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










Publication year range
1.
Sci Rep ; 14(1): 11402, 2024 05 18.
Article in English | MEDLINE | ID: mdl-38762561

ABSTRACT

Despite the therapeutic potential of chemogenetics, the method lacks comprehensive preclinical validation, hindering its progression to human clinical trials. We aimed to validate a robust but simple in vivo efficacy assay in rats which could support chemogenetic drug discovery by providing a quick, simple and reliable animal model. Key methodological parameters such as adeno-associated virus (AAV) serotype, actuator drug, dose, and application routes were investigated by measuring the food-intake-reducing effect of chemogenetic inhibition of the lateral hypothalamus (LH) by hM4D(Gi) designer receptor stimulation. Subcutaneous deschloroclozapine in rats transfected with AAV9 resulted in a substantial reduction of food-intake, comparable to the efficacy of exenatide. We estimated that the effect of deschloroclozapine lasts 1-3 h post-administration. AAV5, oral administration of deschloroclozapine, and clozapine-N-oxide were also effective but with slightly less potency. The strongest effect on food-intake occurred within the first 30 min after re-feeding, suggesting this as the optimal experimental endpoint. This study demonstrates that general chemogenetic silencing of the LH can be utilized as an optimal, fast and reliable in vivo experimental model for conducting preclinical proof-of-concept studies in order to validate the in vivo effectiveness of novel chemogenetic treatments. We also hypothesize based on our results that universal LH silencing with existing and human translatable genetic neuroengineering techniques might be a viable strategy to affect food intake and influence obesity.


Subject(s)
Clozapine , Dependovirus , Eating , Hypothalamic Area, Lateral , Proof of Concept Study , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Rats , Eating/drug effects , Hypothalamic Area, Lateral/drug effects , Dependovirus/genetics , Male , Exenatide/pharmacology , Humans
2.
J Biol Chem ; 300(4): 107155, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479597

ABSTRACT

Despite significant advances in the development of therapeutic interventions targeting autoimmune diseases and chronic inflammatory conditions, lack of effective treatment still poses a high unmet need. Modulating chronically activated T cells through the blockade of the Kv1.3 potassium channel is a promising therapeutic approach; however, developing selective Kv1.3 inhibitors is still an arduous task. Phage display-based high throughput peptide library screening is a rapid and robust approach to develop promising drug candidates; however, it requires solid-phase immobilization of target proteins with their binding site preserved. Historically, the KcsA bacterial channel chimera harboring only the turret region of the human Kv1.3 channel was used for screening campaigns. Nevertheless, literature data suggest that binding to this type of chimera does not correlate well with blocking potency on the native Kv1.3 channels. Therefore, we designed and successfully produced advanced KcsA-Kv1.3, KcsA-Kv1.1, and KcsA-Kv1.2 chimeric proteins in which both the turret and part of the filter regions of the human Kv1.x channels were transferred. These T+F (turret-filter) chimeras showed superior peptide ligand-binding predictivity compared to their T-only versions in novel phage ELISA assays. Phage ELISA binding and competition results supported with electrophysiological data confirmed that the filter region of KcsA-Kv1.x is essential for establishing adequate relative affinity order among selected peptide toxins (Vm24 toxin, Hongotoxin-1, Kaliotoxin-1, Maurotoxin, Stichodactyla toxin) and consequently obtaining more reliable selectivity data. These new findings provide a better screening tool for future drug development efforts and offer insight into the target-ligand interactions of these therapeutically relevant ion channels.


