blocked the effects of this hormone. CONCLUSION: The results of this study showed that stress can cause hair loss by acting through stress hormones. Additionally, these results suggested that a fully functional HPA axis exists in human DPCs and that CRF directly affects human DPCs as well as human hair follicles under stress conditions.


Asunto(s)
Alopecia/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Células Cultivadas/metabolismo , Dermis/citología , Cabello/crecimiento & desarrollo , Folículo Piloso/citología , Humanos , Hidrocortisona/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo
10.
FEBS J ; 287(10): 2087-2104, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32196931

RESUMEN

Meteorin-like (metrnl) is a recently identified adipomyokine that beneficially affects glucose metabolism; however, its underlying mechanism of action is not completely understood. We here show that the level of metrnl increases in vitro under electrical pulse stimulation and in vivo in exercised mice, suggesting that metrnl is secreted during muscle contractions. In addition, metrnl increases glucose uptake via the calcium-dependent AMPKα2 pathway in skeletal muscle cells and increases the phosphorylation of HDAC5, a transcriptional repressor of GLUT4, in an AMPKα2-dependent manner. Phosphorylated HDAC5 interacts with 14-3-3 proteins and sequesters them in the cytoplasm, resulting in the activation of GLUT4 transcription. An intraperitoneal injection of recombinant metrnl improved glucose tolerance in mice with high-fat-diet-induced obesity or type 2 diabetes, but not in AMPK ß1ß2 muscle-specific null mice. Metrnl improves glucose metabolism via AMPKα2 and is a promising therapeutic candidate for glucose-related diseases such as type 2 diabetes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Diabetes Mellitus Tipo 2/genética , Histona Desacetilasas/genética , Factores de Crecimiento Nervioso/genética , Obesidad/genética , Proteínas 14-3-3/genética , Animales , Línea Celular , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa/efectos adversos , Estimulación Eléctrica , Glucosa/genética , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Humanos , Resistencia a la Insulina/genética , Ratones , Contracción Muscular/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Factores de Crecimiento Nervioso/farmacología , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/patología , Condicionamiento Físico Animal , Proteínas Recombinantes/farmacología
11.
J Cosmet Dermatol ; 19(11): 3058-3066, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32056369

RESUMEN

BACKGROUND: There is a growing interest in the relationship among stress hormones, neuroendocrine signaling, and skin diseases, including hair loss. Previous reports showed that stress hormones inhibit human hair growth and induce early catagen transition. Moreover, a CRH receptor antagonist reversed CRH-induced alopecia in a mouse model, suggesting that antagonization of the CRH receptor is a key clinical strategy to treat stress-induced hair loss. OBJECTIVES: The aim of this study was to investigate the protective effect of CRH receptor antagonists from Pulsatilla chinensis on human hair follicles (hHFs) and human dermal papilla cells (hDPCs). METHODS: hHFs were observed and scored by hair cycle. The levels of cAMP, a second messenger, were measured in each group. In addition, the mRNA and protein levels of factors related to the hair cycle were measured. Furthermore, the expression levels of various members of the mitogen-activated protein kinase (MAPK) signaling pathway related to stress were measured. RESULTS: CRH induced early catagen transition in an ex vivo hair organ culture model. In addition, CRH downregulated the levels of alkaline phosphatase (ALP) and hair anagen-related cytokines in cultured hDPCs. Moreover, CRH induced the phosphorylation of JNK, c-Jun, p38, ERK, and Akt in cultured hDPCs. CRH receptor antagonists isolated from P chinensis reversed these CRH-induced modulations in both ex vivo hair follicles (HFs) and cultured hDPCs. CONCLUSIONS: These results indicate that P chinensis effectively blocks CRH receptor function and that saponin derivatives from P chinensis could be a pharmaceutical and cosmetic approach to treat stress-induced hair loss.


