Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Biochem Cell Biol ; 99(5): 519-526, 2021 10.
Article in English | MEDLINE | ID: mdl-33560918

ABSTRACT

Autophagy plays a key role in the metabolism of macromolecules via the degradative abilities of the lysosome. Transcription factor EB (TFEB) regulates autophagosome biogenesis and lysosome function, and promoting TFEB activity has emerged as a potential strategy for the treatment of metabolic disorders. Herein, we report that cetrimonium bromide (CTAB; a quaternary ammonium compound) promotes autophagy and lysosomal biogenesis by inducing the nuclear translocation of TFEB in hepatic cells. Knockdown of TFEB mediated by short hairpin RNA inhibits CTAB-induced autophagy and lysosomal biogenesis. Mechanistically, CTAB treatment inhibits the Akt-mTORC1 signaling pathway. Moreover, CTAB treatment significantly increases lipid metabolism in both palmitate- and oleate-treated HepG2 cells, and this increase was attenuated by knockdown of TFEB. Collectively, our results indicate that CTAB activates the autophagosome-lysosome pathway via inducing the nuclear translocation of TFEB by inhibiting the mTORC1 signaling pathway. These results add to the collective understanding of TFEB function and provide new insights into CTAB-mediated lipid metabolism.


Subject(s)
Autophagosomes/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cetrimonium/pharmacology , Hepatocytes/metabolism , Lysosomes/metabolism , Autophagosomes/drug effects , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Cells, Cultured , Cetrimonium/antagonists & inhibitors , Hepatocytes/drug effects , Humans , Lipid Metabolism/drug effects , Lysosomes/drug effects , RNA, Small Interfering/pharmacology
2.
Chem Res Toxicol ; 34(5): 1240-1244, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33156613

ABSTRACT

The herbicide paraquat (PQ) induces hippocampal neuronal cell loss and cognitive dysfunction after one and repeated treatment. All the mechanisms involved in these effects are not well understood. Single and repeated PQ treatment increased Aß and tau protein levels, through HSP70 and TFEB downregulation and proteasome 20S inhibition, producing cell death in primary hippocampal neurons associated with cognitive decline. Our results reveal the mechanisms through which PQ could induce the accumulation of abnormal proteins and neurodegeneration that could originate the cognitive decline produced by it and could help managing its degenerative effects.


Subject(s)
Paraquat/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Death/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , tau Proteins/antagonists & inhibitors , tau Proteins/metabolism
3.
Nat Commun ; 11(1): 3954, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770072

ABSTRACT

The systematic stabilization of protein-protein interactions (PPI) has great potential as innovative drug discovery strategy to target novel and hard-to-drug protein classes. The current lack of chemical starting points and focused screening opportunities limits the identification of small molecule stabilizers that engage two proteins simultaneously. Starting from our previously described virtual screening strategy to identify inhibitors of 14-3-3 proteins, we report a conceptual molecular docking approach providing concrete entries for discovery and rational optimization of stabilizers for the interaction of 14-3-3 with the carbohydrate-response element-binding protein (ChREBP). X-ray crystallography reveals a distinct difference in the binding modes between weak and general inhibitors of 14-3-3 complexes and a specific, potent stabilizer of the 14-3-3/ChREBP complex. Structure-guided stabilizer optimization results in selective, up to 26-fold enhancement of the 14-3-3/ChREBP interaction. This study demonstrates the potential of rational design approaches for the development of selective PPI stabilizers starting from weak, promiscuous PPI inhibitors.


Subject(s)
14-3-3 Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Drug Design , Drug Discovery , 14-3-3 Proteins/antagonists & inhibitors , 14-3-3 Proteins/ultrastructure , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/ultrastructure , Crystallography, X-Ray , Molecular Docking Simulation , Protein Binding/drug effects , Small Molecule Libraries , Structure-Activity Relationship
4.
Growth Horm IGF Res ; 53-54: 101332, 2020.
Article in English | MEDLINE | ID: mdl-32698101

ABSTRACT

OBJECTIVE: Growth hormone (GH) deficiency has been associated with increased steatosis but the molecular mechanism has not been fully elucidated. We investigated the effect of GH on lipid accumulation of HepG2 cells cultured on an in vitro steatosis model and examined the potential involvement of insulin-like growth factor 1 (IGF-1) as well as lipogenic and lipolytic molecules. METHODS: Control and steatosis conditions were induced by culturing HepG2 cells with 5.5 or 25 mmol/l glucose for 24 h, respectively. Afterward, cells were exposed to 0, 5, 10 or 20 ng/ml GH for another 24 h. Lipid content was quantified as well as mRNA and protein levels of IGF-1, carbohydrate responsive element-binding protein (ChREBP), sterol regulatory element-binding protein 1c (SREBP1c), fatty acid synthase (FAS), carnitine palmitoyltransferase 1A (CPT1A), and peroxisome proliferator-activated receptor alpha (PPAR-alpha) by qPCR and western blot, respectively. Data were analyzed by one-way ANOVA and the Games-Howell post-hoc test. RESULTS: In the steatosis model, HepG2 hepatocytes showed a significant 2-fold increase in lipid amount as compared to control cells. IGF-1 mRNA and protein levels were significantly increased in control cells exposed to 10 ng/ml GH, whereas high glucose abolished this effect. High glucose also significantly increased both mRNA and protein of ChREBP and FAS without having effect on SREBP1c, CPT1A and PPAR-alpha. However, GH inhibited ChREBP and FAS production, even in HepG2 hepatocytes cultured under steatosis conditions. CONCLUSIONS: Growth hormone ameliorates high glucose-induced steatosis in HepG2 cells by suppressing de novo lipogenesis via ChREBP and FAS down-regulation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Fatty Acid Synthases/antagonists & inhibitors , Glucose/adverse effects , Hepatocytes/drug effects , Human Growth Hormone/pharmacology , Lipogenesis , Non-alcoholic Fatty Liver Disease/prevention & control , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Sweetening Agents/adverse effects
5.
Nat Commun ; 11(1): 3612, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681035

ABSTRACT

Bile acid synthesis plays a key role in regulating whole body cholesterol homeostasis. Transcriptional factor EB (TFEB) is a nutrient and stress-sensing transcriptional factor that promotes lysosomal biogenesis. Here we report a role of TFEB in regulating hepatic bile acid synthesis. We show that TFEB induces cholesterol 7α-hydroxylase (CYP7A1) in human hepatocytes and mouse livers and prevents hepatic cholesterol accumulation and hypercholesterolemia in Western diet-fed mice. Furthermore, we find that cholesterol-induced lysosomal stress feed-forward activates TFEB via promoting TFEB nuclear translocation, while bile acid-induced fibroblast growth factor 19 (FGF19), acting via mTOR/ERK signaling and TFEB phosphorylation, feedback inhibits TFEB nuclear translocation in hepatocytes. Consistently, blocking intestinal bile acid uptake by an apical sodium-bile acid transporter (ASBT) inhibitor decreases ileal FGF15, enhances hepatic TFEB nuclear localization and improves cholesterol homeostasis in Western diet-fed mice. This study has identified a TFEB-mediated gut-liver signaling axis that regulates hepatic cholesterol and bile acid homeostasis.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Fibroblast Growth Factors/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Cell Line , Cholesterol 7-alpha-Hydroxylase/metabolism , Diet, Western/adverse effects , Disease Models, Animal , Hep G2 Cells , Humans , Hypercholesterolemia/etiology , Hypercholesterolemia/metabolism , Hypercholesterolemia/prevention & control , Ileum/drug effects , Ileum/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors
6.
Toxicol Appl Pharmacol ; 401: 115090, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32512069

ABSTRACT

Copper dyshomeostasis is involved in the pathogenesis of Alzheimer's disease (AD). Microglia play a major role in the proteolytic clearance of oligomeric ß-amyloid (Aßo). Here, we investigated whether Cu(II) affects microglial Aßo clearance and whether this effect involves autophagy-lysosomal pathway. Microtubule associated protein 1 light chain 3 (LC3)-II and p62 protein levels and autophagic flux in Cu(II)-treated microglia were detected. Aßo clearance was detected by enzyme-linked immunosorbent assay (ELISA) and immunofluorescence. In vivo, Cu(II) and Aßo were injected into mouse hippocampus to evaluate Aß clearance. The results showed that Cu(II) inhibited phagocytic uptake and intracellular degradation of Aßo in microglial cultures. Additionally, Cu(II) elevated LC3-II and p62 protein levels and impaired autophagic flux. It also inhibited transcription factor EB (TFEB) expression and lysosomal biogenesis. Moreover, Cu(II) activated mammalian target of rapamycin kinase (mTOR), an upstream signaling of TFEB. The mTOR inhibitor PP242 ameliorated Cu(II)-impaired TFEB expression, lysosomal biogenesis, autophagic flux, and Aßo clearance in microglia. In vivo, Cu(II) inhibited microglial Aßo clearance in mouse hippocampus, an effect accompanied with activation of mTOR and impairment of TFEB expression and lysosomal biogenesis. Collectively, our results suggest that Cu(II) reduces microglial Aßo clearance through disrupting lysosomal biogenesis and autophagic flux. This effect could involve modulation of mTOR-TFEB axis and was prevented by pharmacological antagonism of mTOR. This study reveals a novel mechanism for Cu(II) involvement in AD. Our results implicate that rescue of Cu(II)-impaired autophagy-mediated lysosomal degradation may provide a new strategy to benefit multiple neurodegenerative disorders.


Subject(s)
Amyloid beta-Peptides/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Copper/toxicity , Lysosomes/metabolism , Microglia/metabolism , Peptide Fragments/metabolism , TOR Serine-Threonine Kinases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Animals, Newborn , Autophagy/drug effects , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Lysosomes/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Peptide Fragments/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/antagonists & inhibitors
7.
Life Sci ; 246: 117418, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32057899

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), which is characterized by disruption of lipid homeostasis, has been the leading cause of chronic liver disease worldwide. However, currently there is no effective therapy for NAFLD. Consequently, it is extremely urgent to explore the specific and effective target functioned as lipids regulator during the pathological process of NAFLD for the drug development. Transcription factor EB (TFEB) plays a crucial role in the regulation of lipid homeostasis through linking autophagy to energy metabolism at the transcriptional level. In this review, we summarize the currently available information regarding the mediation of TFEB in lipid metabolism during the pathological process of NAFLD, and the specific regulatory mechanism of TFEB activity. We further recapitulate TFEB as a promising therapeutic target for NAFLD, primarily through the regulation of lipid homeostasis, energy metabolism as well as immune defense. A better understanding of these key issues will be helpful to promote the development of therapeutic agents which specifically target TFEB to halt or reverse the pathological progression of NAFLD.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Lipid Metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Humans , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/metabolism
8.
Mol Cell ; 77(6): 1251-1264.e9, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32023484

ABSTRACT

Lipid droplets (LDs) store lipids for energy and are central to cellular lipid homeostasis. The mechanisms coordinating lipid storage in LDs with cellular metabolism are unclear but relevant to obesity-related diseases. Here we utilized genome-wide screening to identify genes that modulate lipid storage in macrophages, a cell type involved in metabolic diseases. Among ∼550 identified screen hits is MLX, a basic helix-loop-helix leucine-zipper transcription factor that regulates metabolic processes. We show that MLX and glucose-sensing family members MLXIP/MondoA and MLXIPL/ChREBP bind LDs via C-terminal amphipathic helices. When LDs accumulate in cells, these transcription factors bind to LDs, reducing their availability for transcriptional activity and attenuating the response to glucose. Conversely, the absence of LDs results in hyperactivation of MLX target genes. Our findings uncover a paradigm for a lipid storage response in which binding of MLX transcription factors to LD surfaces adjusts the expression of metabolic genes to lipid storage levels.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Glucose/metabolism , Lipid Droplets/metabolism , Proteome/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Genetic Testing , Humans , Macrophages/cytology , Macrophages/metabolism , Protein Binding , Proteome/analysis , RNA, Small Interfering , Transcription, Genetic
9.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118610, 2020 02.
Article in English | MEDLINE | ID: mdl-31738957

ABSTRACT

OBJECTIVE: In muscle cells, the peroxisome proliferator-activated receptor γ co-activator 1 (PGC-1) signaling network, which has been shown to be disturbed in the skeletal muscle in several chronic diseases, tightly controls mitochondrial biogenesis and oxidative substrate metabolism. Previously, we showed that inactivation of glycogen synthase kinase (GSK)-3ß potently increased Pgc-1α abundance and oxidative metabolism in skeletal muscle cells. The current study aims to unravel the molecular mechanism driving the increase in Pgc-1α mediated by GSK-3ß inactivation. METHODS: GSK-3ß was inactivated genetically or pharmacologically in C2C12 myotubes and the requirement of transcription factors known to be involved in Pgc-1α transcription for increases in Pgc-1α abundance mediated by inactivation of GSK-3ß was examined. RESULTS: Enhanced PGC-1α promoter activation after GSK-3ß inhibition suggested a transcriptionally-controlled mechanism. While myocyte enhancer factor (MEF)2 transcriptional activity was unaltered, GSK-3ß inactivation increased the abundance and activity of the transcription factors estrogen-related receptor (ERR)α and ERRγ. Pharmacological inhibition or knock-down of ERRα and ERRγ however failed to prevent increases in Pgc-1α mRNA mediated by GSK-3ß inactivation. Interestingly, GSK-3ß inactivation activated transcription factor EB (TFEB), evidenced by decreased phosphorylation and enhanced nuclear localization of the TFEB protein. Moreover, knock-down of TFEB completely prevented increases in Pgc-1α gene expression, PGC-1α promoter activity and PGC-1α protein abundance induced by GSK-3ß inactivation. Furthermore, mutation of a specific TFEB binding site on the PGC-1α promoter blocked promoter activation upon inhibition of GSK-3ß. CONCLUSIONS: In skeletal muscle, GSK-3ß inactivation causes dephosphorylation and nuclear translocation of TFEB resulting in TFEB-dependent induction of Pgc-1α expression.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Binding Sites , Cell Line , Cell Nucleus/metabolism , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phosphorylation , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Signal Transduction , Transcriptional Activation , Up-Regulation , ERRalpha Estrogen-Related Receptor
10.
J Nutr Biochem ; 73: 108214, 2019 11.
Article in English | MEDLINE | ID: mdl-31520816

ABSTRACT

Formononetin has been reported to ameliorate hyperlipidemia and obesity, but its effect and mechanism of action in anti-non-alcoholic fatty liver disease (NAFLD) remain unclear. Lipophagy is a critical protective mechanism during steatosis development that results in the decomposition of lipid droplets through autophagy and the prevention of cellular lipid accumulation. This study aimed to investigate the beneficial role of formononetin in treating NAFLD and explore the mechanism of lipophagy in formononetin anti-hepatic steatosis effects. Formononetin treatment significantly ameliorated hepatic steatosis in HFD mice. Consistently, formononetin also reduced FFAs-stimulated lipid accumulation in HepG2 cells and primary mouse hepatocytes. Further analysis revealed that steatosis increased LC3B-II, a marker of autophagy, but caused blockade of autophagic flux associated with a lack of lysosomes. Treatment with formononetin promoted lysosome biogenesis and autophagosome-lysosome fusion, relieving the blockade in autophagic flux and further induced lipophagy. Mechanistically, formononetin activated adenosine monophosphate activated protein kinase (AMPK) and promoted subsequent nuclear translocation of transcription factor EB (TFEB), a key regulator of lysosome biogenesis. TFEB inhibition markedly abolished formononetin-induced lysosome biogenesis, autophagosome-lysosome fusion and lipophagy and concomitantly alleviated lipid accumulation. Formononetin improved hepatic steatosis via TFEB-mediated lysosome biogenesis, which provides new evidence regarding formononetin's anti-NAFLD effects.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Isoflavones/therapeutic use , Lysosomes/physiology , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified/pharmacology , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Isoflavones/pharmacology , Lipid Metabolism/drug effects , Lysosomes/drug effects , Mice , Non-alcoholic Fatty Liver Disease/etiology , Organelle Biogenesis
11.
Biotechnol Appl Biochem ; 66(4): 555-563, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30994947

