Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
World J Diabetes ; 15(5): 988-1000, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38766434

RESUMO

BACKGROUND: Visceral obesity is increasingly prevalent among adolescents and young adults and is commonly recognized as a risk factor for type 2 diabetes. Estrogen [17ß-estradiol (E2)] is known to offer protection against obesity via diverse me-chanisms, while its specific effects on visceral adipose tissue (VAT) remain to be fully elucidated. AIM: To investigate the impact of E2 on the gene expression profile within VAT of a mouse model of prediabetes. METHODS: Metabolic parameters were collected, encompassing body weight, weights of visceral and subcutaneous adipose tissues (VAT and SAT), random blood glucose levels, glucose tolerance, insulin tolerance, and overall body composition. The gene expression profiles of VAT were quantified utilizing the Whole Mouse Genome Oligo Microarray and subsequently analyzed through Agilent Feature Extraction software. Functional and pathway analyses were conducted employing Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses, respectively. RESULTS: Feeding a high-fat diet (HFD) moderately increased the weights of both VAT and SAT, but this increase was mitigated by the protective effect of endogenous E2. Conversely, ovariectomy (OVX) led to a significant increase in VAT weight and the VAT/SAT weight ratio, and this increase was also reversed with E2 treatment. Notably, OVX diminished the expression of genes involved in lipid metabolism compared to HFD feeding alone, signaling a widespread reduction in lipid metabolic activity, which was completely counteracted by E2 administration. This study provides a comprehensive insight into E2's local and direct protective effects against visceral adiposity in VAT at the gene level. CONCLUSION: In conclusion, the present study demonstrated that the HFD-induced over-nutritional challenge disrupted the gene expression profile of visceral fat, leading to a universally decreased lipid metabolic status in E2 deficient mice. E2 treatment effectively reversed this condition, shedding light on the mechanistic role and therapeutic potential of E2 in combating visceral obesity.

3.
J Ethnopharmacol ; 312: 116446, 2023 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-37019162

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Shuxie Compound (SX) combines the composition and efficacy of Suanzaoren decoction and Huanglian Wendan decoction. It can soothe the liver, regulate the qi, nourish the blood and calm the mind. It is used in the clinical treatment of sleep disorder with liver stagnation. Modern studies have proved that circadian rhythm disorder (CRD) can cause sleep deprivation and liver damage, which can be effectively ameliorated by traditional Chinese medicine to soothe the liver stagnation. However, the mechanism of SX is unclear. AIM OF THE STUDY: This study was designed to demonstrate the impact of SX on CRD in vivo, and confirm the molecular mechanisms of SX in vitro. MATERIALS AND METHODS: The quality of SX and drug-containing serum was controlled by UPLC-Q-TOF/MS, which were used in vivo and in vitro experiments, respectively. In vivo, a light deprivation mouse model was used. In vitro, a stable knockdown Bmal1 cell line was used to explore SX mechanism. RESULTS: Low-dose SX (SXL) could restore (1) circadian activity pattern, (2) 24-h basal metabolic pattern, (3) liver injury, and (4) Endoplasmic reticulum (ER) stress in CRD mice. CRD decreased the liver Bmal1 protein at ZT15, which was reversed by SXL treatment. Besides, SXL decreased the mRNA expression of Grp78/ATF4/Chop and the protein expression of ATF4/Chop at ZT11. In vitro experiments, SX reduced the protein expression of thapsigargin (tg)-induced p-eIF2α/ATF4 pathway and increase the viability of AML12 cells by increasing the expression of Bmal1 protein. CONCLUSIONS: SXL relieved CRD-induced ER stress and improve cell viability by up-regulating the expression of Bmal1 protein in the liver and then inhibiting the protein expression of p-eIF2α/ATF4.


Assuntos
Fatores de Transcrição ARNTL , Fator de Iniciação 2 em Eucariotos , Camundongos , Animais , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 2 em Eucariotos/farmacologia , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/farmacologia , Fígado , Ritmo Circadiano , Estresse do Retículo Endoplasmático , Apoptose , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
4.
Front Pharmacol ; 14: 1107507, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36814500

RESUMO

Introduction: Sleep disorders are common clinical psychosomatic disorders that can co-exist with a variety of conditions. In humans and animal models, sleep deprivation (SD) is closely related with gastrointestinal diseases. Shu-Xie Decoction (SX) is a traditional Chinese medicine (TCM) with anti-nociceptive, anti-inflammatory, and antidepressant properties. SX is effective in the clinic for treating patients with abnormal sleep and/or gastrointestinal disorders, but the underlying mechanisms are not known. This study investigated the mechanisms by which SX alleviates SD-induced colon injury in vivo. Methods: C57BL/6 mice were placed on an automated sleep deprivation system for 72 h to generate an acute sleep deprivation (ASD) model, and low-dose SX (SXL), high-dose SX (SXH), or S-zopiclone (S-z) as a positive control using the oral gavage were given during the whole ASD-induced period for one time each day. The colon length was measured and the colon morphology was visualized using hematoxylin and eosin (H&E) staining. ROS and the redox biomarkers include reduced glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) were detected. Quantitative real-time PCR (qRT-PCR), molecular docking, immunofluorescence and western blotting assays were performed to detect the antioxidant signaling pathways. Results: ASD significantly increased FBG levels, decreased colon length, moderately increased the infiltration of inflammatory cells in the colon mucosa, altered the colon mucosal structure, increased the levels of ROS, GSH, MDA, and SOD activity compared with the controls. These adverse effects were significantly alleviated by SX treatment. ASD induced nuclear translocation of NRF2 in the colon mucosal cells and increased the expression levels of p62, NQO1, and HO1 transcripts and proteins, but these effects were reversed by SX treatment. Conclusion: SX decoction ameliorated ASD-induced oxidative stress and colon injury by suppressing the p62/KEAP1/NRF2/HO1/NQO1 signaling pathway. In conclusion, combined clinical experience, SX may be a promising drug for sleep disorder combined with colitis.

5.
FEBS Open Bio ; 12(8): 1475-1488, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35666067

RESUMO

Hypoxia-induced endothelial dysfunction is known to be involved in the pathogenesis of several vascular diseases. However, it remains unclear whether the pentose phosphate pathway (PPP) is involved in regulating the response of endothelial cells to hypoxia. Here, we established an in vitro model by treating EA.hy926 (a hybrid human umbilical vein cell line) with cobalt chloride (CoCl2 ; a chemical mimic that stabilizes HIF-1α, thereby leading to the development of hypoxia), and used this to investigate the involvement of PPP by examining expression of its key enzyme, glucose-6-phosphate dehydrogenase (G6PD). We report that CoCl2 induces the accumulation of HIF-1α, leading to endothelial cell dysfunction characterized by reduced cell viability, proliferation, tube formation, and activation of cytokine production, accompanied with a significant decrease in G6PD expression and activity. The addition of 6-aminonicotinamide (6-AN) to inhibit PPP directly causes endothelial dysfunction. Additionally, N-Acetylcysteine (NAC), a precursor of glutathione, was further evaluated for its protective effects; NAC displayed a protective effect against CoCl2 -induced cell damage by enhancing G6PD activity, and this was abrogated by 6-AN. The effects of CoCl2 and the involvement of G6PD in endothelial dysfunction have been confirmed in primary human aortic endothelial cells. In summary, G6PD was identified as a novel target of CoCl2 -induced damage, which highlighted the involvement of PPP in regulating the response of endothelial cell CoCl2 . Treatment with NAC may be a potential strategy to treat hypoxia or ischemia, which are widely observed in vascular diseases.


Assuntos
Acetilcisteína , Doenças Vasculares , Acetilcisteína/farmacologia , Cobalto , Glucosefosfato Desidrogenase/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipóxia
6.
Front Pharmacol ; 13: 848355, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35462928