Subject(s)
Kv1.3 Potassium Channel , Potassium Channel Blockers , Recombinant Fusion Proteins , Animals , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/chemistry , Ligands , Peptide Library , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Cell Line
3.
J Biol Chem ; 299(9): 104998, 2023 09.
Article in English | MEDLINE | ID: mdl-37394009

ABSTRACT

Chlorotoxin (CTX), a scorpion venom-derived 36-residue miniprotein, binds to and is taken up selectively by glioblastoma cells. Previous studies provided controversial results concerning target protein(s) of CTX. These included CLC3 chloride channel, matrix metalloproteinase 2 (MMP-2), regulators of MMP-2, annexin A2, and neuropilin 1 (NRP1). The present study aimed at clarifying which of the proposed binding partners can really interact with CTX using biochemical methods and recombinant proteins. For this purpose, we established two new binding assays based on anchoring the tested proteins to microbeads and quantifying the binding of CTX by flow cytometry. Screening of His-tagged proteins anchored to cobalt-coated beads indicated strong interaction of CTX with MMP-2 and NRP1, whereas binding to annexin A2 was not confirmed. Similar results were obtained with fluorophore-labeled CTX and CTX-displaying phages. Affinity of CTX to MMP-2 and NRP1 was assessed by the "immunoglobulin-coated bead" test, in which the proteins were anchored to beads by specific antibodies. This assay yielded highly reproducible data using both direct titration and displacement approach. The affinities of labeled and unlabeled CTX appeared to be similar for both MMP-2 and NRP1 with estimated KD values of 0.5 to 0.7 µM. Contrary to previous reports, we found that CTX does not inhibit the activity of MMP-2 and that CTX not only with free carboxyl end but also with carboxamide terminal end binds to NRP1. We conclude that the presented robust assays could also be applied for affinity-improving studies of CTX to its genuine targets using phage display libraries.


Subject(s)
Glioblastoma , Matrix Metalloproteinase 2 , Neuropilin-1 , Scorpion Venoms , Humans , Glioblastoma/metabolism , Matrix Metalloproteinase 2/metabolism , Neuropilin-1/metabolism , Scorpion Venoms/metabolism , Cell Line, Tumor , Protein Binding
4.
Orv Hetil ; 162(15): 563-570, 2021 04 02.
Article in Hungarian | MEDLINE | ID: mdl-33798101

ABSTRACT

Összefoglaló. A koronavírus-betegség 2019 (COVID-19)-pandémia komoly kihívás elé állította nemcsak a mikrobiológiai laboratóriumokat, hanem az eredmények interpretálásában a klinikumban dolgozó kollégákat is. Az orvostudomány specializált világában az immunológiai és a fertozo betegségekkel kapcsolatos ismeretek az antimikrobás terápiás megoldások sikeressége, valamint a széles köru vakcináció miatt az idok folyamán számos szakterületen háttérbe szorultak, felfrissítésük sürgeto és elengedhetetlen része a pandémiával való megküzdésnek. A diagnosztikai vizsgálatok fontos eszközei a járvány megfékezésének, illetve a betegek ellátásának, azonban a vírus és az emberi szervezet interakciójának megértése elengedhetetlenül szükséges a korrekt epidemiológiai és gyógyászati véleményalkotáshoz. Jelen cikkünk az orvosi gyakorlat számára foglalja össze a súlyos akut légzoszervi szindrómát okozó koronavírus-2 (SARS-CoV-2) kimutatására, valamint az immunrendszer specifikus immunválaszának szerológiai vizsgálatára irányuló, gyakorlatban használatos módszereket, azok helyét, szerepét és értékelésük szempontjait a tudomány jelen állása szerint. Orv Hetil. 2021; 162(15): 563-570. Summary. The coronavirus disease 2019 (COVID-19) pandemic posed a serious challenge not only for microbiology laboratories, but also for the clinicians in interpretation of the results. In the specialized world of medicine, knowledge of immunological and infectious diseases has been relegated to the background in many disciplines over time due to the success of antimicrobial therapies and widespread vaccination, so updating them is an urgent and essential part of the fight against the pandemic. Diagnostic tests are important tools for controlling the epidemic and caring for patients, but understanding the interaction between the virus and the human body is essential to form a correct epidemiological and medical opinion. This paper summarizes the medical methods for the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the serological testing of the specific immune response of the immune system, their place, role and criteria of their evaluation according to current scientific knowledge. Orv Hetil. 2021; 162(15): 563-570.


Subject(s)
Antibodies, Viral , Antigens, Viral , COVID-19 Serological Testing , COVID-19 , SARS-CoV-2 , Antibodies, Viral/isolation & purification , Antigens, Viral/isolation & purification , COVID-19/diagnosis , Humans , SARS-CoV-2/immunology
5.
Nucleic Acids Res ; 49(2): e9, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33264395

ABSTRACT

There has been a surge of interest towards targeting protein synthesis to treat diseases and extend lifespan. Despite the progress, few options are available to assess translation in live animals, as their complexity limits the repertoire of experimental tools to monitor and manipulate processes within organs and individual cells. It this study, we developed a labeling-free method for measuring organ- and cell-type-specific translation elongation rates in vivo. It is based on time-resolved delivery of translation initiation and elongation inhibitors in live animals followed by ribosome profiling. It also reports translation initiation sites in an organ-specific manner. Using this method, we found that the elongation rates differ more than 50% among mouse organs and determined them to be 6.8, 5.0 and 4.3 amino acids per second for liver, kidney, and skeletal muscle, respectively. We further found that the elongation rate is reduced by 20% between young adulthood and mid-life. Thus, translation, a major metabolic process in cells, is tightly regulated at the level of elongation of nascent polypeptide chains.


Subject(s)
Aging/metabolism , Kidney/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Peptide Chain Elongation, Translational , Aging/genetics , Animals , Cluster Analysis , Cranial Sinuses , Cycloheximide/administration & dosage , Cycloheximide/pharmacology , Drug Administration Schedule , Harringtonines/administration & dosage , Harringtonines/pharmacology , High-Throughput Nucleotide Sequencing , Injections, Intravenous , Kinetics , Longevity , Macrolides/administration & dosage , Macrolides/pharmacology , Male , Mice , Mice, Inbred C57BL , Orbit , Organ Specificity , Peptide Chain Elongation, Translational/drug effects , Peptide Chain Initiation, Translational , Piperidones/administration & dosage , Piperidones/pharmacology , Ribosomes/metabolism , Tail , Transcriptome
6.
Methods Mol Biol ; 1661: 285-299, 2018.
Article in English | MEDLINE | ID: mdl-28917052

ABSTRACT

The sulfur-containing amino acid methionine (Met) plays critical roles in protein synthesis, methylation, and sulfur metabolism. Both in its free form and in the form of an amino acid residue, it can be oxidized to the R and S diastereomers of methionine sulfoxide (MetO). Organisms evolved methionine sulfoxide reductases (MSRs) to reduce MetO to Met, with the MSRs type A (MSRA) and type B (MSRB) being specific for the S and R forms of MetO, respectively. In mammals, the selenoprotein MSRB1 plays an important protein repair function, and its expression is tightly regulated by dietary selenium. In this chapter, we describe a protocol for determining the concentration of protein-based Met-R-O and its analysis in HEK293 cells using a genetically encoded ratiometric fluorescent biosensor MetROx. We also describe the procedure for quantifying MSR activities in cell extracts using specific substrates and a reverse phase HPLC-based method.


Subject(s)
Biosensing Techniques , Methionine Sulfoxide Reductases/metabolism , Methionine/analogs & derivatives , Cell Line , Chromatography, High Pressure Liquid , Enzyme Activation , Gene Expression , Genes, Reporter , Humans , Methionine/metabolism , Molecular Imaging , Oxidation-Reduction , Oxidative Stress
7.
Free Radic Biol Med ; 116: 41-49, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29278739

ABSTRACT

The p22phox protein is an essential component of the phagocytic- and inner ear NADPH oxidases but its relationship to other Nox proteins is less clear. We have studied the role of p22phox in the TGF-ß1-stimulated H2O2 production of primary human and murine fibroblasts. TGF-ß1 induced H2O2 release of the examined cells, and the response was dependent on the expression of both Nox4 and p22phox. Interestingly, the p22phox protein was present in the absence of any detectable Nox/Duox expression, and the p22phox level was unaffected by TGF-ß1. On the other hand, Nox4 expression was dependent on the presence of p22phox, establishing an asymmetrical relationship between the two proteins. Nox4 and p22phox proteins localized to the endoplasmic reticulum and their distribution was unaffected by TGF-ß1. We used a chemically induced protein dimerization method to study the orientation of p22phox and Nox4 in the endoplasmic reticulum membrane. This technique is based on the rapamycin-mediated heterodimerization of the mammalian FRB domain with the FK506 binding protein. The results of these experiments suggest that the enzyme complex produces H2O2 into the lumen of the endoplasmic reticulum, indicating that Nox4 contributes to the development of the oxidative milieu within this organelle.


Subject(s)
Cytochrome b Group/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/physiology , Multiprotein Complexes/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidases/metabolism , Animals , Cytochrome b Group/genetics , Dimerization , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Mutant Strains , NADPH Oxidase 4/genetics , NADPH Oxidases/genetics , Oxidation-Reduction , Protein Binding , Reactive Oxygen Species/metabolism , Sirolimus/metabolism , Transforming Growth Factor beta1/immunology
8.
Mol Biol Evol ; 33(10): 2670-81, 2016 10.
Article in English | MEDLINE | ID: mdl-27555581

ABSTRACT

Snub-nosed monkeys (genus Rhinopithecus) are a group of endangered colobines endemic to South Asia. Here, we re-sequenced the whole genomes of 38 snub-nosed monkeys representing four species within this genus. By conducting population genomic analyses, we observed a similar load of deleterious variation in snub-nosed monkeys living in both smaller and larger populations and found that genomic diversity was lower than that reported in other primates. Reconstruction of Rhinopithecus evolutionary history suggested that episodes of climatic variation over the past 2 million years, associated with glacial advances and retreats and population isolation, have shaped snub-nosed monkey demography and evolution. We further identified several hypoxia-related genes under selection in R. bieti (black snub-nosed monkey), a species that exploits habitats higher than any other nonhuman primate. These results provide the first detailed and comprehensive genomic insights into genetic diversity, demography, genetic burden, and adaptation in this radiation of endangered primates.


Subject(s)
Adaptation, Physiological/genetics , Colobinae/genetics , Hypoxia/veterinary , Acclimatization/genetics , Adaptation, Biological/genetics , Animals , Base Sequence , Ecosystem , Genetic Variation , Hypoxia/genetics , Hypoxia/metabolism , Metagenomics/methods , Phylogeny , Polymorphism, Genetic , Sequence Analysis, DNA/veterinary
9.
Methods ; 109: 149-157, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27345570

ABSTRACT

In cells, physiological and pathophysiological conditions may lead to the formation of methionine sulfoxide (MetO). This oxidative modification of methionine exists in the form of two diastereomers, R and S, and may occur in both free amino acid and proteins. MetO is reduced back to methionine by methionine sulfoxide reductases (MSRs). Methionine oxidation was thought to be a nonspecific modification affecting protein functions and methionine availability. However, recent findings suggest that cyclic methionine oxidation and reduction is a posttranslational modification that actively regulates protein function akin to redox regulation by cysteine oxidation and phosphorylation. Methionine oxidation is thus an important mechanism that could play out in various physiological contexts. However, detecting MetO generation and MSR functions remains challenging because of the lack of tools and reagents to detect and quantify this protein modification. We recently developed two genetically encoded diasterospecific fluorescent sensors, MetSOx and MetROx, to dynamically monitor MetO in living cells. Here, we provide a detailed procedure for their use in bacterial and mammalian cells using fluorimetric and fluorescent imaging approaches. This method can be adapted to dynamically monitor methionine oxidation in various cell types and under various conditions.


