Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Ther ; 32(6): 1760-1778, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38659223

ABSTRACT

Glaucoma is characterized by the progressive degeneration of retinal ganglion cells (RGCs) and their axons, and its risk increases with aging. Yet comprehensive insights into the complex mechanisms are largely unknown. Here, we found that anti-aging molecule Sirt6 was highly expressed in RGCs. Deleting Sirt6 globally or specifically in RGCs led to progressive RGC loss and optic nerve degeneration during aging, despite normal intraocular pressure (IOP), resembling a phenotype of normal-tension glaucoma. These detrimental effects were potentially mediated by accelerated RGC senescence through Caveolin-1 upregulation and by the induction of mitochondrial dysfunction. In mouse models of high-tension glaucoma, Sirt6 level was decreased after IOP elevation. Genetic overexpression of Sirt6 globally or specifically in RGCs significantly attenuated high tension-induced degeneration of RGCs and their axons, whereas partial or RGC-specific Sirt6 deletion accelerated RGC loss. Importantly, therapeutically targeting Sirt6 with pharmacological activator or AAV2-mediated gene delivery ameliorated high IOP-induced RGC degeneration. Together, our studies reveal a critical role of Sirt6 in preventing RGC and optic nerve degeneration during aging and glaucoma, setting the stage for further exploration of Sirt6 activation as a potential therapy for glaucoma.


Subject(s)
Aging , Disease Models, Animal , Glaucoma , Optic Nerve , Retinal Ganglion Cells , Sirtuins , Animals , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Mice , Sirtuins/metabolism , Sirtuins/genetics , Glaucoma/metabolism , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/etiology , Optic Nerve/metabolism , Optic Nerve/pathology , Aging/metabolism , Aging/genetics , Intraocular Pressure , Humans , Axons/metabolism , Axons/pathology , Mice, Knockout , Nerve Degeneration/metabolism
2.
J Biol Chem ; 298(1): 101431, 2022 01.
Article in English | MEDLINE | ID: mdl-34801552

ABSTRACT

Insulin resistance impairs postprandial glucose uptake through glucose transporter type 4 (GLUT4) and is the primary defect preceding type 2 diabetes. We previously generated an insulin-resistant mouse model with human GLUT4 promoter-driven insulin receptor knockout (GIRKO) in the muscle, adipose, and neuronal subpopulations. However, the rate of diabetes in GIRKO mice remained low prior to 6 months of age on normal chow diet (NCD), suggesting that additional factors/mechanisms are responsible for adverse metabolic effects driving the ultimate progression of overt diabetes. In this study, we characterized the metabolic phenotypes of the adult GIRKO mice acutely switched to high-fat diet (HFD) feeding in order to identify additional metabolic challenges required for disease progression. Distinct from other diet-induced obesity (DIO) and genetic models (e.g., db/db mice), GIRKO mice remained leaner on HFD feeding, but developed other cardinal features of insulin resistance syndrome. GIRKO mice rapidly developed hyperglycemia despite compensatory increases in ß-cell mass and hyperinsulinemia. Furthermore, GIRKO mice also had impaired oral glucose tolerance and a limited glucose-lowering benefit from exendin-4, suggesting that the blunted incretin effect contributed to hyperglycemia. Secondly, GIRKO mice manifested severe dyslipidemia while on HFD due to elevated hepatic lipid secretion, serum triglyceride concentration, and lipid droplet accumulation in hepatocytes. Thirdly, GIRKO mice on HFD had increased inflammatory cues in the gut, which were associated with the HFD-induced microbiome alterations and increased serum lipopolysaccharide (LPS). In conclusion, our studies identified important gene/diet interactions contributing to diabetes progression, which might be leveraged to develop more efficacious therapies.


