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.
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
5.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
J Clin Invest ; 116(1): 101-14, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16374520

ABSTRACT

Insulin receptor substrates, including Irs1 and Irs2, integrate insulin and IGF receptor signals with heterologous pathways to coordinate growth and metabolism. Since Irs2 is thought to be especially important in hepatic nutrient homeostasis, we deleted Irs2 [corrected] from hepatocytes of WT mice (called LKO) or genetically insulin-resistant Irs1-/- mice (called LKO::Irs1-/-). Viable LKO::Irs1-/- mice were 70% smaller than WT or LKO mice, and 40% smaller than Irs1-/- mice. Hepatic insulin receptors were functional in all the mice, but insulin signaling via the Akt-FoxO1 pathway was reduced in Irs1-/- and LKO liver, and undetected in LKO::Irs1-/- liver; however, Gsk3beta phosphorylation (Ser9) and hepatic glycogen stores were nearly normal in all of the mice. LKO and Irs1-/- mice developed insulin resistance and glucose intolerance that never progressed to diabetes, whereas LKO::Irs1-/- mice developed hyperglycemia and hyperinsulinemia immediately after birth. Regardless, few hepatic genes changed expression significantly in Irs1-/- or LKO mice, whereas hundreds of genes changed in LKO::Irs1-/- mice--including elevated levels of Pck1, G6pc, Ppargc1, Pparg, and Igfbp1. Thus, signals delivered by Irs1 or Irs2 regulate hepatic gene expression that coordinates glucose homeostasis and systemic growth.


Subject(s)
Glucose/metabolism , Liver/physiology , Phosphoproteins/physiology , Animals , Apoptosis , Body Weight , Bone Density , Homeostasis , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Liver/cytology , Mice , Mice, Knockout , Organ Size , Pancreas/physiology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Receptor, Insulin/physiology , Signal Transduction
15.
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.

16.
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
17.
Mol Endocrinol ; 21(9): 2294-302, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17579213

ABSTRACT

Multisite phosphorylation of Irs1 on serine and threonine residues regulates insulin signaling that can contribute to insulin resistance. We identified by mass spectrometry the phosphorylation of Ser522 in rat Irs1 (S522(Irs1)). The functional effects of this phosphorylation site were investigated in cultured cells using a sequence-specific phosphoserine antibody. Insulin stimulated the phosphorylation of S522(Irs1) in L6 myoblasts and myotubes. S522(Irs1) phosphorylation was inhibited by wortmannin, whereas PD98059, rapamycin, or glucose-starvation had no effect. Reducing Akt expression with small interfering RNA inhibited insulin-stimulated phosphorylation of S522(Irs1), suggesting the involvement of the phosphatidylinositol 3-kinase--> Akt cascade. A S522(Irs1)-->A522(Irs1) substitution increased insulin-stimulated tyrosine phosphorylation of Irs1 and signaling, whereas a S522(Irs1)-->E522(Irs1) substitution reduced insulin-stimulated Irs1 tyrosine phosphorylation. Together, these results suggest the phosphatidylinositol 3-kinase-->Akt cascade can inhibit insulin signaling through the phosphorylation of S522(Irs1).


Subject(s)
Insulin/metabolism , Phosphoproteins/metabolism , Serine/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Insulin Receptor Substrate Proteins , Phosphoproteins/genetics , Phosphorylation , Rabbits , Serine/genetics , Threonine/genetics , Threonine/metabolism
18.
Mol Cell Biol ; 22(23): 8302-19, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12417732

ABSTRACT

Inhibitors of DNA methyltransferase (Dnmt) and histone deacetylases (HDAC) synergistically activate the methylated metallothionein I gene (MT-I) promoter in mouse lymphosarcoma cells. The cooperative effect of these two classes of inhibitors on MT-I promoter activity was robust following demethylation of only a few CpG dinucleotides by brief exposure to 5-azacytidine (5-AzaC) but persisted even after prolonged treatment with the nucleoside analog. HDAC inhibitors (trichostatin A [TSA] and depsipeptide) either alone or in combination with 5-AzaC did not facilitate demethylation of the MT-I promoter. Treatment of cells with HDAC inhibitors increased accumulation of multiply acetylated forms of H3 and H4 histones that remained unaffected after treatment with 5-AzaC. Chromatin immunoprecipitation (ChIP) assay showed increased association of acetylated histone H4 and lysine 9 (K9)-acetyl H3 with the MT-I promoter after treatment with TSA, which was not affected following treatment with 5-AzaC. In contrast, the association of K9-methyl histone H3 with the MT-I promoter decreased significantly after treatment with 5-AzaC and TSA. ChIP assay with antibodies specific for methyl-CpG binding proteins (MBDs) demonstrated that only methyl-CpG binding protein 2 (MeCP2) was associated with the MT-I promoter, which was significantly enhanced after TSA treatment. Association of histone deacetylase 1 (HDAC1) with the promoter decreased after treatment with TSA or 5-AzaC and was abolished after treatment with both inhibitors. Among the DNA methyltransferases, both Dnmt1 and Dnmt3a were associated with the MT-I promoter in the lymphosarcoma cells, and association of Dnmt1 decreased with time after treatment with 5-AzaC. Treatment of these cells with HDAC inhibitors also increased expression of the MTF-1 (metal transcription factor-1) gene as well as its DNA binding activity. In vivo genomic footprinting studies demonstrated increased occupancy of MTF-1 to metal response elements of the MT-I promoter after treatment with both inhibitors. Analysis of the promoter by mapping with restriction enzymes in vivo showed that the MT-I promoter attained a more open chromatin structure after combined treatment with 5-AzaC and TSA as opposed to treatment with either agent alone. These results implicate involvement of multifarious factors including modified histones, MBDs, and Dnmts in silencing the methylated MT-I promoter in lymphosarcoma cells. The synergistic activation of this promoter by these two types of inhibitors is due to demethylation of the promoter and altered association of different factors that leads to reorganization of the chromatin and the resultant increase in accessibility of the promoter to the activated transcription factor MTF-1.


