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










Publication year range
1.
J Biol Chem ; 298(10): 102325, 2022 10.
Article in English | MEDLINE | ID: mdl-35926710

ABSTRACT

Neurite outgrowth is an integrated whole cell response triggered by the cannabinoid-1 receptor. We sought to identify the many different biochemical pathways that contribute to this whole cell response. To understand underlying mechanisms, we identified subcellular processes (SCPs) composed of one or more biochemical pathways and their interactions required for this response. Differentially expressed genes and proteins were obtained from bulk transcriptomics and proteomic analysis of extracts from cells stimulated with a cannabinoid-1 receptor agonist. We used these differentially expressed genes and proteins to build networks of interacting SCPs by combining the expression data with prior pathway knowledge. From these SCP networks, we identified additional genes that when ablated, experimentally validated the SCP involvement in neurite outgrowth. Our experiments and informatics modeling allowed us to identify diverse SCPs such as those involved in pyrimidine metabolism, lipid biosynthesis, and mRNA splicing and stability, along with more predictable SCPs such as membrane vesicle transport and microtubule dynamics. We find that SCPs required for neurite outgrowth are widely distributed among many biochemical pathways required for constitutive cellular functions, several of which are termed 'deep', since they are distal to signaling pathways and the key SCPs directly involved in extension of the neurite. In contrast, 'proximal' SCPs are involved in microtubule growth and membrane vesicle transport dynamics required for neurite outgrowth. From these bioinformatics and dynamical models based on experimental data, we conclude that receptor-mediated regulation of subcellular functions for neurite outgrowth is both distributed, that is, involves many different biochemical pathways, and deep.


Subject(s)
Cannabinoid Receptor Agonists , Neurites , Neuronal Outgrowth , Proteomics , Receptor, Cannabinoid, CB1 , Neurites/drug effects , Neurites/metabolism , Neuronal Outgrowth/drug effects , Signal Transduction , Receptor, Cannabinoid, CB1/metabolism , Cannabinoid Receptor Agonists/pharmacology , Humans
2.
J Proteomics ; 138: 40-7, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26917471

ABSTRACT

Soluble Guanylyl Cyclase (sGC) is the main receptor for nitric oxide (NO). NO activates sGC to synthesize cGMP, triggering a plethora of signals. Recently, we discovered that NO covalently modifies select sGC cysteines via a post-translational modification termed S-nitrosation or S-nitrosylation. Earlier characterization was conducted on a purified sGC treated with S-nitrosoglutathione, and identified three S-nitrosated cysteines (SNO-Cys). Here we describe a more biologically relevant mapping of sGC SNO-Cys in cells to better understand the multi-faceted interactions between SNO and sGC. Since SNO-Cys are labile during LC/MS/MS, MS analysis of nitrosation typically occurs after a biotin switch reaction, in which a SNO-Cys is converted to a biotin-Cys. Here we report the identification of ten sGC SNO-Cys in rat neonatal cardiomyocytes using an Orbitrap MS. A majority of the SNO-Cys identified is located at the solvent-exposed surface of the sGC, and half of them in the conserved catalytic domain, suggesting biological significance. These findings provide a solid basis for future studies of the regulations and functions of diverse sGC S-nitrosation events in cells.


Subject(s)
Myocytes, Cardiac/enzymology , Nitric Oxide/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Cysteine/analogs & derivatives , Cysteine/chemistry , Cysteine/metabolism , Myocytes, Cardiac/cytology , Nitric Oxide/chemistry , Nitrosation , Rats , Rats, Wistar , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/metabolism , S-Nitrosothiols/chemistry , S-Nitrosothiols/metabolism , Soluble Guanylyl Cyclase/chemistry
3.
Free Radic Biol Med ; 89: 548-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453926

ABSTRACT

Nuclear factor E2-related factor 2 (Nrf2) is a key transcription factor that regulates the expression of a number of antioxidant and detoxifying genes that provide cellular protection against various stressors including reactive oxygen species (ROS). Nrf2 activity is tightly regulated by a cytoplasmic inhibitory protein called Kelch-like ECH-associated protein 1 (Keap1). The mechanism that controls Keap1 expression, however, remains poorly understood. In the present study, we demonstrate that microRNA-7 (miR-7), which is highly expressed in the brain, represses Keap1 expression by targeting the 3'-untranslated region (UTR) of its mRNA in human neuroblastoma cells, SH-SY5Y. Subsequently, this event results in an increased Nrf2 activity, as evidenced by an increase in the expression of its transcriptional targets, heme oxygenase 1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), and an enhanced nuclear localization of Nrf2. In addition, miR-7 decreases the intracellular hydroperoxides level and increases the level of reduced form of glutathione, indicative of oxidative stress relief. We also demonstrate that targeted repression of Keap1 and activation of Nrf2 pathway, in part, underlies the protective effects of miR-7 against 1-methyl-4-phenylpyridinium (MPP+)-induced toxicity in SH-SY5Y and differentiated human neural progenitor cells, ReNcell VM. These findings point to a new mechanism by which miR-7 exerts cytoprotective effects by regulating the Nrf2 pathway.


Subject(s)
Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/biosynthesis , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Blotting, Western , Cell Line , Chromatography, Liquid , Humans , Kelch-Like ECH-Associated Protein 1 , Polymerase Chain Reaction , Signal Transduction/physiology , Tandem Mass Spectrometry , Transfection
4.
Biochim Biophys Acta ; 1854(12): 1816-1822, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410624

ABSTRACT

Thioredoxin 1 (Trx1) is а antioxidant protein that regulates protein disulfide bond reduction, transnitrosylation, denitrosylation and other redox post-translational modifications. In order to better understand how Trx1 modulates downstream protective cellular signaling events following cardiac ischemia, we conducted an expression proteomics study of left ventricles (LVs) after thoracic aortic constriction stress treatment of transgenic mice with cardiac-specific over-expression of Trx1, an animal model that has been proven to withstand more stress than its non-transgenic littermates. Although previous redox post-translational modifications proteomics studies found that several cellular protein networks are regulated by Trx1-mediated disulfide reduction and transnitrosylation, we found that Trx1 regulates the expression of a limited number of proteins. Among the proteins found to be upregulated in this study was SET and MYND domain-containing protein 1 (SMYD1), a lysine methyltransferase highly expressed in cardiac and other muscle tissues and an important regulator of cardiac development. The observation of SMYD1 induction by Trx1 following thoracic aortic constriction stress is consistent with the retrograde fetal gene cardiac protection hypothesis. The results presented here suggest for the first time that, in addition to being a master redox regulator of protein disulfide bonds and nitrosation, Trx1 may also modulate lysine methylation, a non-redox post-translational modification, via the regulation of SMYD1 expression. Such crosstalk between redox signaling and a non-redox PTM regulation may provide novel insights into the functions of Trx1 that are independent from its immediate function as a protein reductase.


Subject(s)
DNA Methylation , DNA-Binding Proteins/physiology , Lysine/metabolism , Muscle Proteins/physiology , Thioredoxins/physiology , Transcription Factors/physiology , Up-Regulation , Animals , Mice , Mice, Transgenic , Oxidation-Reduction
5.
Mol Cell Proteomics ; 13(12): 3507-18, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231459

ABSTRACT

The dysregulation of protein oxidative post-translational modifications has been implicated in stress-related diseases. Trx1 is a key reductase that reduces specific disulfide bonds and other cysteine post-translational modifications. Although commonly in the cytoplasm, Trx1 can also modulate transcription in the nucleus. However, few Trx1 nuclear targets have been identified because of the low Trx1 abundance in the nucleus. Here, we report the large-scale proteomics identification of nuclear Trx1 targets in human neuroblastoma cells using an affinity capture strategy wherein a Trx1C35S mutant is expressed. The wild-type Trx1 contains a conserved C32XXC35 motif, and the C32 thiol initiates the reduction of a target disulfide bond by forming an intermolecular disulfide with one of the oxidized target cysteines, resulting in a transient Trx1-target protein complex. The reduction is rapidly consummated by the donation of a C35 proton to the target molecule, forming a Trx1 C32-C35 disulfide, and results in the concurrent release of the target protein containing reduced thiols. By introducing a point mutation (C35 to S35) in Trx1, we ablated the rapid dissociation of Trx1 from its reduction targets, thereby allowing the identification of 45 putative nuclear Trx1 targets. Unexpectedly, we found that PSIP1, also known as LEDGF, was sensitive to both oxidation and Trx1 reduction at Cys 204. LEDGF is a transcription activator that is vital for regulating cell survival during HIV-1 infection. Overall, this study suggests that Trx1 may play a broader role than previously believed that might include regulating transcription, RNA processing, and nuclear pore function in human cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Cysteine/metabolism , Neurons/metabolism , Thioredoxins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , Cell Line, Tumor , Cysteine/chemistry , Cytoplasm/metabolism , Disulfides/chemistry , Gene Expression Profiling , Gene Expression Regulation , Humans , Molecular Sequence Annotation , Molecular Sequence Data , Mutation , Neurons/cytology , Oxidation-Reduction , Protein Interaction Mapping , Signal Transduction , Thioredoxins/genetics , Transcription Factors/genetics , Transcription, Genetic
6.
J Neurosci ; 34(38): 12725-37, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25232110

ABSTRACT

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Mitochondrial complex I impairment in PD is modeled in vitro by the susceptibility of dopaminergic neurons to the complex I inhibitor 1-methyl-4-phenylpyridinium (MPP+). In the present study, we demonstrate that microRNA-7 (miR-7), which is expressed in tyrosine hydroxylase-positive nigral neurons in mice and humans, protects cells from MPP+-induced toxicity in dopaminergic SH-SY5Y cells, differentiated human neural progenitor ReNcell VM cells, and primary mouse neurons. RelA, a component of nuclear factor-κB (NF-κB), was identified to be downregulated by miR-7 using quantitative proteomic analysis. Through a series of validation experiments, it was confirmed that RelA mRNA is a target of miR-7 and is required for cell death following MPP+ exposure. Further, RelA mediates MPP+-induced suppression of NF-κB activity, which is essential for MPP+-induced cell death. Accordingly, the protective effect of miR-7 is exerted through relieving NF-κB suppression by reducing RelA expression. These findings provide a novel mechanism by which NF-κB suppression, rather than activation, underlies the cell death mechanism following MPP+ toxicity, have implications for the pathogenesis of PD, and suggest miR-7 as a therapeutic target for this disease.


Subject(s)
1-Methyl-4-phenylpyridinium/antagonists & inhibitors , MicroRNAs/physiology , Neuroprotective Agents/metabolism , Parkinson Disease, Secondary/prevention & control , Transcription Factor RelA/biosynthesis , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Down-Regulation , Humans , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , NF-kappa B/biosynthesis , Neurons/drug effects , Parkinson Disease, Secondary/chemically induced , Substantia Nigra/metabolism , Transcription Factor RelA/genetics , Transfection , alpha-Synuclein/genetics
7.
Mol Cell ; 54(4): 639-50, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24813943

ABSTRACT

The Hippo pathway, evolutionarily conserved from flies to mammals, promotes cell death and inhibits cell proliferation to regulate organ size. The core component of this cascade, Mst1 in mammalian cells, is sufficient to promote apoptosis. However, the mechanisms underlying both its activation and its ability to elicit cell death remain largely undefined. We here identify a signaling cassette in cardiac myocytes consisting of K-Ras, the scaffold RASSF1A, and Mst1 that is localized to mitochondria and promotes Mst1 activation in response to oxidative stress. Activated Mst1 phosphorylates Bcl-xL at Ser14, which resides in the BH4 domain, thereby antagonizing Bcl-xL-Bax binding. This, in turn, causes activation of Bax and subsequent mitochondria-mediated apoptotic death. Our findings demonstrate mitochondrial localization of Hippo signaling and identify Bcl-xL as a target that is directly modified to promote apoptosis.


Subject(s)
Apoptosis , Mitochondria/metabolism , Myocytes, Cardiac/physiology , Protein Serine-Threonine Kinases/metabolism , bcl-X Protein/metabolism , Animals , Binding Sites/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Transgenic , Oxidative Stress , Phosphorylation , Rats , Rats, Wistar , Serine/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism , ras Proteins/metabolism
8.
Antimicrob Agents Chemother ; 58(6): 3373-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24687510

ABSTRACT

Invasive pulmonary aspergillosis (IPA) is a life-threatening disease of immunocompromised patients that requires aggressive therapy. Detection of the disease and monitoring of the therapeutic response during IPA are complex, and current molecular diagnostics are not suitably robust. Here, we explored proteomic profiles of bronchoalveolar lavage fluid (BALF) specimens from a persistently neutropenic rabbit model of IPA. Three experimental arms, uninfected control animals, infected untreated animals, and animals infected and treated with ravuconazole/amphotericin B, were studied. Total proteins were evaluated by two-dimensional (2D) gel electrophoresis, followed by matrix-assisted laser desorption ionization-time of flight/time of flight (MALDI-TOF/TOF) mass spectrometry (MS) and quantified by enzyme-linked immunosorbent assay (ELISA). Host-derived proteins haptoglobin (Hp), C-reactive protein (CRP), and annexin A1 (Anx A1) were prominently found in BALF during the IPA infection and showed significant changes in response to antifungal therapy (P < 0.0001). In serum, differences in Hp (P = 0.0001) between infected and treated rabbits were observed. Preliminary in vitro studies revealed that Aspergillus fumigatus-secreted proteases may contribute to the cleavage of Anx A1 during IPA. In summary, host protein biomarkers Hp, CRP, and Anx A1 may have value in monitoring therapeutic response to antifungal agents in IPA patients with confirmed disease.


