Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
IUBMB Life ; 74(4): 339-360, 2022 04.
Article in English | MEDLINE | ID: mdl-34874101

ABSTRACT

Autophagy regulates cellular homeostasis by degrading and recycling cytosolic components and damaged organelles. Disruption of autophagic flux has been shown to induce or facilitate neurodegeneration and accumulation of autophagic vesicles is overt in neurodegenerative diseases. The fruit fly Drosophila has been used as a model system to identify new factors that regulate physiology and disease. Here we provide a historical perspective of how the fly models have offered mechanistic evidence to understand the role of autophagy in neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Charcot-Marie-Tooth neuropathy, and polyglutamine disorders. Autophagy also plays a pivotal role in maintaining tissue homeostasis and protecting organism health. The gastrointestinal tract regulates organism health by modulating food intake, energy balance, and immunity. Growing evidence is strengthening the link between autophagy and digestive tract health in recent years. Here, we also discuss how the fly models have advanced the understanding of digestive physiology regulated by autophagy.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Animals , Autophagy/genetics , Drosophila/genetics , Gastrointestinal Tract , Neurodegenerative Diseases/genetics
2.
J Biomed Sci ; 28(1): 72, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34706729

ABSTRACT

BACKGROUND: During autophagy defense against invading microbes, certain lipid types are indispensable for generating specialized membrane-bound organelles. The lipid composition of autophagosomes remains obscure, as does the issue of how specific lipids and lipid-associated enzymes participate in autophagosome formation and maturation. Helicobacter pylori is auxotrophic for cholesterol and converts cholesterol to cholesteryl glucoside derivatives, including cholesteryl 6'-O-acyl-α-D-glucoside (CAG). We investigated how CAG and its biosynthetic acyltransferase assist H. pylori to escape host-cell autophagy. METHODS: We applied a metabolite-tagging method to obtain fluorophore-containing cholesteryl glucosides that were utilized to understand their intracellular locations. H. pylori 26695 and a cholesteryl glucosyltransferase (CGT)-deletion mutant (ΔCGT) were used as the standard strain and the negative control that contains no cholesterol-derived metabolites, respectively. Bacterial internalization and several autophagy-related assays were conducted to unravel the possible mechanism that H. pylori develops to hijack the host-cell autophagy response. Subcellular fractions of H. pylori-infected AGS cells were obtained and measured for the acyltransferase activity. RESULTS: The imaging studies of fluorophore-labeled cholesteryl glucosides pinpointed their intracellular localization in AGS cells. The result indicated that CAG enhances the internalization of H. pylori in AGS cells. Particularly, CAG, instead of CG and CPG, is able to augment the autophagy response induced by H. pylori. How CAG participates in the autophagy process is multifaceted. CAG was found to intervene in the degradation of autophagosomes and reduce lysosomal biogenesis, supporting the idea that intracellular H. pylori is harbored by autophago-lysosomes in favor of the bacterial survival. Furthermore, we performed the enzyme activity assay of subcellular fractions of H. pylori-infected AGS cells. The analysis showed that the acyltransferase is mainly distributed in autophago-lysosomal compartments. CONCLUSIONS: Our results support the idea that the acyltransferase is mainly distributed in the subcellular compartment consisting of autophagosomes, late endosomes, and lysosomes, in which the acidic environment is beneficial for the maximal acyltransferase activity. The resulting elevated level of CAG can facilitate bacterial internalization, interfere with the autophagy flux, and causes reduced lysosomal biogenesis.


Subject(s)
Acyltransferases/metabolism , Cholesterol/analogs & derivatives , Helicobacter Infections/physiopathology , Helicobacter pylori/physiology , Lysosomes/physiology , Animals , Cholesterol/biosynthesis , Helicobacter Infections/enzymology , Helicobacter Infections/microbiology , Male , Mice , Mice, Inbred C57BL , Specific Pathogen-Free Organisms
3.
EMBO J ; 30(4): 636-51, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21169990

