Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 13(3): 925-947, 2022.
Article in English | MEDLINE | ID: mdl-34890841

ABSTRACT

BACKGROUND & AIMS: Sphingosine 1-phosphate receptors (S1PRs) are a group of G-protein-coupled receptors that confer a broad range of functional effects in chronic inflammatory and metabolic diseases. S1PRs also may mediate the development of nonalcoholic steatohepatitis (NASH), but the specific subtypes involved and the mechanism of action are unclear. METHODS: We investigated which type of S1PR isoforms is activated in various murine models of NASH. The mechanism of action of S1PR4 was examined in hepatic macrophages isolated from high-fat, high-cholesterol diet (HFHCD)-fed mice. We developed a selective S1PR4 functional antagonist by screening the fingolimod (2-amino-2-[2-(4- n -octylphenyl)ethyl]-1,3- propanediol hydrochloride)-like sphingolipid-focused library. RESULTS: The livers of various mouse models of NASH as well as hepatic macrophages showed high expression of S1pr4. Moreover, in a cohort of NASH patients, expression of S1PR4 was 6-fold higher than those of healthy controls. S1pr4+/- mice were protected from HFHCD-induced NASH and hepatic fibrosis without changes in steatosis. S1pr4 depletion in hepatic macrophages inhibited lipopolysaccharide-mediated Ca++ release and deactivated the Nod-like receptor pyrin domain-containning protein 3 (NLRP3) inflammasome. S1P increased the expression of S1pr4 in hepatic macrophages and activated NLRP3 inflammasome through inositol trisphosphate/inositol trisphosphate-receptor-dependent [Ca++] signaling. To further clarify the biological function of S1PR4, we developed SLB736, a novel selective functional antagonist of SIPR4. Similar to S1pr4+/- mice, administration of SLB736 to HFHCD-fed mice prevented the development of NASH and hepatic fibrosis, but not steatosis, by deactivating the NLRP3 inflammasome. CONCLUSIONS: S1PR4 may be a new therapeutic target for NASH that mediates the activation of NLRP3 inflammasome in hepatic macrophages.


Subject(s)
Inflammasomes , Non-alcoholic Fatty Liver Disease , Animals , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Non-alcoholic Fatty Liver Disease/drug therapy , Sphingosine-1-Phosphate Receptors
2.
Autophagy ; 17(5): 1205-1221, 2021 05.
Article in English | MEDLINE | ID: mdl-32400277

ABSTRACT

Although macroautophagy/autophagy deficiency causes degenerative diseases, the deletion of essential autophagy genes in adipocytes paradoxically reduces body weight. Brown adipose tissue (BAT) plays an important role in body weight regulation and metabolic control. However, the key cellular mechanisms that maintain BAT function remain poorly understood. in this study, we showed that global or brown adipocyte-specific deletion of pink1, a Parkinson disease-related gene involved in selective mitochondrial autophagy (mitophagy), induced BAT dysfunction, and obesity-prone type in mice. Defective mitochondrial function is among the upstream signals that activate the NLRP3 inflammasome. NLRP3 was induced in brown adipocyte precursors (BAPs) from pink1 knockout (KO) mice. Unexpectedly, NLRP3 induction did not induce canonical inflammasome activity. Instead, NLRP3 induction led to the differentiation of pink1 KO BAPs into white-like adipocytes by increasing the expression of white adipocyte-specific genes and repressing the expression of brown adipocyte-specific genes. nlrp3 deletion in pink1 knockout mice reversed BAT dysfunction. Conversely, adipose tissue-specific atg7 KO mice showed significantly lower expression of Nlrp3 in their BAT. Overall, our data suggest that the role of mitophagy is different from general autophagy in regulating adipose tissue and whole-body energy metabolism. Our results uncovered a new mitochondria-NLRP3 pathway that induces BAT dysfunction. The ability of the nlrp3 knockouts to rescue BAT dysfunction suggests the transcriptional function of NLRP3 as an unexpected, but a quite specific therapeutic target for obesity-related metabolic diseases.Abbreviations: ACTB: actin, beta; BAPs: brown adipocyte precursors; BAT: brown adipose tissue; BMDMs: bone marrow-derived macrophages; CASP1: caspase 1; CEBPA: CCAAT/enhancer binding protein (C/EBP), alpha; ChIP: chromatin immunoprecipitation; EE: energy expenditure; HFD: high-fat diet; IL1B: interleukin 1 beta; ITT: insulin tolerance test; KO: knockout; LPS: lipopolysaccharide; NLRP3: NLR family, pyrin domain containing 3; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; RD: regular diet; ROS: reactive oxygen species; RT: room temperature; UCP1: uncoupling protein 1 (mitochondrial, proton carrier); WT: wild-type.


Subject(s)
Adipose Tissue, Brown/metabolism , Autophagy/physiology , Inflammasomes/metabolism , Mitophagy/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adipocytes/metabolism , Animals , Energy Metabolism/physiology , Mice, Knockout , Mitochondria/metabolism , Mitophagy/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Reactive Oxygen Species/metabolism
3.
Gut ; 70(10): 1954-1964, 2021 10.
Article in English | MEDLINE | ID: mdl-33208407

ABSTRACT

OBJECTIVE: Lipotoxic hepatocyte injury is a primary event in non-alcoholic steatohepatitis (NASH), but the mechanisms of lipotoxicity are not fully defined. Sphingolipids and free cholesterol (FC) mediate hepatocyte injury, but their link in NASH has not been explored. We examined the role of free cholesterol and sphingomyelin synthases (SMSs) that generate sphingomyelin (SM) and diacylglycerol (DAG) in hepatocyte pyroptosis, a specific form of programmed cell death associated with inflammasome activation, and NASH. DESIGN: Wild-type C57BL/6J mice were fed a high fat and high cholesterol diet (HFHCD) to induce NASH. Hepatic SMS1 and SMS2 expressions were examined in various mouse models including HFHCD-fed mice and patients with NASH. Pyroptosis was estimated by the generation of the gasdermin-D N-terminal fragment. NASH susceptibility and pyroptosis were examined following knockdown of SMS1, protein kinase Cδ (PKCδ), or the NLR family CARD domain-containing protein 4 (NLRC4). RESULTS: HFHCD increased the hepatic levels of SM and DAG while decreasing the level of phosphatidylcholine. Hepatic expression of Sms1 but not Sms2 was higher in mouse models and patients with NASH. FC in hepatocytes induced Sms1 expression, and Sms1 knockdown prevented HFHCD-induced NASH. DAG produced by SMS1 activated PKCδ and NLRC4 inflammasome to induce hepatocyte pyroptosis. Depletion of Nlrc4 prevented hepatocyte pyroptosis and the development of NASH. Conditioned media from pyroptotic hepatocytes activated the NOD-like receptor family pyrin domain containing 3 inflammasome (NLRP3) in Kupffer cells, but Nlrp3 knockout mice were not protected against HFHCD-induced hepatocyte pyroptosis. CONCLUSION: SMS1 mediates hepatocyte pyroptosis through a novel DAG-PKCδ-NLRC4 axis and holds promise as a therapeutic target for NASH.


Subject(s)
Hepatocytes/enzymology , Non-alcoholic Fatty Liver Disease/enzymology , Pyroptosis , Transferases (Other Substituted Phosphate Groups)/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
4.
Exp Mol Med ; 51(7): 1-14, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31285429

ABSTRACT

The administration of mesenchymal stem cells (MSCs) was shown to attenuate overt as well as early diabetic nephropathy in rodents, but the underlying mechanism of this beneficial effect is largely unknown. Inflammation and mitochondrial dysfunction are major pathogenic factors in diabetic nephropathy. In this study, we found that the repeated administration of MSCs prevents albuminuria and injury to tubular epithelial cells (TECs), an important element in the progression of diabetic nephropathy, by improving mitochondrial function. The expression of M1 macrophage markers was significantly increased in diabetic kidneys compared with that in control kidneys. Interestingly, the expression of arginase-1 (Arg1), an important M2 macrophage marker, was reduced in diabetic kidneys and increased by MSC treatment. In cultured TECs, conditioned media from lipopolysaccharide-activated macrophages reduced peroxisomal proliferator-activated receptor gamma coactivator 1α (Pgc1a) expression and impaired mitochondrial function. The coculture of macrophages with MSCs increased and decreased the expression of Arg1 and M1 markers, respectively. Treatment with conditioned media from cocultured macrophages prevented activated macrophage-induced mitochondrial dysfunction in TECs. In the absence of MSC coculture, Arg1 overexpression in macrophages reversed Pgc1a suppression in TECs. These observations suggest that MSCs prevent the progression of diabetic nephropathy by reversing mitochondrial dysfunction in TECs via the induction of Arg1 in macrophages.