Subject(s)
Antifungal Agents/therapeutic use , Aspergillus fumigatus/metabolism , Invasive Pulmonary Aspergillosis/drug therapy , Proteomics , Amphotericin B/therapeutic use , Animals , Annexin A1/metabolism , Aspergillus fumigatus/drug effects , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/microbiology , C-Reactive Protein/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Neutropenia , Rabbits , Thiazoles/therapeutic use , Triazoles/therapeutic use
9.
ASN Neuro ; 5(5): e00131, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24195677

ABSTRACT

OPC (oligodendrocyte progenitor cell) death contributes significantly to the pathology and functional deficits following hypoxic-ischemic injury in the immature brain and to deficits resulting from demyelinating diseases, trauma and degenerative disorders in the adult CNS. Glutamate toxicity is a major cause of oligodendroglial death in diverse CNS disorders, and previous studies have demonstrated that AMPA/kainate receptors require the pro-apoptotic protein Bax in OPCs undergoing apoptosis. The goal of the present study was to define the pro-apoptotic and anti-apoptotic effectors that regulate Bax in healthy OPCs and after exposure to excess glutamate in vitro and following H-I (hypoxia-ischemia) in the immature rat brain. We show that Bax associates with a truncated form of Bid, a BH3-only domain protein, subsequent to glutamate treatment. Furthermore, glutamate exposure reduces Bax association with the anti-apoptotic Bcl family member, Bcl-xL. Cell fractionation studies demonstrated that both Bax and Bid translocate from the cytoplasm to mitochondria during the early stages of cell death consistent with a role for Bid as an activator, whereas Bcl-xL, which normally complexes with both Bax and Bid, disassociates from these complexes when OPCs are exposed to excess glutamate. Bax remained unactivated in the presence of insulin-like growth factor-1, and the Bcl-xL complexes were protected. Our data similarly demonstrate loss of Bcl-xL-Bax association in white matter following H-I and implicate active Bad in Bax-mediated OPC death. To identify other Bax-binding partners, we used proteomics and identified cofilin as a Bax-associated protein in OPCs. Cofilin and Bax associated in healthy OPCs, whereas the Bax-cofilin association was disrupted during glutamate-induced OPC apoptosis.


Subject(s)
Gene Expression Regulation/physiology , Hypoxia-Ischemia, Brain/pathology , Neurotoxicity Syndromes/pathology , Oligodendroglia/metabolism , Stem Cells/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Cerebellum/cytology , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Humans , Insulin-Like Growth Factor I/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Neurotoxicity Syndromes/etiology , Oligodendroglia/chemistry , Oligodendroglia/drug effects , Protein Transport/drug effects , Rats , Rats, Wistar , Stem Cells/drug effects , Time Factors
10.
J Proteome Res ; 12(9): 4221-9, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23855383

ABSTRACT

Mammalian hibernation is a unique strategy for winter survival in response to limited food supply and harsh climate, which includes resistance to cardiac arrhythmias. We previously found that hibernating woodchucks (Marmota monax) exhibit natural resistance to Ca2+ overload-related cardiac dysfunction and nitric oxide (NO)-dependent vasodilation, which maintains myocardial blood flow during hibernation. Since the cellular/molecular mechanisms mediating the protection are less clear, the goal of this study was to investigate changes in the heart proteome and reveal related signaling networks that are involved in establishing cardioprotection in woodchucks during hibernation. This was accomplished using isobaric tags for a relative and absolute quantification (iTRAQ) approach. The most significant changes observed in winter hibernation compared to summer non-hibernation animals were upregulation of the antioxidant catalase and inhibition of endoplasmic reticulum (ER) stress response by downregulation of GRP78, mechanisms which could be responsible for the adaptation and protection in hibernating animals. Furthermore, protein networks pertaining to NO signaling, acute phase response, CREB and NFAT transcriptional regulations, protein kinase A and α-adrenergic signaling were also dramatically upregulated during hibernation. These adaptive mechanisms in hibernators may provide new directions to protect myocardium of non-hibernating animals, especially humans, from cardiac dysfunction induced by hypothermic stress and myocardial ischemia.


Subject(s)
Heart/physiology , Hibernation , Marmota/physiology , Proteome/metabolism , Animals , Endoplasmic Reticulum Chaperone BiP , Female , Intracellular Signaling Peptides and Proteins/metabolism , Male , Protein Interaction Maps , Proteomics , Signal Transduction , Ventricular Pressure
11.
J Neurosci Methods ; 216(2): 118-27, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23558336

ABSTRACT

The dynamic regulation of transcriptional events is fundamental to many aspects of neuronal cell functions. However, proteomics methods have not been routinely used in global neuroproteomics analyses of transcriptional regulators because they are much less abundant than the "house-keeping" proteins in cells and tissues. Recent improvements in both biochemical preparations of nuclear proteins and detection sensitivities of proteomics technologies have made the global analysis of nuclear transcriptional regulators possible. We report here an optimised neuroproteomic method for the analysis of transcriptional regulators in the nuclear extracts of SHSY-5Y neuroblastoma cells by combining an improved nuclear protein extraction procedure with multidimensional peptide separation approaches. We found that rigorous removal of cytoplasmic proteins and solubilisation of DNA-associated proteins improved the number of nuclear proteins identified. Furthermore, we discovered that multidimensional peptide separations by either strong cation exchange (SCX) chromatography or electrostatic repulsion-hydrophilic interaction chromatography (ERLIC) analysis detected more than 1800 nuclear proteins, which constitutes one of the largest datasets of nuclear proteins reported for a neuronal cell. Thus, in-depth analysis of transcriptional regulators for studying neurological diseases are increasingly feasible.


Subject(s)
Neuroblastoma/metabolism , Nuclear Proteins/analysis , Proteomics/methods , Blotting, Western , Cell Line, Tumor , Chromatography, Liquid/methods , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Neoplastic , Humans , Proteome/metabolism , Tandem Mass Spectrometry
12.
Protein Eng Des Sel ; 26(5): 347-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23447652

ABSTRACT

Formation of the ternary complex between GTP-bound form of elongation factor Tu (EF-Tu) and aminoacylated transfer RNA (aa-tRNA) is a key event in protein biosynthesis. Here we show that fluorescently modified Escherichia coli EF-Tu carrying three mutations, C137A, C255V and E348C, and fluorescently modified Phe-tRNA(Phe) form functionally active ternary complex that has properties similar to those of the naturally occurring (unmodified) complex. Similarities include the binding and binding rate constants, behavior in gel retardation assay, as well as activities in tRNA protection and in vitro translation assays. Proper labeling of EF-Tu was demonstrated in MALDI mass spectroscopy experiments. To generate the mutant EF-Tu, a series of genetic constructions were performed. Two native cysteine residues in the wild-type EF-Tu at positions 137 and 255 were replaced by Ala and Val, respectively, and an additional cysteine was introduced either in position 324 or 348. The assembly FRET assay showed a 5- to 7-fold increase of Cy5-labeled EF-Tu E348C mutant fluorescence upon formation of ternary complex with charged tRNA(Phe)(Cy3-labeled) when the complex was excited at 532 nm and monitored at 665 nm. In a control experiment, we did not observe FRET using uncharged tRNA(Phe)(Cy3), nor with wild-type EF-Tu preparation that was allowed to react with Cy5 maleimide, nor in the absence of GTP. The results obtained demonstrate that the EF-Tu:tRNA FRET system described can be used for investigations of ribosomal translation in many types of experiments.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Fluorescence Resonance Energy Transfer/methods , Peptide Elongation Factor Tu/metabolism , RNA, Transfer/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Escherichia coli/genetics , Escherichia coli Proteins/analysis , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Elongation Factor Tu/analysis , Peptide Elongation Factor Tu/genetics , Protein Biosynthesis , RNA, Transfer/analysis , RNA, Transfer/genetics , Ribosomes/genetics
13.
J Biol Chem ; 287(13): 10325-10343, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22262862

ABSTRACT

The intriguing cell biology of apoptotic cell death results in the externalization of numerous autoantigens on the apoptotic cell surface, including protein determinants for specific recognition, linked to immune responses. Apoptotic cells are recognized by phagocytes and trigger an active immunosuppressive response ("innate apoptotic immunity" (IAI)) even in the absence of engulfment. IAI is responsible for the lack of inflammation associated normally with the clearance of apoptotic cells; its failure also has been linked to inflammatory and autoimmune pathology, including systemic lupus erythematosus and rheumatic diseases. Apoptotic recognition determinants underlying IAI have yet to be identified definitively; we argue that these molecules are surface-exposed (during apoptotic cell death), ubiquitously expressed, protease-sensitive, evolutionarily conserved, and resident normally in viable cells (SUPER). Using independent and unbiased quantitative proteomic approaches to characterize apoptotic cell surface proteins and identify candidate SUPER determinants, we made the surprising discovery that components of the glycolytic pathway are enriched on the apoptotic cell surface. Our data demonstrate that glycolytic enzyme externalization is a common and early aspect of cell death in different cell types triggered to die with distinct suicidal stimuli. Exposed glycolytic enzyme molecules meet the criteria for IAI-associated SUPER determinants. In addition, our characterization of the apoptosis-specific externalization of glycolytic enzyme molecules may provide insight into the significance of previously reported cases of plasminogen binding to α-enolase on mammalian cells, as well as mechanisms by which commensal bacteria and pathogens maintain immune privilege.


Subject(s)
Apoptosis , Glycolysis , Immunity, Innate , Animals , Biomarkers/metabolism , Humans , Jurkat Cells , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/pathology , Mice , Proteomics , U937 Cells
14.
J Proteome Res ; 11(3): 1791-803, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22188123

ABSTRACT

Clinically relevant formalin-fixed and paraffin-embedded (FFPE) tissues have not been widely used in neuroproteomic studies because many proteins are presumed to be degraded during tissue preservation. Recent improvements in proteomics technologies, from the 2D gel analysis of intact proteins to the "shotgun" quantification of peptides and the use of isobaric tags for absolute and relative quantification (iTRAQ) method, have made the analysis of FFPE tissues possible. In recent years, iTRAQ has been one of the main methods of choice for high throughput quantitative proteomics analysis, which enables simultaneous comparison of up to eight samples in one experiment. Our objective was to assess the relative merits of iTRAQ analysis of fresh frozen versus FFPE nervous tissues by comparing experimental autoimmune encephalomyelitis (EAE)-induced proteomic changes in FFPE rat spinal cords and frozen tissues. EAE-induced proteomic changes in FFPE tissues were positively correlated with those found in the frozen tissues, albeit with ∼50% less proteome coverage. Subsequent validation of the enrichment of immunoproteasome (IP) activator 1 in EAE spinal cords led us to evaluate other proteasome and IP-specific proteins. We discovered that many IP-specific (as opposed to constitutive) proteasomal proteins were enriched in EAE rat spinal cords, and EAE-induced IP accumulation also occurred in the spinal cords of an independent mouse EAE model in a disability score-dependent manner. Therefore, we conclude that it is feasible to generate useful information from iTRAQ-based neuroproteomics analysis of archived FFPE tissues for studying neurological disease tissues.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteome/metabolism , Spinal Cord/metabolism , Animals , Antigen Presentation , Cysteine Endopeptidases/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Proteomics , Rats , Rats, Inbred Lew , Spinal Cord/enzymology , Spinal Cord/immunology , Up-Regulation
15.
Glia ; 59(11): 1754-69, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21858874

ABSTRACT

Previous work from our laboratory demonstrated that the mammalian target of rapamycin (mTOR) is active during and required for oligodendrocyte progenitor cell (OPC) differentiation. Here, we applied an iTRAQ mass spectrometry-based proteomic approach to identify novel targets of the mTOR pathway during OPC differentiation. Among the 978 proteins identified in this study, 328 (34%) exhibited a greater than 20% change (P < 0.05) in control versus rapamycin-treated cultures following 4 days of differentiation in vitro. Interestingly, 197 (20%) proteins were elevated in rapamycin-treated cultures, while 131 (13%) proteins were downregulated by rapamycin. In support of our previous data, inhibiting mTOR caused a dramatic reduction in the expression of myelin proteins. mTOR also was required for the induction of proteins involved in cholesterol and fatty acid synthesis, as well as the expression of many cytoskeletal proteins, cell signaling components, and nuclear/transcriptional regulators. Of particular interest was the identification of several critical mediators of oligodendrocyte differentiation. Specifically, mTOR activity controls the developmentally programmed upregulation of the prodifferentiation factors Fyn and Quaking, whereas the expression of the differentiation repressor Gpr17 was elevated by mTOR inhibition. These data reveal a distinct signature of mTOR-regulated protein expression during OPC differentiation.


Subject(s)
Cell Differentiation/physiology , Oligodendroglia/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Computational Biology , Culture Media , Cytoskeleton/metabolism , Databases, Protein , Microarray Analysis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Peptides/metabolism , Proteomics , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stem Cells , TOR Serine-Threonine Kinases/genetics , Tandem Mass Spectrometry
16.
Mol Cell Proteomics ; 10(11): M111.010108, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21825280