ABSTRACT

Autophagy is a membrane-mediated degradation process of macromolecule recycling. Although the formation of double-membrane degradation vesicles (autophagosomes) is known to have a central role in autophagy, the mechanism underlying this process remains elusive. The serine/threonine kinase Atg1 has a key role in the induction of autophagy. In this study, we show that overexpression of Drosophila Atg1 promotes the phosphorylation-dependent activation of the actin-associated motor protein myosin II. A novel myosin light chain kinase (MLCK)-like protein, Spaghetti-squash activator (Sqa), was identified as a link between Atg1 and actomyosin activation. Sqa interacts with Atg1 through its kinase domain and is a substrate of Atg1. Significantly, myosin II inhibition or depletion of Sqa compromised the formation of autophagosomes under starvation conditions. In mammalian cells, we found that the Sqa mammalian homologue zipper-interacting protein kinase (ZIPK) and myosin II had a critical role in the regulation of starvation-induced autophagy and mammalian Atg9 (mAtg9) trafficking when cells were deprived of nutrients. Our findings provide evidence of a link between Atg1 and the control of Atg9-mediated autophagosome formation through the myosin II motor protein.


Subject(s)
Autophagy/physiology , Drosophila Proteins/physiology , Myosin Type II/metabolism , Phagosomes/metabolism , Protein Serine-Threonine Kinases/physiology , Starvation/metabolism , Animals , Animals, Genetically Modified , Autophagy/genetics , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Cells, Cultured , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Kinase/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Starvation/genetics , Tissue Distribution
4.
J Cell Sci ; 125(Pt 20): 4841-52, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22825871

ABSTRACT

Protein tyrosine phosphatases (PTPs) are a group of tightly regulated enzymes that coordinate with protein tyrosine kinases to control protein phosphorylation during various cellular processes. Using genetic analysis in Drosophila non-transmembrane PTPs, we identified one role that Myopic (Mop), the Drosophila homolog of the human His domain phosphotyrosine phosphatase (HDPTP), plays in cell adhesion. Depletion of Mop results in aberrant integrin distribution and border cell dissociation during Drosophila oogenesis. Interestingly, Mop phosphatase activity is not required for its role in maintaining border cell cluster integrity. We further identified Rab4 GTPase as a Mop interactor in a yeast two-hybrid screen. Expression of the Rab4 dominant-negative mutant leads to border cell dissociation and suppression of Mop-induced wing-blade adhesion defects, suggesting a critical role of Rab4 in Mop-mediated signaling. In mammals, it has been shown that Rab4-dependent recycling of integrins is necessary for cell adhesion and migration. We found that human HDPTP regulates the spatial distribution of Rab4 and integrin trafficking. Depletion of HDPTP resulted in actin reorganization and increased cell motility. Together, our findings suggest an evolutionarily conserved function of HDPTP-Rab4 in the regulation of endocytic trafficking, cell adhesion and migration.


Subject(s)
Cell Adhesion , Cell Movement , Drosophila Proteins , Protein Tyrosine Phosphatases , rab4 GTP-Binding Proteins , Actins/metabolism , Animals , Cell Adhesion/genetics , Cell Movement/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Mutation , Oogenesis/genetics , Phosphorylation , Protein Transport , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Wings, Animal/growth & development , Wings, Animal/pathology , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism
5.
Biochem Biophys Res Commun ; 452(3): 369-75, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25152394

ABSTRACT

Ubiquitination and the reverse process deubiquitination regulate protein stability and function during animal development. The Drosophila USP5 homolog Leon functions as other family members of unconventional deubiquitinases, disassembling free, substrate-unconjugated polyubiquitin chains to replenish the pool of mono-ubiquitin, and maintaining cellular ubiquitin homeostasis. However, the significance of Leon/USP5 in animal development is still unexplored. In this study, we generated leon mutants to show that Leon is essential for animal viability and tissue integrity during development. Both free and substrate-conjugated polyubiquitin chains accumulate in leon mutants, suggesting that abnormal ubiquitin homeostasis caused tissue disorder and lethality in leon mutants. Further analysis of protein expression profiles in leon mutants shows that the levels of all proteasomal subunits were elevated. Also, proteasomal enzymatic activities were elevated in leon mutants. However, proteasomal degradation of ubiquitinated substrates was impaired. Thus, aberrant ubiquitin homeostasis in leon mutants disrupts normal proteasomal degradation, which is compensated by elevating the levels of proteasomal subunits and activities. Ultimately, the failure to fully compensate the dysfunctional proteasome in leon mutants leads to animal lethality and tissue disorder.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Imaginal Discs/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/growth & development , Genes, Lethal , Homeostasis/genetics , Imaginal Discs/abnormalities , Larva/enzymology , Larva/genetics , Larva/growth & development , Mutation , Polyubiquitin/genetics , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteolysis , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Specific Proteases/metabolism
6.
Autophagy ; 20(6): 1213-1246, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38442890

