Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
bioRxiv ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38853929

ABSTRACT

Batten disease is characterized by early-onset blindness, juvenile dementia and death during the second decade of life. The most common genetic causes are mutations in the CLN3 gene encoding a lysosomal protein. There are currently no therapies targeting the progression of the disease, mostly due to the lack of knowledge about the disease mechanisms. To gain insight into the impact of CLN3 loss on cellular signaling and organelle function, we generated CLN3 knock-out cells in a human cell line (CLN3-KO), and performed RNA sequencing to obtain the cellular transcriptome. Following a multi-dimensional transcriptome analysis, we identified the transcriptional regulator YAP1 as a major driver of the transcriptional changes observed in CLN3-KO cells. We further observed that YAP1 pro-apoptotic signaling is hyperactive as a consequence of CLN3 functional loss in retinal pigment epithelia cells, and in the hippocampus and thalamus of CLN3exΔ7/8 mice, an established model of Batten disease. Loss of CLN3 activates YAP1 by a cascade of events that starts with the inability of releasing glycerophosphodiesthers from CLN3-KO lysosomes, which leads to perturbations in the lipid content of the nuclear envelope and nuclear dysmorphism. This results in increased number of DNA lesions, activating the kinase c-Abl, which phosphorylates YAP1, stimulating its pro-apoptotic signaling. Altogether, our results highlight a novel organelle crosstalk paradigm in which lysosomal metabolites regulate nuclear envelope content, nuclear shape and DNA homeostasis. This novel molecular mechanism underlying the loss of CLN3 in mammalian cells and tissues may open new c-Abl-centric therapeutic strategies to target Batten disease.

2.
Nat Commun ; 14(1): 3911, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37400440

ABSTRACT

Batten disease, one of the most devastating types of neurodegenerative lysosomal storage disorders, is caused by mutations in CLN3. Here, we show that CLN3 is a vesicular trafficking hub connecting the Golgi and lysosome compartments. Proteomic analysis reveals that CLN3 interacts with several endo-lysosomal trafficking proteins, including the cation-independent mannose 6 phosphate receptor (CI-M6PR), which coordinates the targeting of lysosomal enzymes to lysosomes. CLN3 depletion results in mis-trafficking of CI-M6PR, mis-sorting of lysosomal enzymes, and defective autophagic lysosomal reformation. Conversely, CLN3 overexpression promotes the formation of multiple lysosomal tubules, which are autophagy and CI-M6PR-dependent, generating newly formed proto-lysosomes. Together, our findings reveal that CLN3 functions as a link between the M6P-dependent trafficking of lysosomal enzymes and lysosomal reformation pathway, explaining the global impairment of lysosomal function in Batten disease.


Subject(s)
Membrane Glycoproteins , Neuronal Ceroid-Lipofuscinoses , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Proteomics , Molecular Chaperones/metabolism , Lysosomes/metabolism , Hydrolases/metabolism , Autophagy
3.
Nature ; 585(7826): 597-602, 2020 09.
Article in English | MEDLINE | ID: mdl-32612235

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) is a key metabolic hub that controls the cellular response to environmental cues by exerting its kinase activity on multiple substrates1-3. However, whether mTORC1 responds to diverse stimuli by differentially phosphorylating specific substrates is poorly understood. Here we show that transcription factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy4,5, is phosphorylated by mTORC1 via a substrate-specific mechanism that is mediated by Rag GTPases. Owing to this mechanism, the phosphorylation of TFEB-unlike other substrates of mTORC1, such as S6K and 4E-BP1- is strictly dependent on the amino-acid-mediated activation of RagC and RagD GTPases, but is insensitive to RHEB activity induced by growth factors. This mechanism has a crucial role in Birt-Hogg-Dubé syndrome, a disorder that is caused by mutations in the RagC and RagD activator folliculin (FLCN) and is characterized by benign skin tumours, lung and kidney cysts and renal cell carcinoma6,7. We found that constitutive activation of TFEB is the main driver of the kidney abnormalities and mTORC1 hyperactivity in a mouse model of Birt-Hogg-Dubé syndrome. Accordingly, depletion of TFEB in kidneys of these mice fully rescued the disease phenotype and associated lethality, and normalized mTORC1 activity. Our findings identify a mechanism that enables differential phosphorylation of mTORC1 substrates, the dysregulation of which leads to kidney cysts and cancer.