ABSTRACT

Aspergillus fumigatus is a saprophytic fungus that causes a range of diseases in humans including invasive aspergillosis. All forms of disease begin with the inhalation of conidia, which germinate and develop. Four stages of early development were evaluated using the gel free system of isobaric tagging for relative and absolute quantitation to determine the full proteomic profile of the pathogen. A total of 461 proteins were identified at 0, 4, 8, and 16 h and fold changes for each were established. Ten proteins including the hydrophobin rodlet protein RodA and a protein involved in melanin synthesis Abr2 were found to decrease relative to conidia. To generate a more comprehensive view of early development, a whole genome microarray analysis was performed comparing conidia to 8 and 16 h of growth. A total of 1871 genes were found to change significantly at 8 h with 1001 genes up-regulated and 870 down-regulated. At 16 h, 1235 genes changed significantly with 855 up-regulated and 380 down-regulated. When a comparison between the proteomics and microarray data was performed at 8 h, a total of 22 proteins with significant changes also had corresponding genes that changed significantly. When the same comparison was performed at 16 h, 12 protein and gene combinations were found. This study, the most comprehensive to date, provides insights into early pathways activated during growth and development of A. fumigatus. It reveals a pathogen that is gearing up for rapid growth by building translation machinery, generating ATP, and is very much committed to aerobic metabolism.


Subject(s)
Aspergillus fumigatus/growth & development , Fungal Proteins/metabolism , Proteome/metabolism , Aspergillosis/microbiology , Aspergillus fumigatus/genetics , Aspergillus fumigatus/metabolism , Biomarkers/metabolism , Drug Design , Fungal Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal , Genome, Fungal , Mycelium , Oligonucleotide Array Sequence Analysis , Proteome/genetics
17.
J Proteomics ; 74(11): 2498-509, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-21704743

ABSTRACT

S-Nitrosylation is a reversible PTM for regulating protein function. Thioredoxin-1 (Trx1) catalyzes either transnitrosylation or denitrosylation of specific proteins, depending on the redox status of the cysteines within its conserved oxidoreductase CXXC motif. With a disulfide bond formed between the two catalytic cysteines, Trx1 is not only inactive as a denitrosylase, but it may also be nitrosylated at Cys73 and serve as a transnitrosylating agent. Identification of Trx1-mediated transnitrosylation or denitrosylation targets will contribute to a better understanding of Trx1's function. Previous experimental approaches based on the attenuation of CXXC oxidoreductase activity cannot readily distinguish Trx1 transnitrosylation targets from denitrosylation targets. In this study, we used the ICAT method in conjunction with the biotin switch technique to differentiate Trx1 transnitrosylation targets from denitrosylation target proteins from neuroblastoma cells. We demonstrate that the ICAT approach is effective for quantitative identification of putative Trx1 transnitrosylation and denitrosylation target peptides. From these analyses, we confirmed reports that peroxiredoxin 1 is a Trx1 transnitrosylation, but not a denitrosylation target, and we found several other proteins, including cyclophilin A to be modulated in this manner. Unexpectedly, we found that many nitrosylation sites are reversibly regulated by Trx1, suggesting a more prominent role for Trx1 in regulating S-nitrosylation.


Subject(s)
Protein Processing, Post-Translational , Proteins/analysis , Proteins/metabolism , Thioredoxins/metabolism , Thioredoxins/physiology , Amino Acid Sequence , Catalytic Domain , Cells, Cultured , Humans , Isotope Labeling/methods , Models, Biological , Nitrosation/physiology , Oxidation-Reduction , Peptide Mapping , Protein Processing, Post-Translational/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Antimicrob Agents Chemother ; 55(1): 146-54, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974863

ABSTRACT

The proteomic response of Aspergillus fumigatus to caspofungin was evaluated by gel-free isobaric tagging for relative and absolute quantitation (iTRAQ) as a means to determine potential biomarkers of drug action. A cell fractionation approach yielding 4 subcellular compartment fractions was used to enhance the resolution of proteins for proteomic analysis. Using iTRAQ, a total of 471 unique proteins were identified in soluble and cell wall/plasma membrane fractions at 24 and 48 h of growth in rich media in a wild-type drug-susceptible strain. A total of 122 proteins showed at least a 2-fold change in relative abundance following exposure to caspofungin (CSF) at just below the minimum effective concentration (0.12 µg/ml). The largest changes were seen in the mitochondrial hypoxia response domain protein (AFUA_1G12250), the level of which decreased >16-fold in the secreted fraction, and ChiA1, the level of which decreased 12.1-fold in the cell wall/plasma membrane fraction. The level of the major allergen and cytotoxin AspF1 was also shown to decrease by 12.1-fold upon the addition of drug. A subsequent iTRAQ analysis of an echinocandin-resistant strain (fks1-S678P) was used to validate proteins specific to drug action. A total of 103 proteins in the 2 fractions tested by iTRAQ were differentially expressed in the wild-type susceptible strain but not significantly changed in the resistant strain. Of these potential biomarkers, 11 had levels that changed at least 12-fold. Microarray analysis of the susceptible strain was performed to evaluate the correlation between proteomics and genomics, with a total of 117 genes found to be changing at least 2-fold. Of these, a total of 22 proteins with significant changes identified by iTRAQ also showed significant gene expression level changes by microarray. Overall, these data have the potential to identify biomarkers that assess the relative efficacy of echinocandin drug therapy.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/metabolism , Echinocandins/pharmacology , Fungal Proteins/metabolism , Proteome/drug effects , Proteome/metabolism , Caspofungin , Computational Biology , Electrophoresis, Gel, Two-Dimensional , Fungal Proteins/genetics , Lipopeptides , Oligonucleotide Array Sequence Analysis
19.
Mol Cell Proteomics ; 9(10): 2262-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660346