Subject(s)
Diabetes Mellitus, Type 2 , Diet, High-Fat , Glucose Intolerance , Glucose Transporter Type 4 , Hyperglycemia , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/metabolism , Hyperglycemia/blood , Hyperglycemia/etiology , Hyperglycemia/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
BMC Cancer ; 21(1): 1228, 2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34781924

ABSTRACT

BACKGROUND: Competitive Endogenous RNA (ceRNA) may be closely associated with tumor progression. However, studies on ceRNAs and immune cells in LUAD are scarce. METHOD: The profiles of gene expression and clinical data of LUAD patients were extracted from the TCGA database. Bioinformatics methods were used to evaluate differentially-expressed genes (DEGs) and to form a ceRNA network. Preliminary verification of clinical specimens was utilized to detect the expressions of key biomarkers at the tissues. Cox and Lasso regressions were used to identify key genes, and prognosis prediction nomograms were formed. The mRNA levels of 9 genes in the risk score model in independent clinical LUAD samples were detected by qRT-PCR. The interconnection between the risk of cancer and immune cells was evaluated using the CIBERSORT algorithm, while the conformation of notable tumor-infiltrating immune cells (TIICs) in the LUAD tissues of the high and low risk groups was assessed using the RNA transcript subgroup in order to identify tissue types. Finally, co-expression study was used to examine the interconnection between the key genes in the ceRNA networks and the immune cells. RESULT: A ceRNA network of 115 RNAs was established, and nine key genes were identified to construct a Cox proportional-hazard model and create a prognostic nomogram. This risk-assessment model might serve as an independent factor to forecast the prognosis of LUAD, and it was consistent with the preliminary verification of clinical specimens. Survival analysis of clinical samples further validated the potential value of high risk groups in predicting LUAD prognosis. Five immune cells were identified with significant differences in the LUAD tissues of the high and low risk groups. Besides, two pairs of biomarkers associated with the growth of LUAD were found, i.e., E2F7 and macrophage M1 (R = 0.419, p = 1.4e- 08) and DBF4 and macrophage M1 (R = 0.282, p < 2.2 e- 16). CONCLUSION: This study identified several important ceRNAs, i.e. (E2F7 and BNF4) and TIICs (macrophage M1), which might be related to the development and prognosis of LUAD. The established risk-assessment model might be a potential tool in predicting LUAD of prognosis.


Subject(s)
Adenocarcinoma of Lung/genetics , Gene Expression , Gene Regulatory Networks , Lung Neoplasms/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/mortality , Algorithms , Disease Progression , Humans , Immunity, Cellular , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating , MicroRNAs , Nomograms , Regression Analysis , Risk Assessment , Survival Analysis
4.
Photodiagnosis Photodyn Ther ; 36: 102597, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34699982

ABSTRACT

Photodynamic therapy (PDT) utilizes photosensitizers (PSs) together with irradiation light of specific wavelength interacting with oxygen to generate cytotoxic reactive oxygen species (ROS), which could trigger apoptosis and/or necrosis-induced cell death in target tissues. During the past two decades, multifunctional nano-PSs employing nanotechnology and nanomedicine developed, which present not only photosensitizing properties but additionally accurate drug release abilities, efficient response to optical stimuli and hypoxia resistance. Further, nano-PSs have been developed to enhance PDT efficacy by improving the ROS yield. In addition, nano-PSs with additive or synergistic therapies are significant for both currently preclinical study and future clinical practice, given their capability of considerable higher therapeutic efficacy under safer systemic drug dosage. In this review, nano-PSs that allow precise drug delivery for efficient absorption by target cells are introduced. Nano-PSs boosting sensitivity and conversion efficiency to PDT-activating stimuli are highlighted. Nano-PSs developed to address the challenging hypoxia conditions during PDT of deep-sited tumors are summarized. Specifically, PSs capable of synergistic therapy and the emerging novel types with higher ROS yield that further enhance PDT efficacy are presented. Finally, future demands for ideal nano-PSs, emphasizing clinical translation and application are discussed.


Subject(s)
Antineoplastic Agents , Neoplasms , Photochemotherapy , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Humans , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species
5.
Cell Death Dis ; 12(7): 710, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267188