Subject(s)
Albuminuria/prevention & control , Arginase/metabolism , Diabetes Complications/prevention & control , Diabetic Nephropathies/prevention & control , Mesenchymal Stem Cells/metabolism , Animals , Arginase/genetics , Cell Line , Cord Blood Stem Cell Transplantation , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney/metabolism , Kidney/pathology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mice , Mitochondria/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
5.
Biochem Biophys Res Commun ; 486(4): 1014-1020, 2017 05 13.
Article in English | MEDLINE | ID: mdl-28363867

ABSTRACT

Mitochondrial dynamics, including constant fusion and fission, play critical roles in maintaining mitochondrial morphology and function. Here, we report that developmentally regulated GTP-binding protein 2 (DRG2) regulates mitochondrial morphology by modulating the expression of the mitochondrial fission gene dynamin-related protein 1 (Drp1). shRNA-mediated silencing of DRG2 induced mitochondrial swelling, whereas expression of an shRNA-resistant version of DRG2 decreased mitochondrial swelling in DRG2-depleted cells. Analysis of the expression levels of genes involved in mitochondrial fusion and fission revealed that DRG2 depletion significantly decreased the level of Drp1. Overexpression of Drp1 rescued the defect in mitochondrial morphology induced by DRG2 depletion. DRG2 depletion reduced the mitochondrial membrane potential, oxygen consumption rate (OCR), and amount of mitochondrial DNA (mtDNA), whereas it increased reactive oxygen species (ROS) production and apoptosis. Taken together, our data demonstrate that DRG2 acts as a regulator of mitochondrial fission by controlling the expression of Drp1.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Down-Regulation/physiology , Dynamins , HeLa Cells , Humans
6.
Hepatology ; 66(2): 416-431, 2017 08.
Article in English | MEDLINE | ID: mdl-28073164

ABSTRACT

Free cholesterol (FC) accumulation in the liver is an important pathogenic mechanism of nonalcoholic steatohepatitis (NASH). Plasmalogens, key structural components of the cell membrane, act as endogenous antioxidants and are primarily synthesized in the liver. However, the role of hepatic plasmalogens in metabolic liver disease is unclear. In this study, we found that hepatic levels of docosahexaenoic acid (DHA)-containing plasmalogens, expression of glyceronephosphate O-acyltransferase (Gnpat; the rate-limiting enzyme in plasmalogen biosynthesis), and expression of Pparα were lower in mice with NASH caused by accumulation of FC in the liver. Cyclodextrin-induced depletion of FC transactivated Δ-6 desaturase by increasing sterol regulatory element-binding protein 2 expression in cultured hepatocytes. DHA, the major product of Δ-6 desaturase activation, activated GNPAT, thereby explaining the association between high hepatic FC and decreased Gnpat expression. Gnpat small interfering RNA treatment significantly decreased peroxisome proliferator-activated receptor α (Pparα) expression in cultured hepatocytes. In addition to GNPAT, DHA activated PPARα and increased expression of Pparα and its target genes, suggesting that DHA in the DHA-containing plasmalogens contributed to activation of PPARα. Accordingly, administration of the plasmalogen precursor, alkyl glycerol (AG), prevented hepatic steatosis and NASH through a PPARα-dependent increase in fatty acid oxidation. Gnpat+/- mice were more susceptible to hepatic lipid accumulation and less responsive to the preventive effect of fluvastatin on NASH development, suggesting that endogenous plasmalogens prevent hepatic steatosis and NASH. CONCLUSION: Increased hepatic FC in animals with NASH decreased plasmalogens, thereby sensitizing animals to hepatocyte injury and NASH. Our findings uncover a novel link between hepatic FC and plasmalogen homeostasis through GNPAT regulation. Further study of AG or other agents that increase hepatic plasmalogen levels may identify novel therapeutic strategies against NASH. (Hepatology 2017;66:416-431).


Subject(s)
Fatty Liver/metabolism , Glucosamine 6-Phosphate N-Acetyltransferase/metabolism , Mediator Complex Subunit 1/metabolism , Plasmalogens/metabolism , Analysis of Variance , Animals , Biomarkers/metabolism , Biopsy, Needle , Disease Models, Animal , Fatty Acids, Monounsaturated/pharmacology , Fatty Liver/pathology , Fluvastatin , Glucosamine 6-Phosphate N-Acetyltransferase/drug effects , Immunohistochemistry , Indoles/pharmacology , Male , Mediator Complex Subunit 1/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Random Allocation , Sensitivity and Specificity , Signal Transduction
7.
Diabetes ; 65(9): 2516-28, 2016 09.
Article in English | MEDLINE | ID: mdl-27246913