Subject(s)
Chromatin/chemistry , Chromosomal Proteins, Non-Histone , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Histone Deacetylase Inhibitors , Metallothionein/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Acetylation , Animals , Azacitidine/metabolism , Base Sequence , Chromatin/genetics , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Footprinting , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Hydroxamic Acids/metabolism , Metallothionein/metabolism , Methyl-CpG-Binding Protein 2 , Mice , Models, Genetic , Molecular Sequence Data , Repressor Proteins/metabolism , Sequence Analysis, DNA , Tumor Cells, Cultured , Zinc/metabolism , Transcription Factor MTF-1
19.
Zhonghua Nan Ke Xue ; 13(5): 440-3, 2007 May.
Article in Zh | MEDLINE | ID: mdl-17569263

ABSTRACT

OBJECTIVE: To inhibit the expression of connexin43 (Cx43) in the human corpus cavernosum penis smooth muscle cells by small interfering RNA (siRNA) and detect the gap junction intercellular communication (GJIC), and to investigate the application of siRNA technology in the gap junction of corpus cavernosum penis smooth muscle cells and its role in the penile erection process. METHODS: With the help of the software of Ambion Corporation, the specific recombinant plasmids with siRNA targeting human Cx43 gene were constructed. The recombinant plasmids having been stably transferred into human corpus cavernosum penis smooth muscle cells for 48 hours, semi-quantitive reverse transcription polymerase chain reaction (RT-PCR) and Western blotting techniques were used to examine the inhibitory effects of siRNA on the expressions of the Cx43 gene and protein, in comparison with the siRNA negative control and the blank control group, respectively. The GJIC was detected by scrape-loading and fluorescence dye transfer experiments through the fluorescence microscope. RESULTS: The results of enzyme digestion analysis and DNA sequencing showed that the recombinant plasmid pSilencer 1.0-U6-siRNA-Cx43 was successfully constructed. The relative levels of Cx43 mRNA and protein expression in the smooth muscle cells were (0.45 +/- 0.08)% and (0.56 +/- 0.06)% after successful transfer of the recombinant plasmid. However, the expression levels of mRNA and protein were (0.72 +/- 0.04)% and (0.80 +/- 0.08)% in the negative siRNA transfer group, and (0.74 +/- 0.09)% and (0.77 +/- 0.11)% in the blank control, respectively, with a significant difference (P < 0.05). The GJIC also decreased significantly. CONCLUSION: siRNA can significantly inhibit the expression of Cx43 and block the GJIC in the human corpus cavernosum penis smooth muscle cells. siRNA technology plays an important role in penile erection and flaccidity.


Subject(s)
Connexin 43/biosynthesis , Myocytes, Smooth Muscle/physiology , Penis/metabolism , RNA, Small Interfering , Blotting, Northern , Cells, Cultured , Connexin 43/genetics , Humans , Intercellular Junctions , Male , Penis/cytology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
Zhonghua Nan Ke Xue ; 12(5): 405-7, 2006 May.
Article in Zh | MEDLINE | ID: mdl-16755867

ABSTRACT

OBJECTIVE: To study the effects of different immunodepressants on the sperm parameters of kidney transplant recipients. METHODS: In 15 healthy fertile men and 37 kidney transplant recipients, ejaculates were aseptically obtained by masturbation. Thirty-seven patients were divided into two groups, 20 patients were treated with Prograf (FK506) combination with mycophenolate mofetil (MMF) and prednisone; 17 patients were treated with cyclosporine (CsA) combination with azathioprine with prednisone. The sperm viability, mobility parameters such as prorsad percentage motility, straight line velocity (VSL), curve line velocity (VCL), velocity of average path (VAP) and morph were estimated with a computer-assisted sperm analyzer (CASA) provided with a multiple-exposure photography system. RESULTS: There were no significant difference in sperm viability rate [(81.7 +/- 5.7)%, (79.4 +/- 6.8)% and (83.8 +/- 6.0)%], VCL [(24.1 +/- 8.6)%, (23.9 +/- 4.4)%, (24.8 +/- 4.2)% ] and VAP [(19.7 +/- 6.6)%, (18.6 +/- 2.9)%, (21.0 +/- 4.0)%] among groups of FK506, CsA and control, respectively (P > 0.05). The rate of anomaly [(67.8 +/- 5.7)%], the prorsad percentage motility [(46.4 +/- 8.1)%] and VSL [(15.4 +/- 4.6)%] in the group of FK506 were respectively significantly lower and higher than those in the group of CsA [(80.1 +/- 5.6%, (33.3 +/- 6.4)%, (10.2 +/- 2.4)%] (P < 0.05). CONCLUSION: The application of FK506 combined with MMF could help recover the mobility and morphology of the sperm in kidney transplantation recipients.


Subject(s)
Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Kidney Transplantation , Sperm Motility/drug effects , Tacrolimus/pharmacology , Adolescent , Adult , Case-Control Studies , Drug Therapy, Combination , Humans , Male , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Prednisone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL