Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 160
1.
Proc Natl Acad Sci U S A ; 119(31): e2205469119, 2022 08 02.
Article En | MEDLINE | ID: mdl-35895684

T regulatory (Treg) cells are essential for self-tolerance whereas they are detrimental for dampening the host anti-tumor immunity. How Treg cells adapt to environmental signals to orchestrate their homeostasis and functions remains poorly understood. Here, we identified that transcription factor EB (TFEB) is induced by host nutrition deprivation or interleukin (IL)-2 in CD4+ T cells. The loss of TFEB in Treg cells leads to reduced Treg accumulation and impaired Treg function in mouse models of cancer and autoimmune disease. TFEB intrinsically regulates genes involved in Treg cell differentiation and mitochondria function while it suppresses expression of proinflammatory cytokines independently of its established roles in autophagy. This coordinated action is required for mitochondria integrity and appropriate lipid metabolism in Treg cells. These findings identify TFEB as a critical regulator for orchestrating Treg generation and function, which may contribute to the adaptive responses of T cells to local environmental cues.


Adaptation, Physiological , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Mitochondria , Neoplasms , T-Lymphocytes, Regulatory , Adaptation, Physiological/genetics , Adaptation, Physiological/physiology , Animals , Autoimmune Diseases/immunology , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Disease Models, Animal , Interleukin-2/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Xenograft Model Antitumor Assays
2.
J Clin Invest ; 131(22)2021 11 15.
Article En | MEDLINE | ID: mdl-34779410

Growing tumors exist in metabolically compromised environments that require activation of multiple pathways to scavenge nutrients to support accelerated rates of growth. The folliculin (FLCN) tumor suppressor complex (FLCN, FNIP1, FNIP2) is implicated in the regulation of energy homeostasis via 2 metabolic master kinases: AMPK and mTORC1. Loss-of-function mutations of the FLCN tumor suppressor complex have only been reported in renal tumors in patients with the rare Birt-Hogg-Dube syndrome. Here, we revealed that FLCN, FNIP1, and FNIP2 are downregulated in many human cancers, including poor-prognosis invasive basal-like breast carcinomas where AMPK and TFE3 targets are activated compared with the luminal, less aggressive subtypes. FLCN loss in luminal breast cancer promoted tumor growth through TFE3 activation and subsequent induction of several pathways, including autophagy, lysosomal biogenesis, aerobic glycolysis, and angiogenesis. Strikingly, induction of aerobic glycolysis and angiogenesis in FLCN-deficient cells was dictated by the activation of the PGC-1α/HIF-1α pathway, which we showed to be TFE3 dependent, directly linking TFE3 to Warburg metabolic reprogramming and angiogenesis. Conversely, FLCN overexpression in invasive basal-like breast cancer models attenuated TFE3 nuclear localization, TFE3-dependent transcriptional activity, and tumor growth. These findings support a general role of a deregulated FLCN/TFE3 tumor suppressor pathway in human cancers.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Breast Neoplasms/pathology , Neovascularization, Pathologic/prevention & control , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Warburg Effect, Oncologic , AMP-Activated Protein Kinases/physiology , Cell Line, Tumor , Female , Humans , Oxidative Phosphorylation
3.
Mediators Inflamm ; 2021: 2689600, 2021.
Article En | MEDLINE | ID: mdl-34733115

Chronic obstructive pulmonary disease (COPD), a progressive respiratory disease, is characterized by the alveolar epithelium injury and persistent airway inflammation. It is documented that oscillation and dysregulated expression of circadian clock genes, like Bmal1, Per1, and Per2, involved in COPD pathogenies, including chronic inflammation and imbalanced autophagy level, and targeting the associations of circadian rhythm and autophagy is promising strategies in the management and treatment of COPD. Herein, we reviewed the mechanisms of the circadian clock and the unbalance of the autophagic level in COPD, as well as the link between the two, so as to provide further theoretical bases for the study on the pathogenesis of COPD.


Autophagy/physiology , Circadian Clocks/physiology , Pulmonary Disease, Chronic Obstructive/etiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Circadian Clocks/genetics , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Melatonin/physiology
4.
Neurobiol Aging ; 106: 26-36, 2021 10.
Article En | MEDLINE | ID: mdl-34229273

Identification of molecules and molecular pathways that can ameliorate aging-associated decline in cognitive function is crucial. Here we report that the protein levels of transcription factor EB (TFEB) were markedly reduced in both the cytosolic and nuclear fractions of the frontal cortex and hippocampus at 18-months of age relative to 6 months in the normal male wild-type mice. In the transgenic mice with ectopic expression of flag-TFEB in neurons, we observed that the levels of actin-normalized PGC1α and mtTFA were significantly increased in both the cortex and the hippocampus. Additionally, we confirmed increased mitochondria numbers in the flag-TFEB mice by transmission electron microscopy. Most importantly, TFEB expression in the 18-month-old transgenic mice mitigated markers of senescence including P16INK4a, γ-H2AX, and lamin B1, and improved memory skills implying that TFEB may exert an anti-aging effect by modulating neuronal senescence. Taken together these data strongly support that TFEB can be a useful therapeutic target for brain senescent cells to help overcome the age-related issues in cognition and possibly, achieve healthy aging.


Aging/genetics , Aging/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Biomarkers/metabolism , Brain/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression/genetics , Memory Disorders/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Histones/metabolism , Memory Disorders/therapy , Mice, Transgenic , Molecular Targeted Therapy
5.
Int J Mol Sci ; 22(11)2021 May 23.
Article En | MEDLINE | ID: mdl-34071043

A de novo missense variant in Rag GTPase protein C (RagCS75Y) was recently identified in a syndromic dilated cardiomyopathy (DCM) patient. However, its pathogenicity and the related therapeutic strategy remain unclear. We generated a zebrafish RragcS56Y (corresponding to human RagCS75Y) knock-in (KI) line via TALEN technology. The KI fish manifested cardiomyopathy-like phenotypes and poor survival. Overexpression of RagCS75Y via adenovirus infection also led to increased cell size and fetal gene reprogramming in neonatal rat ventricle cardiomyocytes (NRVCMs), indicating a conserved mechanism. Further characterization identified aberrant mammalian target of rapamycin complex 1 (mTORC1) and transcription factor EB (TFEB) signaling, as well as metabolic abnormalities including dysregulated autophagy. However, mTOR inhibition failed to ameliorate cardiac phenotypes in the RagCS75Y cardiomyopathy models, concomitant with a failure to promote TFEB nuclear translocation. This observation was at least partially explained by increased and mTOR-independent physical interaction between RagCS75Y and TFEB in the cytosol. Importantly, TFEB overexpression resulted in more nuclear TFEB and rescued cardiomyopathy phenotypes. These findings suggest that S75Y is a pathogenic gain-of-function mutation in RagC that leads to cardiomyopathy. A primary pathological step of RagCS75Y cardiomyopathy is defective mTOR-TFEB signaling, which can be corrected by TFEB overexpression, but not mTOR inhibition.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Cardiomyopathy, Dilated/genetics , Gain of Function Mutation , Monomeric GTP-Binding Proteins/genetics , Mutation, Missense , Point Mutation , TOR Serine-Threonine Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus , Amino Acid Substitution , Animals , Autophagy , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cardiomyopathy, Dilated/therapy , Cells, Cultured , Gene Knock-In Techniques , Gene Knockout Techniques , Heart Ventricles/cytology , Humans , Mice , Monomeric GTP-Binding Proteins/physiology , Myocytes, Cardiac/metabolism , Phenotype , Rats, Wistar , Recombinant Proteins/metabolism , Signal Transduction , Transcription Activator-Like Effector Nucleases , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
6.
Metabolism ; 120: 154798, 2021 07.
Article En | MEDLINE | ID: mdl-33984335