RESUMO

Introduction: Chronic stress has been shown to cause liver damage in addition to psychological depression. Besides, drug-induced liver injury is frequently caused by antidepressants. Shuxie-1 decoction (SX-1) is a formula of traditional Chinese medicine commonly used in nourishing liver blood, and relieving depression. However, the underlying molecular mechanism remains unclear. Therefore, this study was designed to explore the effects and mechanisms of SX-1 in treating chronic stress-induced depression as well as liver injury. Methods: Chronic unpredictable mild stress (CUMS) was applied to male Wistar rats for 4 weeks, with or without administration of SX-1 at low-dose and high-dose for 6 weeks, using Fluoxetine (Flu) as a positive control. Body weight was monitored once every 2 weeks. In the sixth week, the sugar preference test and open field test were carried out to evaluate the depression status. After that, the serum and liver tissues were collected. The quality control of SX-1 decoctions and drug-containing serum was controlled by UHPLC-QE-MS. The cell viability was measured by Cell Counting Kit-8 (CCK8). Enzyme-linked immunosorbent assay (Elisa), Western Blot and immunohistochemistrical staining was obtained to detect the protein levels in the plasma and the hepatic tissues, respectively. Results: CUMS led to decreased 1) body weight, 2) the preference for sugar water, 3) the desire to explore in open field, and increased serum levels of corticosterone. All these factors were completely reversed by SX-1 treatment. Hematoxylin-eosin staining (HE) showed that SX-1 improved the hepatocyte vacuolization in CUMS treated rats, decreased the serum levels of alanine aminotransferase (ALT) and the deposition of type I collagen (Col I) in hepatocytes as well. CUMS increased the levels of hepatic Interleukin-6 (IL-6), and provoked the activation of Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3), which was abrogated by SX-1 treatment. Cobalt chloride (CoCl2) increased the protein expression of IL-6 and p-STAT3 in AML12 cells. Besides, nuclear pyknosis was observed under electron microscope, which were recovered after rat SX serum. Conclusion: SX-1 effectively ameliorated CUMS-induced depression-like behaviors as well as hepatic injuries, probably by the blockade of hepatic IL-6/JAK2/STAT3 signaling.

7.
Biomed Pharmacother ; 138: 111408, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33684693

RESUMO

PingTang No.5 capsule (PT5), a modified Traditional Chinese Medicine (TCM) formula of Zexie Decoction, is used to treat patients with lipid metabolism disorders in our hospital. The present study was designed to investigate the mechanisms of PT5 in treating non-alcoholic fatty liver disease (NAFLD). PT5 information including ingredients, pharmacological properties, and potential targets was obtained from TCM databases. The candidate targets of PT5 were predicted by network pharmacological analysis, and the possible pathway and mechanism were obtained from DAVID database, followed by experimental validation in NAFLD mice model treated with PT5. Total 328 compounds were selected using the threshold oral bioactivity (OB) > 30% or drug-likeness (DL) > 0.1 of pharmacology characteristic, and 1033 candidate targets obtained to construct the network analysis. The 113 targets were selected from the intersection between candidate targets of PT5 and NAFLD relative gene. These targets were evaluated in diabetic complications, cancer, Hepatitis B, Fluid shear stress and atherosclerosis, and TNF signaling pathway. TNF-α was the important factor in protein interaction analysis of STRING and involved in the lipid regulation and oxidative stress in NAFLD. When administrated to the NAFLD mice, PT5 reduced weight, blood fatty acids, decreased the adipocyte size, and improved the metabolism. Besides, the molecular verification of lipid metabolism increased and oxidative stress reduced that interpreted the mechanism of PT5 preventing liver cell from lipid accumulation and injury of NAFLD. These results presented PT5 have the potential therapy as an alternative treatment for NAFLD.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Metabolismo dos Lipídeos/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Animais , Cápsulas , Bases de Dados Factuais/tendências , Metabolismo dos Lipídeos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Estresse Oxidativo/fisiologia , Reprodutibilidade dos Testes
8.
BMC Gastroenterol ; 20(1): 366, 2020 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-33143650

RESUMO

BACKGROUND: The occurrence of non-alcoholic fatty liver disease (NAFLD) is found to be higher in patients with obstructive sleep apnea (OSA), which is characterized by intermittent hypoxia. Activation of hypoxia-inducible factors has been shown in the development and progression of NAFLD, implying a cause and effects relationship between NAFLD and hypoxia. The present study was designed to investigate the interaction of lipotoxicity and hypoxia in the pathogenesis of NAFLD using mice model with high-fat diet (HFD) feeding or hypoxic treatment. METHODS: NAFLD model was induced in mice by HFD feeding, and in cultured primary hepatocytes by administration of palmitate acid. Mouse hypoxic model was produced by placing the mice in a Animal incubator with oxygen concentration at 75% followed by a 21% oxygen supplement. Hypoxic condition was mimicked by treating the hepatocytes with cobalt chloride (CoCl2) or 1% oxygen supply. Pimonidazole assay was conducted to evaluate hypoxia. Lipid metabolic genes were measured by real-time polymerase-chain reaction. HIF-1α and HIF-2α genes were silenced by siRNA. RESULTS: HFD feeding and palmitate acid treatment provoked severe hepatic hypoxia along with TG accumulation in mice and in cultured primary hepatocytes respectively. Conversely, hypoxia induced hepatic TG accumulation in mice and in cultured primary hepatocytes. Hypoxic treatment inhibited the expression of lipolytic genes, while increased the expression of lipogenicgenes in mice. Although both lipotoxicity and hypoxia could activate hepatic hypoxia-induced factor 1α and 2α, while neither lipotoxicity- nor hypoxia- induced hepatic steatosis was affected when HIF was knocked down. CONCLUSIONS: HFD resulted in hepatic TG accumulation and concomitant hypoxia. Conversely, hypoxia induced hepatic TG accumulation in mice and in cultured heptocytes. Thus lipotoxicity and hypoxia might work as reciprocal causation and orchestrate to promote the development of NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Dieta Hiperlipídica/efeitos adversos , Hepatócitos , Humanos , Hipóxia , Fígado , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia
9.
Biomed Pharmacother ; 118: 109407, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31545290