ABSTRACT

Alcohol-related liver disease (ALD), a condition caused by alcohol overconsumption, occurs in three stages of liver injury including steatosis, hepatitis, and cirrhosis. DEP domain-containing protein 5 (DEPDC5), a component of GAP activities towards Rags 1 (GATOR1) complex, is a repressor of amino acid-sensing branch of the mammalian target of rapamycin complex 1 (mTORC1) pathway. In the current study, we found that aberrant activation of mTORC1 was likely attributed to the reduction of DEPDC5 in the livers of ethanol-fed mice or ALD patients. To further define the in vivo role of DEPDC5 in ALD development, we generated Depdc5 hepatocyte-specific knockout mouse model (Depdc5-LKO) in which mTORC1 pathway was constitutively activated through loss of the inhibitory effect of GATOR1. Hepatic Depdc5 ablation leads to mild hepatomegaly and liver injury and protects against diet-induced liver steatosis. In contrast, ethanol-fed Depdc5-LKO mice developed severe hepatic steatosis and inflammation. Pharmacological intervention with Torin 1 suppressed mTORC1 activity and remarkably ameliorated ethanol-induced hepatic steatosis and inflammation in both control and Depdc5-LKO mice. The pathological effect of sustained mTORC1 activity in ALD may be attributed to the suppression of peroxisome proliferator activated receptor α (PPARα), the master regulator of fatty acid oxidation in hepatocytes, because fenofibrate (PPARα agonist) treatment reverses ethanol-induced liver steatosis and inflammation in Depdc5-LKO mice. These findings provide novel insights into the in vivo role of hepatic DEPDC5 in the development of ALD.


Subject(s)
Fatty Liver, Alcoholic/metabolism , GTPase-Activating Proteins/deficiency , Liver/metabolism , PPAR alpha/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/pathology , Fatty Liver, Alcoholic/prevention & control , Female , GTPase-Activating Proteins/genetics , Inflammation Mediators , Liver/drug effects , Liver/ultrastructure , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , Naphthyridines/pharmacology , Oxidation-Reduction , Oxidative Stress , PPAR alpha/genetics , Signal Transduction
6.
Antioxid Redox Signal ; 35(9): 689-717, 2021 09 20.
Article in English | MEDLINE | ID: mdl-33906425

ABSTRACT

Significance: Fatty liver disease is a major liver disorder in the modern societies. Comprehensive understanding of the pathophysiology and molecular mechanisms is essential for the prevention and treatment of the disease. Recent Advances: Remarkable progress has been made in the recent years in basic and translational research in the field of fatty liver disease. Multiple signaling pathways have been implicated in the development of fatty liver disease, including AMP-activated protein kinase, mechanistic target of rapamycin kinase, endoplasmic reticulum stress, oxidative stress, inflammation, transforming growth factor ß, and yes1-associated transcriptional regulator/transcriptional coactivator with PDZ-binding motif (YAP/TAZ). In addition, critical molecular regulations at the transcriptional and epigenetic levels have been linked to the pathogenesis of fatty liver disease. Critical Issues: Some critical issues remain to be solved so that research findings can be translated into clinical applications. Robust and reliable biomarkers are needed for diagnosis of different stages of the fatty liver disease. Effective and safe molecular targets remain to be identified and validated. Prevention strategies require solid scientific evidence and population-wide feasibility. Future Directions: As more data are generated with time, integrative approaches are needed to comprehensively understand the disease pathophysiology and mechanisms at multiple levels from population, organismal system, organ/tissue, to cell. The interactions between genes and environmental factors require deeper investigation for the purposes of prevention and personalized treatment of fatty liver disease. Antioxid. Redox Signal. 35, 689-717.


Subject(s)
Signal Transduction , Transcription Factors , Endoplasmic Reticulum Stress , Liver/metabolism , Oxidation-Reduction , Oxidative Stress , Transcription Factors/metabolism
7.
J Hepatol ; 73(2): 263-276, 2020 08.
Article in English | MEDLINE | ID: mdl-32165250