BACKGROUND: Recent studies have shown that dysregulation of autophagy is involved in the development of nonalcoholic fatty liver disease (NAFLD). Transcription factors E3 (TFE3) and EB (TFEB) are master regulators of the transcriptional response of basic cellular processes such as lysosomal biogenesis and autophagy. Here, we investigated the role of fenofibrate, a PPARα agonist, in promotion of intracellular lipid clearance by upregulation of TFEB/TFE3. METHODS: We investigated whether the effects of fenofibrate on livers were dependent on TFEB in high fat diet (HFD)-fed mice and in vivo Tfeb knockdown mice. These mice were analyzed for characteristics of obesity and diabetes; the effects of fenofibrate on hepatic fat content, glucose sensitivity, insulin resistance, and autophagy functional dependence on TFEB were investigated. HepG2, Hep3B, TSC2+/+ and tsc2-/- MEFs, tfeb wild type- and tfeb knockout-HeLa cells were used for in vitro experiments. RESULTS: Fenofibrate treatment activated autophagy and TFEB/TFE3 and reduced hepatic fat accumulation in an mTOR-independent manner. Knockdown of TFEB offset the effects of fenofibrate on autophagy and hepatic fat accumulation. In addition, fenofibrate treatment induced lysosomal Ca2+ release through mucolipin 1, activated calcineurin and the CaMKKß-AMPK-ULK1 pathway, subsequently promoted TFEB and TFE3 dephosphorylation and nuclear translocation. Treatment with calcium chelator or knockdown of mucolipin 1 in hepatocytes offset the effects of fenofibrate treatment on autophagy and hepatic fat accumulation. CONCLUSION: Activation of PPARα ameliorates hepatic fat accumulation via activation of TFEB and lipophagy induction. Lysosomal calcium signaling appears to play a critical role in this process. In addition, activation of TFEB by modulating nuclear receptors including PPARα with currently available drugs or new molecules might be a therapeutic target for treatment of NAFLD and other cardiometabolic diseases.


Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Fenofibrate/pharmacology , Liver/drug effects , Animals , Autophagy/genetics , Cells, Cultured , HeLa Cells , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/metabolism , Mice , Mice, Knockout , PPAR alpha/agonists , Signal Transduction/drug effects , Up-Regulation/drug effects
7.
Autophagy ; 17(11): 3740-3752, 2021 11.
Article En | MEDLINE | ID: mdl-33706671

The autophagy-lysosome system is an important cellular degradation pathway that recycles dysfunctional organelles and cytotoxic protein aggregates. A decline in this system is pathogenic in many human diseases including neurodegenerative disorders, fatty liver disease, and atherosclerosis. Thus there is intense interest in discovering therapeutics aimed at stimulating the autophagy-lysosome system. Trehalose is a natural disaccharide composed of two glucose molecules linked by a ɑ-1,1-glycosidic bond with the unique ability to induce cellular macroautophagy/autophagy and with reported efficacy on mitigating several diseases where autophagy is dysfunctional. Interestingly, the mechanism by which trehalose induces autophagy is unknown. One suggested mechanism is its ability to activate TFEB (transcription factor EB), the master transcriptional regulator of autophagy-lysosomal biogenesis. Here we describe a potential mechanism involving direct trehalose action on the lysosome. We find trehalose is endocytically taken up by cells and accumulates within the endolysosomal system. This leads to a low-grade lysosomal stress with mild elevation of lysosomal pH, which acts as a potent stimulus for TFEB activation and nuclear translocation. This process appears to involve inactivation of MTORC1, a known negative regulator of TFEB which is sensitive to perturbations in lysosomal pH. Taken together, our data show the trehalose can act as a weak inhibitor of the lysosome which serves as a trigger for TFEB activation. Our work not only sheds light on trehalose action but suggests that mild alternation of lysosomal pH can be a novel method of inducing the autophagy-lysosome system.Abbreviations: ASO: antisense oligonucleotide; AU: arbitrary units; BMDM: bone marrow-derived macrophages; CLFs: crude lysosomal fractions; CTSD: cathepsin D; LAMP: lysosomal associated membrane protein; LIPA/LAL: lipase A, lysosomal acid type; MAP1LC3: microtubule-associated protein 1 light chain 3; MFI: mean fluorescence intensity; MTORC1: mechanistic target of rapamycin kinase complex 1; pMAC: peritoneal macrophages; SLC2A8/GLUT8: solute carrier family 2, (facilitated glucose transporter), member 8; TFEB: transcription factor EB; TMR: tetramethylrhodamine; TREH: trehalase.


Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Trehalose/metabolism , Animals , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Blotting, Western , Endocytosis , Fluorescent Antibody Technique , Gas Chromatography-Mass Spectrometry , Lysosomes/physiology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Trehalose/physiology
8.
Autophagy ; 17(11): 3776-3793, 2021 11.
Article En | MEDLINE | ID: mdl-33706682

Preconditioning with a mild stressor such as fasting is a promising way to reduce severe side effects from subsequent chemo- or radiotherapy. However, the underlying mechanisms have been largely unexplored. Here, we demonstrate that the TP53/p53-FBXO22-TFEB (transcription factor EB) axis plays an essential role in this process through upregulating basal macroautophagy/autophagy. Mild stress-activated TP53 transcriptionally induced FBXO22, which in turn ubiquitinated KDM4B (lysine-specific demethylase 4B) complexed with MYC-NCOR1 suppressors for degradation, leading to transcriptional induction of TFEB. Upregulation of autophagy-related genes by increased TFEB dramatically enhanced autophagic activity and cell survival upon following a severe stressor. Mitogen-induced AKT1 activation counteracted this process through the phosphorylation of KDM4B, which inhibited FBXO22-mediated ubiquitination. Additionally, fbxo22-/- mice died within 10 h of birth, and their mouse embryonic fibroblasts (MEFs) showed a lowered basal autophagy, whereas FBXO22-overexpressing mice were resistant to chemotherapy. Taken together, these results suggest that TP53 upregulates basal autophagy through the FBXO22-TFEB axis, which governs the hormetic effect in chemotherapy.Abbreviations: BBC3/PUMA: BCL2 binding component 3; CDKN1A/p21: cyclin dependent kinase inhibitor 1A; ChIP-seq: chromatin immunoprecipitation followed by sequencing; DDB2: damage specific DNA binding protein 2; DRAM: DNA damage regulated autophagy modulator; ESR/ER: estrogen receptor 1; FMD: fasting mimicking diet; HCQ: hydroxychloroquine; KDM4B: lysine-specific demethylase 4B; MAP1LC3/LC3: microtubule associated protein 1 light chain 3 alpha; MEFs: mouse embryonic fibroblasts; MTOR: mechanistic target of rapamycin kinase; NCOR1: nuclear receptor corepressor 1; SCF: SKP1-CUL-F-box protein; SQSTM1: sequestosome 1; TFEB: transcription factor EB.


Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , F-Box Proteins/metabolism , Hormesis , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Cells, Cultured , F-Box Proteins/physiology , Female , Fibroblasts/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/physiology , Tumor Suppressor Protein p53/physiology , Ubiquitination
9.
Dev Dyn ; 250(7): 943-954, 2021 07.
Article En | MEDLINE | ID: mdl-33410225

BACKGROUND: Persistent elevated concentrations of urinary protein can destroy proximal tubule epithelial cells (PTECs) by inducing lysosomal abnormalities, thereby aggravating PTEC damage and renal fibrosis. However, the specific mechanisms of these serial biochemical events and methods for treating or preventing PTEC damage upon proteinuria need further investigation. RESULTS: In this study, electron microscopy and dual-labeled immunofluorescence analysis for identifying lysosome type revealed inadequate primary lysosome biogenesis and secondary lysosome accumulation in the PTECs of patients with minimal change nephrotic syndrome or membranous nephropathy who suffered from proteinuria. In vitro studies on HK-2 cells indicated that this abnormality was associated with decreased expression of transcription factor EB (TFEB). In contrast, TFEB overexpressing HK-2 cells under urinary protein overload exhibited significantly reduced accumulation of secondary lysosomes and increased proportion and quantity of primary lysosomes as indicated by dual-labeled immunofluorescence. Further, these cells could upregulate lysosomal degradation functions, as determined using Cathepsin L activity assays and flow cytometry for dye quenched-albumin. CONCLUSIONS: These results indicate that abnormal TFEB expression is a key mechanism of lysosomal dyshomeostasis caused by protein overload in PTECs. TFEB is thus a potential therapeutic target for the treatment of urinary protein-related kidney disease.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Kidney Tubules, Proximal/metabolism , Lysosomes/metabolism , Proteinuria/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney Tubules, Proximal/pathology , Lysosomes/pathology , Proteins/metabolism , Proteinuria/complications , Proteinuria/genetics , Proteinuria/metabolism
10.
Elife ; 92020 12 10.
Article En | MEDLINE | ID: mdl-33300868

Disrupted nucleocytoplasmic transport (NCT) has been implicated in neurodegenerative disease pathogenesis; however, the mechanisms by which disrupted NCT causes neurodegeneration remain unclear. In a Drosophila screen, we identified ref(2)P/p62, a key regulator of autophagy, as a potent suppressor of neurodegeneration caused by the GGGGCC hexanucleotide repeat expansion (G4C2 HRE) in C9orf72 that causes amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). We found that p62 is increased and forms ubiquitinated aggregates due to decreased autophagic cargo degradation. Immunofluorescence and electron microscopy of Drosophila tissues demonstrate an accumulation of lysosome-like organelles that precedes neurodegeneration. These phenotypes are partially caused by cytoplasmic mislocalization of Mitf/TFEB, a key transcriptional regulator of autophagolysosomal function. Additionally, TFEB is mislocalized and downregulated in human cells expressing GGGGCC repeats and in C9-ALS patient motor cortex. Our data suggest that the C9orf72-HRE impairs Mitf/TFEB nuclear import, thereby disrupting autophagy and exacerbating proteostasis defects in C9-ALS/FTD.


Active Transport, Cell Nucleus/genetics , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Microphthalmia-Associated Transcription Factor/physiology , Amyotrophic Lateral Sclerosis/genetics , Animals , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blotting, Western , C9orf72 Protein/genetics , Disease Models, Animal , Drosophila melanogaster , Female , Fluorescent Antibody Technique , Frontotemporal Dementia/genetics , HeLa Cells , Humans , Lysosomes/genetics , Male , Microphthalmia-Associated Transcription Factor/metabolism , Microscopy, Electron, Transmission , Motor Cortex/metabolism
11.
Nat Cell Biol ; 22(10): 1252-1263, 2020 10.
Article En | MEDLINE | ID: mdl-32989250

Sensing and clearance of dysfunctional lysosomes is critical for cellular homeostasis. Here we show that transcription factor EB (TFEB)-a master transcriptional regulator of lysosomal biogenesis and autophagy-is activated during the lysosomal damage response, and its activation is dependent on the function of the ATG conjugation system, which mediates LC3 lipidation. In addition, lysosomal damage triggers LC3 recruitment on lysosomes, where lipidated LC3 interacts with the lysosomal calcium channel TRPML1, facilitating calcium efflux essential for TFEB activation. Furthermore, we demonstrate the presence and importance of this TFEB activation mechanism in kidneys in a mouse model of oxalate nephropathy accompanying lysosomal damage. A proximal tubule-specific TFEB-knockout mouse exhibited progression of kidney injury induced by oxalate crystals. Together, our results reveal unexpected mechanisms of TFEB activation by LC3 lipidation and their physiological relevance during the lysosomal damage response.


Acute Kidney Injury/pathology , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Lipids/chemistry , Lysosomes/pathology , Microtubule-Associated Proteins/metabolism , Acute Kidney Injury/metabolism , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Calcium/metabolism , HeLa Cells , Homeostasis , Humans , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/genetics
12.
PLoS One ; 15(9): e0238546, 2020.
Article En | MEDLINE | ID: mdl-32881934

The MITF, TFEB, TFE3 and TFEC (MiT-TFE) proteins belong to the basic helix-loop-helix family of leucine zipper transcription factors. MITF is crucial for melanocyte development and differentiation, and has been termed a lineage-specific oncogene in melanoma. The three related proteins MITF, TFEB and TFE3 have been shown to be involved in the biogenesis and function of lysosomes and autophagosomes, regulating cellular clearance pathways. Here we investigated the cross-regulatory relationship of MITF and TFEB in melanoma cells. Like MITF, the TFEB and TFE3 genes are expressed in melanoma cells as well as in melanoma tumors, albeit at lower levels. We show that the MITF and TFEB proteins, but not TFE3, directly affect each other's mRNA and protein expression. In addition, the subcellular localization of MITF and TFEB is subject to regulation by the mTOR signaling pathway, which impacts their cross-regulatory relationship at the transcriptional level. Our work shows that the relationship between MITF and TFEB is multifaceted and that the cross-regulatory interactions of these factors need to be taken into account when considering pathways regulated by these proteins.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Signal Transduction , TOR Serine-Threonine Kinases/physiology
13.
Circulation ; 142(5): 483-498, 2020 08 04.
Article En | MEDLINE | ID: mdl-32354235

BACKGROUND: Abdominal aortic aneurysm (AAA) is a severe aortic disease with a high mortality rate in the event of rupture. Pharmacological therapy is needed to inhibit AAA expansion and prevent aneurysm rupture. Transcription factor EB (TFEB), a master regulator of autophagy and lysosome biogenesis, is critical to maintain cell homeostasis. In this study, we aim to investigate the role of vascular smooth muscle cell (VSMC) TFEB in the development of AAA and establish TFEB as a novel target to treat AAA. METHODS: The expression of TFEB was measured in human and mouse aortic aneurysm samples. We used loss/gain-of-function approaches to understand the role of TFEB in VSMC survival and explored the underlying mechanisms through transcriptome and functional studies. Using VSMC-selective Tfeb knockout mice and different mouse AAA models, we determined the role of VSMC TFEB and a TFEB activator in AAA in vivo. RESULTS: We found that TFEB is downregulated in both human and mouse aortic aneurysm lesions. TFEB potently inhibits apoptosis in VSMCs, and transcriptome analysis revealed that TFEB regulates apoptotic signaling pathways, especially apoptosis inhibitor B-cell lymphoma 2. B-cell lymphoma 2 is significantly upregulated by TFEB and is required for TFEB to inhibit VSMC apoptosis. We consistently observed that TFEB deficiency increases VSMC apoptosis and promotes AAA formation in different mouse AAA models. Furthermore, we demonstrated that 2-hydroxypropyl-ß-cyclodextrin, a clinical agent used to enhance the solubility of drugs, activates TFEB and inhibits AAA formation and progression in mice. Last, we found that 2-hydroxypropyl-ß-cyclodextrin inhibits AAA in a VSMC TFEB-dependent manner in mouse models. CONCLUSIONS: Our study demonstrated that TFEB protects against VSMC apoptosis and AAA. TFEB activation by 2-hydroxypropyl-ß-cyclodextrin may be a promising therapeutic strategy for the prevention and treatment of AAA.


2-Hydroxypropyl-beta-cyclodextrin/therapeutic use , Aortic Aneurysm, Abdominal/prevention & control , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Disease Models, Animal , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , 2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Aminopropionitrile/toxicity , Aneurysm, Ruptured/etiology , Angiotensin II/toxicity , Animals , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Apoptosis/drug effects , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/deficiency , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cholesterol/metabolism , Down-Regulation , Drug Evaluation, Preclinical , Gain of Function Mutation , Gene Expression Regulation , Genetic Vectors/toxicity , Humans , Loss of Function Mutation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Transcriptome/drug effects
14.
Elife ; 92020 01 14.
Article En | MEDLINE | ID: mdl-31934850

Intermittent hypoxia (IH) is the predominant pathophysiological disturbance in obstructive sleep apnea (OSA), known to be independently associated with cardiovascular diseases. However, the effect of IH on cardiac fibrosis and molecular events involved in this process are unclear. Here, we tested IH in angiotensin II (Ang II)-induced cardiac fibrosis and signaling linked to fibroblast activation. IH triggered cardiac fibrosis and aggravated Ang II-induced cardiac dysfunction in mice. Plasma thrombospondin-1 (TSP1) content was upregulated in both IH-exposed mice and OSA patients. Moreover, both in vivo and in vitro results showed IH-induced cardiac fibroblast activation and increased TSP1 expression in cardiac fibroblasts. Mechanistically, phosphorylation of STAT3 at Tyr705 mediated the IH-induced TSP1 expression and fibroblast activation. Finally, STAT3 inhibitor S3I-201 or AAV9 carrying a periostin promoter driving the expression of shRNA targeting Stat3 significantly attenuated the synergistic effects of IH and Ang II on cardiac fibrosis in mice. This work suggests a potential therapeutic strategy for OSA-related fibrotic heart disease.


Fibroblasts/cytology , Heart/physiology , Hypoxia/metabolism , Myocardium/pathology , STAT3 Transcription Factor/metabolism , Thrombospondin 1/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Echocardiography , Fibrosis , Gene Expression Regulation , Gene Silencing , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Signal Transduction , Tyrosine/chemistry
15.
Biochem J ; 477(1): 137-160, 2020 01 17.
Article En | MEDLINE | ID: mdl-31820786

Transcription factor EB (TFEB) is a master regulator of lysosomal biogenesis and autophagy with critical roles in several cancers. Lysosomal autophagy promotes cancer survival through the degradation of toxic molecules and the maintenance of adequate nutrient supply. Doxorubicin (DOX) is the standard of care treatment for triple-negative breast cancer (TNBC); however, chemoresistance at lower doses and toxicity at higher doses limit its usefulness. By targeting pathways of survival, DOX can become an effective antitumor agent. In this study, we examined the role of TFEB in TNBC and its relationship with autophagy and DNA damage induced by DOX. In TNBC cells, TFEB was hypo-phosphorylated and localized to the nucleus upon DOX treatment. TFEB knockdown decreased the viability of TNBC cells while increasing caspase-3 dependent apoptosis. Additionally, inhibition of the TFEB-phosphatase calcineurin sensitized cells to DOX-induced apoptosis in a TFEB dependent fashion. Regulation of apoptosis by TFEB was not a consequence of altered lysosomal function, as TFEB continued to protect against apoptosis in the presence of lysosomal inhibitors. RNA-Seq analysis of MDA-MB-231 cells with TFEB silencing identified a down-regulation in cell cycle and homologous recombination genes while interferon-γ and death receptor signaling genes were up-regulated. In consequence, TFEB knockdown disrupted DNA repair following DOX, as evidenced by persistent γH2A.X detection. Together, these findings describe in TNBC a novel lysosomal independent function for TFEB in responding to DNA damage.


Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , DNA Repair , Lysosomes/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Apoptosis/drug effects , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Doxorubicin/pharmacology , Gene Knockdown Techniques , Humans
16.
Infect Immun ; 88(3)2020 02 20.
Article En | MEDLINE | ID: mdl-31818957

Coxiella burnetii is an obligate intracellular bacterial pathogen that replicates inside the lysosome-derived Coxiella-containing vacuole (CCV). To establish this unique niche, C. burnetii requires the Dot/Icm type IV secretion system (T4SS) to translocate a cohort of effector proteins into the host cell, which modulate multiple cellular processes. To characterize the host-pathogen interactions that occur during C. burnetii infection, stable-isotope labeling by amino acids in cell culture (SILAC)-based proteomics was used to identify changes in the host proteome during infection of a human-derived macrophage cell line. These data revealed that the abundances of many proteins involved in host cell autophagy and lysosome biogenesis were increased in infected cells. Thus, the role of the host transcription factors TFEB and TFE3, which regulate the expression of a network of genes involved in autophagy and lysosomal biogenesis, were examined in the context of C. burnetii infection. During infection with C. burnetii, both TFEB and TFE3 were activated, as demonstrated by the transport of these proteins from the cytoplasm into the nucleus. The nuclear translocation of these transcription factors was shown to be dependent on the T4SS, as a Dot/Icm mutant showed reduced nuclear translocation of TFEB and TFE3. This was supported by the observation that blocking bacterial translation with chloramphenicol resulted in the movement of TFEB and TFE3 back into the cytoplasm. Silencing of the TFEB and TFE3 genes, alone or in combination, significantly reduced the size of the CCV, which indicates that these host transcription factors facilitate the expansion and maintenance of the organelle that supports C. burnetii intracellular replication.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Coxiella burnetii/physiology , Host-Pathogen Interactions/physiology , Active Transport, Cell Nucleus/physiology , Gene Expression Regulation/physiology , Humans , Macrophages/metabolism , Proteome/metabolism
17.
Plant J ; 102(1): 138-152, 2020 04.
Article En | MEDLINE | ID: mdl-31755159

Jasmonates are key regulators of the balance between defence and growth in plants. However, the molecular mechanisms by which activation of defence reduces growth are not yet fully understood. Here, we analyze the role of MYC transcription factors (TFs) and jasmonic acid (JA) in photomorphogenic growth. We found that multiple myc mutants share light-associated phenotypes with mutants of the phytochrome B photoreceptor, such as delayed seed germination in the dark and long hypocotyl growth. Overexpression of MYC2 in a phyB background partially suppressed its long hypocotyl phenotype. Transcriptomic analysis of multiple myc mutants confirmed that MYCs are required for full expression of red (R) light-regulated genes, including the master regulator HY5. ChIP-seq analyses revealed that MYC2 and MYC3 bind directly to the promoter of HY5 and that HY5 gene expression and protein levels are compromised in multiple myc mutants. Altogether, our results pinpoint MYCs as photomorphogenic TFs that control phytochrome responses by activating HY5 expression. This has important implications in understanding the trade-off between growth and defence as the same TFs that activate defence responses are photomorphogenic growth regulators.


Arabidopsis Proteins/genetics , Arabidopsis Proteins/physiology , Arabidopsis/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/physiology , Cyclopentanes/metabolism , Oxylipins/metabolism , Phototropism , Plant Growth Regulators/metabolism , Signal Transduction , Arabidopsis/growth & development , Arabidopsis Proteins/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Plant , Genes, myc , Phototropism/genetics , Phototropism/physiology
18.
J Nutr Biochem ; 73: 108214, 2019 11.
Article En | MEDLINE | ID: mdl-31520816

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.


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
19.
FASEB J ; 33(11): 12374-12391, 2019 11.
Article En | MEDLINE | ID: mdl-31404503

AMPK is a central regulator of energy homeostasis. AMPK not only elicits acute metabolic responses but also promotes metabolic reprogramming and adaptations in the long-term through regulation of specific transcription factors and coactivators. We performed a whole-genome transcriptome profiling in wild-type (WT) and AMPK-deficient mouse embryonic fibroblasts (MEFs) and primary hepatocytes that had been treated with 2 distinct classes of small-molecule AMPK activators. We identified unique compound-dependent gene expression signatures and several AMPK-regulated genes, including folliculin (Flcn), which encodes the tumor suppressor FLCN. Bioinformatics analysis highlighted the lysosomal pathway and the associated transcription factor EB (TFEB) as a key transcriptional mediator responsible for AMPK responses. AMPK-induced Flcn expression was abolished in MEFs lacking TFEB and transcription factor E3, 2 transcription factors with partially redundant function; additionally, the promoter activity of Flcn was profoundly reduced when its putative TFEB-binding site was mutated. The AMPK-TFEB-FLCN axis is conserved across species; swimming exercise in WT zebrafish induced Flcn expression in muscle, which was significantly reduced in AMPK-deficient zebrafish. Mechanistically, we have found that AMPK promotes dephosphorylation and nuclear localization of TFEB independently of mammalian target of rapamycin activity. Collectively, we identified the novel AMPK-TFEB-FLCN axis, which may function as a key cascade for cellular and metabolic adaptations.-Collodet, C., Foretz, M., Deak, M., Bultot, L., Metairon, S., Viollet, B., Lefebvre, G., Raymond, F., Parisi, A., Civiletto, G., Gut, P., Descombes, P., Sakamoto, K. AMPK promotes induction of the tumor suppressor FLCN through activation of TFEB independently of mTOR.


AMP-Activated Protein Kinases/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Proto-Oncogene Proteins/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Active Transport, Cell Nucleus , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Gene Expression Profiling , Hepatocytes/metabolism , Mice , Phosphorylation , Ribonucleotides/pharmacology , Zebrafish
20.
Autophagy ; 15(10): 1854-1856, 2019 10.
Article En | MEDLINE | ID: mdl-31318631

Most essential physiological functions in mammals show a 24-h rhythmic pattern, which includes sleep-wake, feeding-non-feeding cycles and energy metabolism. Recent studies indicate that macroautophagy/autophagy also displays a robust circadian rhythmicity following the daily feeding pattern in adult mammals. We discovered that MiT-TFE transcription factors TFEB and TFE3, master regulators of autophagy and lysosomal biogenesis, are activated in a circadian manner and drive the expression of NR1D1/REV-ERBα, a key component of the core clockwork, thus revealing a molecular link between the nutrient-driven circadian cycle and the light-induced molecular clock. The dynamic balance between TFEB and TFE3 activation and NR1D1 expression is responsible for the modulation and oscillation of autophagy and metabolism genes.


Autophagy/physiology , Circadian Rhythm/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Biological Clocks/genetics , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Time Factors
...