RESUMO

The purpose of this study was to observe the functions of preconditioning with endoplasmic reticulum stress (ERS) whether alleviated heart ischemia/reperfusion injury (HI/RI) via modulating IRE1/ATF6/RACK1/PERK and PGC-1α expressions in diabetes mellitus (DM) or not. Diabetic rats were pretreated with 0.6 mg/kg tunicamycin (TM, 0.6 mg/kg tunicamycin was administered via intraperitoneal injection 30 minutes prior to the I/R procedures), and then subjected to 45 minutes of ischemia and 3 hours of reperfusion. Blood and myocardial tissues were collected, myocardial pathological injuries were investigated, serum creatine kinase-MB (CK-MB) and cardiac troponin T (cTnT) levels were measured, left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP), maximum rate of left ventricular pressure rise (+dp/dtmax) and maximum rate of left ventricular pressure drop (-dp/dtmax) were evaluated, reactive oxygen species (ROS) and caspase-3 levels were observed, ΔΨm level and ROS expression were measured, and activated transcript factor 6 (ATF6), receptor for activated C kinase 1 (RACK1), PRK-like ER kinase (PERK), glucose regulated protein 78 (GRP78) and peroxisome proliferator-activated receptor γ co-activator 1-α (PGC-1α) expressions were assessed. The TM ameliorated the pathological damages, reduced myocardial oxidative stress damages, restrained apoptosis, and upregulated the expressions of ATF6, RACK1, PERK, GRP78 and PGC-1α compared with those of the ischemia/reperfusion (I/R) group in DM. This study suggested the preconditioning with endoplasmic reticulum stress (TM) strategy that could enhance protection against HI/RI in DM in clinical myocardial diseases.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Diabetes Mellitus Experimental/patologia , Estresse do Retículo Endoplasmático , Proteínas de Membrana/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Quinase C Ativada/metabolismo , eIF-2 Quinase/metabolismo , Animais , Creatina Quinase/sangue , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Masculino , Potencial da Membrana Mitocondrial , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/complicações , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Troponina T/metabolismo
10.
Cancer Manag Res ; 11: 263-272, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30636898

RESUMO

PURPOSE: We aimed to explore the association of plasma TP53-induced glycolysis and apoptosis regulator (TIGAR) level with colorectal cancer (CRC) metastasis. METHODS: A cross-sectional study of 126 CRC patients was conducted in Xiamen, China. Multivariable logistic regression was used to calculate adjusted OR and 95% CIs of plasma TIGAR concentration for CRC metastasis in different models with adjustment for potential confounders. Area under the curve (AUC) of receiver operating characteristic (ROC) curve was used to evaluate the diagnostic value. RESULTS: CRC patients with metastasis showed significantly decreased plasma TIGAR concentration compared to their controls (1.97±0.64 vs 2.49±0.69 ng/mL [log transformed], P=0.002). Higher plasma TIGAR was significantly associated with the decreased risk of CRC metastasis, and the adjusted OR (95% CI) was 0.134 (0.027-0.676, P=0.015) for per SD increase in plasma TIGAR concentration, and the trend test for increasing tertiles showed a negative trend of plasma TIGAR on risk of CRC metastasis (P for trend test: 0.005). Pearson correlation coefficients of plasma TIGAR with other cancer biomarkers (carbohydrate antigen 50 [CA50], carbohydrate antigen 199 [CA199], carbohydrate antigen 125 [CA125], carbohydrate antigen 724 [CA724], carcinoembryonic antigen [CEA], and ferritin [FER]) was low (P>0.05). AUC (95% CI) of ROC curve of plasma TIGAR for CRC metastasis was comparable to the values of cancer biomarkers (all P-values <0.05). CONCLUSION: Higher level of plasma TIGAR was significantly and independently associated with lower risk of CRC metastasis, and its prognostic value on CRC metastasis was comparable to the common cancer biomarkers.