ABSTRACT

Although cilostazol was proved to have antitumor biological effects, its function in myocardial ischemia and reperfusion (I/R) injury and the underlying mechanisms were not fully illustrated yet. In this study, a rat model of I/R injury was constructed and quantitative real-time PCR, Western blot, and immunofluorescence (IF) assay were performed. Our results showed that cilostazol increased LC3 II/LC3 I ratio, reduced p62 abundance, and promoted the expressions of LAMP1, LAMP2, cathepsin B, and cathepsin D, indicating that cilostazol could activate autophagy and elevated lysosome activation. Following analysis showed that cilostazol enhanced nuclear protein expression of transcription factor EB (TFEB), an important regulator of autophagy-lysosome pathway. Furthermore, CCI-779, an inhibitor of TFEB, could reverse the effects of cilostazol on autophagic activity and lysosome activation. Importantly, cilostazol suppressed I/R injury-induced apoptosis by decreasing the cleavage of caspase 3 and PARP. Enzyme-linked immunosorbent assay showed that cilostazol reduced the serum levels of CTn1 and CK-MB and decreased infract size caused by I/R injuries. Altogether this study suggested that cilostazol protects against I/R injury by regulating autophagy, lysosome, and apoptosis in a rat model of I/R injury. The protective mechanism of cilostazol was partially through increasing the transcriptional activity of TFEB.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Cilostazol/pharmacology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Animals , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Disease Models, Animal , Lysosomes/drug effects , Myocardial Reperfusion Injury/surgery , Rats , Rats, Sprague-Dawley , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
12.
Cell Death Dis ; 10(3): 242, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858361

ABSTRACT

Cystic fibrosis is a disease caused by defective function of a chloride channel coupled to a blockade of autophagic flux. It has been proposed to use synthetic chloride transporters as pharmacological agents to compensate insufficient chloride fluxes. Here, we report that such chloride anionophores block autophagic flux in spite of the fact that they activate the pro-autophagic transcription factor EB (TFEB) coupled to the inhibition of the autophagy-suppressive mTORC1 kinase activity. Two synthetic chloride transporters (SQ1 and SQ2) caused a partially TFEB-dependent relocation of the autophagic marker LC3 to the Golgi apparatus. Inhibition of TFEB activation using a calcium chelator or calcineurin inhibitors reduced the formation of LC3 puncta in cells, yet did not affect the cytotoxic action of SQ1 and SQ2 that could be observed after prolonged incubation. In conclusion, the squaramide-based synthetic chloride transporters studied in this work (which can also dissipate pH gradients) are probably not appropriate for the treatment of cystic fibrosis yet might be used for other indications such as cancer.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Hydrocarbons, Fluorinated/pharmacology , Ion Transport/drug effects , TOR Serine-Threonine Kinases/metabolism , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Death , Cell Line, Tumor , Golgi Apparatus/drug effects , Humans , Hydrocarbons, Fluorinated/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/genetics , Up-Regulation
13.
Autophagy ; 15(3): 493-509, 2019 03.
Article in English | MEDLINE | ID: mdl-30304977