Subject(s)
Biosensing Techniques/methods , Methionine Sulfoxide Reductases/chemistry , Methionine/analogs & derivatives , Molecular Imaging/methods , Animals , Bacteria/chemistry , Humans , Mammals , Methionine/chemistry , Methionine/isolation & purification , Methionine Sulfoxide Reductases/genetics , Oxidation-Reduction , Protein Processing, Post-Translational/genetics , Stereoisomerism
10.
Nat Chem Biol ; 11(5): 332-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25799144

ABSTRACT

Methionine can be reversibly oxidized to methionine sulfoxide (MetO) under physiological and pathophysiological conditions, but its use as a redox marker suffers from the lack of tools to detect and quantify MetO within cells. In this work, we created a pair of complementary stereospecific genetically encoded mechanism-based ratiometric fluorescent sensors of MetO by inserting a circularly permuted yellow fluorescent protein between yeast methionine sulfoxide reductases and thioredoxins. The two sensors, respectively named MetSOx and MetROx for their ability to detect S and R forms of MetO, were used for targeted analysis of protein oxidation, regulation and repair as well as for monitoring MetO in bacterial and mammalian cells, analyzing compartment-specific changes in MetO and examining responses to physiological stimuli.


Subject(s)
Methionine/analogs & derivatives , Methionine/chemistry , Escherichia coli/drug effects , Fluorescence , HEK293 Cells , Humans , Methionine/analysis , Oxidants/pharmacology , Oxidation-Reduction , Sodium Hypochlorite/pharmacology , Stereoisomerism
11.
Free Radic Biol Med ; 83: 273-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25708780

ABSTRACT

Basement membranes provide structural support and convey regulatory signals to cells in diverse tissues. Assembly of collagen IV into a sheet-like network is a fundamental mechanism during the formation of basement membranes. Peroxidasin (PXDN) was recently described to catalyze crosslinking of collagen IV through the formation of sulfilimine bonds. Despite the significance of this pathway in tissue genesis, our understanding of PXDN function is far from complete. In this work we demonstrate that collagen IV crosslinking is a physiological function of mammalian PXDN. Moreover, we carried out structure-function analysis of PXDN to gain a better insight into its role in collagen IV synthesis. We identify conserved cysteines in PXDN that mediate the oligomerization of the protein into a trimeric complex. We also demonstrate that oligomerization is not an absolute requirement for enzymatic activity, but optimal collagen IV coupling is only catalyzed by the PXDN trimers. Localization experiments of different PXDN mutants in two different cell models revealed that PXDN oligomers, but not monomers, adhere on the cell surface in "hot spots," which represent previously unknown locations of collagen IV crosslinking.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Basement Membrane/metabolism , Collagen Type IV/chemistry , Extracellular Matrix Proteins/physiology , Extracellular Matrix/metabolism , Peroxidase/physiology , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Animals , Antigens, Neoplasm/genetics , Apoptosis , Blotting, Western , Catalysis , Cell Proliferation , Cells, Cultured , Collagen Type IV/metabolism , Cross-Linking Reagents/pharmacology , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Mice , Mice, Knockout , Peroxidases , Protein Conformation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Peroxidasin
12.
Trends Biochem Sci ; 39(7): 305-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24924147

ABSTRACT

Stabilization of extracellular matrix by protein crosslinking is a universal and essential process in multicellular organisms. Recent findings revealed that peroxidasin, a unique heme-peroxidase, produces hypohalides to support matrix synthesis. Unexpectedly, the highly reactive and potentially damaging hypohalides mediate the formation of sulfilimine bonds between adjacent collagen IV protomers. This crosslink is a fundamental feature of basal membranes, defining peroxidasin-dependent oxidant generation and sulfilimine crosslink formation as an elemental mechanism of tissue biogenesis.


Subject(s)
Basement Membrane/metabolism , Cross-Linking Reagents/metabolism , Extracellular Matrix Proteins/metabolism , Peroxidase/metabolism , Animals , Collagen Type IV/chemistry , Humans , Sulfur Compounds/chemistry , Peroxidasin
13.
Cardiovasc Res ; 101(3): 393-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24253521

ABSTRACT

AIMS: Peroxidases serve diverse biological functions including well-characterized activities in host defence and hormone biosynthesis. More recently, peroxidasin (PXDN) was found to be involved in collagen IV cross-linking in the extracellular matrix (ECM). The aim of this study was to characterize the expression and function of peroxidasin-like protein (PXDNL), a previously unknown peroxidase homologue. METHODS AND RESULTS: We cloned the PXDNL cDNA from the human heart and identified its expression pattern by northern blot, in situ hybridization, and immunohistochemistry. PXDNL is expressed exclusively in the heart and it has evolved to lose its peroxidase activity. The protein is produced by cardiomyocytes and localizes to cell-cell junctions. We also demonstrate that PXDNL can form a complex with PXDN and antagonizes its peroxidase activity. Furthermore, we show an increased expression of PXDNL in the failing myocardium. CONCLUSION: PXDNL is a unique component of the heart with a recently evolved inactivation of peroxidase function. The elevation of PXDNL levels in the failing heart may contribute to ECM dysregulation due to its antagonism of PXDN function.


Subject(s)
Extracellular Matrix Proteins/pharmacology , Gene Expression Regulation , Heart/drug effects , Peroxidase/pharmacology , Animals , Cells, Cultured , Extracellular Matrix/metabolism , Heart Failure/metabolism , Humans , In Situ Hybridization/methods , Oxidation-Reduction/drug effects , RNA, Messenger/metabolism , Peroxidasin
14.
J Mol Signal ; 8: 8, 2013.
Article in English | MEDLINE | ID: mdl-23924390

ABSTRACT

BACKGROUND: Tks5/FISH is a scaffold protein comprising of five SH3 domains and one PX domain. Tks5 is a substrate of the tyrosine kinase Src and is required for the organization of podosomes/invadopodia implicated in invasion of tumor cells. Recent data have suggested that a close homologue of Tks5, Tks4, is implicated in the EGF signaling. RESULTS: Here, we report that Tks5 is a component of the EGF signaling pathway. In EGF-treated cells, Tks5 is tyrosine phosphorylated within minutes and the level of phosphorylation is sustained for at least 2 hours. Using specific kinase inhibitors, we demonstrate that tyrosine phosphorylation of Tks5 is catalyzed by Src tyrosine kinase. We show that treatment of cells with EGF results in plasma membrane translocation of Tks5. In addition, treatment of cells with LY294002, an inhibitor of PI 3-kinase, or mutation of the PX domain reduces tyrosine phosphorylation and membrane translocation of Tks5. CONCLUSIONS: Our results identify Tks5 as a novel component of the EGF signaling pathway.

15.
Mol Cell ; 51(3): 397-404, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23911929

ABSTRACT

Redox control of protein function involves oxidation and reduction of amino acid residues, but the mechanisms and regulators involved are insufficiently understood. Here, we report that in conjunction with Mical proteins, methionine-R-sulfoxide reductase B1 (MsrB1) regulates mammalian actin assembly via stereoselective methionine oxidation and reduction in a reversible, site-specific manner. Two methionine residues in actin are specifically converted to methionine-R-sulfoxide by Mical1 and Mical2 and reduced back to methionine by selenoprotein MsrB1, supporting actin disassembly and assembly, respectively. Macrophages utilize this redox control during cellular activation by stimulating MsrB1 expression and activity as a part of innate immunity. We identified the regulatory role of MsrB1 as a Mical antagonist in orchestrating actin dynamics and macrophage function. More generally, our study shows that proteins can be regulated by reversible site-specific methionine-R-sulfoxidation.


Subject(s)
Actins/metabolism , Macrophages/metabolism , Methionine Sulfoxide Reductases/genetics , Methionine/metabolism , Microtubule-Associated Proteins/metabolism , Mixed Function Oxygenases/metabolism , Oxidoreductases/metabolism , Animals , Cells, Cultured , Mice , Mice, Knockout , Microfilament Proteins , Oxidation-Reduction , Oxidative Stress , Oxidoreductases/genetics
16.
PLoS One ; 6(8): e23653, 2011.
Article in English | MEDLINE | ID: mdl-21886807

ABSTRACT

Motility of normal and transformed cells within and across tissues requires specialized subcellular structures, e.g. membrane ruffles, lamellipodia and podosomes, which are generated by dynamic rearrangements of the actin cytoskeleton. Because the formation of these sub-cellular structures is complex and relatively poorly understood, we evaluated the role of the adapter protein SH3PXD2B [HOFI, fad49, Tks4], which plays a role in the development of the eye, skeleton and adipose tissue. Surprisingly, we find that SH3PXD2B is requisite for the development of EGF-induced membrane ruffles and lamellipodia, as well as for efficient cellular attachment and spreading of HeLa cells. Furthermore, SH3PXD2B is present in a complex with the non-receptor protein tyrosine kinase Src, phosphorylated by Src, which is consistent with SH3PXD2B accumulating in Src-induced podosomes. Furthermore, SH3PXD2B closely follows the subcellular relocalization of cortactin to Src-induced podosomes, EGF-induced membrane ruffles and lamellipodia. Because SH3PXD2B also forms a complex with the C-terminal region of cortactin, we propose that SH3PXD2B is a scaffold protein that plays a key role in regulating the actin cytoskeleton via Src and cortactin.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Pseudopodia/metabolism , Sequence Homology, Amino Acid , src Homology Domains , Actins/metabolism , Cortactin/metabolism , ErbB Receptors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Phosphatidylinositols/metabolism , Protein Binding , Protein Transport
17.
Free Radic Biol Med ; 49(12): 2040-8, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21146788

ABSTRACT

Hydrogen peroxide (H(2)O(2)) has important messenger and effector functions in the plant and animal kingdom. Phagocytes produce H(2)O(2) to kill pathogens, and epithelial cells of large airways have also been reported to produce H(2)O(2) for signaling and host defense purposes. In this report, we show for the first time that urothelial cells produce H(2)O(2) in response to a calcium signal. Using a gene-deficient mouse model we also demonstrate that H(2)O(2) is produced by the NADPH oxidase Duox1, which is expressed in the mouse urothelium. In contrast, we found no evidence for the expression of lactoperoxidase, an enzyme that has been shown to cooperate with Duox enzymes. We also found that specific activation of TRPV4 calcium channels elicits a calcium signal and stimulates H(2)O(2) production in urothelial cells. Furthermore, we detected altered pressure responses in the urinary bladders of Duox1 knockout animals. Our results raise the possibility that mechanosensing in epithelial cells involves calcium-dependent H(2)O(2) production similar to that observed in plants.


Subject(s)
Epithelial Cells/enzymology , Hydrogen Peroxide/metabolism , NADPH Oxidases/metabolism , Urinary Bladder/cytology , Animals , Calcium Signaling/drug effects , Dual Oxidases , Enzyme Activation , Epithelial Cells/metabolism , Escherichia coli/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , NADPH Oxidases/genetics , TRPV Cation Channels/metabolism , Thapsigargin/pharmacology , Urinary Bladder/microbiology , Urinary Bladder/physiology , Urothelium/cytology , Urothelium/physiology
18.
Am J Pathol ; 175(2): 725-35, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590037

ABSTRACT

Mammalian peroxidases are heme-containing enzymes that serve diverse biological roles, such as host defense and hormone biosynthesis. A mammalian homolog of Drosophila peroxidasin belongs to the peroxidase family; however, its function is currently unknown. In this study, we show that peroxidasin is present in the endoplasmic reticulum of human primary pulmonary and dermal fibroblasts, and the expression of this protein is increased during transforming growth factor-beta1-induced myofibroblast differentiation. Myofibroblasts secrete peroxidasin into the extracellular space where it becomes organized into a fibril-like network and colocalizes with fibronectin, thus helping to form the extracellular matrix. We also demonstrate that peroxidasin expression is increased in a murine model of kidney fibrosis and that peroxidasin localizes to the peritubular space in fibrotic kidneys. In addition, we show that this novel pathway of extracellular matrix formation is unlikely mediated by the peroxidase activity of the protein. Our data indicate that peroxidasin secretion represents a previously unknown pathway in extracellular matrix formation with a potentially important role in the physiological and pathological fibrogenic response.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Kidney/pathology , Myoblasts/metabolism , Peroxidase/metabolism , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Fibrosis , Humans , Kidney/metabolism , Mice , Peroxidasin
19.
Mol Biol Cell ; 18(3): 1083-97, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17215519

ABSTRACT

Epithelial-mesenchymal-myofibroblast transition (EMT), a key feature in organ fibrosis, is regulated by the state of intercellular contacts. Our recent studies have shown that an initial injury of cell-cell junctions is a prerequisite for transforming growth factor-beta1 (TGF-beta1)-induced transdifferentiation of kidney tubular cells into alpha-smooth muscle actin (SMA)-expressing myofibroblasts. Here we analyzed the underlying contact-dependent mechanisms. Ca(2+) removal-induced disruption of intercellular junctions provoked Rho/Rho kinase (ROK)-mediated myosin light chain (MLC) phosphorylation and Rho/ROK-dependent SMA promoter activation. Importantly, myosin-based contractility itself played a causal role, because the myosin ATPase inhibitor blebbistatin or a nonphosphorylatable, dominant negative MLC (DN-MLC) abolished the contact disruption-triggered SMA promoter activation, eliminated the synergy between contact injury and TGF-beta1, and suppressed SMA expression. To explore the responsible mechanisms, we investigated the localization of the main SMA-inducing transcription factors, serum response factor (SRF), and its coactivator myocardin-related transcription factor (MRTF). Contact injury enhanced nuclear accumulation of SRF and MRTF. These processes were inhibited by DN-Rho or DN-MLC. TGF-beta1 strongly facilitated nuclear accumulation of MRTF in cells with reduced contacts but not in intact epithelia. DN-myocardin abrogated the Ca(2+)-removal- +/- TGF-beta1-induced promoter activation. These studies define a new mechanism whereby cell contacts regulate epithelial-myofibroblast transition via Rho-ROK-phospho-MLC-dependent nuclear accumulation of MRTF.


Subject(s)
Cell Communication , Epithelial Cells/cytology , Fibroblasts/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Myosins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Actins/genetics , Animals , CHO Cells , Calcium/metabolism , Cell Nucleus/metabolism , Cricetinae , Cricetulus , Cytoplasm/metabolism , Humans , Kidney Tubules/cytology , Muscle, Smooth/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , Protein Transport , Serum Response Factor/metabolism , Trans-Activators/metabolism , rho-Associated Kinases
20.
Philos Trans R Soc Lond B Biol Sci ; 360(1464): 2301-8, 2005 Dec 29.
Article in English | MEDLINE | ID: mdl-16321800

ABSTRACT

Reactive oxygen species (ROS) have an important role in various physiological processes including host defence, mitogenesis, hormone biosynthesis, apoptosis and fertilization. Currently, the most characterized ROS-producing system operates in phagocytic cells, where ROS generated during phagocytosis act in host defence. Recently, several novel homologues of the phagocytic oxidase have been discovered and this protein family is now designated as the NOX/DUOX family of NADPH oxidases. NOX/DUOX enzymes function in a variety of tissues, including colon, kidney, thyroid gland, testis, salivary glands, airways and lymphoid organs. Importantly, members of the enzyme family are also found in non-mammalian species, including Caenorhabditis elegans and sea urchin. The physiological functions of novel NADPH oxidase enzymes are currently largely unknown. This review focuses on our current knowledge about dual oxidases.


Subject(s)
Autoantigens/metabolism , Flavoproteins/metabolism , Iodide Peroxidase/metabolism , Iron-Binding Proteins/metabolism , Models, Molecular , NADPH Oxidases/metabolism , Phagocytosis/physiology , Reactive Oxygen Species/metabolism , Amino Acid Sequence , Autoantigens/genetics , Dual Oxidases , Extracellular Matrix/enzymology , Flavoproteins/genetics , Humans , Iodide Peroxidase/genetics , Iron-Binding Proteins/genetics , Molecular Sequence Data , Mucous Membrane/enzymology , NADPH Oxidases/genetics , Salivary Glands/enzymology , Thyroid Gland/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...