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic fatty liver disease (NAFLD), type 2 diabetes (T2D) and obesity are epidemiologically correlated with each other but the causal inter-relationships between them remain incompletely understood. We aimed to explore the causal relationships between the 3 diseases. METHODS: Using both UK Biobank and publicly available genome-wide association study data, we performed a 2-sample bidirectional Mendelian randomization analysis to test the causal inter-relationships between NAFLD, T2D, and obesity. Transgenic mice expressing the human PNPLA3-I148M isoforms (TghPNPLA3-I148M) were used as an example to validate causal effects and explore underlying mechanisms. RESULTS: Genetically driven NAFLD significantly increased the risk of T2D and central obesity but not insulin resistance or generalized obesity, while genetically driven T2D, body mass index and WHRadjBMI causally increased NAFLD risk. The animal study focusing on PNPLA3 corroborated these causal effects: compared to the TghPNPLA3-I148I controls, the TghPNPLA3-I148M mice developed glucose intolerance and increased visceral fat, but maintained normal insulin sensitivity, reduced body weight, and decreased circulating total cholesterol. Mechanistically, the TghPNPLA3-I148M mice demonstrated decreased pancreatic insulin but increased glucagon secretion, which was associated with increased pancreatic inflammation. In addition, transcription of hepatic cholesterol biosynthesis pathway genes was significantly suppressed, while transcription of thermogenic pathway genes was activated in subcutaneous and brown adipose tissues but not in visceral fat in TghPNPLA3-I148M mice. CONCLUSIONS: Our study suggests that lifelong, genetically driven NAFLD causally promotes T2D with a late-onset type 1-like diabetic subphenotype and central obesity; while genetically driven T2D, obesity, and central obesity all causally increase the risk of NAFLD. This causal relationship revealed new insights into how nature and nurture drive these diseases, providing novel hypotheses for disease subphenotyping. LAY SUMMARY: Non-alcoholic fatty liver disease, type 2 diabetes and obesity are epidemiologically correlated with each other, but their causal relationships were incompletely understood. Herein, we identified causal relationships between these conditions, which suggest that each of these closely related diseases should be further stratified into subtypes. This is important for accurate diagnosis, prevention and treatment of these diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Lipase/genetics , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease , Obesity, Abdominal , Animals , Causality , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Europe/epidemiology , Founder Effect , Genome-Wide Association Study , Humans , Mendelian Randomization Analysis , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/genetics , Obesity, Abdominal/epidemiology , Obesity, Abdominal/genetics , Protein Isoforms
8.
J Hepatol ; 71(5): 960-969, 2019 11.
Article in English | MEDLINE | ID: mdl-31295533

ABSTRACT

BACKGROUND & AIMS: As a nicotinamide adenine dinucleotide-dependent deacetylase and a key epigenetic regulator, sirtuin 6 (SIRT6) has been implicated in the regulation of metabolism, DNA repair, and inflammation. However, the role of SIRT6 in alcohol-related liver disease (ALD) remains unclear. The aim of this study was to investigate the function and mechanism of SIRT6 in ALD pathogenesis. METHODS: We developed and characterized Sirt6 knockout (KO) and transgenic mouse models that were treated with either control or ethanol diet. Hepatic steatosis, inflammation, and oxidative stress were analyzed using biochemical and histological methods. Gene regulation was analyzed by luciferase reporter and chromatin immunoprecipitation assays. RESULTS: The Sirt6 KO mice developed severe liver injury characterized by a remarkable increase of oxidative stress and inflammation, whereas the Sirt6 transgenic mice were protected from ALD via normalization of hepatic lipids, inflammatory response, and oxidative stress. Our molecular analysis has identified a number of novel Sirt6-regulated genes that are involved in antioxidative stress, including metallothionein 1 and 2 (Mt1 and Mt2). Mt1/2 genes were downregulated in the livers of Sirt6 KO mice and patients with alcoholic hepatitis. Overexpression of Mt1 in the liver of Sirt6 KO mice improved ALD by reducing hepatic oxidative stress and inflammation. We also identified a critical link between SIRT6 and metal regulatory transcription factor 1 (Mtf1) via a physical interaction and functional coactivation. Mt1/2 promoter reporter assays showed a strong synergistic effect of SIRT6 on the transcriptional activity of Mtf1. CONCLUSIONS: Our data suggest that SIRT6 plays a critical protective role against ALD and it may serve as a potential therapeutic target for ALD. LAY SUMMARY: The liver, the primary organ for ethanol metabolism, can be damaged by the byproducts of ethanol metabolism, including reactive oxygen species. In this study, we have identified a key epigenetic regulator SIRT6 that plays a critical role in protecting the liver from oxidative stress-induced liver injury. Thus, our data suggest that SIRT6 may be a potential therapeutic target for alcohol-related liver disease.