Asunto(s)
Preparaciones Farmacéuticas , Pulsatilla , Cabello , Folículo Piloso , Humanos , Receptores de Hormona Liberadora de Corticotropina
12.
Metabolism ; 105: 154171, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32006557

RESUMEN

BACKGROUND: Based on the metabolic effect of exogenous ATPase inhibitory factor 1 (IF1) on glucose metabolism, we tested whether IF1 treatment is effective in ameliorating weight gain and whether its effects are sex specific. METHODS: HFD-fed C57BL/6 mice were treated with IF1 (5 mg/kg body weight, injected intraperitoneally). The underlying mechanisms of effect of IF1 on body weight were investigated in vitro and in vivo. Associations between genotypes of IF1 and obesity and relevant phenotype were further tested at the population level. RESULTS: Chronic treatment with IF1 significantly decreased body weight gain by regulating food intake of HFD-fed male mice. IF1 activated the AKT/mTORC pathway and modulated the expression of appetite genes in the hypothalamus of HFD-fed male mice and its effect was confirmed in hypothalamic cell lines as well as hypothalamic primary cells. This required the interaction of IF1 with ß-F1-ATPase on the plasma membrane of hypothalamic cells, which led to an increase in extracellular ATP production. In addition, IF1 treatment showed sympathetic nerve activation as measured by serum norepinephrine levels and UCP-1 expression in the subcutaneous fat of HFD-fed male mice. Notably, administration of recombinant IF1 to HFD-fed ovariectomized female mice showed remarkable reductions in food intake as well as body weight, which was not observed in wild-type 5-week female mice. Lastly, sex-specific genotype associations of IF1 with obesity prevalence and metabolic traits were demonstrated at the population level in humans. IF1 genetic variant (rs3767303) was significantly associated with lower prevalence of obesity and lower levels of body mass index, waist circumference, hemoglobin A1c, and glucose response area only in male participants. CONCLUSION: IF1 is involved in weight regulation by controlling food intake and potentially sympathetic nerve activation in a sex-specific manner.


Asunto(s)
Peso Corporal/efectos de los fármacos , Obesidad/genética , Proteínas/genética , Proteínas/farmacología , Animales , Apetito/genética , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Femenino , Variación Genética , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Persona de Mediana Edad , Obesidad/epidemiología , Ovariectomía , Prevalencia , Caracteres Sexuales , Aumento de Peso/efectos de los fármacos , Proteína Inhibidora ATPasa
13.
J Mol Cell Cardiol ; 138: 244-255, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31866378

RESUMEN

BACKGROUND: Survivin has an anti-apoptotic effect against anthracycline-induced cardiotoxicity. Clinically, statin use is associated with a lower risk for heart failure in breast cancer patients with anthracycline chemotherapy. So, the purpose of our study was to investigate whether survivin mediates the protective effect of statin against anthracycline-induced cardiotoxicity. METHODS: Mice were treated once a week with 5 mg/kg doxorubicin for 4 weeks with or without atorvastatin 20 mg/kg every day then heart tissues were analyzed. Molecular and cellular biology analyses were performed with H9c2 cell lysates. RESULTS: Doxorubicin suppressed survivin expression via activation of FOXO1 in H9c2 cardiomyocytes. Whereas, atorvastatin inhibited FOXO1 by increasing phosphorylation and inhibiting nuclear localization. Doxorubicin induced FOXO1 binding to STAT3 and prevented STAT3 from interacting with Sp1. However, atorvastatin inhibited these interactions and stabilized STAT3/Sp1 transcription complex. Chromatin immunoprecipitation analysis demonstrated that doxorubicin decreased STAT3/Sp1 complex binding to survivin promoter, whereas atorvastatin stabilized this binding. In mouse model, atorvastatin rescued doxorubicin-induced reduction of survivin expression and of heart function measured by cardiac magnetic resonance imaging. CONCLUSIONS: Our study suggested a new pathophysiologic mechanism that survivin mediated protective effect of atorvastatin against doxorubicin-induced cardiotoxicity via FOXO1/STAT3/Sp1 transcriptional network.