Subject(s)
Birt-Hogg-Dube Syndrome/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/deficiency , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Birt-Hogg-Dube Syndrome/genetics , Birt-Hogg-Dube Syndrome/pathology , Cell Line , Disease Models, Animal , Enzyme Activation , HeLa Cells , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Substrate Specificity , Tuberous Sclerosis Complex 2 Protein/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
4.
Nat Commun ; 11(1): 2461, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424153

ABSTRACT

It is well established that pluripotent stem cells in fetal and postnatal liver (LPCs) can differentiate into both hepatocytes and cholangiocytes. However, the signaling pathways implicated in the differentiation of LPCs are still incompletely understood. Transcription Factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, is known to be involved in osteoblast and myeloid differentiation, but its role in lineage commitment in the liver has not been investigated. Here we show that during development and upon regeneration TFEB drives the differentiation status of murine LPCs into the progenitor/cholangiocyte lineage while inhibiting hepatocyte differentiation. Genetic interaction studies show that Sox9, a marker of precursor and biliary cells, is a direct transcriptional target of TFEB and a primary mediator of its effects on liver cell fate. In summary, our findings identify an unexplored pathway that controls liver cell lineage commitment and whose dysregulation may play a role in biliary cancer.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Lineage , Liver/cytology , Liver/physiology , Regeneration/physiology , Animals , Bile Duct Neoplasms/pathology , Bile Ducts/metabolism , Cell Differentiation , Cell Proliferation , Cholangiocarcinoma/pathology , Down-Regulation/genetics , Hepatocytes/cytology , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Spheroids, Cellular/cytology , Stem Cells/cytology , Stem Cells/metabolism , Up-Regulation/genetics
5.
J Cell Biol ; 218(12): 3954-3966, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31694921

ABSTRACT

Endosomal recycling maintains the cell surface abundance of nutrient transporters for nutrient uptake, but how the cell integrates nutrient availability with recycling is less well understood. Here, in studying the recycling of human glutamine transporters ASCT2 (SLC1A5), LAT1 (SLC7A5), SNAT1 (SLC38A1), and SNAT2 (SLC38A2), we establish that following amino acid restriction, the adaptive delivery of SNAT2 to the cell surface relies on retromer, a master conductor of endosomal recycling. Upon complete amino acid starvation or selective glutamine depletion, we establish that retromer expression is upregulated by transcription factor EB (TFEB) and other members of the MiTF/TFE family of transcription factors through association with CLEAR elements in the promoters of the retromer genes VPS35 and VPS26A TFEB regulation of retromer expression therefore supports adaptive nutrient acquisition through endosomal recycling.


Subject(s)
Amino Acid Transport System A/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Endosomes/metabolism , Nutrients , Amino Acid Transport System ASC/metabolism , Animals , Cell Membrane/metabolism , Glutamine/metabolism , HEK293 Cells , HeLa Cells , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens/metabolism , Promoter Regions, Genetic , Signal Transduction , Up-Regulation , Vesicular Transport Proteins/metabolism
6.
J Pathol ; 246(4): 470-484, 2018 12.
Article in English | MEDLINE | ID: mdl-30073645

ABSTRACT

Obesity and dyslipidaemia are features of the metabolic syndrome and risk factors for chronic kidney disease. The cellular mechanisms connecting metabolic syndrome with chronic kidney disease onset and progression remain largely unclear. We show that proximal tubular epithelium is a target site for lipid deposition upon overnutrition with a cholesterol-rich Western-type diet. Affected proximal tubule epithelial cells displayed giant vacuoles of lysosomal or autophagosomal origin, harbouring oxidised lipoproteins and concentric membrane layer structures (multilamellar bodies), reminiscent of lysosomal storage diseases. Additionally, lipidomic analysis revealed renal deposition of cholesterol and phospholipids, including lysosomal phospholipids. Proteomic profiles of renal multilamellar bodies were distinct from those of epidermis or lung multilamellar bodies and of cytoplasmic lipid droplets. Tubular multilamellar bodies were observed in kidney biopsies of obese hypercholesterolaemic patients, and the concentration of the phospholipidosis marker di-docosahexaenoyl (22:6)-bis(monoacylglycerol) phosphate was doubled in urine from individuals with metabolic syndrome and chronic kidney disease. The enrichment of proximal tubule epithelial cells with phospholipids and multilamellar bodies was accompanied by enhanced inflammation, fibrosis, tubular damage markers, and higher urinary electrolyte content. Concomitantly to the intralysosomal lipid storage, a renal transcriptional response was initiated to enhance lysosomal degradation and lipid synthesis. In cultured proximal tubule epithelial cells, inhibition of cholesterol efflux transport or oxysterol treatment induced effects very similar to the in vivo situation, such as multilamellar body and phospholipid amassing, and induction of damage, inflammatory, fibrotic, and lipogenic molecules. The onset of phospholipidosis in proximal tubule epithelial cells is a novel pathological trait in metabolic syndrome-related chronic kidney disease, and emphasises the importance of healthy lysosomes and nutrition for kidney well-being. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Hypercholesterolemia/complications , Kidney Tubules, Proximal/metabolism , Lysosomes/metabolism , Obesity/complications , Phospholipids/adverse effects , Renal Insufficiency, Chronic/etiology , Animals , Case-Control Studies , Cell Line , Cholesterol, Dietary/metabolism , Disease Models, Animal , Fibrosis , Kidney Tubules, Proximal/ultrastructure , Lysosomes/ultrastructure , Male , Mice, Inbred C57BL , Mice, Transgenic , Phospholipids/metabolism , Proteomics/methods , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
7.
Science ; 356(6343): 1188-1192, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28619945

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) is recruited to the lysosome by Rag guanosine triphosphatases (GTPases) and regulates anabolic pathways in response to nutrients. We found that MiT/TFE transcription factors-master regulators of lysosomal and melanosomal biogenesis and autophagy-control mTORC1 lysosomal recruitment and activity by directly regulating the expression of RagD. In mice, this mechanism mediated adaptation to food availability after starvation and physical exercise and played an important role in cancer growth. Up-regulation of MiT/TFE genes in cells and tissues from patients and murine models of renal cell carcinoma, pancreatic ductal adenocarcinoma, and melanoma triggered RagD-mediated mTORC1 induction, resulting in cell hyperproliferation and cancer growth. Thus, this transcriptional regulatory mechanism enables cellular adaptation to nutrient availability and supports the energy-demanding metabolism of cancer cells.


Subject(s)
Feedback, Physiological/physiology , Gene Expression Regulation, Neoplastic , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/physiopathology , Animals , Caloric Restriction , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Liver/enzymology , Liver/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL , Neoplasms/enzymology , Signal Transduction
8.
Elife ; 52016 Sep 26.
Article in English | MEDLINE | ID: mdl-27668431

ABSTRACT

TFE-fusion renal cell carcinomas (TFE-fusion RCCs) are caused by chromosomal translocations that lead to overexpression of the TFEB and TFE3 genes (Kauffman et al., 2014). The mechanisms leading to kidney tumor development remain uncharacterized and effective therapies are yet to be identified. Hence, the need to model these diseases in an experimental animal system (Kauffman et al., 2014). Here, we show that kidney-specific TFEB overexpression in transgenic mice, resulted in renal clear cells, multi-layered basement membranes, severe cystic pathology, and ultimately papillary carcinomas with hepatic metastases. These features closely recapitulate those observed in both TFEB- and TFE3-mediated human kidney tumors. Analysis of kidney samples revealed transcriptional induction and enhanced signaling of the WNT ß-catenin pathway. WNT signaling inhibitors normalized the proliferation rate of primary kidney cells and significantly rescued the disease phenotype in vivo. These data shed new light on the mechanisms underlying TFE-fusion RCCs and suggest a possible therapeutic strategy based on the inhibition of the WNT pathway.