Subject(s)
Epigenesis, Genetic/genetics , Ethanol/metabolism , Liver Diseases, Alcoholic/metabolism , Oxidative Stress/genetics , Sirtuins/genetics , Sirtuins/metabolism , Adult , Animals , Disease Models, Animal , Down-Regulation/genetics , Ethanol/adverse effects , Fatty Liver/metabolism , Female , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Knockout , Middle Aged , Reactive Oxygen Species/metabolism
9.
Biochim Biophys Acta Mol Basis Dis ; 1865(10): 2685-2693, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31351129

ABSTRACT

Sestrin 3 (Sesn3) belongs to a small protein family that has been implicated in multiple biological processes including anti-oxidative stress, anti-aging, cell signaling, and metabolic homeostasis. However, the role of Sesn3 in hepatocellular carcinoma (HCC) remains unclear. Here we generated a Sesn3 knockout mouse model and induced HCC development by a combination of a single dose of diethylnitrosamine and chronic feeding of a choline deficient-high fat diet. After 6 months of the dietary treatment, Sesn3 knockout mice developed more severe HCC with higher levels of alpha-fetoprotein, arginase 1, and cytokeratin 19, but also higher metastatic rates than wild-type mice. Histological analysis revealed elevated extracellular matrix and cancer stem cell markers including Acta2, Cd44, and Cd133. Signaling analysis showed activated IL6-Stat3 and Akt pathways. Biochemical and microscopic analyses uncovered a novel inhibitory regulation of Gli2, a downstream transcription factor of the hedgehog signaling, by Sesn3. Two of the Gli2-regulated genes - Pdgfrb and Cd44 were upregulated in the Sesn3-deficient liver tissue. In conclusion, our data suggest that Sesn3 plays a critical tumor suppressor role in the liver partly through the inhibition of the hedgehog signaling.


Subject(s)
Carcinogens/metabolism , Carcinoma, Hepatocellular/metabolism , Genetic Predisposition to Disease/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hedgehog Proteins/metabolism , Liver Neoplasms/metabolism , AC133 Antigen/metabolism , Actins/metabolism , Animals , Arginase/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Interleukin-6/metabolism , Keratin-19/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation , Zinc Finger Protein Gli2/metabolism , alpha-Fetoproteins/metabolism
10.
Front Med (Lausanne) ; 6: 304, 2019.
Article in English | MEDLINE | ID: mdl-31921875

ABSTRACT

Patatin-like phospholipase domain-containing protein 3 (PNPLA3) is a lipid droplet-associated protein that has been shown to have hydrolase activity toward triglycerides and retinyl esters. The first evidence of PNPLA3 being associated with fatty liver disease was revealed by a genome-wide association study (GWAS) of Hispanic, African American, and European American individuals in the Dallas Heart Study back in 2008. Since then, numerous GWAS reports have shown that PNPLA3 rs738409[G] (148M) variant is associated with hepatic triglyceride accumulation (steatosis), inflammation, fibrosis, cirrhosis, and even hepatocellular carcinoma regardless of etiologies including alcohol- or obesity-related and others. The frequency of PNPLA3(148M) variant ranges from 17% in African Americans, 23% in European Americans, to 49% in Hispanics in the Dallas Heart Study. Due to high prevalence of obesity and alcohol consumption in modern societies, the PNPLA3(148M) gene variant and environment interaction poses a serious concern for public health, especially chronic liver diseases including alcohol-related liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD). Therefore, PNPLA3(148M) variant is a potential therapeutic target for chronic liver disease in the rs738409 allele carriers. Currently, there is no approved drug specifically targeting the PNPLA3(148M) variant yet. With additional mechanistic studies, novel therapeutic strategies are expected to be developed for the treatment of the PNPLA3(148M) variant-associated chronic liver diseases in the near future.