ABSTRACT

Transnitrosylation and denitrosylation are emerging as key post-translational modification events in regulating both normal physiology and a wide spectrum of human diseases. Thioredoxin 1 (Trx1) is a conserved antioxidant that functions as a classic disulfide reductase. It also catalyzes the transnitrosylation or denitrosylation of caspase 3 (Casp3), underscoring its central role in determining Casp3 nitrosylation specificity. However, the mechanisms that regulate Trx1 transnitrosylation and denitrosylation of specific targets are unresolved. Here we used an optimized mass spectrometric method to demonstrate that Trx1 is itself nitrosylated by S-nitrosoglutathione at Cys(73) only after the formation of a Cys(32)-Cys(35) disulfide bond upon which the disulfide reductase and denitrosylase activities of Trx1 are attenuated. Following nitrosylation, Trx1 subsequently transnitrosylates Casp3. Overexpression of Trx1(C32S/C35S) (a mutant Trx1 with both Cys(32) and Cys(35) replaced by serine to mimic the disulfide reductase-inactive Trx1) in HeLa cells promoted the nitrosylation of specific target proteins. Using a global proteomics approach, we identified 47 novel Trx1 transnitrosylation target protein candidates. From further bioinformatics analysis of this set of nitrosylated peptides, we identified consensus motifs that are likely to be the determinants of Trx1-mediated transnitrosylation specificity. Among these proteins, we confirmed that Trx1 directly transnitrosylates peroxiredoxin 1 at Cys(173) and Cys(83) and protects it from H(2)O(2)-induced overoxidation. Functionally, we found that Cys(73)-mediated Trx1 transnitrosylation of target proteins is important for protecting HeLa cells from apoptosis. These data demonstrate that the ability of Trx1 to transnitrosylate target proteins is regulated by a crucial stepwise oxidative and nitrosative modification of specific cysteines, suggesting that Trx1, as a master regulator of redox signaling, can modulate target proteins via alternating modalities of reduction and nitrosylation.


Subject(s)
Nitroso Compounds/metabolism , Thioredoxins/metabolism , Amino Acid Sequence , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Mass Spectrometry , Oxidation-Reduction
20.
Proteome Sci ; 7: 25, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19607715

ABSTRACT

BACKGROUND: Abnormal activation of protease activities during experimental autoimmune encephalomyelitis (EAE) in rats, a rodent model of multiple sclerosis, have been implicated in either the direct destruction of myelin components or the intracellular signal transduction pathways that lead to lymphocyte infiltration, oligodendrocyte destruction, neuronal dysfunctions and axonal degeneration. The identification of changes in regulated proteolytic events during EAE is crucial for uncovering activated proteases that may underline the pathological features such as inflammation and demyelination. We searched for either non-tryptic or semi-tryptic peptides from a previous shotgun proteomics study using isobaric tags for relative and absolute quantification (iTRAQ) to compare the proteomes of normal and EAE rat lumbar spinal cords. RESULTS: We discovered that several proteins, such as alpha1-macroglobulin, a protease inhibitor, alpha1B-glycoprotein, beta2-microglobulin, neurofilament light polypeptide and sulfated glycoprotein 1 had non-tryptic peptide iTRAQ ratios that were substantially different from the overall protein iTRAQ ratios, suggesting that such peptides may be markers for the proteolytic products generated by the protease(s) altered during EAE. Indeed, subsequent Western blotting confirmed the dysregulation of specific protein cleavages in EAE tissues. Additional proteolytic changes in alpha2-macroglobulin, another protease inhibitor similar to alpha1-macroglobulin was also observed. CONCLUSION: The results from this study revealed changes among both neuronal protein processing and endogenous proteolysis modulators in EAE animals. This information may provide a rationale for protease inhibitor-based therapeutic interventions for multiple sclerosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...