ABSTRACT

Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results.Abbreviation: 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.


Subject(s)
Autophagy , Ferroptosis , Ferroptosis/physiology , Humans , Autophagy/physiology , Animals , Consensus
7.
J Proteome Res ; 12(5): 2138-50, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23517121

ABSTRACT

Although stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics was first developed as a cell culture-based technique, stable isotope-labeled amino acids have since been successfully introduced in vivo into select multicellular model organisms by manipulating the feeding diets. An earlier study by others has demonstrated that heavy lysine labeled Drosophila melanogaster can be derived by feeding with an exclusive heavy lysine labeled yeast diet. In this work, we have further evaluated the use of heavy lysine and/or arginine for metabolic labeling of fruit flies, with an aim to determine its respective quantification accuracy and versatility. In vivo conversion of heavy lysine and/or heavy arginine to several nonessential amino acids was observed in labeled flies, leading to distorted isotope pattern and underestimated heavy to light ratio. These quantification defects can nonetheless be rectified at protein level using the normalization function. The only caveat is that such a normalization strategy may not be suitable for every biological application, particularly when modified peptides need to be individually quantified at peptide level. In such cases, we showed that peptide ratios calculated from the summed intensities of all isotope peaks are less affected by the heavy amino acid conversion and therefore less sequence-dependent and more reliable. Applying either the single Lys8 or double Lys6/Arg10 metabolic labeling strategy to flies, we quantitatively mapped the proteomic changes during the onset of metamorphosis and upon amino acid deprivation. The expression of a number of steroid hormone 20-hydroxyecdysone regulated proteins was found to be changed significantly during larval-pupa transition, while several subunits of the V-ATPase complex and components regulating actomyosin were up-regulated under starvation-induced autophagy conditions.


Subject(s)
Amino Acids/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Proteome/metabolism , Amino Acids/chemistry , Animals , Drosophila Proteins/chemistry , Drosophila melanogaster/growth & development , Fat Body/growth & development , Fat Body/metabolism , Food Deprivation , Isotope Labeling/methods , Male , Metamorphosis, Biological , Proteome/chemistry , Proteomics , Pupa/growth & development , Pupa/metabolism , Stress, Physiological , Tandem Mass Spectrometry
8.
Structure ; 31(12): 1567-1577.e5, 2023 12 07.
Article in English | MEDLINE | ID: mdl-37794594

ABSTRACT

The structure determination of protein tyrosine phosphatase (PTP): phospho-protein complexes, which is essential to understand how specificity is achieved at the amino acid level, remains a significant challenge for protein crystallography and cryoEM due to the transient nature of binding interactions. Using rPTPεD1 and phospho-SrcKD as a model system, we have established an integrative workflow to address this problem, by means of which we generate a protein:phospho-protein complex model using predetermined protein structures, SAXS and pTyr-tailored MD simulations. Our model reveals transient protein-protein interactions between rPTPεD1 and phospho-SrcKD and is supported by three independent experimental validations. Measurements of the association rate between rPTPεD1 and phospho-SrcKD showed that mutations on the rPTPεD1: SrcKD complex interface disrupts these transient interactions, resulting in a reduction in protein-protein association rate and, eventually, phosphatase activity. This integrative approach is applicable to other PTP: phospho-protein complexes and the characterization of transient protein-protein interface interactions.


Subject(s)
Proteins , Scattering, Small Angle , X-Ray Diffraction , Phosphorylation
9.
Cell Death Dis ; 14(8): 540, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37607937

ABSTRACT

Accumulating evidence has shown that the quality of proteins must be tightly monitored and controlled to maintain cellular proteostasis. Misfolded proteins and protein aggregates are targeted for degradation through the ubiquitin proteasome (UPS) and autophagy-lysosome systems. The ubiquitination and deubiquitinating enzymes (DUBs) have been reported to play pivotal roles in the regulation of the UPS system. However, the function of DUBs in the regulation of autophagy remain to be elucidated. In this study, we found that knockdown of Leon/USP5 caused a marked increase in the formation of autophagosomes and autophagic flux under well-fed conditions. Genetic analysis revealed that overexpression of Leon suppressed Atg1-induced cell death in Drosophila. Immunoblotting assays further showed a strong interaction between Leon/USP5 and the autophagy initiating kinase Atg1/ULK1. Depletion of Leon/USP5 led to increased levels of Atg1/ULK1. Our findings indicate that Leon/USP5 is an autophagic DUB that interacts with Atg1/ULK1, negatively regulating the autophagic process.


Subject(s)
Autophagy , Drosophila Proteins , Animals , Autophagy/genetics , Autophagosomes , Cell Death , Drosophila , Lysosomes , Proteasome Endopeptidase Complex , Ubiquitin , Deubiquitinating Enzymes , Autophagy-Related Protein-1 Homolog/genetics , Drosophila Proteins/genetics , Ubiquitin-Specific Proteases/genetics
10.
J Biomed Sci ; 19: 52, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22621211

ABSTRACT

BACKGROUND: Autophagy and molecular chaperones both regulate protein homeostasis and maintain important physiological functions. Atg7 (autophagy-related gene 7) and Hsp27 (heat shock protein 27) are involved in the regulation of neurodegeneration and aging. However, the genetic connection between Atg7 and Hsp27 is not known. METHODS: The appearances of the fly eyes from the different genetic interactions with or without polyglutamine toxicity were examined by light microscopy and scanning electronic microscopy. Immunofluorescence was used to check the effect of Atg7 and Hsp27 knockdown on the formation of autophagosomes. The lifespan of altered expression of Hsp27 or Atg7 and that of the combination of the two different gene expression were measured. RESULTS: We used the Drosophila eye as a model system to examine the epistatic relationship between Hsp27 and Atg7. We found that both genes are involved in normal eye development, and that overexpression of Atg7 could eliminate the need for Hsp27 but Hsp27 could not rescue Atg7 deficient phenotypes. Using a polyglutamine toxicity assay (41Q) to model neurodegeneration, we showed that both Atg7 and Hsp27 can suppress weak, toxic effect by 41Q, and that overexpression of Atg7 improves the worsened mosaic eyes by the knockdown of Hsp27 under 41Q. We also showed that overexpression of Atg7 extends lifespan and the knockdown of Atg7 or Hsp27 by RNAi reduces lifespan. RNAi-knockdown of Atg7 expression can block the extended lifespan phenotype by Hsp27 overexpression, and overexpression of Atg7 can extend lifespan even under Hsp27 knockdown by RNAi. CONCLUSIONS: We propose that Atg7 acts downstream of Hsp27 in the regulation of eye morphology, polyglutamine toxicity, and lifespan in Drosophila.


Subject(s)
Drosophila Proteins/genetics , Drosophila , Eye , HSP27 Heat-Shock Proteins/genetics , Animals , Autophagy/genetics , Autophagy-Related Protein 7 , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Epistasis, Genetic/genetics , Eye/anatomy & histology , Eye/growth & development , Eye/metabolism , Gene Expression Regulation, Developmental , HSP27 Heat-Shock Proteins/physiology , Peptides/toxicity , RNA Interference
11.
Autophagy ; 18(8): 2008-2010, 2022 08.
Article in English | MEDLINE | ID: mdl-35380918

ABSTRACT

High levels of reactive oxygen species (ROS) result in oxidative stress, which damages cells and leads to the development of many diseases. Macroautophagy/autophagy plays an important role in protecting cells from diverse stress stimuli including oxidative stress. However, the molecular mechanisms of autophagy activation in response to oxidative stress remain largely unclear. In this study, we showed that TRAF6 mediates oxidative stress-induced ATG9A ubiquitination at two C-terminal lysine residues (K581 and K838). ATG9A ubiquitination promotes its association with BECN1, BECN1-PIK3C3/VPS34-UVRAG complex assembly and PIK3C3/VPS34 activation, thereby activating autophagy and endocytic trafficking. We also identified TNFAIP3/A20 as a negative regulator of oxidative-induced autophagy by counteracting TRAF6-mediated ATG9A ubiquitination. Moreover, ATG9A depletion attenuates LPS-induced autophagy and causes aberrant TLR4 signaling and inflammatory responses. Our findings revealed a critical role of ATG9A ubiquitination in oxidative stress-induced autophagy, endocytic trafficking and innate immunity.


Subject(s)
Autophagy , TNF Receptor-Associated Factor 6 , Autophagy/physiology , Class III Phosphatidylinositol 3-Kinases , Oxidative Stress , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination
12.
Cell Rep ; 38(8): 110354, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35196483

ABSTRACT

Excessive generation and accumulation of highly reactive oxidizing molecules causes oxidative stress and oxidative damage to cellular components. Accumulating evidence indicates that autophagy diminishes oxidative damage in cells and maintains redox homeostasis by degrading and recycling intracellular damaged components. Here, we show that TRAF6 E3 ubiquitin ligase and A20 deubiquitinase coordinate to regulate ATG9A ubiquitination and autophagy activation in cells responding to oxidative stress. The ROS-dependent TRAF6-mediated non-proteolytic, K48/63-linked ubiquitination of ATG9A enhances its association with Beclin 1 and the assembly of VPS34-UVRAG complex, thereby stimulating autophagy. Notably, expression of the ATG9A ubiquitination mutants impairs ROS-induced VPS34 activation and autophagy. We further find that lipopolysaccharide (LPS)-induced ROS production also stimulates TRAF6-mediated ATG9A ubiquitination. Ablation of ATG9A causes aberrant TLR4 endosomal trafficking and decreases IRF-3 phosphorylation in LPS-stimulated macrophages. Our findings provide important insights into how K48/K63-linked ubiquitination of ATG9A contributes to the regulation of oxidative stress-induced autophagy.


Subject(s)
TNF Receptor-Associated Factor 6 , Ubiquitin-Protein Ligases , Autophagy/physiology , Oxidative Stress , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
13.
Autophagy ; 17(10): 2750-2765, 2021 10.
Article in English | MEDLINE | ID: mdl-33112705

ABSTRACT

Macroautophagy/autophagy is an evolutionarily conserved intracellular pathway for the degradation of cytoplasmic materials. Under stress conditions, autophagy is upregulated and double-membrane autophagosomes are formed by the expansion of phagophores. The ATG16L1 precursor fusion contributes to development of phagophore structures and is critical for the biogenesis of autophagosomes. Here, we discovered a novel role of the protein tyrosine phosphatase PTPN9 in the regulation of homotypic ATG16L1 vesicle fusion and early autophagosome formation. Depletion of PTPN9 and its Drosophila homolog Ptpmeg2 impaired autophagosome formation and autophagic flux. PTPN9 colocalized with ATG16L1 and was essential for homotypic fusion of ATG16L1+ vesicles during starvation-induced autophagy. We further identified the Q-SNARE VTI1B as a substrate target of PTPN9 phosphatase. Like PTPN9, the VTI1B nonphosphorylatable mutant but not the phosphomimetic mutant enhanced SNARE complex assembly and autophagic flux. Our findings highlight the important role of PTPN9 in the regulation of ATG16L1+ autophagosome precursor fusion and autophagosome biogenesis through modulation of VTI1B phosphorylation status.Abbreviations: csw: corkscrew; EBSS: Earle's balanced salt solution; ERGIC: ER-Golgi intermediate compartment; ESCRT: endosomal sorting complexes required for transport; mop: myopic; NSF: N-ethylmaleimide-sensitive factor; PAS: phagophore assembly site; PolyQ: polyglutamine; PtdIns3P: phosphatidylinositol-3-phosphate; PTK: protein tyrosine kinase; PTM: posttranslational modification; PTP: protein tyrosine phosphatase; PTPN23/HD-PTP: protein tyrosine phosphatase non-receptor type 23; SNARE: soluble N-ethylmaleimide sensitive factor attachment protein receptor; STX7: syntaxin 7; STX8: syntaxin 8; STX17: syntaxin 17; VAMP3: vesicle associated membrane protein 3; VAMP7: vesicle associated membrane protein 7; VTI1B: vesicle transport through interaction with t-SNAREs 1B; YKT6: YKT6 v-SNARE homolog; ZFYVE1/DFCP1: zinc finger FYVE-type containing 1.


Subject(s)
Autophagosomes , Autophagy-Related Proteins , Macroautophagy , Protein Tyrosine Phosphatases, Non-Receptor , Qb-SNARE Proteins , Autophagosomes/metabolism , Autophagy/physiology , Autophagy-Related Proteins/metabolism , HeLa Cells , Humans , Membrane Fusion , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Qb-SNARE Proteins/metabolism
14.
Nat Commun ; 12(1): 1322, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637724

ABSTRACT

The ubiquitin-proteasome system (UPS) and autophagy are two major quality control processes whose impairment is linked to a wide variety of diseases. The coordination between UPS and autophagy remains incompletely understood. Here, we show that ubiquitin ligase UBE3C and deubiquitinating enzyme TRABID reciprocally regulate K29/K48-branched ubiquitination of VPS34. We find that this ubiquitination enhances the binding of VPS34 to proteasomes for degradation, thereby suppressing autophagosome formation and maturation. Under ER and proteotoxic stresses, UBE3C recruitment to phagophores is compromised with a concomitant increase of its association with proteasomes. This switch attenuates the action of UBE3C on VPS34, thereby elevating autophagy activity to facilitate proteostasis, ER quality control and cell survival. Specifically in the liver, we show that TRABID-mediated VPS34 stabilization is critical for lipid metabolism and is downregulated during the pathogenesis of steatosis. This study identifies a ubiquitination type on VPS34 and elucidates its cellular fate and physiological functions in proteostasis and liver metabolism.


Subject(s)
Autophagy/physiology , Class III Phosphatidylinositol 3-Kinases/metabolism , Liver/metabolism , Proteostasis/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Animals , Autophagosomes/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Diet, High-Fat/adverse effects , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
15.
Aging Cell ; 19(8): e13191, 2020 08.
Article in English | MEDLINE | ID: mdl-32666649

ABSTRACT

Changes in mitochondrial dynamics (fusion and fission) are known to occur during stem cell differentiation; however, the role of this phenomenon in tissue aging remains unclear. Here, we report that mitochondrial dynamics are shifted toward fission during aging of Drosophila ovarian germline stem cells (GSCs), and this shift contributes to aging-related GSC loss. We found that as GSCs age, mitochondrial fragmentation and expression of the mitochondrial fission regulator, Dynamin-related protein (Drp1), are both increased, while mitochondrial membrane potential is reduced. Moreover, preventing mitochondrial fusion in GSCs results in highly fragmented depolarized mitochondria, decreased BMP stemness signaling, impaired fatty acid metabolism, and GSC loss. Conversely, forcing mitochondrial elongation promotes GSC attachment to the niche. Importantly, maintenance of aging GSCs can be enhanced by suppressing Drp1 expression to prevent mitochondrial fission or treating with rapamycin, which is known to promote autophagy via TOR inhibition. Overall, our results show that mitochondrial dynamics are altered during physiological aging, affecting stem cell homeostasis via coordinated changes in stemness signaling, niche contact, and cellular metabolism. Such effects may also be highly relevant to other stem cell types and aging-induced tissue degeneration.


Subject(s)
Adult Germline Stem Cells/metabolism , Mitochondrial Dynamics/genetics , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Drosophila , Female , Male , Signal Transduction
16.
Oncogene ; 38(44): 7002-7016, 2019 10.
Article in English | MEDLINE | ID: mdl-31406243

ABSTRACT

Cancer cell migration plays a crucial role during the metastatic process. Reversible tyrosine phosphorylation by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) have been implicated in the regulation of cancer cell migration and invasion. However, the underlying mechanisms have not been fully elucidated. Here, we show that depletion of the FERM and PDZ domain-containing protein tyrosine phosphatase PTPN3 enhances lung cancer cell migration/invasion and metastasis by promoting actin filament assembly and focal adhesion dynamics. We further identified Src and DAAM1 (dishevelled associated activator of morphogenesis 1) as interactors of PTPN3. DAAM1 is a formin-like protein involved in the regulation of actin cytoskeletal remodeling. PTPN3 inhibits Src activity and Src-mediated phosphorylation of Tyr652 on DAAM1. The tyrosine phosphorylation of DAAM1 is essential for DAAM1 homodimer formation and actin polymerization. Ectopic expression of a DAAM1 phosphodeficient mutant inhibited F-actin assembly and suppressed lung cancer cell migration and invasion. Our findings reveal a novel mechanism by which reversible tyrosine phosphorylation of DAAM1 by Src and PTPN3 regulates actin dynamics and lung cancer invasiveness.


Subject(s)
Actins/metabolism , Lung Neoplasms/pathology , Microfilament Proteins/metabolism , Neoplasm Invasiveness , Protein Tyrosine Phosphatase, Non-Receptor Type 3/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , rho GTP-Binding Proteins/metabolism , Focal Adhesions , Humans , Polymerization
17.
Autophagy ; 15(10): 1829-1833, 2019 10.
Article in English | MEDLINE | ID: mdl-31234750

ABSTRACT

The NIH-funded center for autophagy research named Autophagy, Inflammation, and Metabolism (AIM) Center of Biomedical Research Excellence, located at the University of New Mexico Health Science Center is now completing its second year as a working center with a mission to promote autophagy research locally, nationally, and internationally. The center has thus far supported a cadre of 6 junior faculty (mentored PIs; mPIs) at a near-R01 level of funding. Two mPIs have graduated by obtaining their independent R01 funding and 3 of the remaining 4 have won significant funding from NIH in the form of R21 and R56 awards. The first year and a half of setting up the center has been punctuated by completion of renovations and acquisition and upgrades for equipment supporting autophagy, inflammation and metabolism studies. The scientific cores usage, and the growth of new studies is promoted through pilot grants and several types of enablement initiatives. The intent to cultivate AIM as a scholarly hub for autophagy and related studies is manifested in its Vibrant Campus Initiative, and the Tuesday AIM Seminar series, as well as by hosting a major scientific event, the 2019 AIM symposium, with nearly one third of the faculty from the International Council of Affiliate Members being present and leading sessions, giving talks, and conducting workshop activities. These and other events are often videostreamed for a worldwide scientific audience, and information about events at AIM and elsewhere are disseminated on Twitter and can be followed on the AIM web site. AIM intends to invigorate research on overlapping areas between autophagy, inflammation and metabolism with a number of new initiatives to promote metabolomic research. With the turnover of mPIs as they obtain their independent funding, new junior faculty are recruited and appointed as mPIs. All these activities are in keeping with AIM's intention to enable the next generation of autophagy researchers and help anchor, disseminate, and convey the depth and excitement of the autophagy field.


Subject(s)
Autophagy/physiology , Biomedical Research/organization & administration , Inflammation , Metabolism/physiology , Societies, Scientific , Biomedical Research/economics , Biomedical Research/trends , Faculty, Medical/economics , Faculty, Medical/education , Financing, Government , Financing, Organized/economics , History, 21st Century , Humans , Inflammation/etiology , Inflammation/pathology , Mentors , National Institutes of Health (U.S.)/economics , New Mexico , Research Personnel/economics , Research Personnel/education , Societies, Scientific/economics , Societies, Scientific/organization & administration , Societies, Scientific/standards , Societies, Scientific/trends , United States
18.
Dev Comp Immunol ; 32(7): 814-24, 2008.
Article in English | MEDLINE | ID: mdl-18187191

ABSTRACT

In invertebrates, the JAK-STAT signaling pathway is involved in the anti-bacterial response and is part of an anti-viral response in Drosophila. In this study, we show that two STAT transcripts are generated by alternative splicing and encode two isoforms of Sf-STAT with different C-terminal ends. These two isoforms were produced and purified using the recombinant baculovirus technology. Both purified isoforms showed similar DNA-binding activity and displayed weak but significant transactivation potential toward a Drosophila promoter that contained a STAT-binding motif. No significant activation of the Sf-STAT protein in Sf9 cells was found by infection with baculovirus AcMNPV.


Subject(s)
Gene Expression , STAT Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/metabolism , DNA/metabolism , Humans , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic/genetics , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , STAT Transcription Factors/chemistry , STAT Transcription Factors/genetics , STAT Transcription Factors/isolation & purification , Sequence Alignment , Sequence Homology, Amino Acid , Spodoptera , Transcriptional Activation/genetics , raf Kinases/genetics , raf Kinases/metabolism
19.
Mol Cell Biol ; 25(3): 979-87, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15657426

ABSTRACT

Paxillin is a prominent focal adhesion docking protein that regulates cell adhesion and migration. Although numerous paxillin-binding proteins have been identified and paxillin is required for normal embryogenesis, the precise mechanism by which paxillin functions in vivo has not yet been determined. We identified an ortholog of mammalian paxillin in Drosophila (Dpax) and have undertaken a genetic analysis of paxillin function during development. Overexpression of Dpax disrupted leg and wing development, suggesting a role for paxillin in imaginal disc morphogenesis. These defects may reflect a function for paxillin in regulation of Rho family GTPase signaling as paxillin interacts genetically with Rac and Rho in the developing eye. Moreover, a gain-of-function suppressor screen identified a genetic interaction between Dpax and cdi in wing development. cdi belongs to the cofilin kinase family, which includes the downstream Rho target, LIM kinase (LIMK). Significantly, strong genetic interactions were detected between Dpax and Dlimk, as well as downstream effectors of Dlimk. Supporting these genetic data, biochemical studies indicate that paxillin regulates Rac and Rho activity, positively regulating Rac and negatively regulating Rho. Taken together, these data indicate the importance of paxillin modulation of Rho family GTPases during development and identify the LIMK pathway as a critical target of paxillin-mediated Rho regulation.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Drosophila/metabolism , Phosphoproteins/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Actin Depolymerizing Factors , Actins/metabolism , Amino Acid Sequence , Animals , Cytoskeletal Proteins/genetics , Drosophila/growth & development , Drosophila Proteins/metabolism , Embryo, Nonmammalian/metabolism , Lim Kinases , Microfilament Proteins/metabolism , Molecular Sequence Data , Paxillin , Phosphoproteins/genetics , Protein Kinases/metabolism , Signal Transduction/physiology , Wings, Animal/growth & development
20.
Autophagy ; 14(6): 925-929, 2018.
Article in English | MEDLINE | ID: mdl-29938597

ABSTRACT

Recently, NIH has funded a center for autophagy research named the Autophagy, Inflammation, and Metabolism (AIM) Center of Biomedical Research Excellence, located at the University of New Mexico Health Science Center (UNM HSC), with aspirations to promote autophagy research locally, nationally, and internationally. The center has 3 major missions: (i) to support junior faculty in their endeavors to develop investigations in this area and obtain independent funding; (ii) to develop and provide technological platforms to advance autophagy research with emphasis on cellular approaches for high quality reproducible research; and (iii) to foster international collaborations through the formation of an International Council of Affiliate Members and through hosting national and international workshops and symposia. Scientifically, the AIM center is focused on autophagy and its intersections with other processes, with emphasis on both fundamental discoveries and applied translational research.


Subject(s)
Autophagy , Biomedical Research , Inflammation/pathology , International Cooperation , Research Personnel , Congresses as Topic , Information Dissemination
SELECTION OF CITATIONS
SEARCH DETAIL