13.
Onco Targets Ther ; 9: 1241-9, 2016.
Article in English | MEDLINE | ID: mdl-27022282

ABSTRACT

OBJECTIVE: We conducted a systematic review and meta-analysis aiming to assess the relationship between apolipoprotein E (APOE) gene ε2/ε3/ε4 polymorphism and breast cancer risk. METHODS: Yun-Long Liu and Hao-Min Zhang independently completed literature retrieval and data collection, and statistical analyses were performed by Stata. Individual odds ratio (OR) and 95% confidence interval (CI) were pooled in a random-effects model using the DerSimonian-Laird method. Heterogeneity was evaluated by I (2) statistic at a significance level of 50%. Publication bias was assessed by Egger's test. RESULTS: Eleven articles including 2,074 breast cancer patients and 2,372 controls were summarized. Using the most common allele ε3 as a reference, the ε2 (OR =0.87, 95% CI =0.72-1.05, P=0.154, I (2)=0.0%) and ε4 (OR =1.07, 95% CI =0.80-1.42, P=0.654, I (2)=71.8%) alleles were not found to be significantly associated with breast cancer risk in the overall analyses. Subgroup analyses revealed that the comparison of allele ε4 with ε3 was significant in Asians (OR =1.58, 95% CI =1.17-6.32, P=0.003, I (2)=12.1%) and in studies that used the restriction fragment length polymorphism (RFLP) genotyping method (OR =1.27; 95% CI =1.01-1.61, P=0.045, I (2)=34.3%), and was marginally significant in hospital-based studies (OR =1.33; 95% CI =0.98-1.79, P=0.065, I (2)=30.2%), without heterogeneity. Moreover, the presence of the ε2 allele was significantly associated with breast cancer in small studies (total sample size <500) (OR =0.73, 95% CI =0.54-1.00, P=0.052, I (2)=0.0%) without heterogeneity. The Egger's test indicated low probabilities of publication bias. CONCLUSION: We observed a significant association between APOE gene ε4 allele and breast cancer risk in Asian populations. Moreover, the findings of our subgroup analyses suggest that source of controls, genotyping platform, and sample size might be the potential causes of heterogeneity.

14.
Expert Opin Ther Targets ; 19(8): 1011-5, 2015.
Article in English | MEDLINE | ID: mdl-25944222

ABSTRACT

Sestrins (Sesn1/2/3) belong to a small protein family that has versatile biological functions. In addition to initially characterized oxidoreductase activity, sestrins also have oxidoreductase-independent functions, including activation of AMP-activated protein kinase, inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) and activation of mTORC2. As these kinases are important for metabolic regulation, sestrins have a favorable profile as potential therapeutic targets for metabolic diseases such as diabetes. Recent data are in line with such a notion. In this editorial, I have attempted to provide a brief update on the major findings in regard to sestrins in metabolism.


Subject(s)
Diabetes Mellitus/drug therapy , Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Diabetes Mellitus/physiopathology , Humans , Molecular Targeted Therapy
15.
J Clin Invest ; 123(12): 5319-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24177427

ABSTRACT

The induction of autophagy in the mammalian heart during the perinatal period is an essential adaptation required to survive early neonatal starvation; however, the mechanisms that mediate autophagy suppression once feeding is established are not known. Insulin signaling in the heart is transduced via insulin and IGF-1 receptors (IGF-1Rs). We disrupted insulin and IGF-1R signaling by generating mice with combined cardiomyocyte-specific deletion of Irs1 and Irs2. Here we show that loss of IRS signaling prevented the physiological suppression of autophagy that normally parallels the postnatal increase in circulating insulin. This resulted in unrestrained autophagy in cardiomyocytes, which contributed to myocyte loss, heart failure, and premature death. This process was ameliorated either by activation of mTOR with aa supplementation or by genetic suppression of autophagic activation. Loss of IRS1 and IRS2 signaling also increased apoptosis and precipitated mitochondrial dysfunction, which were not reduced when autophagic flux was normalized. Together, these data indicate that in addition to prosurvival signaling, insulin action in early life mediates the physiological postnatal suppression of autophagy, thereby linking nutrient sensing to postnatal cardiac development.


Subject(s)
Autophagy , Heart/growth & development , Insulin Receptor Substrate Proteins/physiology , Myocytes, Cardiac/metabolism , Amino Acids/pharmacology , Animals , Apoptosis , Apoptosis Regulatory Proteins/deficiency , Autophagy/genetics , Autophagy/physiology , Beclin-1 , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Fetal Heart/pathology , Heart Failure/etiology , Heart Failure/pathology , Insulin/physiology , Insulin Receptor Substrate Proteins/deficiency , Insulin-Like Growth Factor I/physiology , Mice , Mitochondria, Heart/physiology , Oxidative Phosphorylation , Phosphorylation , Protein Processing, Post-Translational , Receptor, IGF Type 1/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology
16.
PLoS One ; 8(8): e71997, 2013.
Article in English | MEDLINE | ID: mdl-23940800