ABSTRACT

Mounting attention has been focused on defects in macroautophagy/autophagy and the autophagy-lysosomal pathway (ALP) in cerebral ischemia. TFEB (transcription factor EB)-mediated induction of ALP has been recently considered as the common mechanism in ameliorating the pathological lesion of myocardial ischemia and neurodegenerative diseases. Here we explored the vital role of TFEB in permanent middle cerebral artery occlusion (pMCAO)-mediated dysfunction of ALP and ischemic insult in rats. The results showed that ALP function was first enhanced in the early stage of the ischemic process, especially in neurons of the cortex, and this was accompanied by increased TFEB expression and translocation to the nucleus, which was mediated at least in part through activation by PPP3/calcineurin. At the later stages of ischemia, a gradual decrease in the level of nuclear TFEB was coupled with a progressive decline in lysosomal activity, accumulation of autophagosomes and autophagy substrates, and exacerbation of the ischemic injury. Notably, neuron-specific overexpression of TFEB significantly enhanced ALP function and rescued the ischemic damage, starting as early as 6 h and even lasting to 48 h after ischemia. Furthermore, neuron-specific knockdown of TFEB markedly reversed the activation of ALP and further aggravated the neurological deficits and ischemic outcome at the early stage of pMCAO. These results highlight neuronal-targeted TFEB as one of the key players in the pMCAO-mediated dysfunction of ALP and ischemic injury, and identify TFEB as a promising target for therapies aimed at neuroprotection in cerebral ischemia. Abbreviations: AAV, adeno-associated virus; AIF1/IBA1, allograft inflammatory factor 1; ALP, autophagy-lysosomal pathway; CQ, chloroquine; CTSB, cathepsin B; CTSD, cathepsin D; CsA, cyclosporin A; GFAP, glial fibrillary acidic protein; LAMP, lysosomal-associated membrane protein; LC3, microtubule-associated protein 1 light chain 3; MAP2, microtubule-associated protein 2; mNSS, modified Neurological Severity Score; MTOR, mechanistic target of rapamycin kinase; OGD, oxygen and glucose deprivation; pMCAO, permanent middle cerebral artery occlusion; RBFOX3/NeuN, RNA binding fox-1 homolog 3; SQSTM1, sequestosome1; TFEB, transcription factor EB; TTC, 2,3,5-triphenyltetrazolium chloride.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Brain Ischemia/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Lysosomes/metabolism , Neurons/metabolism , Animals , Autophagosomes/metabolism , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Brain/enzymology , Brain/pathology , Brain Ischemia/diagnostic imaging , Brain Ischemia/enzymology , Brain Ischemia/pathology , Calcineurin/metabolism , Cell Nucleus/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/surgery , Lysosomes/enzymology , Male , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation
14.
Clin Cancer Res ; 24(23): 5977-5989, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30061365

ABSTRACT

PURPOSE: Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease. EXPERIMENTAL DESIGN: We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models. RESULTS: The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3-tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation. CONCLUSIONS: These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3-tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3-tRCC.


Subject(s)
Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/metabolism , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Kidney Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Animals , Antineoplastic Agents/therapeutic use , Binding Sites , Biomarkers, Tumor , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Male , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Binding , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Sci Rep ; 8(1): 10064, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968736

ABSTRACT

MYC is a key player in tumor development, but unfortunately no specific MYC-targeting drugs are clinically available. MYC is strictly dependent on heterodimerization with MAX for transcription activation. Aiming at targeting this interaction, we identified MYCMI-6 in a cell-based protein interaction screen for small inhibitory molecules. MYCMI-6 exhibits strong selective inhibition of MYC:MAX interaction in cells and in vitro at single-digit micromolar concentrations, as validated by split Gaussia luciferase, in situ proximity ligation, microscale thermophoresis and surface plasmon resonance (SPR) assays. Further, MYCMI-6 blocks MYC-driven transcription and binds selectively to the MYC bHLHZip domain with a KD of 1.6 ± 0.5 µM as demonstrated by SPR. MYCMI-6 inhibits tumor cell growth in a MYC-dependent manner with IC50 concentrations as low as 0.5 µM, while sparing normal cells. The response to MYCMI-6 correlates with MYC expression based on data from 60 human tumor cell lines and is abrogated by MYC depletion. Further, it inhibits MYC:MAX interaction, reduces proliferation and induces massive apoptosis in tumor tissue from a MYC-driven xenograft tumor model without severe side effects. Since MYCMI-6 does not affect MYC expression, it is a unique molecular tool to specifically target MYC:MAX pharmacologically and it has good potential for drug development.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Diamines/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Pyridines/pharmacology , Animals , Apoptosis/physiology , COS Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Drug Screening Assays, Antitumor , HEK293 Cells , HeLa Cells , High-Throughput Screening Assays/methods , Humans , MCF-7 Cells , Mice , Mice, Nude , Protein Binding/drug effects , Small Molecule Libraries/pharmacology , Transcriptional Activation , Xenograft Model Antitumor Assays
16.
Oxid Med Cell Longev ; 2018: 8067592, 2018.
Article in English | MEDLINE | ID: mdl-29765503