Asunto(s)
Atorvastatina/farmacología , Cardiotónicos/farmacología , Citoprotección , Doxorrubicina/toxicidad , Proteína Forkhead Box O1/antagonistas & inhibidores , Miocitos Cardíacos/metabolismo , Survivin/metabolismo , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Proteína Forkhead Box O1/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
14.
Nutrients ; 11(12)2019 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-31842363

RESUMEN

Since arginase has been shown to compete with nitric oxide (NO) synthase, emerging evidence has reported that arginase inhibition improves obesity by increasing NO production. Semen cuscutae (SC), which is a well-known Chinese medicine, has multiple biological functions such as anti-oxidant function and immune regulation. In this study, we investigated whether the SC as a natural arginase inhibitor influences hepatic lipid abnormalities and whole-body adiposity in high-fat diet (HFD)-induced obese mice. The lipid accumulation was significantly reduced by SC treatment in oleic acid-induced hepatic steatosis in vitro. Additionally, SC supplementation substantially lowered HFD-induced increases in arginase activity and weights of liver and visceral fat tissue, while increasing hepatic NO. Furthermore, elevated mRNA expressions of sterol regulatory element-binding transcription factor 1 (SREBP-1c), fatty-acid synthase (FAS), peroxisome proliferator-activated receptor-gamma (PPAR-γ)1, and PPAR-γ2 in HFD-fed mice were significantly attenuated by SC supplementation. Taken together, SC, as a novel natural arginase inhibitor, showed anti-obesity properties by modulating hepatic arginase and NO production and metabolic pathways related to hepatic triglyceride (TG) metabolism.


Asunto(s)
Adiposidad/efectos de los fármacos , Cuscuta , Suplementos Dietéticos , Medicamentos Herbarios Chinos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Obesidad/metabolismo , Animales , Arginasa/antagonistas & inhibidores , Técnicas de Cultivo de Célula , Dieta Alta en Grasa , Acido Graso Sintasa Tipo I/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Óxido Nítrico/biosíntesis , Obesidad/terapia , PPAR gamma/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
15.
FASEB J ; 33(12): 14825-14840, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31670977

RESUMEN

ATPase inhibitory factor 1 (IF1) is an ATP synthase-interacting protein that suppresses the hydrolysis activity of ATP synthase. In this study, we observed that the expression of IF1 was up-regulated in response to electrical pulse stimulation of skeletal muscle cells and in exercized mice and healthy men. IF1 stimulates glucose uptake via AMPK in skeletal muscle cells and primary cultured myoblasts. Reactive oxygen species and Rac family small GTPase 1 (Rac1) function in the upstream and downstream of AMPK, respectively, in IF1-mediated glucose uptake. In diabetic animal models, the administration of recombinant IF1 improved glucose tolerance and down-regulated blood glucose level. In addition, IF1 inhibits ATP hydrolysis by ß-F1-ATPase in plasma membrane, thereby increasing extracellular ATP and activating the protein kinase B (Akt) pathway, ultimately leading to glucose uptake. Thus, we suggest that IF1 is a novel myokine and propose a mechanism by which AMPK and Akt contribute independently to IF1-mediated improvement of glucose tolerance impairment. These results demonstrate the importance of IF1 as a potential antidiabetic agent.-Lee, H. J., Moon, J., Chung, I., Chung, J. H., Park, C., Lee, J. O., Han, J. A., Kang, M. J., Yoo, E. H., Kwak, S.-Y., Jo, G., Park, W., Park, J., Kim, K. M., Lim, S., Ngoei, K. R. W., Ling, N. X. Y., Oakhill, J. S., Galic, S., Murray-Segal, L., Kemp, B. E., Mantzoros, C. S., Krauss, R. M., Shin, M.-J., Kim, H. S. ATP synthase inhibitory factor 1 (IF1), a novel myokine, regulates glucose metabolism by AMPK and Akt dual pathways.


Asunto(s)
Glucosa/metabolismo , Mioblastos/metabolismo , Proteínas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Adenosina Trifosfato/metabolismo , Adulto , Animales , Línea Celular , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Hipoglucemiantes/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas/metabolismo , Proteínas/genética , Proteínas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/uso terapéutico , Proteína Inhibidora ATPasa
16.
BMC Mol Cell Biol ; 20(1): 54, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775627