ABSTRACT

Pyruvate dehydrogenase kinases (PDK1-4) play a critical role in the inhibition of the mitochondrial pyruvate dehydrogenase complex especially when blood glucose levels are low and pyruvate can be conserved for gluconeogenesis. Under diabetic conditions, the Pdk genes, particularly Pdk4, are often induced, and the elevation of the Pdk4 gene expression has been implicated in the increased gluconeogenesis in the liver and the decreased glucose utilization in the peripheral tissues. However, there is no direct evidence yet to show to what extent that the dysregulation of hepatic Pdk genes attributes to hyperglycemia and insulin resistance in vivo. To address this question, we crossed Pdk2 or Pdk4 null mice with a diabetic model that is deficient in hepatic insulin receptor substrates 1 and 2 (Irs1/2). Metabolic analyses reveal that deletion of the Pdk4 gene had better improvement in hyperglycemia and glucose tolerance than knockout of the Pdk2 gene whereas the Pdk2 gene deletion showed better insulin tolerance as compared to the Pdk4 gene inactivation on the Irs1/2 knockout genetic background. To examine the specific hepatic effects of Pdks on diabetes, we also knocked down the Pdk2 or Pdk4 gene using specific shRNAs. The data also indicate that the Pdk4 gene knockdown led to better glucose tolerance than the Pdk2 gene knockdown. In conclusion, our data suggest that hepatic Pdk4 may be critically involved in the pathogenesis of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Insulin Resistance/genetics , Liver/metabolism , Protein Serine-Threonine Kinases/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Gene Expression Regulation, Enzymologic , Gene Silencing , Glucose Intolerance/genetics , Glucose Tolerance Test , Insulin Receptor Substrate Proteins/genetics , Mice , Mice, Knockout , Organ Specificity/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
17.
J Hepatol ; 58(5): 993-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23339953

ABSTRACT

BACKGROUND & AIMS: Pharmacological approaches can potentially improve fatty liver condition in alcoholic and non-alcoholic fatty liver diseases. The salutary effects of reducing lipid synthesis or promoting lipid oxidation have been well reported, but the benefits of increasing lipid degradation have yet to be well explored. Macroautophagy is a cellular degradation process that can remove subcellular organelles including lipid droplets. We thus investigated whether pharmacological modulation of macroautophagy could be an effective approach to alleviate fatty liver condition and liver injury. METHODS: C57BL/6 mice were given ethanol via intraperitoneal injection (acute) or by a 4-week oral feeding regime (chronic), or high fat diet for 12 weeks. An autophagy enhancer, carbamazepine or rapamycin, or an autophagy inhibitor, chloroquine, was given before sacrifice. Activation of autophagy, level of hepatic steatosis, and blood levels of triglycerides, liver enzyme, glucose and insulin were measured. RESULTS: In both acute and chronic ethanol condition, macroautophagy was activated. Carbamazepine, as well as rapamycin, enhanced ethanol-induced macroautophagy in hepatocytes in vitro and in vivo. Hepatic steatosis and liver injury were exacerbated by chloroquine, but alleviated by carbamazepine. The protective effects of carbamazepine and rapamycin in reducing steatosis and in improving insulin sensitivity were also demonstrated in high fat diet-induced non-alcoholic fatty liver condition. CONCLUSIONS: These findings indicate that pharmacological modulation of macroautophagy in the liver can be an effective strategy for reducing fatty liver condition and liver injury.


Subject(s)
Autophagy/drug effects , Carbamazepine/pharmacology , Fatty Liver, Alcoholic/prevention & control , Fatty Liver/prevention & control , Sirolimus/pharmacology , Animals , Autophagy/physiology , Biomarkers/metabolism , Carbamazepine/therapeutic use , Cells, Cultured , Chloroquine/pharmacology , Dietary Fats/adverse effects , Disease Models, Animal , Ethanol/adverse effects , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , In Vitro Techniques , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease , Sirolimus/therapeutic use
18.
J Am Chem Soc ; 134(43): 18116-24, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23075115