ABSTRACT

Fibrosis of adipose tissue induces ectopic fat accumulation and insulin resistance by inhibiting adipose tissue expandability. Mechanisms responsible for the induction of adipose tissue fibrosis may provide therapeutic targets but are poorly understood. In this study, high-fat diet (HFD)-fed wild-type (WT) and iNOS(-/-) mice were used to examine the relationship between nitric oxide (NO) produced by macrophages and adipose tissue fibrosis. In contrast to WT mice, iNOS(-/-) mice fed an HFD were protected from infiltration of proinflammatory macrophages and adipose tissue fibrosis. Hypoxia-inducible factor 1α (HIF-1α) protein level was increased in adipose tissue of HFD-fed WT mice, but not iNOS(-/-) mice. In contrast, the expression of mitochondrial biogenesis factors was decreased in HFD-fed WT mice, but not iNOS(-/-) mice. In studies with cultured cells, macrophage-derived NO decreased the expression of mitochondrial biogenesis factors, and increased HIF-1α protein level, DNA damage, and phosphorylated p53 in preadipocytes. By activating p53 signaling, NO suppressed peroxisome proliferator-activated receptor γ coactivator 1α expression, which induced mitochondrial dysfunction and inhibited preadipocyte differentiation in adipocytes. The effects of NO were blocked by rosiglitazone. The findings suggest that NO produced by macrophages induces mitochondrial dysfunction in preadipocytes by activating p53 signaling, which in turn increases HIF-1α protein level and promotes a profibrogenic response in preadipocytes that results in adipose tissue fibrosis.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Fibrosis/metabolism , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Adipocytes/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibrosis/etiology , Fluorescent Antibody Technique , Glucose Tolerance Test , Isothiuronium/analogs & derivatives , Isothiuronium/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/antagonists & inhibitors , RAW 264.7 Cells
8.
Diabetes Metab J ; 40(5): 376-385, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27098507

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease is the most common form of chronic liver disease in industrialized countries. Recent studies have highlighted the association between peroxisomal dysfunction and hepatic steatosis. Peroxisomes are intracellular organelles that contribute to several crucial metabolic processes, such as facilitation of mitochondrial fatty acid oxidation (FAO) and removal of reactive oxygen species through catalase or plasmalogen synthesis. Statins are known to prevent hepatic steatosis and non-alcoholic steatohepatitis (NASH), but underlying mechanisms of this prevention are largely unknown. METHODS: Seven-week-old C57BL/6J mice were given normal chow or a methionine- and choline-deficient diet (MCDD) with or without various statins, fluvastatin, pravastatin, simvastatin, atorvastatin, and rosuvastatin (15 mg/kg/day), for 6 weeks. Histological lesions were analyzed by grading and staging systems of NASH. We also measured mitochondrial and peroxisomal FAO in the liver. RESULTS: Statin treatment prevented the development of MCDD-induced NASH. Both steatosis and inflammation or fibrosis grades were significantly improved by statins compared with MCDD-fed mice. Gene expression levels of peroxisomal proliferator-activated receptor α (PPARα) were decreased by MCDD and recovered by statin treatment. MCDD-induced suppression of mitochondrial and peroxisomal FAO was restored by statins. Each statin's effect on increasing FAO and improving NASH was independent on its effect of decreasing cholesterol levels. CONCLUSION: Statins prevented NASH and increased mitochondrial and peroxisomal FAO via induction of PPARα. The ability to increase hepatic FAO is likely the major determinant of NASH prevention by statins. Improvement of peroxisomal function by statins may contribute to the prevention of NASH.

9.
Mol Biol Cell ; 27(2): 334-48, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26582392

ABSTRACT

The small GTPase Rab5 regulates the early endocytic pathway of transferrin (Tfn), and Rab5 deactivation is required for Tfn recycling. Rab5 deactivation is achieved by RabGAP5, a GTPase-activating protein, on the endosomes. Here we report that recruitment of RabGAP5 is insufficient to deactivate Rab5 and that developmentally regulated GTP-binding protein 2 (DRG2) is required for Rab5 deactivation and Tfn recycling. DRG2 was associated with phosphatidylinositol 3-phosphate-containing endosomes. It colocalized and interacted with EEA1 and Rab5 on endosomes in a phosphatidylinositol 3-kinase-dependent manner. DRG2 depletion did not affect Tfn uptake and recruitment of RabGAP5 and Rac1 to Rab5 endosomes. However, it resulted in impairment of interaction between Rab5 and RabGAP5, Rab5 deactivation on endosomes, and Tfn recycling. Ectopic expression of shRNA-resistant DRG2 rescued Tfn recycling in DRG2-depleted cells. Our results demonstrate that DRG2 is an endosomal protein and a key regulator of Rab5 deactivation and Tfn recycling.


Subject(s)
GTP-Binding Proteins/metabolism , Transferrin/metabolism , rab5 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Endocytosis/physiology , Endosomes/metabolism , Female , GTP-Binding Proteins/genetics , GTPase-Activating Proteins/metabolism , HeLa Cells , Humans , MCF-7 Cells , Male , Membrane Fusion , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Structure, Tertiary , Vesicular Transport Proteins/metabolism
10.
J Endocrinol ; 225(3): 147-58, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25869616

ABSTRACT

Mitochondrial dysfunction in hypertrophic adipocytes can reduce adiponectin synthesis. We investigated whether 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) expression is increased in hypertrophic adipocytes and whether this is responsible for mitochondrial dysfunction and reduced adiponectin synthesis. Differentiated 3T3L1 adipocytes were cultured for up to 21 days. The effect of AZD6925, a selective 11ß-HSD1 inhibitor, on metabolism was examined. db/db mice were administered 600 mg/kg AZD6925 daily for 4 weeks via gastric lavage. Mitochondrial DNA (mtDNA) content, mRNA expression levels of 11 ß -H sd1 and mitochondrial biogenesis factors, adiponectin synthesis, fatty acid oxidation (FAO), oxygen consumption rate and glycolysis were measured. Adipocyte hypertrophy in 3T3L1 cells exposed to a long duration of culture was associated with increased 11 ß -Hsd1 mRNA expression and reduced mtDNA content, mitochondrial biogenesis factor expression and adiponectin synthesis. These cells displayed reduced mitochondrial respiration and increased glycolysis. Treatment of these cells with AZD6925 increased adiponectin synthesis and mitochondrial respiration. Inhibition of FAO by etomoxir blocked the AZD6925-induced increase in adiponectin synthesis, indicating that 11ß-HSD1-mediated reductions in FAO are responsible for the reduction in adiponectin synthesis. The expression level of 11 ß -Hsd1 was higher in adipose tissues of db/db mice. Administration of AZD6925 to db/db mice increased the plasma adiponectin level and adipose tissue FAO. In conclusion, increased 11ß-HSD1 expression contributes to reduced mitochondrial respiration and adiponectin synthesis in hypertrophic adipocytes.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adiponectin/metabolism , Adipose Tissue, White/metabolism , Energy Metabolism , Gene Expression Regulation , Obesity/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 3T3-L1 Cells , Adamantane/analogs & derivatives , Adamantane/therapeutic use , Adiponectin/blood , Adiponectin/genetics , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Animals , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Hypertrophy , Lipid Metabolism/drug effects , Lipotropic Agents/pharmacology , Lipotropic Agents/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Mutant Strains , Mitochondrial Dynamics , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/drug therapy , Obesity/pathology , Obesity/physiopathology , Oxygen Consumption/drug effects
11.
Oncol Rep ; 32(4): 1537-42, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25109698

ABSTRACT

The purpose of the present study was to identify a predictive marker associated with tumor progression or recurrence. We investigated the expression of p53, Ki-67, Bax, Bcl-2, vascular endothelial growth factor (VEGF)-A, VEGFR-1, VEGFR-2 and neuropilin-1 (NRP-1) in pituitary adenomas (PAs) with/without tumor progression during follow-up periods. We compared the expression of these molecules in primary and recurrent specimens to identify a predictive marker associated with tumor progression. Nineteen patients had no progression for more than 5-years of follow-up. Nine patients had tumor progression within 5 years of their first transsphenoidal surgery (TSS) surgery and underwent re-TSS for treating progression of adenoma. Tumor size was larger and involvement of the cavernous sinus was more frequent in the progression group than these variables in the no progression group. A strong association was observed between NRP-1 expression and tumor progression. No significant risk for developing tumor progression was associated with Ki-67, p53, Bax, Bcl-2, VEGFR-1, VEGFR-2, or VEGF-A expression. Four of nine patients showed strong NRP-1 immunoreactivity in progression specimens. Negative NRP-1 immunoreactivity in the initial specimens was converted into strong positivity in the progression specimens of five patients. NRP-1 could be a relevant PA marker of progression and could be a potential target for medical therapy.


Subject(s)
Adenoma/metabolism , Neoplasm Recurrence, Local/metabolism , Neuropilin-1/metabolism , Pituitary Neoplasms/metabolism , Adenoma/pathology , Adenoma/surgery , Adult , Aged , Cell Line, Tumor , Disease Progression , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Middle Aged , Neuropilin-1/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/surgery , Prognosis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
12.
Clin Immunol ; 150(2): 225-35, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24463315

ABSTRACT

Developmentally regulated GTP-binding protein 2 (DRG2) represents a novel subclass of GTP-binding proteins. We here report that transgenic overexpression of DRG2 in mice ameliorates experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). The protective effect of DRG2 in EAE was mediated by the inhibition of the development of T(H)17 cells. DRG2 enhanced the activity of PPARγ, which led to an inhibition of the nuclear factor kappa B (NF-κB) activity and IL-6 production in antigen presenting cells and an inhibition of the development of T(H)17 cells. Our results demonstrate that DRG2 is an essential modulator of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , GTP-Binding Proteins/genetics , Th17 Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Differentiation , Co-Repressor Proteins/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , GTP-Binding Proteins/metabolism , Gene Expression , Genotype , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , PPAR gamma/metabolism , Promoter Regions, Genetic , Protein Binding , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism
13.
Exp Mol Med ; 44(9): 562-70, 2012 Sep 30.
Article in English | MEDLINE | ID: mdl-22809900

ABSTRACT

Mitochondrial dysfunction and endoplasmic reticulum (ER) stress are considered the key determinants of insulin resistance. Impaired mitochondrial function in obese animals was shown to induce the ER stress response, resulting in reduced adiponectin synthesis in adipocytes. The expression of inducible nitric oxide synthase (iNOS) is increased in adipose tissues in genetic and dietary models of obesity. In this study, we examined whether activation of iNOS is responsible for palmitate-induced mitochondrial dysfunction, ER stress, and decreased adiponectin synthesis in 3T3L1 adipocytes. As expected, palmitate increased the expression levels of iNOS and ER stress response markers, and decreased mitochondrial contents. Treatment with iNOS inhibitor increased adiponectin synthesis and reversed the palmitate-induced ER stress response. However, the iNOS inhibitor did not affect the palmitate-induced decrease in mitochondrial contents. Chemicals that inhibit mitochondrial function increased iNOS expression and the ER stress response, whereas measures that increase mitochondrial biogenesis (rosiglitazone and adenoviral overexpression of nuclear respiratory factor-1) reversed them. Inhibition of mitochondrial biogenesis prevented the rosiglitazone-induced decrease in iNOS expression and increase in adiponectin synthesis. These results suggest that palmitate-induced mitochondrial dysfunction is the primary event that leads to iNOS induction, ER stress, and decreased adiponectin synthesis in cultured adipocytes.


Subject(s)
Adipocytes , Mitochondria/metabolism , Nitric Oxide Synthase Type II , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/biosynthesis , Adipose Tissue/metabolism , Animals , Endoplasmic Reticulum Stress/drug effects , Insulin Resistance/genetics , Mice , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Turnover/drug effects , Mitochondrial Turnover/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nuclear Respiratory Factor 1 , Obesity/genetics , Obesity/metabolism , Palmitic Acid/pharmacology , Rosiglitazone , Thiazolidinediones/pharmacology
14.
PLoS One ; 6(7): e22362, 2011.
Article in English | MEDLINE | ID: mdl-21814578

ABSTRACT

The nonvirion (NV) protein of infectious hematopoietic necrosis virus (IHNV) has been previously reported to be essential for efficient growth and pathogenicity of IHNV. However, little is known about the mechanism by which the NV supports the viral growth. In this study, cellular localization of NV and its role in IHNV growth in host cells was investigated. Through transient transfection in RTG-2 cells of NV fused to green fluorescent protein (GFP), a nuclear localization of NV was demonstrated. Deletion analyses showed that the (32)EGDL(35) residues were essential for nuclear localization of NV protein, and fusion of these 4 amino acids to GFP directed its transport to the nucleus. We generated a recombinant IHNV, rIHNV-NV-ΔEGDL in which the (32)EGDL(35) was deleted from the NV. rIHNVs with wild-type NV (rIHNV-NV) or with the NV gene replaced with GFP (rIHNV-ΔNV-GFP) were used as controls. RTG-2 cells infected with rIHNV-ΔNV-GFP and rIHNV-NV-ΔEGDL yielded 12- and 5-fold less infectious virion, respectively, than wild type rIHNV-infected cells at 48 h post-infection (p.i.). While treatment with poly I∶C at 24 h p.i. did not inhibit replication of wild-type rIHNVs, replication rates of rIHNV-ΔNV-GFP and rIHNV-NV-ΔEGDL were inhibited by poly I∶C. In addition, both rIHNV-ΔNV and rIHNV-NV-ΔEGDL induced higher levels of expressions of both IFN1 and Mx1 than wild-type rIHNV. These data suggest that the IHNV NV may support the growth of IHNV through inhibition of the INF system and the amino acid residues of (32)EGDL(35) responsible for nuclear localization are important for the inhibitory activity of NV.