RESUMEN

BACKGROUND: Reactive oxygen species (ROS) produce different lesions in DNA by ROS-induced DNA damage. Detection and quantification of 8-oxo-7,8-dihydroguanine (8-oxoG) within cells are important for study. Human ribosomal protein S3 (hRpS3) has a high binding affinity to 8-oxoG. In this study, we developed an imaging probe to detect 8-oxoG using a specific peptide from hRpS3. Transactivator (TAT) proteins are known to have cell-penetrating properties. Therefore, we developed a TAT-S3 probe by attaching a TAT peptide to our imaging probe. RESULTS: A DNA binding assay was conducted to confirm that our probe bound to 8-oxoG and apurinic/apyrimidinic (AP) sites. We confirmed that the TAT-S3 probe localized in the mitochondria, without permeabilization, and fluoresced in H2O2-treated HeLa cells and zebrafish embryos. Treatment with Mitoquinone (MitoQ), a mitochondria-targeted antioxidant, reduced TAT-S3 probe fluorescence. Additionally, treatment with O8, an inhibitor of OGG1, increased probe fluorescence. A competition assay was conducted with an aldehyde reaction probe (ARP) and methoxyamine (MX) to confirm binding of TAT-S3 to the AP sites. The TAT-S3 probe showed competitive binding to AP sites with ARP and MX. CONCLUSIONS: These results revealed that the TAT-S3 probe successfully detected the presence of 8-oxoG and AP sites in damaged cells. The TAT-S3 probe may have applications for the detection of diseases caused by reactive oxygen species.


Asunto(s)
ADN/análisis , Colorantes Fluorescentes , Guanina/análogos & derivados , Animales , Sitios de Unión , ADN/química , Daño del ADN , ADN Mitocondrial , ADN-(Sitio Apurínico o Apirimidínico) Liasa/análisis , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , Citometría de Flujo , Colorantes Fluorescentes/síntesis química , Guanina/análisis , Guanina/metabolismo , Células HeLa , Humanos , Microscopía Confocal , Mitocondrias/patología , Unión Proteica , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Transactivadores/química , Pez Cebra
17.
Oncol Lett ; 13(5): 3681-3687, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28521470

RESUMEN

Human ribosomal protein S3 (hRpS3) is a component of the 40S ribosomal subunit that associated in protein synthesis. hRpS3 has additional ribosomal functions such as DNA repair, transcription, metastasis, and apoptosis via interaction with numerous signaling molecules and has different modifications. Cyclin-dependent kinases (CDKs) are heterodimeric serine/threonine protein kinases that regulate cell cycle progression. Among its members, the Cdk1-cyclin B complex is known to control cell progression in the G2/M phase, while Cdk2-cyclin E/A complexes function in G1/S and S/G2 transition. In our previous study, we observed interaction between hRpS3 and Cdk1. The present study investigated the interaction between hRpS3 and Cdk2. Cdk2 phosphorylated hRps3 at amino acid residues S6 and T221 during the S-phase. Furthermore, hRpS3 knockdown delayed cell cycle progression by modulating the expression of cell cycle-related proteins, including cyclin B1 and cyclin E1. These findings suggest that hRpS3 is involved in Cdk2-mediated cell cycle regulation.

18.
Biochem Pharmacol ; 115: 85-93, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27208887

RESUMEN

Rosiglitazone is a selective ligand for peroxisome proliferator-activated receptor-gamma (PPAR-γ), which serves diverse biological functions. A number of autoimmune disease models have been used to examine the anti-inflammatory and immunosuppressive effects of tolerogenic dendritic cells (tDCs). The aim of the present study was to investigate whether rosiglitazone-mediated DC (Rosi-DC) therapy suppressed arthritis in a collagen-induced arthritis (CIA) mouse model. Rosi-DCs were generated by treating immature DCs with TNF-α, type II collagen, and rosiglitazone. CIA mice then received subcutaneously (s.c.) two injections of Rosi-DCs. The severity of arthritis was then assessed histopathologically. The phenotypes of the DC and regulatory T (Treg) cell populations in CIA mice were determined by flow cytometry and the effect of Rosi-DCs on the secretion of autoimmunity-inducing cytokines was examined by ELISA. Rosi-DCs expressed lower levels of DC-related surface markers than mature DCs. Histopathological examination revealed that the degree of inflammation in the paws of Rosi-DC-treated mice was much lower than that in the paws of PBS-treated CIA mice. Taken together, these results clearly show that rosiglitazone-mediated DCs ameliorate CIA, most likely via the induction of antigen-specific Treg cells.