ABSTRACT

Protein tyrosine phosphatases (PTPs) constitute a large family of signaling enzymes that control the cellular levels of protein tyrosine phosphorylation. A detailed understanding of PTP functions in normal physiology and in pathogenic conditions has been hampered by the absence of PTP-specific, cell-permeable small-molecule agents. We present a stepwise focused library approach that transforms a weak and general non-hydrolyzable pTyr mimetic (F(2)Pmp, phosphonodifluoromethyl phenylalanine) into a highly potent and selective inhibitor of PTP-MEG2, an antagonist of hepatic insulin signaling. The crystal structures of the PTP-MEG2-inhibitor complexes provide direct evidence that potent and selective PTP inhibitors can be obtained by introducing molecular diversity into the F(2)Pmp scaffold to engage both the active site and unique nearby peripheral binding pockets. Importantly, the PTP-MEG2 inhibitor possesses highly efficacious cellular activity and is capable of augmenting insulin signaling and improving insulin sensitivity and glucose homeostasis in diet-induced obese mice. The results indicate that F(2)Pmp can be converted into highly potent and selective PTP inhibitory agents with excellent in vivo efficacy. Given the general nature of the approach, this strategy should be applicable to other members of the PTP superfamily.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Phenylalanine/analogs & derivatives , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Molecular , Molecular Structure , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Phenylalanine/pharmacology , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
19.
J Clin Invest ; 121(10): 4070-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926467

ABSTRACT

Aging is a major risk factor for the progression of neurodegenerative diseases, including Huntington disease (HD). Reduced neuronal IGF1 or Irs2 signaling have been shown to extend life span in mice. To determine whether Irs2 signaling modulates neurodegeneration in HD, we genetically modulated Irs2 concentrations in the R6/2 mouse model of HD. Increasing Irs2 levels in the brains of R6/2 mice significantly reduced life span and increased neuronal oxidative stress and mitochondrial dysfunction. In contrast, reducing Irs2 levels throughout the body (except in ß cells, where Irs2 expression is needed to prevent diabetes onset; R6/2•Irs2+/-•Irs2ßtg mice) improved motor performance and extended life span. The slower progression of HD-like symptoms was associated with increased nuclear localization of the transcription factor FoxO1 and increased expression of FoxO1-dependent genes that promote autophagy, mitochondrial function, and resistance to oxidative stress. Mitochondrial function improved and the number of autophagosomes increased in R6/2•Irs2+/-•Irs2ßtg mice, whereas aggregate formation and oxidative stress decreased. Thus, our study suggests that Irs2 signaling can modulate HD progression. Since we found the expression of Irs2 to be normal in grade II HD patients, our results suggest that decreasing IRS2 signaling could be part of a therapeutic approach to slow the progression of HD.


Subject(s)
Huntington Disease/physiopathology , Insulin Receptor Substrate Proteins/physiology , Mitochondria/physiology , Aging/genetics , Aging/physiology , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Disease Progression , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Insulin Receptor Substrate Proteins/deficiency , Insulin Receptor Substrate Proteins/genetics , Longevity/genetics , Longevity/physiology , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Oxidative Stress , Signal Transduction
20.
Int J Urol ; 18(2): 126-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21198943

ABSTRACT

OBJECTIVES: Treatment of staghorn calculus is challenging. We evaluated the feasibility and efficacy of the retroperitoneal laparoscopic approach for the management of large staghorn renal calculi. METHODS: Patients with staghorn renal calculi unsuitable for percutaneous nephrolithotomy were analyzed. They underwent retroperitoneal laparoscopic anatrophic nephrolithotomy, involving control of the renal artery, stone removal through a nephrotomy incision on the Brodel's line and closure with continuous sutures. RESULTS: A total of 11 patients with renal stones were included in the present study. Mean patient age was 55 years (range 42-68) and stone size was 52 mm (range 43-61). Warm ischemia time and operative duration were 31 (range 23-38) and 139 min (range 105-160), respectively. No blood transfusion was needed during or after operation. An 8-mm residual calculus remained in the lower calyces in one patient who was successfully treated by using shock wave lithotripsy. Intravenous pyelogram after surgery showed a functional corresponding renal unit, with an improvement in obstruction in all patients. CONCLUSIONS: Retroperitoneal laparoscopic technique can be applied for patients who are candidates for anatrophic nephrolithotomy. Larger studies with a longer follow up are needed to confirm these findings.


Subject(s)
Kidney Calculi/surgery , Laparoscopy/methods , Nephrostomy, Percutaneous , Adult , Aged , Feasibility Studies , Humans , Middle Aged , Retroperitoneal Space/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...