Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Autophagy ; 20(6): 1462-1464, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38382540

ABSTRACT

Ribosomes are conserved macromolecular machines that are responsible for protein synthesis in all cells. While our knowledge of ribosome biogenesis and function has increased significantly in recent years, little is known about how ribosomes are degraded under specific cellular conditions. We recently uncovered that ribosomes are efficiently turned over by selective macroautophagy/autophagy during oncogene-induced senescence (OIS). By profiling the ribosome interactome in human fibroblasts undergoing OIS, we discovered a key role for the de-ubiquitinating enzyme USP10 in guiding this process. Release of USP10 from ribosomes during senescence leads to their enhanced ubiquitination and selective sequestering by autophagy through the SQSTM1/p62 receptor protein. This process is important for sustaining senescence-associated metabolome and secretome alterations.


Subject(s)
Autophagy , Cellular Senescence , Oncogenes , Ribosomes , Humans , Ribosomes/metabolism , Cellular Senescence/physiology , Autophagy/physiology , Models, Biological , Animals
2.
Cell Rep ; 42(11): 113381, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37930887

ABSTRACT

Oncogene-induced senescence (OIS) is a persistent anti-proliferative response that acts as a barrier against malignant transformation. During OIS, cells undergo dynamic remodeling, which involves alterations in protein and organelle homeostasis through autophagy. Here, we show that ribosomes are selectively targeted for degradation by autophagy during OIS. By characterizing senescence-dependent alterations in the ribosomal interactome, we find that the deubiquitinase USP10 dissociates from the ribosome during the transition to OIS. This release of USP10 leads to an enhanced ribosome ubiquitination, particularly of small subunit proteins, including lysine 275 on RPS2. Both reinforcement of the USP10-ribosome interaction and mutation of RPS2 K275 abrogate ribosomal delivery to lysosomes without affecting bulk autophagy. We show that the selective recruitment of ubiquitinated ribosomes to autophagosomes is mediated by the p62 receptor. While ribophagy is not required for the establishment of senescence per se, it contributes to senescence-related metabolome alterations and facilitates the senescence-associated secretory phenotype.


Subject(s)
Ribosomes , Ubiquitin , Ribosomes/metabolism , Ubiquitination , Ubiquitin/metabolism , Autophagy/physiology , Oncogenes , Cellular Senescence
3.
Cell Death Differ ; 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37558732

ABSTRACT

Autophagy is an essential recycling and quality control pathway which preserves cellular and organismal homeostasis. As a catabolic process, autophagy degrades damaged and aged intracellular components in response to conditions of stress, including nutrient deprivation, oxidative and genotoxic stress. Autophagy is a highly adaptive and dynamic process which requires an intricately coordinated molecular control. Here we provide an overview of how autophagy is regulated post-transcriptionally, through RNA processing events, epitranscriptomic modifications and non-coding RNAs. We further discuss newly revealed RNA-binding properties of core autophagy machinery proteins and review recent indications of autophagy's ability to impact cellular RNA homeostasis. From a physiological perspective, we examine the biological implications of these emerging regulatory layers of autophagy, particularly in the context of nutrient deprivation and tumorigenesis.

4.
Cell Metab ; 34(12): 2036-2046.e8, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36384144

ABSTRACT

Impairment of translation can lead to collisions of ribosomes, which constitute an activation platform for several ribosomal stress-surveillance pathways. Among these is the ribotoxic stress response (RSR), where ribosomal sensing by the MAP3K ZAKα leads to activation of p38 and JNK kinases. Despite these insights, the physiological ramifications of ribosomal impairment and downstream RSR signaling remain elusive. Here, we show that stalling of ribosomes is sufficient to activate ZAKα. In response to amino acid deprivation and full nutrient starvation, RSR impacts on the ensuing metabolic responses in cells, nematodes, and mice. The RSR-regulated responses in these model systems include regulation of AMPK and mTOR signaling, survival under starvation conditions, stress hormone production, and regulation of blood sugar control. In addition, ZAK-/- male mice present a lean phenotype. Our work highlights impaired ribosomes as metabolic signals and demonstrates a role for RSR signaling in metabolic regulation.