Asunto(s)
Artritis Experimental/terapia , Células Dendríticas/efectos de los fármacos , Células Dendríticas/trasplante , Tiazolidinedionas/uso terapéutico , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/inmunología , Células Cultivadas , Técnicas de Cocultivo , Colágeno , Células Dendríticas/inmunología , Femenino , Ratones , Ratones Endogámicos DBA , Rosiglitazona , Linfocitos T Reguladores/inmunología
19.
Yonsei Med J ; 56(6): 1678-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26446654

RESUMEN

PURPOSE: To investigate the effects of resveratrol on the expression of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) in human adult retinal pigment epithelial (ARPE-19) cells, and on experimental choroidal neovascularization (CNV) in mice. MATERIALS AND METHODS: ARPE-19 cells were treated with different concentrations of resveratrol and then incubated under hypoxic conditions with subsequent evaluation of cell viability, expression of HIF-1α, and expression of VEGF. The effects of resveratrol on the synthesis and degradation of hypoxia-induced HIF-1α were evaluated using inhibitors of the PI3K/Akt/mTOR and the ubiquitin proteasome pathways. In animal studies, CNV lesions were induced in C57BL/6 mice by laser photocoagulation. After 7 days of oral administration of resveratrol or vehicle, which began one day after CNV induction, image analysis was used to measure CNV areas on choroidal flat mounts stained with isolectin IB4. RESULTS: In ARPE-19 cells, resveratrol significantly inhibited HIF-1α and VEGF in a dose-dependent manner, by blocking the PI3K/Akt/mTOR signaling pathway and by promoting proteasomal HIF-1α degradation. In mice experiments, orally administered resveratrol significantly inhibited CNV growth in a dose-dependent manner. CONCLUSION: Resveratrol may have therapeutic value in the management of diseases involving pathological neovascularization.


Asunto(s)
Neovascularización Coroidal/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Estilbenos/farmacología , Serina-Treonina Quinasas TOR/fisiología , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Adulto , Animales , Supervivencia Celular/efectos de los fármacos , Neovascularización Coroidal/patología , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidores de las Quinasa Fosfoinosítidos-3 , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Resveratrol , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal , Estilbenos/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ubiquitina , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
PLoS One ; 10(8): e0135438, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26271039

RESUMEN

Insulin inhibits ischemia/reperfusion-induced myocardial apoptosis through the PI3K/Akt/mTOR pathway. Survivin is a key regulator of anti-apoptosis against doxorubicin-induced cardiotoxicity. Insulin increases survivin expression in cardiac myocytes to mediate cytoprotection. However, the mechanism by which survivin mediates the protective effect of insulin against doxorubicin-associated injury remains to be determined. In this study, we demonstrated that pretreatment of H9c2 cardiac myocytes with insulin resulted in a significant decrease in doxorubicin-induced apoptotic cell death by reducing cytochrome c release and caspase-3 activation. Doxorubicin-induced reduction of survivin mRNA and protein levels was also significantly perturbed by insulin pretreatment. Reducing survivin expression with survivin siRNA abrogated insulin-mediated inhibition of caspase-3 activation, suggesting that insulin signals to survivin inhibited caspase-3 activation. Interestingly, pretreatment of H9c2 cells with insulin or MG132, a proteasome inhibitor, inhibited doxorubicin-induced degradation of the transcription factor Sp1. ChIP assay showed that pretreatment with insulin inhibited doxorubicin-stimulated Sp1 dissociation from the survivin promoter. Finally using pharmacological inhibitors of the PI3K pathway, we showed that insulin-mediated activation of the PI3K/Akt/mTORC1 pathway prevented doxorubicin-induced proteasome-mediated degradation of Sp1. Taken together, insulin pretreatment confers a protective effect against doxorubicin-induced cardiotoxicity by promoting Sp1-mediated transactivation of survivin to inhibit apoptosis. Our study is the first to define a role for survivin in cellular protection by insulin against doxorubicin-associated injury and show that Sp1 is a critical factor in the transcriptional regulation of survivin.


Asunto(s)
Doxorrubicina/farmacología , Insulina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/metabolismo , Interferencia de ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Survivin
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...