Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Ther Nucleic Acids ; 31: 353-366, 2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36817728

ABSTRACT

The abnormal aggregation of TDP-43 into cytoplasmic inclusions in affected neurons is a pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although how TDP-43 forms cytoplasmic aggregates and causes neurodegeneration in patients with ALS/FTD remains unclear, reducing cellular TDP-43 levels is likely to prevent aggregation and to rescue neurons from TDP-43 toxicity. To address this issue, here we developed gapmer-type antisense oligonucleotides (ASOs) against human TDP-43 using 2'-O,4'-C-ethylene nucleic acids (ENAs), which are modified nucleic acids with high stability, and tested the therapeutic potential of lowering TDP-43 levels using ENA-modified ASOs. We demonstrated that intracerebroventricular administration of ENA-modified ASOs into a mouse model of ALS/FTD expressing human TDP-43 results in the efficient reduction of TDP-43 levels in the brain and spinal cord. Surprisingly, a single injection of ENA-modified ASOs into TDP-43 mice led to long-lasting improvement of behavioral abnormalities and the suppression of cytoplasmic TDP-43 aggregation, even after TDP-43 levels had returned to the initial levels. Our results demonstrate that transient reduction of TDP-43 using ENA-modified ASOs leads to sustained therapeutic benefits in vivo, indicating the possibility of a disease-modifying therapy by lowering TDP-43 levels for the treatment of the TDP-43 proteinopathies, including ALS/FTD.

3.
PLoS Genet ; 18(6): e1010264, 2022 06.
Article in English | MEDLINE | ID: mdl-35771772

ABSTRACT

Autophagy is an indispensable process that degrades cytoplasmic materials to maintain cellular homeostasis. During autophagy, double-membrane autophagosomes surround cytoplasmic materials and either fuse with endosomes (called amphisomes) and then lysosomes, or directly fuse with lysosomes, in both cases generating autolysosomes that degrade their contents by lysosomal hydrolases. However, it remains unclear if there are specific mechanisms and/or conditions which distinguish these alternate routes. Here, we identified PACSIN1 as a novel autophagy regulator. PACSIN1 deletion markedly decreased autophagic activity under basal nutrient-rich conditions but not starvation conditions, and led to amphisome accumulation as demonstrated by electron microscopic and co-localization analysis, indicating inhibition of lysosome fusion. PACSIN1 interacted with SNAP29, an autophagic SNARE, and was required for proper assembly of the STX17 and YKT6 complexes. Moreover, PACSIN1 was required for lysophagy, aggrephagy but not mitophagy, suggesting cargo-specific fusion mechanisms. In C. elegans, deletion of sdpn-1, a homolog of PACSINs, inhibited basal autophagy and impaired clearance of aggregated protein, implying a conserved role of PACSIN1. Taken together, our results demonstrate the amphisome-lysosome fusion process is preferentially regulated in response to nutrient state and stress, and PACSIN1 is a key to specificity during autophagy.


Subject(s)
Caenorhabditis elegans , Macroautophagy , Animals , Autophagosomes/metabolism , Autophagy/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Lysosomes/metabolism , Macroautophagy/genetics , SNARE Proteins/metabolism
4.
Nat Cell Biol ; 22(10): 1252-1263, 2020 10.
Article in English | MEDLINE | ID: mdl-32989250

ABSTRACT

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.


Subject(s)
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
5.
Mol Cell ; 73(2): 339-353.e6, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30581147

ABSTRACT

Membrane targeting of the BECN1-containing class III PI 3-kinase (PI3KC3) complexes is pivotal to the regulation of autophagy. The interaction of PI3KC3 complex II and its ubiquitously expressed inhibitor, Rubicon, was mapped to the first ß sheet of the BECN1 BARA domain and the UVRAG BARA2 domain by hydrogen-deuterium exchange and cryo-EM. These data suggest that the BARA ß sheet 1 unfolds to directly engage the membrane. This mechanism was confirmed using protein engineering, giant unilamellar vesicle assays, and molecular simulations. Using this mechanism, a BECN1 ß sheet-1 derived peptide activates both PI3KC3 complexes I and II, while HIV-1 Nef inhibits complex II. These data reveal how BECN1 switches on and off PI3KC3 binding to membranes. The observations explain how PI3KC3 inhibition by Rubicon, activation by autophagy-inducing BECN1 peptides, and inhibition by HIV-1 Nef are mediated by the switchable ability of the BECN1 BARA domain to partially unfold and insert into membranes.


Subject(s)
Autophagy , Beclin-1/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Autophagy-Related Proteins , Beclin-1/chemistry , Beclin-1/genetics , Binding Sites , Class III Phosphatidylinositol 3-Kinases/chemistry , Class III Phosphatidylinositol 3-Kinases/genetics , Cryoelectron Microscopy , Enzyme Activation , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Dynamics Simulation , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Signal Transduction , Structure-Activity Relationship , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/metabolism
6.
J Cell Sci ; 129(20): 3781-3791, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27587839

ABSTRACT

Autophagy is an intracellular degradation pathway conserved in eukaryotes. Among core autophagy-related (Atg) proteins, mammalian Atg9A is the sole multi-spanning transmembrane protein, and both of its N- and C-terminal domains are exposed to the cytoplasm. It is known that Atg9A travels through the trans-Golgi network (TGN) and the endosomal system under nutrient-rich conditions, and transiently localizes to the autophagosome upon autophagy induction. However, the significance of Atg9A trafficking for autophagosome formation remains elusive. Here, we identified sorting motifs in the N-terminal cytosolic stretch of Atg9A that interact with the adaptor protein AP-2. Atg9A with mutations in the sorting motifs could not execute autophagy and was abnormally accumulated at the recycling endosomes. The combination of defects in autophagy and Atg9A accumulation in the recycling endosomes was also found upon the knockdown of TRAPPC8, a specific subunit of the TRAPPIII complex. These results show directly that the trafficking of Atg9A through the recycling endosomes is an essential step for autophagosome formation.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Endocytosis , Endosomes/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Protein Complex 2/metabolism , Amino Acid Motifs , Animals , Autophagy , Autophagy-Related Proteins/chemistry , Gene Knockdown Techniques , HeLa Cells , Humans , Membrane Proteins/chemistry , Mice, Knockout , Models, Biological , Protein Transport , Salmonella/metabolism , Structure-Activity Relationship , Tyrosine/metabolism , Vesicular Transport Proteins/chemistry , trans-Golgi Network/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL