Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Life Sci Alliance ; 5(3)2022 03.
Article in English | MEDLINE | ID: mdl-34933920

ABSTRACT

The autophagy-lysosomal pathway is impaired in many neurodegenerative diseases characterized by protein aggregation, but the link between aggregation and lysosomal dysfunction remains poorly understood. Here, we combine cryo-electron tomography, proteomics, and cell biology studies to investigate the effects of protein aggregates in primary neurons. We use artificial amyloid-like ß-sheet proteins (ß proteins) to focus on the gain-of-function aspect of aggregation. These proteins form fibrillar aggregates and cause neurotoxicity. We show that late stages of autophagy are impaired by the aggregates, resulting in lysosomal alterations reminiscent of lysosomal storage disorders. Mechanistically, ß proteins interact with and sequester AP-3 µ1, a subunit of the AP-3 adaptor complex involved in protein trafficking to lysosomal organelles. This leads to destabilization of the AP-3 complex, missorting of AP-3 cargo, and lysosomal defects. Restoring AP-3µ1 expression ameliorates neurotoxicity caused by ß proteins. Altogether, our results highlight the link between protein aggregation, lysosomal impairments, and neurotoxicity.


Subject(s)
Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/metabolism , Gain of Function Mutation , Neurons/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Amyloidogenic Proteins/ultrastructure , Cell Survival/genetics , Gene Expression , Lysosomes/metabolism , Lysosomes/ultrastructure , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurons/ultrastructure , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Signal Transduction
2.
Autophagy ; 15(9): 1572-1591, 2019 09.
Article in English | MEDLINE | ID: mdl-30917721

ABSTRACT

Mitochondria are key organelles for cellular metabolism, and regulate several processes including cell death and macroautophagy/autophagy. Here, we show that mitochondrial respiratory chain (RC) deficiency deactivates AMP-activated protein kinase (AMPK, a key regulator of energy homeostasis) signaling in tissue and in cultured cells. The deactivation of AMPK in RC-deficiency is due to increased expression of the AMPK-inhibiting protein FLCN (folliculin). AMPK is found to be necessary for basal lysosomal function, and AMPK deactivation in RC-deficiency inhibits lysosomal function by decreasing the activity of the lysosomal Ca2+ channel MCOLN1 (mucolipin 1). MCOLN1 is regulated by phosphoinositide kinase PIKFYVE and its product PtdIns(3,5)P2, which is also decreased in RC-deficiency. Notably, reactivation of AMPK, in a PIKFYVE-dependent manner, or of MCOLN1 in RC-deficient cells, restores lysosomal hydrolytic capacity. Building on these data and the literature, we propose that downregulation of the AMPK-PIKFYVE-PtdIns(3,5)P2-MCOLN1 pathway causes lysosomal Ca2+ accumulation and impaired lysosomal catabolism. Besides unveiling a novel role of AMPK in lysosomal function, this study points to the mechanism that links mitochondrial malfunction to impaired lysosomal catabolism, underscoring the importance of AMPK and the complexity of organelle cross-talk in the regulation of cellular homeostasis. Abbreviation: ΔΨm: mitochondrial transmembrane potential; AMP: adenosine monophosphate; AMPK: AMP-activated protein kinase; ATG5: autophagy related 5; ATP: adenosine triphosphate; ATP6V0A1: ATPase, H+ transporting, lysosomal, V0 subbunit A1; ATP6V1A: ATPase, H+ transporting, lysosomal, V0 subbunit A; BSA: bovine serum albumin; CCCP: carbonyl cyanide-m-chlorophenylhydrazone; CREB1: cAMP response element binding protein 1; CTSD: cathepsin D; CTSF: cathepsin F; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; EBSS: Earl's balanced salt solution; ER: endoplasmic reticulum; FBS: fetal bovine serum; FCCP: carbonyl cyanide-p-trifluoromethoxyphenolhydrazone; GFP: green fluorescent protein; GPN: glycyl-L-phenylalanine 2-naphthylamide; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MCOLN1/TRPML1: mucolipin 1; MEF: mouse embryonic fibroblast; MITF: melanocyte inducing transcription factor; ML1N*2-GFP: probe used to detect PtdIns(3,5)P2 based on the transmembrane domain of MCOLN1; MTORC1: mechanistic target of rapamycin kinase complex 1; NDUFS4: NADH:ubiquinone oxidoreductase subunit S4; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; pcDNA: plasmid cytomegalovirus promoter DNA; PCR: polymerase chain reaction; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns(3,5)P2: phosphatidylinositol-3,5-bisphosphate; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; P/S: penicillin-streptomycin; PVDF: polyvinylidene fluoride; qPCR: quantitative real time polymerase chain reaction; RFP: red fluorescent protein; RNA: ribonucleic acid; SDS-PAGE: sodium dodecyl sulfate polyacrylamide gel electrophoresis; shRNA: short hairpin RNA; siRNA: small interfering RNA; TFEB: transcription factor EB; TFE3: transcription factor binding to IGHM enhancer 3; TMRM: tetramethylrhodamine, methyl ester, perchlorate; ULK1: unc-51 like autophagy activating kinase 1; ULK2: unc-51 like autophagy activating kinase 2; UQCRC1: ubiquinol-cytochrome c reductase core protein 1; v-ATPase: vacuolar-type H+-translocating ATPase; WT: wild-type.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagosomes/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Transient Receptor Potential Channels/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Animals , Autophagosomes/drug effects , Autophagosomes/ultrastructure , Calcium/metabolism , Cell Death/drug effects , Cell Death/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Fibroblasts , HEK293 Cells , HeLa Cells , Humans , Lysosomes/drug effects , Lysosomes/enzymology , Lysosomes/ultrastructure , Mice , Mitochondria/drug effects , Mitochondria/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Elife ; 82019 02 18.
Article in English | MEDLINE | ID: mdl-30775969

ABSTRACT

Perturbations in mitochondrial function and homeostasis are pervasive in lysosomal storage diseases, but the underlying mechanisms remain unknown. Here, we report a transcriptional program that represses mitochondrial biogenesis and function in lysosomal storage diseases Niemann-Pick type C (NPC) and acid sphingomyelinase deficiency (ASM), in patient cells and mouse tissues. This mechanism is mediated by the transcription factors KLF2 and ETV1, which are both induced in NPC and ASM patient cells. Mitochondrial biogenesis and function defects in these cells are rescued by the silencing of KLF2 or ETV1. Increased ETV1 expression is regulated by KLF2, while the increase of KLF2 protein levels in NPC and ASM stems from impaired signaling downstream sphingosine-1-phosphate receptor 1 (S1PR1), which normally represses KLF2. In patient cells, S1PR1 is barely detectable at the plasma membrane and thus unable to repress KLF2. This manuscript provides a mechanistic pathway for the prevalent mitochondrial defects in lysosomal storage diseases. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Subject(s)
Lipids/genetics , Lysosomal Storage Diseases/genetics , Organelle Biogenesis , Transcription, Genetic , Animals , Brain/metabolism , Cell Respiration , Down-Regulation/genetics , Electron Transport , Fibroblasts/metabolism , Genes, Mitochondrial , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , MAP Kinase Signaling System , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Sphingomyelin Phosphodiesterase/metabolism , Transcription Factors/metabolism , Up-Regulation/genetics
4.
Orphanet J Rare Dis ; 13(1): 52, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29631605

ABSTRACT

BACKGROUND: Fabry disease is a multisystemic lysosomal storage disorder caused by the impairment of α-galactosidase A. The incidence of this rare disease is underestimated due to delayed diagnosis. Moreover, the management of the identified subjects is often complicated by the detection of variants of unclear diagnostic interpretation, usually identified in screening studies. We performed an observational study based on biochemical and genetic analysis of 805 dried blood spot samples from patients with clinical symptoms or family history of this pathology, which were collected from 109 Spanish hospitals, all over the country. RESULTS: We identified 77 new diagnosed patients with mutations related to classical Fabry disease, as well as 2 subjects with c.374A > T; p.His125Leu, a possible new mutation that need to be confirmed. Additionally, we detected 8 subjects carrying genetic variants possibly linked to late onset Fabry disease (p.Arg118Cys and p.Ala143Thr), 4 cases with polymorphism p.Asp313Tyr and 36 individuals with single nucleotide polymorphisms in intronic regions of GLA. Five of the identified mutations (c.431delG; c.1182delA; c.374A > T; c.932 T > C; c.125 T > A; c.778G > A), which were associated with a classical phenotype have not been previously described. Moreover 3 subjects presenting complex haplotypes made up by the association of intronic variants presented impaired levels of GLA transcripts and Gb3 deposits in skin biopsy. CONCLUSIONS: Enzymatic screening for Fabry Disease in risk population (2 or more clinical manifestations or family history of the disease) helped to identify undiagnosed patients and unravel the impairment of GLA expression in some subjects with complex haplotypes.


Subject(s)
Fabry Disease/diagnosis , Fabry Disease/epidemiology , alpha-Galactosidase/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Fabry Disease/genetics , Female , Genetic Predisposition to Disease , Humans , Infant , Male , Middle Aged , Spain/epidemiology , Young Adult
5.
Biochem Biophys Res Commun ; 500(1): 87-93, 2018 05 27.
Article in English | MEDLINE | ID: mdl-28456629

ABSTRACT

Mitochondria are constantly communicating with the rest of the cell. Defects in mitochondria underlie severe pathologies, whose mechanisms remain poorly understood. It is becoming increasingly evident that mitochondrial malfunction resonates in other organelles, perturbing their function and their biogenesis. In this manuscript, we review the current knowledge on the cross-talk between mitochondria and other organelles, particularly lysosomes, peroxisomes and the endoplasmic reticulum. Several organelle interactions are mediated by transcriptional programs, and other signaling mechanisms are likely mediating organelle dysfunction downstream of mitochondrial impairments. Many of these organelle crosstalk pathways are likely to have a role in pathological processes.


Subject(s)
Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Peroxisomes/metabolism , Zellweger Syndrome/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomes/pathology , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Peroxisomes/pathology , Signal Transduction , Transcription, Genetic , Zellweger Syndrome/genetics , Zellweger Syndrome/pathology
6.
Sci Rep ; 7: 45076, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345620

ABSTRACT

Mitochondria are key cellular signaling platforms, affecting fundamental processes such as cell proliferation, differentiation and death. However, it remains unclear how mitochondrial signaling affects other organelles, particularly lysosomes. Here, we demonstrate that mitochondrial respiratory chain (RC) impairments elicit a stress signaling pathway that regulates lysosomal biogenesis via the microphtalmia transcription factor family. Interestingly, the effect of mitochondrial stress over lysosomal biogenesis depends on the timeframe of the stress elicited: while RC inhibition with rotenone or uncoupling with CCCP initially triggers lysosomal biogenesis, the effect peaks after few hours and returns to baseline. Long-term RC inhibition by long-term treatment with rotenone, or patient mutations in fibroblasts and in a mouse model result in repression of lysosomal biogenesis. The induction of lysosomal biogenesis by short-term mitochondrial stress is dependent on TFEB and MITF, requires AMPK signaling and is independent of calcineurin signaling. These results reveal an integrated view of how mitochondrial signaling affects lysosomes, which is essential to fully comprehend the consequences of mitochondrial malfunction, particularly in the context of mitochondrial diseases.


Subject(s)
Electron Transport , Lysosomes/metabolism , Mitochondrial Diseases/metabolism , Organelle Biogenesis , AMP-Activated Protein Kinase Kinases , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Protein Kinases/metabolism , Rotenone/pharmacology , Uncoupling Agents/pharmacology
7.
Int J Biochem Cell Biol ; 79: 345-349, 2016 10.
Article in English | MEDLINE | ID: mdl-27613573

ABSTRACT

Mitochondria and lysosomes have long been studied in the context of their classic functions: energy factory and recycle bin, respectively. In the last twenty years, it became evident that these organelles are much more than simple industrial units, and are indeed in charge of many of cellular processes. Both mitochondria and lysosomes are now recognized as far-reaching signaling platforms, regulating many key aspects of cell and tissue physiology. It has furthermore become clear that mitochondria and lysosomes impact each other. The mechanisms underlying the cross-talk between these organelles are only now starting to be addressed. In this review, we briefly summarize how mitochondria, lysosomes and the lysosome-related process of autophagy affect each other in physiology and pathology.


Subject(s)
Lysosomes/metabolism , Mitochondria/metabolism , Animals , Autophagy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...