Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Proc Natl Acad Sci U S A ; 114(23): E4621-E4630, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28536193

ABSTRACT

Podocytes form the outer part of the glomerular filter, where they have to withstand enormous transcapillary filtration forces driving glomerular filtration. Detachment of podocytes from the glomerular basement membrane precedes most glomerular diseases. However, little is known about the regulation of podocyte adhesion in vivo. Thus, we systematically screened for podocyte-specific focal adhesome (FA) components, using genetic reporter models in combination with iTRAQ-based mass spectrometry. This approach led to the identification of FERM domain protein EPB41L5 as a highly enriched podocyte-specific FA component in vivo. Genetic deletion of Epb41l5 resulted in severe proteinuria, detachment of podocytes, and development of focal segmental glomerulosclerosis. Remarkably, by binding and recruiting the RhoGEF ARGHEF18 to the leading edge, EPB41L5 directly controls actomyosin contractility and subsequent maturation of focal adhesions, cell spreading, and migration. Furthermore, EPB41L5 controls matrix-dependent outside-in signaling by regulating the focal adhesome composition. Thus, by linking extracellular matrix sensing and signaling, focal adhesion maturation, and actomyosin activation EPB41L5 ensures the mechanical stability required for podocytes at the kidney filtration barrier. Finally, a diminution of EPB41L5-dependent signaling programs appears to be a common theme of podocyte disease, and therefore offers unexpected interventional therapeutic strategies to prevent podocyte loss and kidney disease progression.


Subject(s)
Actomyosin/metabolism , Cytoskeletal Proteins/metabolism , Focal Adhesions/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , Animals , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Female , Focal Adhesions/pathology , Gene Knockout Techniques , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Nephrotic Syndrome/etiology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Podocytes/pathology , Pregnancy , Proteomics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction
2.
EMBO J ; 32(23): 3041-54, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24129513

ABSTRACT

Malfunctioning of the protein α-synuclein is critically involved in the demise of dopaminergic neurons relevant to Parkinson's disease. Nonetheless, the precise mechanisms explaining this pathogenic neuronal cell death remain elusive. Endonuclease G (EndoG) is a mitochondrially localized nuclease that triggers DNA degradation and cell death upon translocation from mitochondria to the nucleus. Here, we show that EndoG displays cytotoxic nuclear localization in dopaminergic neurons of human Parkinson-diseased patients, while EndoG depletion largely reduces α-synuclein-induced cell death in human neuroblastoma cells. Xenogenic expression of human α-synuclein in yeast cells triggers mitochondria-nuclear translocation of EndoG and EndoG-mediated DNA degradation through a mechanism that requires a functional kynurenine pathway and the permeability transition pore. In nematodes and flies, EndoG is essential for the α-synuclein-driven degeneration of dopaminergic neurons. Moreover, the locomotion and survival of α-synuclein-expressing flies is compromised, but reinstalled by parallel depletion of EndoG. In sum, we unravel a phylogenetically conserved pathway that involves EndoG as a critical downstream executor of α-synuclein cytotoxicity.


Subject(s)
Apoptosis , Endodeoxyribonucleases/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Parkinson Disease/pathology , Substantia Nigra/pathology , alpha-Synuclein/metabolism , Aged , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , DNA Damage/genetics , Dopamine/pharmacology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Endodeoxyribonucleases/genetics , Humans , Immunoblotting , Immunoenzyme Techniques , Mitochondria/metabolism , Mitochondria/pathology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neurons/cytology , Oxidative Stress , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substantia Nigra/metabolism , Tumor Cells, Cultured , alpha-Synuclein/genetics
3.
Mol Syst Biol ; 9: 657, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23591773

ABSTRACT

The mammalian cellular microenvironment is shaped by soluble factors and structural components, the extracellular matrix, providing physical support, regulating adhesion and signalling. A global, quantitative mass spectrometry strategy, combined with bioinformatics data processing, was developed to assess proteome differences in the microenvironment of primary human fibroblasts. We studied secreted proteins of fibroblasts from normal and pathologically altered skin and their post-translational modifications. The influence of collagen VII, an important structural component, which is lost in genetic skin fragility, was used as model. Loss of collagen VII had a global impact on the cellular microenvironment and was associated with proteome alterations highly relevant for disease pathogenesis including decrease in basement membrane components, increase in dermal matrix proteins, TGF-ß and metalloproteases, but not higher protease activity. The definition of the proteome of fibroblast microenvironment and its plasticity in health and disease identified novel disease mechanisms and potential targets of intervention.


Subject(s)
Basement Membrane/metabolism , Cellular Microenvironment/genetics , Collagen Type VII/genetics , Dermis/metabolism , Epidermolysis Bullosa Dystrophica/genetics , Extracellular Matrix/genetics , Fibroblasts/metabolism , Basement Membrane/pathology , Case-Control Studies , Cell Communication , Collagen Type VII/deficiency , Dermis/pathology , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Extracellular Matrix/pathology , Female , Fibroblasts/pathology , Gene Expression , Humans , Infant , Infant, Newborn , Male , Metalloproteases/genetics , Metalloproteases/metabolism , Primary Cell Culture , Protein Processing, Post-Translational , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
EMBO Mol Med ; 14(5): e13952, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35373908

ABSTRACT

Amyloid beta 42 (Abeta42) is the principal trigger of neurodegeneration during Alzheimer's disease (AD). However, the etiology of its noxious cellular effects remains elusive. In a combinatory genetic and proteomic approach using a yeast model to study aspects of intracellular Abeta42 toxicity, we here identify the HSP40 family member Ydj1, the yeast orthologue of human DnaJA1, as a crucial factor in Abeta42-mediated cell death. We demonstrate that Ydj1/DnaJA1 physically interacts with Abeta42 (in yeast and mouse), stabilizes Abeta42 oligomers, and mediates their translocation to mitochondria. Consequently, deletion of YDJ1 strongly reduces co-purification of Abeta42 with mitochondria and prevents Abeta42-induced mitochondria-dependent cell death. Consistently, purified DnaJ chaperone delays Abeta42 fibrillization in vitro, and heterologous expression of human DnaJA1 induces formation of Abeta42 oligomers and their deleterious translocation to mitochondria in vivo. Finally, downregulation of the Ydj1 fly homologue, Droj2, improves stress resistance, mitochondrial morphology, and memory performance in a Drosophila melanogaster AD model. These data reveal an unexpected and detrimental role for specific HSP40s in promoting hallmarks of Abeta42 toxicity.


Subject(s)
Alzheimer Disease , Saccharomyces cerevisiae Proteins , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Drosophila melanogaster/metabolism , HSP40 Heat-Shock Proteins/genetics , Mice , Molecular Chaperones , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Proteomics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
5.
Biochim Biophys Acta Rev Cancer ; 1873(2): 188340, 2020 04.
Article in English | MEDLINE | ID: mdl-31926290

ABSTRACT

Tumor-derived exosomes are nano-sized vesicles acting as multi-signal devices influencing tumor growth at local and distant sites. Exosomes are derived from the endolysosomal compartment and can shuttle diverse biomolecules like nucleic acids (microRNAs and DNA fragments), lipids, proteins, and even pharmacological compounds from a donor cell to recipient cells. The transfer of cargo to recipient cells enables tumor-derived exosomes to influence diverse cellular functions like proliferation, cell survival, and migration in recipient cells, highlighting tumor-derived exosomes as important players in communication within the tumor microenvironment and at distant sites. In this review, we discuss the mechanisms associated with exosome biogenesis and cargo sorting. In addition, we highlight the communication of tumor-derived exosomes in the tumor microenvironment during different phases of tumor development, focusing on angiogenesis, immune escape mechanisms, drug resistance, and metastasis.


Subject(s)
Carcinogenesis/pathology , Cell Communication , Exosomes/metabolism , Neoplasms/pathology , Tumor Microenvironment , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Movement , Cell Proliferation , Cell Survival , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm , Humans , Lipid Metabolism , MicroRNAs/metabolism , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Tumor Escape
6.
Science ; 361(6409)2018 09 28.
Article in English | MEDLINE | ID: mdl-30262472

ABSTRACT

Cancer cells from a primary tumor can disseminate to other tissues, remaining dormant and clinically undetectable for many years. Little is known about the cues that cause these dormant cells to awaken, resume proliferating, and develop into metastases. Studying mouse models, we found that sustained lung inflammation caused by tobacco smoke exposure or nasal instillation of lipopolysaccharide converted disseminated, dormant cancer cells to aggressively growing metastases. Sustained inflammation induced the formation of neutrophil extracellular traps (NETs), and these were required for awakening dormant cancer. Mechanistic analysis revealed that two NET-associated proteases, neutrophil elastase and matrix metalloproteinase 9, sequentially cleaved laminin. The proteolytically remodeled laminin induced proliferation of dormant cancer cells by activating integrin α3ß1 signaling. Antibodies against NET-remodeled laminin prevented awakening of dormant cells. Therapies aimed at preventing dormant cell awakening could potentially prolong the survival of cancer patients.


Subject(s)
Carcinogenesis/metabolism , Extracellular Traps/enzymology , Lamins/metabolism , Lung Neoplasms/pathology , Neutrophils/enzymology , Pneumonia/pathology , Animals , DNA/metabolism , Humans , Inflammation/chemically induced , Inflammation/microbiology , Integrin alpha3beta1/metabolism , Leukocyte Elastase/metabolism , Lipopolysaccharides , Lung/pathology , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Pneumonia/chemically induced , Pneumonia/microbiology , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/pathology , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases/antagonists & inhibitors , Protein-Arginine Deiminases/metabolism , Proteolysis , Rats , Signal Transduction , Smoking , Nicotiana
7.
Cell Rep ; 23(8): 2495-2508, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791858

ABSTRACT

Damage to and loss of glomerular podocytes has been identified as the culprit lesion in progressive kidney diseases. Here, we combine mass spectrometry-based proteomics with mRNA sequencing, bioinformatics, and hypothesis-driven studies to provide a comprehensive and quantitative map of mammalian podocytes that identifies unanticipated signaling pathways. Comparison of the in vivo datasets with proteomics data from podocyte cell cultures showed a limited value of available cell culture models. Moreover, in vivo stable isotope labeling by amino acids uncovered surprisingly rapid synthesis of mitochondrial proteins under steady-state conditions that was perturbed under autophagy-deficient, disease-susceptible conditions. Integration of acquired omics dimensions suggested FARP1 as a candidate essential for podocyte function, which could be substantiated by genetic analysis in humans and knockdown experiments in zebrafish. This work exemplifies how the integration of multi-omics datasets can identify a framework of cell-type-specific features relevant for organ health and disease.


Subject(s)
Gene Expression Regulation , Genetic Association Studies , Kidney Diseases/genetics , Podocytes/metabolism , Animals , Base Sequence , Cells, Cultured , Humans , Mice , Proteome/metabolism , Transcriptome/genetics , Zebrafish
8.
Oncotarget ; 8(23): 37478-37490, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28415582

ABSTRACT

The anthraquinone emodin has been shown to have antineoplastic properties and a wealth of unconnected effects have been linked to its use, most of which are likely secondary outcomes of the drug treatment. The primary activity of emodin on cells has remained unknown. In the present study we demonstrate dramatic and extensive effects of emodin on the redox state of cells and on mitochondrial homeostasis, irrespectively of the cell type and organism, ranging from the yeast Saccharomyces cerevisiae to human cell lines and primary cells. Emodin binds to redox-active enzymes and its effectiveness depends on the oxidative and respiratory status of cells. We show that cells with efficient respiratory metabolism are less susceptible to emodin, whereas cells under glycolytic metabolism are more vulnerable to the compound. Our findings indicate that emodin acts in a similar way as known uncouplers of the mitochondrial electron transport chain and causes oxidative stress that particularly disturbs cancer cells.


Subject(s)
Cell Proliferation/drug effects , Emodin/pharmacology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , A549 Cells , Caco-2 Cells , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Humans , MCF-7 Cells , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Proteomics/methods , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism
9.
Autophagy ; 13(6): 1064-1075, 2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28453381

ABSTRACT

Macroautophagy is regarded as a nonspecific bulk degradation process of cytoplasmic material within the lysosome. However, the process has mainly been studied by nonspecific bulk degradation assays using radiolabeling. In the present study we monitor protein turnover and degradation by global, unbiased approaches relying on quantitative mass spectrometry-based proteomics. Macroautophagy is induced by rapamycin treatment, and by amino acid and glucose starvation in differentially, metabolically labeled cells. Protein dynamics are linked to image-based models of autophagosome turnover. Depending on the inducing stimulus, protein as well as organelle turnover differ. Amino acid starvation-induced macroautophagy leads to selective degradation of proteins important for protein translation. Thus, protein dynamics reflect cellular conditions in the respective treatment indicating stimulus-specific pathways in stress-induced macroautophagy.


Subject(s)
Amino Acids/deficiency , Autophagy , Protein Biosynthesis , Proteolysis , Autophagosomes/metabolism , Humans , Isotope Labeling , MCF-7 Cells
10.
Sci Transl Med ; 8(361): 361ra138, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798263

ABSTRACT

Neutrophils, the most abundant type of leukocytes in blood, can form neutrophil extracellular traps (NETs). These are pathogen-trapping structures generated by expulsion of the neutrophil's DNA with associated proteolytic enzymes. NETs produced by infection can promote cancer metastasis. We show that metastatic breast cancer cells can induce neutrophils to form metastasis-supporting NETs in the absence of infection. Using intravital imaging, we observed NET-like structures around metastatic 4T1 cancer cells that had reached the lungs of mice. We also found NETs in clinical samples of triple-negative human breast cancer. The formation of NETs stimulated the invasion and migration of breast cancer cells in vitro. Inhibiting NET formation or digesting NETs with deoxyribonuclease I (DNase I) blocked these processes. Treatment with NET-digesting, DNase I-coated nanoparticles markedly reduced lung metastases in mice. Our data suggest that induction of NETs by cancer cells is a previously unidentified metastasis-promoting tumor-host interaction and a potential therapeutic target.


Subject(s)
Extracellular Traps , Neoplasm Metastasis , Neutrophils/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Deoxyribonuclease I/chemistry , Humans , Lung/pathology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neutrophils/cytology
11.
Cell Metab ; 22(4): 721-33, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26387864

ABSTRACT

The essential oxidoreductase Mia40/CHCHD4 mediates disulfide bond formation and protein folding in the mitochondrial intermembrane space. Here, we investigated the interactome of Mia40 thereby revealing links between thiol-oxidation and apoptosis, energy metabolism, and Ca(2+) signaling. Among the interaction partners of Mia40 is MICU1-the regulator of the mitochondrial Ca(2+) uniporter (MCU), which transfers Ca(2+) across the inner membrane. We examined the biogenesis of MICU1 and find that Mia40 introduces an intermolecular disulfide bond that links MICU1 and its inhibitory paralog MICU2 in a heterodimer. Absence of this disulfide bond results in increased receptor-induced mitochondrial Ca(2+) uptake. In the presence of the disulfide bond, MICU1-MICU2 heterodimer binding to MCU is controlled by Ca(2+) levels: the dimer associates with MCU at low levels of Ca(2+) and dissociates upon high Ca(2+) concentrations. Our findings support a model in which mitochondrial Ca(2+) uptake is regulated by a Ca(2+)-dependent remodeling of the uniporter complex.


Subject(s)
Calcium Channels/metabolism , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Calcium Channels/chemistry , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Dimerization , Disulfides/chemistry , Disulfides/metabolism , HEK293 Cells , HeLa Cells , Humans , Ions/chemistry , Ions/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , RNA Interference , RNA, Small Interfering/metabolism
12.
J Invest Dermatol ; 134(9): 2381-2389, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24732400

ABSTRACT

Absence of collagen VII leads to widespread cellular and tissue phenotypes. However, the underlying molecular mechanisms are not well understood. To gain insights into cellular responses to loss of collagen VII, we undertook a quantitative disease proteomics approach. By using recessive dystrophic epidermolysis bullosa (RDEB), a skin blistering disease caused by collagen VII deficiency, as a genetic model, collagen VII-dependent differences in cellular protein abundances and protein-protein interactions were analyzed. Absence of collagen VII led to alterations of intracellular protein compositions and to perturbations in cell adhesion, protein trafficking, and the turnover pathway autophagy. A potential linker of the different cellular phenotypes is transglutaminase 2 (TGM2), a multifunctional enzyme important for protein cross-linking. TGM2 was identified as a stable interaction partner of collagen VII. In RDEB, both abundance and activity of TGM2 were reduced, accounting not only for diminished adhesion and perturbed autophagy but also for reduced cross-linking of the extracellular matrix and for decreased epidermal-dermal integrity in RDEB.


Subject(s)
Collagen Type VII/genetics , Dermis/enzymology , Epidermis/enzymology , Epidermolysis Bullosa Dystrophica , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism , Autophagy/physiology , Cell Adhesion/physiology , Collagen Type VII/isolation & purification , Collagen Type VII/metabolism , Dermis/cytology , Enzyme Activation/physiology , Epidermal Cells , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Fibroblasts/cytology , Fibroblasts/enzymology , Humans , Infant, Newborn , Phenotype , Primary Cell Culture , Protein Glutamine gamma Glutamyltransferase 2 , Proteome
13.
J Invest Dermatol ; 134(9): 2321-2330, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24496236

ABSTRACT

Aging is a common risk factor of many disorders. With age, the level of insoluble extracellular matrix increases leading to increased stiffness of a number of tissues. Matrix accumulation can also be observed in fibrotic disorders, such as systemic sclerosis (SSc). Although the intrinsic aging process in skin is phenotypically distinct from SSc, here we demonstrate similar behavior of aged and SSc skin fibroblasts in culture. We have used quantitative proteomics to characterize the phenotype of dermal fibroblasts from healthy subjects of various ages and from patients with SSc. Our results demonstrate that proteins involved in DNA and RNA processing decrease with age and in SSc, whereas those involved in mitochondrial and other metabolic processes behave the opposite. Specifically, minichromosome maintenance (MCM) helicase proteins are less abundant with age and SSc, and they exhibit an altered subcellular distribution. We observed that lower levels of MCM7 correlate with reduced cell proliferation, lower autophagic capacity, and higher intracellular protein abundance phenotypes of aged and SSc cells. In addition, we show that SSc fibroblasts exhibit higher levels of senescence compared with their healthy counterparts, suggesting further similarities between the fibrotic disorder and the aging process. Hence, at the molecular level, SSc fibroblasts exhibit intrinsic characteristics of fibroblasts from aged skin.


Subject(s)
Autophagy/physiology , Minichromosome Maintenance Complex Component 7/metabolism , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Skin Aging/physiology , Adolescent , Adult , Aged , Cells, Cultured , Child, Preschool , Dermis/cytology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Infant, Newborn , Male , Middle Aged , Minichromosome Maintenance Complex Component 7/genetics , Phenotype , Proteomics , RNA, Messenger/metabolism , Scleroderma, Systemic/genetics , Young Adult
14.
Cell Metab ; 19(3): 431-44, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24606900

ABSTRACT

Healthy aging depends on removal of damaged cellular material that is in part mediated by autophagy. The nutritional status of cells affects both aging and autophagy through as-yet-elusive metabolic circuitries. Here, we show that nucleocytosolic acetyl-coenzyme A (AcCoA) production is a metabolic repressor of autophagy during aging in yeast. Blocking the mitochondrial route to AcCoA by deletion of the CoA-transferase ACH1 caused cytosolic accumulation of the AcCoA precursor acetate. This led to hyperactivation of nucleocytosolic AcCoA-synthetase Acs2p, triggering histone acetylation, repression of autophagy genes, and an age-dependent defect in autophagic flux, culminating in a reduced lifespan. Inhibition of nutrient signaling failed to restore, while simultaneous knockdown of ACS2 reinstated, autophagy and survival of ach1 mutant. Brain-specific knockdown of Drosophila AcCoA synthetase was sufficient to enhance autophagic protein clearance and prolong lifespan. Since AcCoA integrates various nutrition pathways, our findings may explain diet-dependent lifespan and autophagy regulation.


Subject(s)
Autophagy , Coenzyme A Ligases/metabolism , Drosophila Proteins/metabolism , Longevity , Acetyl Coenzyme A/biosynthesis , Acetylation , Aging , Animals , Autophagy-Related Protein 7 , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/genetics , Drosophila/enzymology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Energy Metabolism , Histones/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Up-Regulation
15.
Methods Mol Biol ; 961: 179-91, 2013.
Article in English | MEDLINE | ID: mdl-23325643

ABSTRACT

Physiological functions of skin cells are often altered in diseases. Since the underlying molecular mechanisms are generally executed by proteins, it is of interest to assess protein dynamics in normal and pathologically altered cells. These can be readily analyzed in relevant cell culture models by quantitative mass spectrometry (MS)-based proteomics, which is the method of choice to track the concerted action and spatial relocation of unknown involved factors in an unbiased way. Different quantitative MS strategies have been used to characterize protein dynamics. In this chapter we describe in detail the use of stable isotope labeling by amino acids in cell culture for an unbiased quantitative analysis of protein dynamics in the two major cell types of the skin, keratinocytes and fibroblasts.