Subject(s)
MAP Kinase Kinase Kinases , Protein Biosynthesis , Ribosomes , Stress, Physiological , Animals , Male , Mice , MAP Kinase Kinase Kinases/metabolism
5.
Methods Mol Biol ; 2445: 99-115, 2022.
Article in English | MEDLINE | ID: mdl-34972988

ABSTRACT

Autophagy and autophagy-associated genes are implicated in a growing list of cellular, physiological, and pathophysiological processes and conditions. Therefore, it is ever more important to be able to reliably monitor and quantify autophagic activity. Whereas autophagic markers, such as LC3 can provide general indications about autophagy, specific and accurate detection of autophagic activity requires assessment of autophagic cargo flux. Here, we provide protocols on how to monitor bulk and selective autophagy by the use of inducible expression of exogenous probes based on the fluorescent coral protein Keima. To exemplify and demonstrate the power of this system, we provide data obtained by analyses of cytosolic and mitochondrially targeted Keima probes in human retinal epithelial cells treated with the mTOR-inhibitor Torin1 or with the iron chelator deferiprone (DFP). Our data indicate that Torin1 induces autophagic flux of cytosol and mitochondria to a similar degree, that is, compatible with induction of bulk autophagy, whereas DFP induces a highly selective form of mitophagy that efficiently excludes cytosol.


Subject(s)
Autophagy , Microtubule-Associated Proteins , Autophagy/physiology , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitophagy
6.
Front Mol Biosci ; 9: 1074701, 2022.
Article in English | MEDLINE | ID: mdl-36601581

ABSTRACT

During autophagy, the ATG8 family proteins have several well-characterized roles in facilitating early, mid, and late steps of autophagy, including autophagosome expansion, cargo recruitment and autophagosome-lysosome fusion. Their discovery has importantly allowed for precise experimental monitoring of the pathway, bringing about a huge expansion of research in the field over the last decades. In this review, we discuss both canonical and non-canonical roles of the autophagic lipidation machinery, with particular focus on the ATG8 proteins, their post-translational modifications and their increasingly uncovered alternative roles mediated through their anchoring at different membranes. These include endosomes, macropinosomes, phagosomes and the plasma membrane, to which ATG8 proteins can bind through canonical or alternative lipidation. Beyond new ATG8 binding partners and cargo types, we also explore several open questions related to alternative outcomes of autophagic machinery engagement beyond degradation. These include their roles in plasma membrane repair and secretion of selected substrates as well as the physiological implications hereof in health and disease.

7.
Nat Commun ; 12(1): 7113, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876567

ABSTRACT

Dynamic change in subcellular localization of signaling proteins is a general concept that eukaryotic cells evolved for eliciting a coordinated response to stimuli. Mass spectrometry-based proteomics in combination with subcellular fractionation can provide comprehensive maps of spatio-temporal regulation of protein networks in cells, but involves laborious workflows that does not cover the phospho-proteome level. Here we present a high-throughput workflow based on sequential cell fractionation to profile the global proteome and phospho-proteome dynamics across six distinct subcellular fractions. We benchmark the workflow by studying spatio-temporal EGFR phospho-signaling dynamics in vitro in HeLa cells and in vivo in mouse tissues. Finally, we investigate the spatio-temporal stress signaling, revealing cellular relocation of ribosomal proteins in response to hypertonicity and muscle contraction. Proteomics data generated in this study can be explored through https://SpatialProteoDynamics.github.io .


Subject(s)
Proteome/metabolism , Proteomics , Signal Transduction , Animals , Biological Phenomena , Cell Fractionation , HeLa Cells , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Osmotic Pressure , Phosphorylation , Subcellular Fractions/metabolism , Workflow
8.
Autophagy ; 17(12): 4504-4505, 2021 12.
Article in English | MEDLINE | ID: mdl-34643458

ABSTRACT

EIF4A3 (eukaryotic translation initiation factor 4A3) is an RNA helicase and core component of the exon junction complex. While this RNA-binding protein (RBP) is well-characterized for its crucial roles in splicing, RNA trafficking and nonsense-mediated decay, its role in the regulation of metabolic signaling pathways remains elusive. In a recent study, we describe a new role for EIF4A3 as a negative regulator of macroautophagy/autophagy. Mechanistically, we report that EIF4A3, through its ability to safeguard splicing, can maintain low basal levels of autophagy through the cytosolic retention of the key autophagy transcription factor TFEB. Upon EIF4A3 depletion, the shuttling of TFEB to the nucleus results in an integrated transcriptional response, which induces both early and late steps of the autophagy pathway and enhances autophagic flux. We further report the upregulation of EIF4A3 across multiple cancer types and highlight the relevance of this newly identified EIF4A3-TFEB signaling axis in human tumors.


Subject(s)
DEAD-box RNA Helicases , Eukaryotic Initiation Factor-4A , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Humans , RNA Splicing/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Sci Adv ; 7(27)2021 Jul.
Article in English | MEDLINE | ID: mdl-34215587

ABSTRACT

The plasma membrane shapes and protects the eukaryotic cell from its surroundings and is crucial for cell life. Although initial repair mechanisms to reseal injured membranes are well established, less is known about how cells restructure damaged membranes in the aftermath to restore homeostasis. Here, we show that cells respond to plasma membrane injury by activating proteins associated with macropinocytosis specifically at the damaged membrane. Subsequent to membrane resealing, cells form large macropinosomes originating from the repair site, which eventually become positive for autophagy-related LC3B protein. This process occurs independent of ULK1, ATG13, and WIPI2 but dependent on ATG7, p62, and Rubicon. Internalized macropinosomes shrink in the cytoplasm, likely by osmotic draining, and eventually fuse with lysosomes. We propose that a form of macropinocytosis coupled to noncanonical autophagy, which we term LC3-associated macropinocytosis (LAM) functions to remove damaged material from the plasma membrane and restore membrane integrity upon injury.

10.
Cell Death Differ ; 28(12): 3344-3356, 2021 12.
Article in English | MEDLINE | ID: mdl-34158631

ABSTRACT

During autophagy, the coordinated actions of autophagosomes and lysosomes result in the controlled removal of damaged intracellular organelles and superfluous substrates. The evolutionary conservation of this process and its requirement for maintaining cellular homeostasis emphasizes the need to better dissect the pathways governing its molecular regulation. In our previously performed high-content screen, we assessed the effect of 1530 RNA-binding proteins on autophagy. Among the top regulators, we identified the eukaryotic translation initiation factor 4A-3 (eIF4A3). Here we show that depletion of eIF4A3 leads to a potent increase in autophagosome and lysosome biogenesis and an enhanced autophagic flux. This is mediated by the key autophagy transcription factor, TFEB, which becomes dephosphorylated and translocates from the cytoplasm to the nucleus where it elicits an integrated transcriptional response. We further identified an exon-skipping event in the transcript encoding for the direct TFEB kinase, GSK3B, which leads to a reduction in GSK3B expression and activity. Through analysis of TCGA data, we found a significant upregulation of eIF4A3 expression across several cancer types and confirmed the potential relevance of this newly identified signaling axis in human tumors. Hence, our data suggest a previously unrecognized role for eIF4A3 as a gatekeeper of autophagy through the control of TFEB activation, revealing a new mechanism for autophagy regulation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Transcription Factors/metabolism , Autophagy , Humans , Transfection
11.
Trends Biochem Sci ; 45(10): 845-857, 2020 10.
Article in English | MEDLINE | ID: mdl-32828649

ABSTRACT

Autophagy is a highly conserved degradation pathway that ensures nutrient recycling and removal of unwanted substrates. This process has a fundamental role in stress adaptation and maintenance of cellular homeostasis. Here, we discuss emerging aspects of the autophagy-RNA interplay, including autophagy-mediated degradation of RNA, RNA-binding proteins (RBPs), and ribonucleoprotein (RNP) complexes. Beyond degradation, we review new roles for autophagy players in the secretion and intracellular transport of RNA and related complexes. We discuss the physiological importance of these events for RNA homeostasis and gene expression programs, as well as their implications for disease, including cancer and neurodegeneration. Lastly, we examine how post-transcriptional regulation of autophagy, through specialized processing and selective translation of key transcripts, challenges and updates our current view of autophagy complexity.