ABSTRACT

Age-associated diseases such as neurodegenerative and cardiovascular disorders are characterized by increased oxidative stress associated with autophagy dysfunction. Oleuropein aglycone (OA), the main polyphenol found in olive oil, was recently characterized as an autophagy inducer and a promising agent against neurodegeneration. It is presently unknown whether OA can have beneficial effects in a model of cardiac stress characterized by autophagy dysfunction. Here, we explored the effects of OA in cardiomyocytes with overexpression of monoamine oxidase-A (MAO-A). This enzyme, by degrading catecholamine and serotonin, produces hydrogen peroxide (H2O2), which causes oxidative stress, autophagic flux blockade, and cell necrosis. We observed that OA treatment counteracted the cytotoxic effects of MAO-A through autophagy activation, as displayed by the increase of autophagic vacuoles and autophagy-specific markers (Beclin1 and LC3-II). Moreover, the decrease in autophagosomes and the increase in autolysosomes, indicative of autophagosome-lysosome fusion, suggested a restoration of the defective autophagic flux. Most interestingly, we found that the ability of OA to confer cardioprotection through autophagy induction involved nuclear translocation and activation of the transcriptional factor EB (TFEB). Our data provide strong evidence of the beneficial effects of OA, suggesting its potential use as a nutraceutical agent against age-related pathologies involving autophagy dysfunction, including cardiovascular diseases.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Iridoids/pharmacology , Monoamine Oxidase/metabolism , Protective Agents/pharmacology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Nucleus/metabolism , Iridoid Glucosides , Microscopy, Fluorescence , Monoamine Oxidase/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Reactive Oxygen Species/metabolism , Tyramine/pharmacology
17.
Diabetes ; 67(3): 461-472, 2018 03.
Article in English | MEDLINE | ID: mdl-29282201

ABSTRACT

Although the mechanisms by which glucose regulates insulin secretion from pancreatic ß-cells are now well described, the way glucose modulates gene expression in such cells needs more understanding. Here, we demonstrate that MondoA, but not its paralog carbohydrate-responsive element-binding protein, is the predominant glucose-responsive transcription factor in human pancreatic ß-EndoC-ßH1 cells and in human islets. In high-glucose conditions, MondoA shuttles to the nucleus where it is required for the induction of the glucose-responsive genes arrestin domain-containing protein 4 (ARRDC4) and thioredoxin interacting protein (TXNIP), the latter being a protein strongly linked to ß-cell dysfunction and diabetes. Importantly, increasing cAMP signaling in human ß-cells, using forskolin or the glucagon-like peptide 1 mimetic Exendin-4, inhibits the shuttling of MondoA and potently inhibits TXNIP and ARRDC4 expression. Furthermore, we demonstrate that silencing MondoA expression improves glucose uptake in EndoC-ßH1 cells. These results highlight MondoA as a novel target in ß-cells that coordinates transcriptional response to elevated glucose levels.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Second Messenger Systems , Active Transport, Cell Nucleus/drug effects , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cyclic AMP/metabolism , Exenatide , Gene Expression Regulation/drug effects , Humans , Incretins/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Peptides/pharmacology , RNA Interference , Second Messenger Systems/drug effects , Thioredoxins/genetics , Thioredoxins/metabolism , Tissue Culture Techniques , Venoms/pharmacology
18.
Autophagy ; 14(2): 269-282, 2018.
Article in English | MEDLINE | ID: mdl-29172997

ABSTRACT

Bleomycin is a clinically potent anticancer drug used for the treatment of germ-cell tumors, lymphomas and squamous-cell carcinomas. Unfortunately, the therapeutic efficacy of bleomycin is severely hampered by the development of pulmonary fibrosis. However, the mechanisms underlying bleomycin-induced pulmonary fibrosis, particularly the molecular target of bleomycin, remains unknown. Here, using a chemical proteomics approach, we identify ANXA2 (annexin A2) as a direct binding target of bleomycin. The interaction of bleomycin with ANXA2 was corroborated both in vitro and in vivo. Genetic depletion of anxa2 in mice mitigates bleomycin-induced pulmonary fibrosis. We further demonstrate that Glu139 (E139) of ANXA2 is required for bleomycin binding in lung epithelial cells. A CRISPR-Cas9-engineered ANXA2E139A mutation in lung epithelial cells ablates bleomycin binding and activates TFEB (transcription factor EB), a master regulator of macroautophagy/autophagy, resulting in substantial acceleration of autophagic flux. Pharmacological activation of TFEB elevates bleomycin-initiated autophagic flux, inhibits apoptosis and proliferation of epithelial cells, and ameliorates pulmonary fibrosis in bleomycin-treated mice. Notably, we observe lowered TFEB and LC3B levels in human pulmonary fibrosis tissues compared to normal controls, suggesting a critical role of TFEB-mediated autophagy in pulmonary fibrosis. Collectively, our data demonstrate that ANXA2 is a specific bleomycin target, and bleomycin binding with ANXA2 impedes TFEB-induced autophagic flux, leading to induction of pulmonary fibrosis. Our findings provide insight into the mechanisms of bleomycin-induced fibrosis and may facilitate development of optimized bleomycin therapeutics devoid of lung toxicity.


Subject(s)
Annexin A2/metabolism , Antibiotics, Antineoplastic/adverse effects , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Bleomycin/adverse effects , Pulmonary Fibrosis/chemically induced , A549 Cells , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Annexin A2/genetics , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Proteomics
19.
Acta Pharmacol Sin ; 38(9): 1305-1316, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28603284

ABSTRACT

Transcription factor EB (TFEB) is a master regulator of autophagy activity and lysosomal biogenesis, but its role in autophagy-mediated cell survival and chemotherapy resistance is not completely understood. In this study, we explored whether TFEB played an important role in autophagy-mediated chemotherapy resistance in human cancer LoVo and HeLa cells in vitro. Treatment of human colon cancer LoVo cells with doxorubicin (0.5 µmol/L) induced autophagy activation and nuclear translocation of TFEB, which resulted from inactivation of the mTOR pathway. In both LoVo and HeLa cells, overexpression of TFEB enhanced doxorubicin-induced autophagy activation and significantly decreased doxorubicin-induced cell death, whereas knockdown of TFEB with small interfering RNA blocked doxorubicin-induced autophagy and significantly enhanced the cytotoxicity of doxorubicin. In LoVo cells, autophagy inhibition by 3-methyladenine (3-MA) or knockdown of autophagy-related gene Atg5 increased cell death in response to doxorubicin, and abolished TFEB overexpression-induced chemotherapy resistance, suggesting that the inhibition of autophagy made cancer cells more sensitive to doxorubicin. The results demonstrate that TFEB-mediated autophagy activation decreases the sensitivity of cancer cells to doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Antibiotics, Antineoplastic/chemistry , Apoptosis/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/analysis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Cell Survival/drug effects , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
20.
Exp Biol Med (Maywood) ; 242(11): 1185-1197, 2017 06.
Article in English | MEDLINE | ID: mdl-28474989

ABSTRACT

The Wnt/ß-catenin signaling pathway typically shows aberrant activation in various cancer cells, especially colorectal cancer cells. This signaling pathway regulates the expression of a variety of tumor-related proteins, including c-myc and cyclin D1, and plays essential roles in tumorigenesis and in the development of many cancers. Small molecules that block the interactions between ß-catenin and Tcf4, a downstream stage of activation of the Wnt/ß-catenin signaling pathway, could efficiently cut off this signal transduction and thereby act as a novel class of anticancer drugs. This paper reviews the currently reported inhibitors that target ß-catenin/Tcf4 interactions, focusing on the discovery approaches taken in the design of these inhibitors and their bioactivities. A brief perspective is then shared on the future discovery and development of this class of inhibitors. Impact statement This mini-review summarized the current knowledge of inhibitors of interactions of beta-catenin/Tcf4 published to date according to their discovery approaches, and discussed their in vitro and in vivo activities, selectivities, and pharmacokinetic properties. Several reviews presently available now in this field describe modulators of the Wnt/beta-catenin pathway, but are generally focused on the bioactivities of these inhibitors. By contrast, this review focused on the drug discovery approaches taken in identifying these types of inhibitors and provided our perspective on further strategies for future drug discoveries. This review also integrated many recently published and important works on highly selective inhibitors as well as rational drug design. We believe that the findings and strategies summarized in this review have broad implications and will be of interest throughout the biochemical and pharmaceutical research community.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Drug Discovery , Transcription Factors/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Animals , Drug Discovery/trends , Humans , Protein Binding/drug effects , Transcription Factor 4
SELECTION OF CITATIONS
SEARCH DETAIL
...