Subject(s)
Cell Nucleus/metabolism , Infectious hematopoietic necrosis virus/growth & development , Infectious hematopoietic necrosis virus/pathogenicity , Rhabdoviridae Infections/virology , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Cells, Cultured , Cyprinidae , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Nuclear Localization Signals , Oncorhynchus mykiss , Poly I-C/genetics , Promoter Regions, Genetic , RNA, Viral , Rhabdoviridae Infections/metabolism , Salmon , Subcellular Fractions
15.
J Biol Chem ; 286(28): 24735-42, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21606497

ABSTRACT

Nicotine inhibits the release of TNF-α from macrophage through activation of STAT3. Tristetraprolin (TTP) is known to destabilize pro-inflammatory transcripts containing AU-rich elements (ARE) in 3'-untranslated region (3'-UTR). Here we show that in LPS-stimulated human macrophages the anti-inflammatory action of nicotine is mediated by TTP. Nicotine induced activation of STAT3 enhanced STAT3 binding to the TTP promoter, increased TTP promoter activity, and increased TTP expression resulting in the suppression of LPS-stimulated TNF-α production. Overexpression of a dominant negative mutant of STAT3 (R382W) or down-regulation of STAT3 by siRNA abolished nicotine-induced TTP expression and suppression of LPS-stimulated TNF-α production. Nicotine enhanced the decay of TNF-α mRNA and decreased luciferase expression of a TNF-α 3'-UTR reporter plasmid in U937 cells. However, siRNA to TTP abrogated these effects of nicotine. In this experiment, we are reporting for the first time the involvement of TTP in the cholinergic anti-inflammatory cascade consisting of nicotine-STAT3-TTP-dampening inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Macrophages/metabolism , Nicotine/pharmacology , Promoter Regions, Genetic , Tristetraprolin/metabolism , 3' Untranslated Regions/genetics , Amino Acid Substitution , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Mutation, Missense , Nicotinic Agonists/pharmacology , Protein Binding , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/biosynthesis , U937 Cells
16.
J Biol Chem ; 286(24): 21577-87, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21507959

ABSTRACT

Tristetraprolin (TTP) is an AU-rich element-binding protein that regulates mRNA stability. We previously showed that TTP acts as a negative regulator of VEGF gene expression in colon cancer cells. The p38 MAPK pathway is known to suppress the TTP activity. However, until now the signaling pathway to enhance TTP function is not well known. Here, we show that casein kinase 2 (CK2) enhances the TTP function in the regulation of the VEGF expression in colon cancer cells. CK2 increased TTP protein levels and enhanced VEGF mRNA decaying activity of TTP. TTP was not a direct target of CK2. Instead, CK2 increased the phosphorylation of MKP-1, which led to a decrease in the phosphorylation of p38 MAPK. Inhibition of MKP-1 by siRNA attenuated the increase in TTP function and the decrease of p38 phosphorylation induced by CK2α overexpression. TGF-ß1 increased the expressions of CK2 and TTP and the TTP function. The siRNA against CK2α or TTP reversed TGF-ß1-induced increases in the expression of CK2 and TTP and the TTP function. Our data suggest that CK2 enhances the protein level and activity of TTP via the modulation of the MKP-1-p38 MAPK signaling pathway and that TGF-ß1 enhances the activity of CK2.


Subject(s)
Casein Kinase II/metabolism , Dual Specificity Phosphatase 1/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Tristetraprolin/chemistry , Cell Line, Tumor , Colonic Neoplasms/metabolism , Humans , MAP Kinase Signaling System , Models, Biological , Phosphorylation , RNA Stability/genetics , RNA, Small Interfering/metabolism , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/metabolism
17.
Biochim Biophys Acta ; 1809(3): 184-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21296692

ABSTRACT

Developmentally regulated GTP-binding protein 2 (DRG2) is an evolutionarily conserved GTP-binding protein. DRG2 mRNA expression has been confirmed in many animal and human tissues. DRG2 is thought to play an essential role in the control of cell growth and differentiation. However, transcriptional regulation of DRG2 is largely unknown. To investigate the mechanisms controlling DRG2 expression, we cloned 1509bp of the 5'-flanking sequence of this gene. Deletion analysis showed that the region between -113 and -70 is essential for the basal level expression of the DRG2 gene in K562 human erythroleukemic cells. Mutation of a putative stimulating protein 1 (Sp1) regulatory site located at position -108 resulted in a significant decline in DRG2 promoter activity. Electrophoretic mobility shift assay and chromatin immunoprecipitation analysis revealed that Sp1 binds to this site. Knockdown of Sp1 expression using siRNA inhibited the promoter activation as well as the endogenous DRG2 transcriptional level. Taken together, these results demonstrate that basal expression level of DRG2 is regulated by the Sp1 transcription factor.


Subject(s)
GTP-Binding Proteins/genetics , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , Cell Survival , DNA Mutational Analysis , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Gene Silencing , Humans , K562 Cells , Molecular Sequence Data , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Sequence Deletion
18.
J Biol Chem ; 285(23): 17329-37, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20335167

ABSTRACT

LATS2 is a tumor suppressor gene implicated in the control of cell growth and the cell cycle. Here, we investigated the post-transcriptional regulation of LATS2 expression by tristetraprolin (TTP). Our results show that the expression level of LATS2 is inversely correlated with TTP expression in human cancer cell lines. Overexpression of TTP reduced the expression level of LATS2. Conversely, treatment with small interfering RNA against TTP increased the expression level of LATS2 through stabilization of LATS2 mRNA and suppressed the proliferation of A549 human lung cancer cells. LATS2 mRNA contains AU-rich elements (AREs) within the 3'-untranslated region, and TTP destabilized a luciferase mRNA containing LATS2 ARE. In addition, RNA electrophoretic mobility shift assay revealed that TTP directly bound to the ARE of LATS2 mRNA. These results establish LATS2 mRNA as a physiological target of TTP and suggest the possibility that TTP controls cell growth through regulation of LATS2 mRNA stability.


Subject(s)
Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/metabolism , Tristetraprolin/metabolism , Tumor Suppressor Proteins/metabolism , 3' Untranslated Regions , Base Sequence , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/metabolism , Models, Biological , Molecular Sequence Data , RNA Stability/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
FEBS Lett ; 580(1): 311-8, 2006 Jan 09.
Article in English | MEDLINE | ID: mdl-16376337

ABSTRACT

This study demonstrates a requirement for NF-kappaB activation in cis-diamminedichloroplatinum (cisplatin)-induced apoptosis in human head and neck squamous cell carcinoma (HNSCC) cell lines. This conclusion was supported by the following observations: cisplatin induced IkappaBalpha degradation and NF-kappaB-dependent transcriptional activation prior to cell death; pyrrolidine dithiocarbamate (PDTC), a chemical inhibitor of NF-kappaB activation, prevented apoptosis; lactacystin, an inhibitor of IkappaBalpha degradation, also prevented apoptosis; and finally, the expression of a super-repressor mutant IkappaBalpha blocked apoptosis. The expression of tumor necrosis factor alpha (TNFalpha) was promoted by cisplatin treatment and was suppressed by PDTC treatment. In addition, a neutralizing antibody against TNFalpha protected cells from cisplatin-induced apoptosis. These findings suggest that NF-kappaB activation is required for cisplatin-induced apoptosis and TNFalpha may play an important role in NF-kappaB-mediated apoptosis in cisplatin-treated HNSCC cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Cisplatin/pharmacology , Head and Neck Neoplasms/metabolism , NF-kappa B/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Autocrine Communication/drug effects , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Humans , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Thiocarbamates/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
20.
Stem Cells ; 23(10): 1502-13, 2005.
Article in English | MEDLINE | ID: mdl-16100000

ABSTRACT

The cell-surface markers used routinely to define the undifferentiated state and pluripotency of human embryonic stem cells (hESCs) are those used in mouse embryonic stem cells (mESCs) because of a lack of markers directly originated from hESC itself. To identify more hESC-specific cell-surface markers, we generated a panel of monoclonal antibodies (MAbs) by immunizing the irradiated cell clumps of hESC line Miz-hES1, and selected 26 MAbs that were able to bind to Miz-hES1 cells but not to mESCs, mouse embryonic fibroblast cells, and STO cells. Most antibodies did not bind to human neural progenitor cells derived from the Miz-hES1 cells, either. Of these, MAb 20-202S (IgG1, kappa) immunoprecipitated a cell-surface protein of 72-kDa from the lysate of biotin-labeled Miz-hES1 cells, which was identified to be heat shock 70-kDa protein 8 isoform 1 (HSPA8) by quadrupole time-of-flight tandem mass spectrometry. Immunocytochemical analyses proved that the HSPA8 protein was also present on the surface of hESC lines Miz-hES4, Miz-hES6, and HSF6. Two-color flow cytometric analysis of Miz-hES1 and HSF6 showed the coexpression of the HSPA8 protein with other hESC markers such as stage-specific embryonic antigen 3 (SSEA3), SSEA4, TRA-1-60, and TRA-1-81. Flow cytometric and Western blot analyses using various cells showed that MAb 20-202S specifically bound to the HSPA8 protein on the surface of Miz-hES1, contrary to other anti-HSP70 antibodies examined. Furthermore, the surface expression of the HSPA8 protein on Miz-hES1 was markedly downregulated upon differentiation. These data indicate that a novel MAb 20-202S recognizes the HSPA8 protein on the surface of hESCs and suggest that the HSPA8 protein is a putative cell-surface marker for undifferentiated hESCs.


Subject(s)
Cell Differentiation , Down-Regulation , HSP70 Heat-Shock Proteins/biosynthesis , Protein Isoforms/biosynthesis , Stem Cells/metabolism , Antibodies, Monoclonal/metabolism , Antigens, Surface/metabolism , Cell Line , Embryo Research , HeLa Cells , Humans , Immunohistochemistry , Mass Spectrometry , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...