11.
Artigo em Inglês | MEDLINE | ID: mdl-30108656

RESUMO

Danggui Sini decoction (DSD) is a traditional Chinese decoction, which is wildly applied and showed to be effective in ameliorating ischemia-related symptoms. However, the mechanisms of DSD action in ischemic damage remain to be fully clarified. Pancreatic islet endothelial cells are pivotal constituent of islet microvasculature, with high vulnerability to hypoxic injuries. Here, using MST1 cell, a pancreatic islet endothelial cell-line, as a model, we investigated the effects of DSD on hypoxia-stimulated endothelial cell lesions and its underlying mechanisms. We found that DSD-Containing Serum (DSD-CS), collected from DSD-treated rats, could efficiently protect MST1 survival and proliferation from Cobalt chloride (CoCl2) induced damage, including cell viability, proliferation, and tube formation. Furthermore, DSD-CS restored the activity of PI3K/Akt/eNOS signaling inhibited by CoCl2 in MST1 cells. The protective effect of DSD-CS could be blocked by the specific PI3K/Akt/eNOS inhibitor LY294002, suggesting that DSD-CS protection of MST1 cell survival from hypoxia was mediated by PI3K/Akt/eNOS pathway. In conclusion, DSD treatment protected MST1 survival from hypoxic injuries via PI3K/Akt/eNOS pathway, indicating its role in protecting microvascular endothelial cells.

12.
JCI Insight ; 3(12)2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29925687

RESUMO

Androgen excess predisposes women to type 2 diabetes (T2D), but the mechanism of this is poorly understood. We report that female mice fed a Western diet and exposed to chronic androgen excess using dihydrotestosterone (DHT) exhibit hyperinsulinemia and insulin resistance associated with secondary pancreatic ß cell failure, leading to hyperglycemia. These abnormalities are not observed in mice lacking the androgen receptor (AR) in ß cells and partially in neurons of the mediobasal hypothalamus (MBH) as well as in mice lacking AR selectively in neurons. Accordingly, i.c.v. infusion of DHT produces hyperinsulinemia and insulin resistance in female WT mice. We observe that acute DHT produces insulin hypersecretion in response to glucose in cultured female mouse and human pancreatic islets in an AR-dependent manner via a cAMP- and mTOR-dependent pathway. Acute DHT exposure increases mitochondrial respiration and oxygen consumption in female cultured islets. As a result, chronic DHT exposure in vivo promotes islet oxidative damage and susceptibility to additional stress induced by streptozotocin via AR in ß cells. This study suggests that excess androgen predisposes female mice to T2D following AR activation in neurons, producing peripheral insulin resistance, and in pancreatic ß cells, promoting insulin hypersecretion, oxidative injury, and secondary ß cell failure.


Assuntos
Androgênios/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Neurônios/metabolismo , Animais , Dieta Ocidental , Di-Hidrotestosterona/metabolismo , Feminino , Glucose/metabolismo , Humanos , Hiperinsulinismo , Hipotálamo , Resistência à Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Estreptozocina/farmacologia
13.
Int J Nanomedicine ; 13: 2507-2520, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29719397

RESUMO

INTRODUCTION: Insulin has shown antioxidation and cytoprotective effects to decrease heart ischemia/reperfusion injury (HI/RI) in diabetes mellitus (DM), but the role of insulin/poly(ethylene glycol)-carboxymethyl chitosan (PEG-CMCS) on HI/RI in DM is not known. This research explored whether insulin/PEG-CMCS revealed a protective effect on HI/RI in DM through ornithine decarboxylase (ODC)/polyamine systems. MATERIALS AND METHODS: Diabetes was induced via streptozotocin (STZ) in Sprague Dawley (SD) rats, which suffered from HI via blocking the left circumflex artery for 45 minutes, followed by 2 hours of reperfusion. α-Difluoromethylornithine-ethylglyoxal bis (guanylhydrazone) (DFMO-EGBG) and insulin/PEG-CMCS were administered to diabetic rats to explore their roles on severity of HI/RI. RESULTS: Insulin could be fleetly and efficiently loaded via the nanocarrier PEG-CMCS at pH =6, showing efficient loading and stable release. In addition, insulin/PEG-CMCS showed significant hypoglycemic activity in diabetic rats. On the other hand, ischemia/reperfusion obviously augmented the contents of creatine kinase (CK), lactic dehydrogenase (LDH), putrescine (Pu), myocardial infarct size, and NF-κB and spermidine/spermine N'-acetyltransferase (SSAT) expressions and decreased the levels of spermine (Sp), polyamine pools (PAs), heart rate (HR), coronary blood flow (CF), left ventricular developed pressure (LVDP), and ODC expression, compared with Sham. Administration of insulin and insulin/PEG-CMCS both reduced the contents of CK, LDH, Pu, myocardial infarct size, and NF-κB and SSAT expressions and increased the levels of Sp, PAs, HR, CF, LVDP, and ODC expression, while insulin/PEG-CMCS significantly indicated the protective results, and DFMO-EGBG showed the opposite effects. CONCLUSION: The research showed that insulin/PEG-CMCS could play a protective effect on HR/RI in diabetic rats via its antioxidative, antiapoptotic, and anti-inflammatory roles and modulating ODC/polyamine systems.


Assuntos
Insulina/análogos & derivados , Precondicionamento Isquêmico Miocárdico/métodos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Ornitina Descarboxilase/metabolismo , Poliaminas/metabolismo , Animais , Apoptose , Cardiotônicos/administração & dosagem , Cardiotônicos/farmacologia , Quitosana/análogos & derivados , Quitosana/química , Vasos Coronários/fisiopatologia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/fisiopatologia , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacologia , Insulina/administração & dosagem , Insulina/farmacologia , Masculino , Infarto do Miocárdio/patologia , Nanopartículas/administração & dosagem , Nanopartículas/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Ratos Sprague-Dawley
14.
Diabetes ; 67(7): 1345-1355, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29716892

RESUMO

We investigated how human proislet peptide (HIP) regulates differentiation of human fetus-derived pancreatic progenitor cells (HFPPCs) and explored the potential link between HIP signaling and the menin pathway, which is key to regulating pancreatic islet differentiation. The data show that HIP promoted expression of proislet transcription factors (TFs), including PDX-1, MAFA, and NKX6.1, as well as other maturation markers of ß-cells, such as insulin, GLUT2, KIR6.2, SUR1, and VDCC. Moreover, HIP increased insulin content and promoted the ability of HFPPCs to normalize blood glucose in diabetic mice. HIP inhibited the TF FOXO1 by increasing AKT-mediated phosphorylation. HIP-induced repression of FOXO1 suppressed menin expression, leading to reducing menin binding to the promoter of the three key proislet TFs, decreasing recruitment of H3K9 methyltransferase SUV39H1, and thus reducing repressive H3K9me3 at the promoter. These coordinated actions lead to increased expression of the proislet TFs, resulting in induction of HFPPC differentiation. Consistently, constitutive activation of FOXO1 blocks HIP-induced transcription of these TFs. Together, these studies unravel the crucial role of the HIP/AKT/FOXO/menin axis in epigenetically controlling expression of proislet TFs, regulating the differentiation of HFPPCs, and normalizing blood glucose in diabetic mice.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/genética , Epigênese Genética/efeitos dos fármacos , Proteína Forkhead Box O1/genética , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas/genética , Células-Tronco/efeitos dos fármacos , Animais , Diferenciação Celular/genética , Células Cultivadas , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/terapia , Células HEK293 , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células-Tronco/fisiologia
15.
Am J Transl Res ; 10(3): 957-965, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29636885

RESUMO

Acute liver injury is a destructive liver disorder resulting from overwhelming liver inflammation, oxidative stress and hepatocyte death. Puerarin is a natural flavonoid compound isolated from the traditional Chinese herb radix puerariae. This study investigated the protective effects of puerarin against lipopolysaccharide (LPS)/D-galactosamine (D-Gal)-induced liver injury and the potential mechanisms in mice. Mice were given an intraperitoneal administration of puerarin 200 mg/kg 2 h prior to LPS (50 µg/kg)/D-Gal (400 mg/kg) injection and were sacrificed 6 h post LPS/D-Gal treatment. The results showed that administration of puerarin substantially alleviated LPS/D-Gal-induced acute liver injury in mice by increased survival rates, improved liver histopathology, reduced plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, alleviated production of pro-inflammatory cytokines, and suppressed hepatocyte apoptosis. Moreover, puerarin pretreatment activated autophagy by increased the ratio of LC3B-II/I and the protein levels of Beclin-1, decreased the levels of p62 protein expression. Taken together, these findings demonstrated that puerarin could prevent the LPS/D-Gal-induced liver injury in mice, and its mechanisms might be associated with the increments of autophagy and suppression of apoptosis.