Subject(s)
Autophagy , RNA/metabolism , Biological Transport , Homeostasis , Hydrolysis , Lysosomes/metabolism , Ribonucleoproteins/metabolism
12.
Cell Death Differ ; 27(3): 1134-1153, 2020 03.
Article in English | MEDLINE | ID: mdl-31409894

ABSTRACT

Autophagy is an evolutionarily conserved process that captures aberrant intracellular proteins and/or damaged organelles for delivery to lysosomes, with implications for cellular and organismal homeostasis, aging and diverse pathologies, including cancer. During cancer development, autophagy may play both tumour-supporting and tumour-suppressing roles. Any relationships of autophagy to the established oncogene-induced replication stress (RS) and the ensuing DNA damage response (DDR)-mediated anti-cancer barrier in early tumorigenesis remain to be elucidated. Here, assessing potential links between autophagy, RS and DDR, we found that autophagy is enhanced in both early and advanced stages of human urinary bladder and prostate tumorigenesis. Furthermore, a high-content, single-cell-level microscopy analysis of human cellular models exposed to diverse genotoxic insults showed that autophagy is enhanced in cells that experienced robust DNA damage, independently of the cell-cycle position. Oncogene- and drug-induced RS triggered first DDR and later autophagy. Unexpectedly, genetic inactivation of autophagy resulted in RS, despite cellular retention of functional mitochondria and normal ROS levels. Moreover, recovery from experimentally induced RS required autophagy to support DNA synthesis. Consistently, RS due to the absence of autophagy could be partly alleviated by exogenous supply of deoxynucleosides. Our results highlight the importance of autophagy for DNA synthesis, suggesting that autophagy may support cancer progression, at least in part, by facilitating tumour cell survival and fitness under replication stress, a feature shared by most malignancies. These findings have implications for better understanding of the role of autophagy in tumorigenesis, as well as for attempts to manipulate autophagy as an anti-tumour therapeutic strategy.


Subject(s)
Autophagy , DNA Replication , Oncogenes , Stress, Physiological , Autophagosomes/drug effects , Autophagosomes/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , DNA Replication/drug effects , Humans , Models, Biological , Stress, Physiological/drug effects
13.
Nat Commun ; 10(1): 4176, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519908

ABSTRACT

The centrosome is the master orchestrator of mitotic spindle formation and chromosome segregation in animal cells. Centrosome abnormalities are frequently observed in cancer, but little is known of their origin and about pathways affecting centrosome homeostasis. Here we show that autophagy preserves centrosome organization and stability through selective turnover of centriolar satellite components, a process we termed doryphagy. Autophagy targets the satellite organizer PCM1 by interacting with GABARAPs via a C-terminal LIR motif. Accordingly, autophagy deficiency results in accumulation of large abnormal centriolar satellites and a resultant dysregulation of centrosome composition. These alterations have critical impact on centrosome stability and lead to mitotic centrosome fragmentation and unbalanced chromosome segregation. Our findings identify doryphagy as an important centrosome-regulating pathway and bring mechanistic insights to the link between autophagy dysfunction and chromosomal instability. In addition, we highlight the vital role of centriolar satellites in maintaining centrosome integrity.


Subject(s)
Autophagy/physiology , Centrioles/metabolism , Centrosome/metabolism , Mitosis/physiology , Autophagy/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Chromatography, Liquid , Humans , Immunoblotting , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microscopy, Fluorescence , Microtubules/metabolism , Mitosis/genetics , Molecular Dynamics Simulation
14.
Oncogene ; 38(26): 5127-5141, 2019 06.
Article in English | MEDLINE | ID: mdl-30872794

ABSTRACT

Autophagy is a conserved degradation process that occurs in all eukaryotic cells and its dysfunction has been associated with various diseases including cancer. While a number of large-scale attempts have recently identified new molecular players in autophagy regulation, including proteins and microRNAs, little is known regarding the function of long non-coding RNAs (lncRNAs) in the regulation of this process. To identify new long non-coding RNAs with functional implications in autophagy, we performed a high-throughput RNAi screen targeting more than 600 lncRNA transcripts and monitored their effects on autophagy in MCF-7 cells. We identified 63 lncRNAs that affected GFP-LC3B puncta numbers significantly. We validated the strongest hit, the lncRNA DRAIC previously shown to impact cell proliferation, and revealed a novel role for this lncRNA in the regulation of autophagic flux. Interestingly, we find DRAIC's pro-proliferative effects to be autophagy-independent. This study serves as a valuable resource for researchers from both the lncRNA and autophagy fields as it advances the current understanding of autophagy regulation by non-coding RNAs.