Subject(s)
Fibroblasts/pathology , Keratinocytes/pathology , Proteome/analysis , Proteomics/methods , Cell Line , Cell Line, Tumor , Cell Separation/methods , Cells, Cultured , Fibroblasts/metabolism , Humans , Isotope Labeling/methods , Keratinocytes/metabolism , Male , Mass Spectrometry/methods , Proteome/metabolism
16.
Mol Biol Cell ; 24(14): 2160-70, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23676665

ABSTRACT

Oxidation of cysteine residues to disulfides drives import of many proteins into the intermembrane space of mitochondria. Recent studies in yeast unraveled the basic principles of mitochondrial protein oxidation, but the kinetics under physiological conditions is unknown. We developed assays to follow protein oxidation in living mammalian cells, which reveal that import and oxidative folding of proteins are kinetically and functionally coupled and depend on the oxidoreductase Mia40, the sulfhydryl oxidase augmenter of liver regeneration (ALR), and the intracellular glutathione pool. Kinetics of substrate oxidation depends on the amount of Mia40 and requires tightly balanced amounts of ALR. Mia40-dependent import of Cox19 in human cells depends on the inner membrane potential. Our observations reveal considerable differences in the velocities of mitochondrial import pathways: whereas preproteins with bipartite targeting sequences are imported within seconds, substrates of Mia40 remain in the cytosol for several minutes and apparently escape premature degradation and oxidation.


Subject(s)
Cytochrome Reductases/metabolism , Fibroblasts/metabolism , Glutathione/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Animals , Biological Assay , Cytochrome Reductases/genetics , Disulfides/chemistry , Disulfides/metabolism , Fibroblasts/cytology , Gene Expression Regulation , HEK293 Cells , Humans , Kinetics , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors , Protein Folding , Protein Stability , Protein Transport , Proteolysis , Signal Transduction , Sulfur Radioisotopes
17.
PLoS One ; 7(7): e41897, 2012.
Article in English | MEDLINE | ID: mdl-22860026

ABSTRACT

Mass Spectrometry (MS) is becoming a preferred method to identify class I and class II peptides presented on major histocompability complexes (MHC) on antigen presenting cells (APC). We describe a combined computational and MS approach to identify exogenous MHC II peptides presented on mouse spleen dendritic cells (DCs). This approach enables rapid, effective screening of a large number of possible peptides by a computer-assisted strategy that utilizes the extraordinary human ability for pattern recognition. To test the efficacy of the approach, a mixture of epitope peptide mimics (mimetopes) from HIV gag p24 sequence were added exogenously to Fms-like tyrosine kinase 3 ligand (Flt3L)-mobilized splenic DCs. We identified the exogenously added peptide, VDRFYKTLRAEQASQ, and a second peptide, DRFYKLTRAEQASQ, derived from the original exogenously added 15-mer peptide. Furthermore, we demonstrated that our strategy works efficiently with HIV gag p24 protein when delivered, as vaccine protein, to Flt3L expanded mouse splenic DCs in vitro through the DEC-205 receptor. We found that the same MHC II-bound HIV gag p24 peptides, VDRFYKTLRAEQASQ and DRFYKLTRAEQASQ, were naturally processed from anti-DEC-205 HIV gag p24 protein and presented on DCs. The two identified VDRFYKTLRAEQASQ and DRFYKLTRAEQASQ MHC II-bound HIV gag p24 peptides elicited CD4(+) T-cell mediated responses in vitro. Their presentation by DCs to antigen-specific T cells was inhibited by chloroquine (CQ), indicating that optimal presentation of these exogenously added peptides required uptake and vesicular trafficking in mature DCs. These results support the application of our strategy to identify and characterize peptide epitopes derived from vaccine proteins processed by DCs and thus has the potential to greatly accelerate DC-based vaccine development.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , HIV Core Protein p24/immunology , Amino Acid Sequence , Animals , Antigen Presentation , Antigens, Viral/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/metabolism , HIV Core Protein p24/metabolism , Histocompatibility Antigens Class II/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Fragments/metabolism
18.
Mol Biosyst ; 6(9): 1579-82, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20454738

ABSTRACT

We monitor proteome changes of primary human skin fibroblasts and keratinocytes during cell culture and compare them to respective immortalized cell lines using stable isotope labeling by amino acids to address their suitability as cell models for clinical disease proteomics.


Subject(s)
Cellular Senescence/physiology , Fibroblasts/metabolism , Keratinocytes/metabolism , Proteome/metabolism , Cells, Cultured , Humans
SELECTION OF CITATIONS
SEARCH DETAIL