16.
Mol Med Rep ; 17(5): 7149-7155, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29568862

RESUMO

Damage to pancreatic ß­cells is closely associated with diabetes. However, the mechanism underlying injury to pancreatic ß­cells remains unclear, although hypoxia is considered as one of the leading causes. Appoptosin is a mitochondrial protein that promotes neuronal apoptosis. Studies conducted on appoptosin thus far have primarily focused on Alzheimer's disease, and have demonstrated that the expression of appoptosin is significantly increased in ischemic­reperfused rat brains, which indicates its close association with hypoxia. However, the role of appoptosin in pancreatic ß­cells, which are sensitive to hypoxia, remains unknown. Therefore, the current study aimed to investigate the function of appoptosin in pancreatic ß­cells in a hypoxic environment. Cobalt chloride (CoCl2) was used to mimic the hypoxic status of the cells. The results of a terminal deoxynucleotidyl transferase dUTP nick­end labeling assay demonstrated that CoCl2 promoted apoptosis in MIN6 mouse insulinoma cells, and western blotting and reverse transcription­quantitative polymerase chain reaction results demonstrated that the activation of appoptosin was induced, promoting mitochondrial damage and caspase 3 activation. Silencing of appoptosin using short hairpin RNA significantly reduced CoCl2­induced apoptosis in MIN6 cells. In conclusion, CoCl2 increased the expression of appoptosin, which aggravated mitochondrial damage in MIN6 cells. Therefore, inhibiting the expression of appoptosin may benefit pancreatic ß-cells survival during islet transplantation.


Assuntos
Apoptose , Células Secretoras de Insulina/patologia , Mitocôndrias/patologia , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas Carreadoras de Solutos/genética , Regulação para Cima , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular , Cobalto/efeitos adversos , Ativação Enzimática/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Regulação para Cima/efeitos dos fármacos
17.
Mol Med Rep ; 17(5): 7281-7286, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29568901

RESUMO

Type 1 diabetes is caused by destruction of the pancreatic ß­cells and, to date, no cure has been developed. Promoting the survival of pancreatic ß­cells may be beneficial for patients with type 1 diabetes. Puerarin is an estrogen analogue that been demonstrated in previous studies to be able to decreased blood glucose in patients with type 1 diabetes. Similar results were demonstrated in previous studies which additionally demonstrated that puerarin was able to decreased blood glucose in type 1 diabetic mice by protecting pancreatic ß­cells. However, the mechanism underlying the function of puerarin in pancreatic ß­cells remains unclear. Therefore, the present study sought to investigate the detailed function of puerarin in pancreatic ß­cells. In the present study, H2O2 was used to induce apoptosis. It was observed that puerarin significantly decreased H2O2­induced apoptosis in mouse insulinoma MIN6 cells. It was additionally observed that puerarin decreased the levels of intracellular reactive oxygen species and mitochondrial superoxide in MIN6 cells. The protective effect of puerarin was markedly decreased by 6­aminonicotinamide, an inhibitor of glucose­6­phosphate dehydrogenase (G6PD). In conclusion, the results of the present study suggested that puerarin may increase the activity of G6PD, decreased the level of oxidative stress in MIN6 cells, protect mitochondria and promote MIN6 cell survival. Investigating the mechanism underlying the effect of puerarin in MIN6 cells may provide a novel approach for development of a cure for type 1 diabetes.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Isoflavonas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Peróxido de Hidrogênio/metabolismo , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Camundongos
18.
Exp Cell Res ; 364(2): 152-159, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29391151

RESUMO

Diabetic vascular complications are often defined by vascular endothelial lesions. However, as a plastic cell type, whether endothelial cells could transit from quiescence to hyper-active status and hamper vascular stability upon hyperglycemia stimulation and whether this process is involved in diabetic vascular complications remain obscure. Survivin has been identified as an anti-apoptotic protein in tumor or epithelial cells by either promoting proliferation or inhibiting apoptosis. Therefore, this study aims at investigating the effects of hyperglycemia on endothelial cell status and the potential involvement of survivin. We found that high glucose (25 mM) did not cause endothelial injuries, instead, it evidently promotes endothelial proliferation and tube formation capacity indicating endothelial cell dysfunction upon hyperglycemia characterized by its preference to hyper-active status. Concomitantly, an upregulation of survivin was detected accompanied by the key component elevations of autophagy pathway including LC3, Beclin1, and p62. YM155, a specific inhibitor of survivin, could abrogate hyperglycemia-induced endothelial hyper-activation. Application of the autophagy inhibitor (3MA) and agonist (rapamycin) supported that survivin could be as a downstream effect or of autophagy. Thus, our results suggested that survivin/autophagy axis a potential therapeutic target in treatment of diabetic vascular complications.