Subject(s)
Autophagy/genetics , RNA, Long Noncoding/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , MCF-7 Cells , Microarray Analysis , RNA Interference/physiology , Sequence Analysis, RNA
15.
Front Cell Dev Biol ; 7: 373, 2019.
Article in English | MEDLINE | ID: mdl-32039200

ABSTRACT

The eukaryotic cell has developed intricate machineries that monitor and maintain proteome homeostasis in order to ensure cellular functionality. This involves the carefully coordinated balance between protein synthesis and degradation pathways, which are dynamically regulated in order to meet the constantly changing demands of the cell. Ribosomes, together with the endoplasmic reticulum (ER), are the key drivers of protein synthesis, folding, maturation and sorting, while the proteasome plays a pivotal role in terminating the existence of thousands of proteins that are misfolded, damaged or otherwise obsolete. The synthesis, structure and function of these dedicated machines has been studied for decades, however, much less is understood about the mechanisms that control and execute their own turnover. Autophagy, an evolutionarily conserved catabolic pathway, mediates degradation of a large variety of cytosolic substrates, ranging from single proteins to entire organelles or multi-subunit macromolecular complexes. In this review, we focus on selective autophagy of three key components of the protein homeostasis machinery: ribosomes, ER and proteasomes, through the selective autophagy pathways of ribophagy, ER-phagy, and proteaphagy. We discuss newly discovered mechanisms for the selective clearance of these substrates, which are often stress-dependent and involve specialized signals for cargo recognition by a growing number of receptors. We further discuss the interplay between these pathways and their biological impact on key aspects of proteome homeostasis and cellular function in health and disease.

16.
Autophagy ; 14(7): 1288-1289, 2018.
Article in English | MEDLINE | ID: mdl-29973124

ABSTRACT

The core macroautophagy/autophagy machinery consists of a large group of autophagy-related (ATG) proteins, that mediate highly controlled, step-wise execution of this conserved intracellular degradation process. Whereas ATG proteins have been intensely studied in terms of protein interactions, post-translational modifications and transcriptional regulation, the mechanisms ensuring efficient translation of ATG proteins are not well understood. In a recent study, we describe an evolutionarily conserved role for EIF5A (eukaryotic translation initiation factor 5A) in autophagy. We demonstrate that EIF5A mediates Atg8-family protein lipidation and autophagosome formation via translation of the E2-like ATG3 protein. Moreover, we identify a particular motif in ATG3 causing EIF5A-dependency for its efficient translation.


Subject(s)
Autophagy-Related Protein 5/metabolism , Autophagy , Peptide Initiation Factors/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Animals , Cell Line , Humans , Models, Biological , Eukaryotic Translation Initiation Factor 5A
17.
EMBO Rep ; 19(6)2018 06.
Article in English | MEDLINE | ID: mdl-29712776

ABSTRACT

Autophagy is an essential catabolic process responsible for recycling of intracellular material and preserving cellular fidelity. Key to the autophagy pathway is the ubiquitin-like conjugation system mediating lipidation of Atg8 proteins and their anchoring to autophagosomal membranes. While regulation of autophagy has been characterized at the level of transcription, protein interactions and post-translational modifications, its translational regulation remains elusive. Here we describe a role for the conserved eukaryotic translation initiation factor 5A (eIF5A) in autophagy. Identified from a high-throughput screen, we find that eIF5A is required for lipidation of LC3B and its paralogs and promotes autophagosome formation. This feature is evolutionarily conserved and results from the translation of the E2-like ATG3 protein. Mechanistically, we identify an amino acid motif in ATG3 causing eIF5A dependency for its efficient translation. Our study identifies eIF5A as a key requirement for autophagosome formation and demonstrates the importance of translation in mediating efficient autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Peptide Initiation Factors/physiology , Protein Biosynthesis , RNA-Binding Proteins/physiology , Ubiquitin-Conjugating Enzymes/metabolism , Autophagy-Related Proteins/genetics , Humans , MCF-7 Cells , Microtubule-Associated Proteins/metabolism , Protein Processing, Post-Translational , Proteomics , Ubiquitin-Conjugating Enzymes/genetics , Eukaryotic Translation Initiation Factor 5A
18.
Oncogenesis ; 7(2): 14, 2018 Feb 03.
Article in English | MEDLINE | ID: mdl-29396433

ABSTRACT

Cancer cells utilize lysosomes for invasion and metastasis. Myeloid Zinc Finger1 (MZF1) is an ErbB2-responsive transcription factor that promotes invasion of breast cancer cells via upregulation of lysosomal cathepsins B and L. Here we identify let-7 microRNA, a well-known tumor suppressor in breast cancer, as a direct negative regulator of MZF1. Analysis of primary breast cancer tissues reveals a gradual upregulation of MZF1 from normal breast epithelium to invasive ductal carcinoma and a negative correlation between several let-7 family members and MZF1 mRNA, suggesting that the inverse regulatory relationship between let-7 and MZF1 may play a role in the development of invasive breast cancer. Furthermore, we show that MZF1 regulates lysosome trafficking in ErbB2-positive breast cancer cells. In line with this, MZF1 depletion or let-7 expression inhibits invasion-promoting anterograde trafficking of lysosomes and invasion of ErbB2-expressing MCF7 spheres. The results presented here link MZF1 and let-7 to lysosomal processes in ErbB2-positive breast cancer cells that in non-cancerous cells have primarily been connected to the transcription factor EB. Identifying MZF1 and let-7 as regulators of lysosome distribution in invasive breast cancer cells, uncouples cancer-associated, invasion-promoting lysosomal alterations from normal lysosomal functions and thus opens up new possibilities for the therapeutic targeting of cancer lysosomes.

19.
Autophagy ; 13(1): 3-23, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27715443

ABSTRACT

Macroautophagy/autophagy is a key catabolic process, essential for maintaining cellular homeostasis and survival through the removal and recycling of unwanted cellular material. Emerging evidence has revealed intricate connections between the RNA and autophagy research fields. While a majority of studies have focused on protein, lipid and carbohydrate catabolism via autophagy, accumulating data supports the view that several types of RNA and associated ribonucleoprotein complexes are specifically recruited to phagophores (precursors to autophagosomes) and subsequently degraded in the lysosome/vacuole. Moreover, recent studies have revealed a substantial number of novel autophagy regulators with RNA-related functions, indicating roles for RNA and associated proteins not only as cargo, but also as regulators of this process. In this review, we discuss widespread evidence of RNA catabolism via autophagy in yeast, plants and animals, reviewing the molecular mechanisms and biological importance in normal physiology, stress and disease. In addition, we explore emerging evidence of core autophagy regulation mediated by RNA-binding proteins and noncoding RNAs, and point to gaps in our current knowledge of the connection between RNA and autophagy. Finally, we discuss the pathological implications of RNA-protein aggregation, primarily in the context of neurodegenerative disease.


Subject(s)
Autophagy/physiology , RNA/metabolism , Animals , Arabidopsis , Carbohydrate Metabolism , Drosophila melanogaster , Genome , HEK293 Cells , HeLa Cells , Humans , Lipid Metabolism , Lysosomes/metabolism , Metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Proteins/metabolism , RNA, Long Noncoding/metabolism , RNA, Transfer/metabolism , RNA, Viral/genetics , RNA-Binding Proteins/metabolism , Tetrahymena , Vacuoles/metabolism , Zebrafish
20.
Nat Commun ; 5: 5840, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25524633

ABSTRACT

Sulfatases are key enzymatic regulators of sulfate homeostasis with several biological functions including degradation of glycosaminoglycans (GAGs) and other macromolecules in lysosomes. In a severe lysosomal storage disorder, multiple sulfatase deficiency (MSD), global sulfatase activity is deficient due to mutations in the sulfatase-modifying factor 1 (SUMF1) gene, encoding the essential activator of all sulfatases. We identify a novel regulatory layer of sulfate metabolism mediated by a microRNA. miR-95 depletes SUMF1 protein levels and suppresses sulfatase activity, causing the disruption of proteoglycan catabolism and lysosomal function. This blocks autophagy-mediated degradation, causing cytoplasmic accumulation of autophagosomes and autophagic substrates. By targeting miR-95 in cells from MSD patients, we can effectively increase residual SUMF1 expression, allowing for reactivation of sulfatase activity and increased clearance of sulfated GAGs. The identification of this regulatory mechanism opens the opportunity for a unique therapeutic approach in MSD patients where the need for exogenous enzyme replacement is circumvented.


Subject(s)
Lysosomes/metabolism , MicroRNAs/metabolism , Multiple Sulfatase Deficiency Disease/metabolism , Sulfates/metabolism , Autophagy , Glycosaminoglycans/metabolism , Humans , Lysosomes/enzymology , MicroRNAs/genetics , Multiple Sulfatase Deficiency Disease/enzymology , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/physiopathology , Oxidoreductases Acting on Sulfur Group Donors , Sulfatases/genetics , Sulfatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...