9.
Cell Signal ; 26(2): 363-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24308962

ABSTRACT

The E3 Ubiquitin ligase TRIM50 promotes the formation and clearance of aggresome-associated polyubiquitinated proteins through HDAC6 interaction, a tubulin specific deacetylase that regulates microtubule-dependent aggresome formation. In this report we showed that TRIM50 is a target of HDAC6 with Lys-372 as a critical residue for acetylation. We identified p300 and PCAF as two TRIM50 acetyltransferases and we further showed that a balance between ubiquitination and acetylation regulates TRIM50 degradation.


Subject(s)
Histone Deacetylases/metabolism , Ubiquitin-Protein Ligases/metabolism , Acetylation , Animals , Cell Line , HEK293 Cells , HeLa Cells , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Mice , Microtubules/metabolism , Protein Binding , Protein Structure, Tertiary , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/chemistry , Ubiquitination , p300-CBP Transcription Factors/metabolism
10.
PLoS One ; 7(7): e40440, 2012.
Article in English | MEDLINE | ID: mdl-22792322

ABSTRACT

In this study we report that, in response to proteasome inhibition, the E3-Ubiquitin ligase TRIM50 localizes to and promotes the recruitment and aggregation of polyubiquitinated proteins to the aggresome. Using Hdac6-deficient mouse embryo fibroblasts (MEF) we show that this localization is mediated by the histone deacetylase 6, HDAC6. Whereas Trim50-deficient MEFs allow pinpointing that the TRIM50 ubiquitin-ligase regulates the clearance of polyubiquitinated proteins localized to the aggresome. Finally we demonstrate that TRIM50 colocalizes, interacts with and increases the level of p62, a multifunctional adaptor protein implicated in various cellular processes including the autophagy clearance of polyubiquitinated protein aggregates. We speculate that when the proteasome activity is impaired, TRIM50 fails to drive its substrates to the proteasome-mediated degradation, and promotes their storage in the aggresome for successive clearance.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Histone Deacetylases/metabolism , Inclusion Bodies/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitinated Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy , Female , Histone Deacetylase 6 , Humans , Lysosomes/metabolism , Male , Mice , Mice, Inbred C3H , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Transport , Proteolysis , Sequestosome-1 Protein , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
11.
Orphanet J Rare Dis ; 6: 38, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21658225

ABSTRACT

BACKGROUND: Kabuki syndrome (Niikawa-Kuroki syndrome) is a rare, multiple congenital anomalies/mental retardation syndrome characterized by a peculiar face, short stature, skeletal, visceral and dermatoglyphic abnormalities, cardiac anomalies, and immunological defects. Recently mutations in the histone methyl transferase MLL2 gene have been identified as its underlying cause. METHODS: Genomic DNAs were extracted from 62 index patients clinically diagnosed as affected by Kabuki syndrome. Sanger sequencing was performed to analyze the whole coding region of the MLL2 gene including intron-exon junctions. The putative causal and possible functional effect of each nucleotide variant identified was estimated by in silico prediction tools. RESULTS: We identified 45 patients with MLL2 nucleotide variants. 38 out of the 42 variants were never described before. Consistently with previous reports, the majority are nonsense or frameshift mutations predicted to generate a truncated polypeptide. We also identified 3 indel, 7 missense and 3 splice site. CONCLUSIONS: This study emphasizes the relevance of mutational screening of the MLL2 gene among patients diagnosed with Kabuki syndrome. The identification of a large spectrum of MLL2 mutations possibly offers the opportunity to improve the actual knowledge on the clinical basis of this multiple congenital anomalies/mental retardation syndrome, design functional studies to understand the molecular mechanisms underlying this disease, establish genotype-phenotype correlations and improve clinical management.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Hematologic Diseases/genetics , Hematologic Diseases/pathology , Neoplasm Proteins/genetics , Vestibular Diseases/genetics , Vestibular Diseases/pathology , Abnormalities, Multiple/diagnosis , Child , Child, Preschool , Codon, Nonsense/genetics , Cohort Studies , Developmental Disabilities/genetics , Face/abnormalities , Face/pathology , Female , Frameshift Mutation/genetics , Genetic Association Studies , Humans , Intellectual Disability/genetics , Male , Phenotype , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...