Assuntos
Autofagia , Células Endoteliais/metabolismo , Hiperglicemia/metabolismo , Survivina/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Células Endoteliais/patologia , Humanos , Hiperglicemia/patologia
19.
J Diabetes ; 10(7): 581-588, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29194974

RESUMO

BACKGROUND: Fetuin-B impairs insulin action in myotubes and hepatocytes and causes glucose intolerance in mice. This study explored the correlation between serum fetuin-B and intrahepatic triglyceride (IHTG) content, and the association between fetuin-B and the risk of insulin resistance in the general adult population. METHODS: A cross-sectional study of 1318 obese adults who underwent serum fetuin-B testing and hepatic ultrasonography was conducted in Xiamen, China. The IHTG content was determined in 428 subjects by magnetic resonance spectroscopy. RESULTS: Non-alcoholic fatty liver disease prevalence was significantly higher in those with the highest serum fetuin-B concentrations and the highest IHTC content (Tertile 3) than in subjects in Tertiles 1 and 2 (62.6% vs 60.7% and 54.3%, respectively [P = 0.032], and 15.3% vs 12.8% and 12.7%, respectively [P = 0.049]). There was a significant association between increasing serum fetuin-B tertiles and both increasing fasting insulin concentrations (mean [± SD] 11.9 ± 6.8, 12.7 ± 7.6, and 13.3 ± 6.4 mIU/L in Tertiles 1, 2 and 3, respectively; P = 0.006) and prevalence of insulin resistance (54.4%, 58.9%, and 64.5% in Tertiles 1, 2 and 3, respectively; P = 0.010). In linear regression analysis, IHTG content was independently and positively correlated with serum fetuin-B (regression coefficient 0.015; P = 0.045). With adjustment for potential confounders, serum fetuin-B was independently associated with increased risk of insulin resistance, with an adjusted odds ratio per standard deviation increase in fetuin-B of 1.14 (95% confidence interval 1.01-1.30; P = 0.031). CONCLUSIONS: The results demonstrate the role of fetuin-B linking liver fat accumulation to insulin resistance in humans.


Assuntos
Biomarcadores/sangue , Fetuína-B/análise , Intolerância à Glucose/diagnóstico , Resistência à Insulina , Fígado/metabolismo , Obesidade/fisiopatologia , Triglicerídeos/metabolismo , Adulto , China/epidemiologia , Estudos Transversais , Feminino , Seguimentos , Intolerância à Glucose/sangue , Intolerância à Glucose/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/sangue , Prognóstico , Fatores de Risco
20.
Adv Exp Med Biol ; 1043: 385-399, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224104

RESUMO

In rodent models of insulin-deficient diabetes, 17ß-estradiol (E2) protects pancreatic insulin-producing ß-cells against oxidative stress, amyloid polypeptide toxicity, gluco-lipotoxicity, and apoptosis. Three estrogen receptors (ERs)-ERα, ERß, and the G protein-coupled ER (GPER)-have been identified in rodent and human ß-cells. This chapter describes recent advances in our understanding of the role of ERs in islet ß-cell function, nutrient homeostasis, survival from pro-apoptotic stimuli, and proliferation. We discuss why and how ERs represent potential therapeutic targets for the maintenance of functional ß-cell mass.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Terapia de Reposição de Estrogênios , Estrogênios/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Apoptose , Glicemia/metabolismo , Proliferação de Células , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 1/terapia , Estradiol/efeitos adversos , Terapia de Reposição de Estrogênios/efeitos adversos , Estrogênios/efeitos adversos , Humanos , Hipoglicemiantes/uso terapêutico , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Ilhotas Pancreáticas/cirurgia , Transplante das Ilhotas Pancreáticas , Ligantes , Camundongos Endogâmicos NOD , Estado Nutricional , Receptores de Estrogênio/efeitos dos fármacos , Fatores de Risco , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA