1
|
Nakao S, Shirakado K, Tamura K, Koga R, Ikeda-Imafuku M, Ishima Y, Nakagata N, Takeo T. Oxidation of thiol groups in membrane proteins inhibits the fertilization ability and motility of sperm by suppressing calcium influx†. Biol Reprod 2025; 112:563-571. [PMID: 39689237 DOI: 10.1093/biolre/ioae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/19/2024] Open
Abstract
The redox state of thiol groups derived from cysteine residues in proteins regulates cellular functions. Changes in the redox state of thiol groups in the epididymis are involved in sperm maturation. Furthermore, the redox state of thiol groups in proteins changes during the process of sperm capacitation. However, the effect of the redox state of thiol groups in sperm membrane proteins on the fertilization ability of sperm has not been studied. Therefore, in this study, we oxidized thiol groups in sperm membrane proteins using 5,5-dithiobis-(2-nitrobenzoic acid) (DTNB), which is a thiol-selective oxidizing agent, and examined the effect of oxidation of these thiol groups on the fertilization ability of sperm. Oocytes and sperm were obtained from C57BL/6 J mice, and Jcl:ICR mice were used as recipients for embryo transfer. Oxidation of the thiol groups by DTNB decreased the in vitro fertilization rate, and removal of the zona pellucida recovered the fertilization rate. DTNB treatment decreased the amplitude of the lateral head, which is an indicator of hyperactivation, and suppressed an increase in the intracellular calcium ion concentration, which is essential for hyperactivation. These findings suggest that oxidation of thiol groups in sperm membrane proteins can decrease the fertility of sperm by suppressing calcium ion influx and hyperactivation.
Collapse
Affiliation(s)
- Satohiro Nakao
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Kazuki Shirakado
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Kana Tamura
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Reiri Koga
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Mayumi Ikeda-Imafuku
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Yu Ishima
- Laboratory of Biopharmaceutics, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Biotechnology and Innovation, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
2
|
Saei AA, Lundin A, Lyu H, Gharibi H, Luo H, Teppo J, Zhang X, Gaetani M, Végvári Á, Holmdahl R, Gygi SP, Zubarev RA. Multifaceted Proteome Analysis at Solubility, Redox, and Expression Dimensions for Target Identification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401502. [PMID: 39120068 PMCID: PMC11481203 DOI: 10.1002/advs.202401502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 07/24/2024] [Indexed: 08/10/2024]
Abstract
Multifaceted interrogation of the proteome deepens the system-wide understanding of biological systems; however, mapping the redox changes in the proteome has so far been significantly more challenging than expression and solubility/stability analyses. Here, the first high-throughput redox proteomics approach integrated with expression analysis (REX) is devised and combined with the Proteome Integral Solubility Alteration (PISA) assay. The whole PISA-REX experiment with up to four biological replicates can be multiplexed into a single tandem mass tag TMTpro set. For benchmarking this compact tool, HCT116 cells treated with auranofin are analyzed, showing great improvement compared with previous studies. PISA-REX is then applied to study proteome remodeling upon stimulation of human monocytes by interferon α (IFN-α). Applying this tool to study the proteome changes in plasmacytoid dendritic cells (pDCs) isolated from wild-type versus Ncf1-mutant mice treated with interferon α, shows that NCF1 deficiency enhances the STAT1 pathway and modulates the expression, solubility, and redox state of interferon-induced proteins. Providing comprehensive multifaceted information on the proteome, the compact PISA-REX has the potential to become an industry standard in proteomics and to open new windows into the biology of health and disease.
Collapse
Affiliation(s)
- Amir A. Saei
- Department of Cell BiologyHarvard Medical SchoolBostonMA02115USA
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
- BiozentrumUniversity of BaselBasel4056Switzerland
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholm17165Sweden
| | - Albin Lundin
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
| | - Hezheng Lyu
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
| | - Hassan Gharibi
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
| | - Huqiao Luo
- Division of Immunology, Medical Inflammation Research Group, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSE‐17 177Sweden
| | - Jaakko Teppo
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
- Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Xuepei Zhang
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
| | - Massimiliano Gaetani
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
- SciLifeLabStockholmSE‐17 177Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
| | - Rikard Holmdahl
- Division of Immunology, Medical Inflammation Research Group, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSE‐17 177Sweden
| | - Steven P. Gygi
- Department of Cell BiologyHarvard Medical SchoolBostonMA02115USA
| | - Roman A. Zubarev
- Division of Chemistry I, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSE‐17 177Sweden
- SciLifeLabStockholmSE‐17 177Sweden
| |
Collapse
|
3
|
Cadenas-Garrido P, Schonvandt-Alarcos A, Herrera-Quintana L, Vázquez-Lorente H, Santamaría-Quiles A, Ruiz de Francisco J, Moya-Escudero M, Martín-Oliva D, Martín-Guerrero SM, Rodríguez-Santana C, Aragón-Vela J, Plaza-Diaz J. Using Redox Proteomics to Gain New Insights into Neurodegenerative Disease and Protein Modification. Antioxidants (Basel) 2024; 13:127. [PMID: 38275652 PMCID: PMC10812581 DOI: 10.3390/antiox13010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Antioxidant defenses in biological systems ensure redox homeostasis, regulating baseline levels of reactive oxygen and nitrogen species (ROS and RNS). Oxidative stress (OS), characterized by a lack of antioxidant defenses or an elevation in ROS and RNS, may cause a modification of biomolecules, ROS being primarily absorbed by proteins. As a result of both genome and environment interactions, proteomics provides complete information about a cell's proteome, which changes continuously. Besides measuring protein expression levels, proteomics can also be used to identify protein modifications, localizations, the effects of added agents, and the interactions between proteins. Several oxidative processes are frequently used to modify proteins post-translationally, including carbonylation, oxidation of amino acid side chains, glycation, or lipid peroxidation, which produces highly reactive alkenals. Reactive alkenals, such as 4-hydroxy-2-nonenal, are added to cysteine (Cys), lysine (Lys), or histidine (His) residues by a Michael addition, and tyrosine (Tyr) residues are nitrated and Cys residues are nitrosylated by a Michael addition. Oxidative and nitrosative stress have been implicated in many neurodegenerative diseases as a result of oxidative damage to the brain, which may be especially vulnerable due to the large consumption of dioxygen. Therefore, the current methods applied for the detection, identification, and quantification in redox proteomics are of great interest. This review describes the main protein modifications classified as chemical reactions. Finally, we discuss the importance of redox proteomics to health and describe the analytical methods used in redox proteomics.
Collapse
Affiliation(s)
- Paula Cadenas-Garrido
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain; (P.C.-G.); (A.S.-A.); (A.S.-Q.); (J.R.d.F.); (M.M.-E.)
| | - Ailén Schonvandt-Alarcos
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain; (P.C.-G.); (A.S.-A.); (A.S.-Q.); (J.R.d.F.); (M.M.-E.)
| | - Lourdes Herrera-Quintana
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.); (C.R.-S.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Héctor Vázquez-Lorente
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.); (C.R.-S.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Alicia Santamaría-Quiles
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain; (P.C.-G.); (A.S.-A.); (A.S.-Q.); (J.R.d.F.); (M.M.-E.)
| | - Jon Ruiz de Francisco
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain; (P.C.-G.); (A.S.-A.); (A.S.-Q.); (J.R.d.F.); (M.M.-E.)
| | - Marina Moya-Escudero
- Research and Advances in Molecular and Cellular Immunology, Center of Biomedical Research, University of Granada, Avda, del Conocimiento s/n, 18016 Armilla, Spain; (P.C.-G.); (A.S.-A.); (A.S.-Q.); (J.R.d.F.); (M.M.-E.)
| | - David Martín-Oliva
- Department of Cell Biology, Faculty of Science, University of Granada, 18071 Granada, Spain;
| | - Sandra M. Martín-Guerrero
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
| | - César Rodríguez-Santana
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.); (C.R.-S.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Jerónimo Aragón-Vela
- Department of Health Sciences, Area of Physiology, Building B3, Campus s/n “Las Lagunillas”, University of Jaén, 23071 Jaén, Spain
| | - Julio Plaza-Diaz
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
| |
Collapse
|
4
|
Coelho F, Saidjalolov S, Moreau D, Thorn-Seshold O, Matile S. Inhibition of Cell Motility by Cell-Penetrating Dynamic Covalent Cascade Exchangers: Integrins Participate in Thiol-Mediated Uptake. JACS AU 2023; 3:1010-1016. [PMID: 37124287 PMCID: PMC10131202 DOI: 10.1021/jacsau.3c00113] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 05/03/2023]
Abstract
Integrins are cell surface proteins responsible for cell motility. Inspired by the rich disulfide exchange chemistry of integrins, we show here the inhibition of cell migration by cascade exchangers (CAXs), which also enable and inhibit cell penetration by thiol-mediated uptake. Fast-moving CAXs such as reversible Michael acceptor dimers, dithiabismepanes, and bioinspired epidithiodiketopiperazines are best, much better than Ellman's reagent. The implication that integrins participate in thiol-mediated uptake is confirmed by reduced uptake in integrin-knockdown cells. Although thiol-mediated uptake is increasingly emerging as a unifying pathway to bring matter into cells, its molecular basis is essentially unknown. These results identify the integrin superfamily as experimentally validated general cellular partners in the dynamic covalent exchange cascades that are likely to account for thiol-mediated uptake. The patterns identified testify to the complexity of the dynamic covalent networks involved. This work also provides chemistry tools to explore cell motility and expands the drug discovery potential of CAXs from antiviral toward antithrombotic and antitumor perspectives.
Collapse
Affiliation(s)
- Filipe Coelho
- Department
of Organic Chemistry, University of Geneva, 1211 Geneva, Switzerland
| | | | - Dimitri Moreau
- Department
of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Oliver Thorn-Seshold
- Department
of Pharmacy, Ludwig-Maximilians University
of Munich, 81377 Munich, Germany
| | - Stefan Matile
- Department
of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
5
|
Auf der Maur P, Trefny MP, Baumann Z, Vulin M, Correia AL, Diepenbruck M, Kramer N, Volkmann K, Preca BT, Ramos P, Leroy C, Eichlisberger T, Buczak K, Zilli F, Okamoto R, Rad R, Jensen MR, Fritsch C, Zippelius A, Stadler MB, Bentires-Alj M. N-acetylcysteine overcomes NF1 loss-driven resistance to PI3Kα inhibition in breast cancer. Cell Rep Med 2023; 4:101002. [PMID: 37044095 PMCID: PMC10140479 DOI: 10.1016/j.xcrm.2023.101002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023]
Abstract
A genome-wide PiggyBac transposon-mediated screen and a resistance screen in a PIK3CAH1047R-mutated murine tumor model reveal NF1 loss in mammary tumors resistant to the phosphatidylinositol 3-kinase α (PI3Kα)-selective inhibitor alpelisib. Depletion of NF1 in PIK3CAH1047R breast cancer cell lines and a patient-derived organoid model shows that NF1 loss reduces sensitivity to PI3Kα inhibition and correlates with enhanced glycolysis and lower levels of reactive oxygen species (ROS). Unexpectedly, the antioxidant N-acetylcysteine (NAC) sensitizes NF1 knockout cells to PI3Kα inhibition and reverts their glycolytic phenotype. Global phospho-proteomics indicates that combination with NAC enhances the inhibitory effect of alpelisib on mTOR signaling. In public datasets of human breast cancer, we find that NF1 is frequently mutated and that such mutations are enriched in metastases, an indication for which use of PI3Kα inhibitors has been approved. Our results raise the attractive possibility of combining PI3Kα inhibition with NAC supplementation, especially in patients with drug-resistant metastases associated with NF1 loss.
Collapse
Affiliation(s)
- Priska Auf der Maur
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Marcel P Trefny
- Cancer Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Zora Baumann
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Milica Vulin
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ana Luisa Correia
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Maren Diepenbruck
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nicolas Kramer
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Katrin Volkmann
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Bogdan-Tiberius Preca
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Pedro Ramos
- Oncology Research, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Cedric Leroy
- Oncology Research, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Katarzyna Buczak
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Federica Zilli
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ryoko Okamoto
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine, Technische Universität München, München, Germany; Center for Translational Cancer Research (TranslaTUM), TUM School of Medicine, Technische Universität München, München, Germany; Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Christine Fritsch
- Oncology Research, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; Swiss Institute of Bioinformatics, Basel, Switzerland; Faculty of Science, University of Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
6
|
Caliandro MF, Schmalbein F, Todesca LM, Mörgelin M, Rezaei M, Meißner J, Siepe I, Grosche J, Schwab A, Eble JA. A redox-dependent thiol-switch and a Ca 2+ binding site within the hinge region hierarchically depend on each other in α7β1 integrin regulation. Free Radic Biol Med 2022; 187:38-49. [PMID: 35605898 DOI: 10.1016/j.freeradbiomed.2022.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022]
Abstract
Integrin-mediated cell contacts with the extracellular matrix (ECM) are essential for cellular adhesion, force transmission, and migration. Several effectors, such as divalent cations and redox-active compounds, regulate ligand binding activities of integrins and influence their cellular functions. To study the role of the Ca2+ binding site within the hinge region of the integrin α7 subunit, we genetically abrogated it in the α7hiΔCa mutant. This mutant folded correctly, associated with the β1 subunit and was exposed on the cell surface, but showed reduced ligand binding and weaker cell adhesion to laminin-111. Thus, it resembles the α7hiΔSS mutant, in which the redox-regulated pair of cysteines, closeby to the Ca2+ binding site within the hinge, was abrogated. Comparing both mutants in adhesion strength and cell migration revealed that both Ca2+ complexation and redox-regulation within the hinge interdepend on each other. Moreover, protein-chemical analyses of soluble integrin ectodomains containing the same α7 hinge mutations suggest that integrin activation via the subunit α hinge is primed by the formation of the cysteine pair-based crosslinkage. Then, this allows Ca2+ complexation within the hinge, which is another essential step for integrin activation and ligand binding. Thus, the α hinge is an allosteric integrin regulation site, in which both effectors, Ca2+ and redox-active compounds, synergistically and hierarchically induce far-ranging conformational changes, such as the extension of the integrin ectodomain, resulting in integrin activation of ECM ligand binding and altered integrin-mediated cell functions.
Collapse
Affiliation(s)
- Michele F Caliandro
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Felix Schmalbein
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Luca Matteo Todesca
- University of Münster, Institute of Physiology II, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | | | - Maryam Rezaei
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Juliane Meißner
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Isabel Siepe
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Julius Grosche
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Albrecht Schwab
- University of Münster, Institute of Physiology II, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Johannes A Eble
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany.
| |
Collapse
|
7
|
Brücksken KA, Loreto Palacio P, Hanschmann EM. Thiol Modifications in the Extracellular Space-Key Proteins in Inflammation and Viral Infection. Front Immunol 2022; 13:932525. [PMID: 35833136 PMCID: PMC9271835 DOI: 10.3389/fimmu.2022.932525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Posttranslational modifications (PTMs) allow to control molecular and cellular functions in response to specific signals and changes in the microenvironment of cells. They regulate structure, localization, stability, and function of proteins in a spatial and temporal manner. Among them, specific thiol modifications of cysteine (Cys) residues facilitate rapid signal transduction. In fact, Cys is unique because it contains the highly reactive thiol group that can undergo different reversible and irreversible modifications. Upon inflammation and changes in the cellular microenvironment, many extracellular soluble and membrane proteins undergo thiol modifications, particularly dithiol-disulfide exchange, S-glutathionylation, and S-nitrosylation. Among others, these thiol switches are essential for inflammatory signaling, regulation of gene expression, cytokine release, immunoglobulin function and isoform variation, and antigen presentation. Interestingly, also the redox state of bacterial and viral proteins depends on host cell-mediated redox reactions that are critical for invasion and infection. Here, we highlight mechanistic thiol switches in inflammatory pathways and infections including cholera, diphtheria, hepatitis, human immunodeficiency virus (HIV), influenza, and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
| | | | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
8
|
Nosareva ОL, Stepovaya EA, Shakhristova EV, Karpov RM. The Role of Redox Status Changes in Dexamethasone-Induced Apoptosis in Jurkat Tumor Cells. Bull Exp Biol Med 2022; 172:464-466. [PMID: 35175483 DOI: 10.1007/s10517-022-05414-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Indexed: 10/19/2022]
Abstract
The apoptotic death and its regulation was studied in intact Jurkat tumor cells and under the influence of buthionine-sulfoximine (de novo glutathione synthesis inhibitor; 1 mM) and/or apoptosis inducer dexamethasone (10 μM). The role of glutathione system components in dexamethasone-induced apoptosis in Jurkat tumor cells (both receptor-mediated and mitochondrial pathways) was analyzed. Under conditions of dexamethasone-induced apoptosis, glutathione system blockage mostly affects presentation of TNF RI- and Fas-receptors in Jurkat tumor cells, as well as change in content of transcription factors Apaf-1 and NF-κB, thereby promoting cell death. The decrease in the content of oxidized glutathione produced a potentiating effect on dexamethasone-induced apoptotic death of Jurkat tumor cells.
Collapse
Affiliation(s)
- О L Nosareva
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - E A Stepovaya
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - E V Shakhristova
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - R M Karpov
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| |
Collapse
|
9
|
Ramos AP, Sebinelli HG, Ciancaglini P, Rosato N, Mebarek S, Buchet R, Millán JL, Bottini M. The functional role of soluble proteins acquired by extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e34. [PMID: 38938684 PMCID: PMC11080634 DOI: 10.1002/jex2.34] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanosized particles released by all cell types during physiological as well as pathophysiological processes to carry out diverse biological functions, including acting as sources of cellular dumping, signalosomes and mineralisation nanoreactors. The ability of EVs to perform specific biological functions is due to their biochemical machinery. Among the components of the EVs' biochemical machinery, surface proteins are of critical functional significance as they mediate the interactions of EVs with components of the extracellular milieu, the extracellular matrix and neighbouring cells. Surface proteins are thought to be native, that is, pre-assembled on the EVs' surface by the parent cells before the vesicles are released. However, numerous pieces of evidence have suggested that soluble proteins are acquired by the EVs' surface from the extracellular milieu and further modulate the biological functions of EVs during innate and adaptive immune responses, autoimmune disorders, complement activation, coagulation, viral infection and biomineralisation. Herein, we will describe the methods currently used to identify the EVs' surface proteins and discuss recent knowledge on the functional relevance of the soluble proteins acquired by EVs.
Collapse
Affiliation(s)
- Ana Paula Ramos
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Heitor Gobbi Sebinelli
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Pietro Ciancaglini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Nicola Rosato
- Dipartimento di Medicina SperimentaleUniversita’ di Roma “Tor Vergata”RomeItaly
| | - Saida Mebarek
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | - Rene Buchet
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | | | - Massimo Bottini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
- Sanford Burnham PrebysLa JollaCaliforniaUSA
| |
Collapse
|
10
|
Pedre B, Barayeu U, Ezeriņa D, Dick TP. The mechanism of action of N-acetylcysteine (NAC): The emerging role of H 2S and sulfane sulfur species. Pharmacol Ther 2021; 228:107916. [PMID: 34171332 DOI: 10.1016/j.pharmthera.2021.107916] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022]
Abstract
Initially adopted as a mucolytic about 60 years ago, the cysteine prodrug N-acetylcysteine (NAC) is the standard of care to treat paracetamol intoxication, and is included on the World Health Organization's list of essential medicines. Additionally, NAC increasingly became the epitome of an "antioxidant". Arguably, it is the most widely used "antioxidant" in experimental cell and animal biology, as well as clinical studies. Most investigators use and test NAC with the idea that it prevents or attenuates oxidative stress. Conventionally, it is assumed that NAC acts as (i) a reductant of disulfide bonds, (ii) a scavenger of reactive oxygen species and/or (iii) a precursor for glutathione biosynthesis. While these mechanisms may apply under specific circumstances, they cannot be generalized to explain the effects of NAC in a majority of settings and situations. In most cases the mechanism of action has remained unclear and untested. In this review, we discuss the validity of conventional assumptions and the scope of a newly discovered mechanism of action, namely the conversion of NAC into hydrogen sulfide and sulfane sulfur species. The antioxidative and cytoprotective activities of per- and polysulfides may explain many of the effects that have previously been ascribed to NAC or NAC-derived glutathione.
Collapse
Affiliation(s)
- Brandán Pedre
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Daria Ezeriņa
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
11
|
Caserta S, Ghezzi P. Release of redox enzymes and micro-RNAs in extracellular vesicles, during infection and inflammation. Free Radic Biol Med 2021; 169:248-257. [PMID: 33862160 DOI: 10.1016/j.freeradbiomed.2021.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/27/2021] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
Many studies reported that redox enzymes, particularly thioredoxin and peroxiredoxin, can be released by cells and act as soluble mediators in immunity. Recently, it became clear that peroxiredoxins can be secreted via the exosome-release route, yet it remains unclear how this exactly happens and why. This review will first introduce briefly the possible redox states of protein cysteines and the role of redox enzymes in their regulation. We will then discuss the studies on the extracellular forms of some of these enzymes, their association with exosomes/extracellular vesicles and with exosome micro-RNAs (miRNAs)/mRNAs involved in oxidative processes, relevant in infection and inflammation.
Collapse
Affiliation(s)
- Stefano Caserta
- Department of Biomedical Sciences, Hardy Building, The University of Hull, Hull, HU6 7RX, United Kingdom
| | - Pietro Ghezzi
- Department of Clinical Experimental Medicine, Brighton & Sussex Medical School, Brighton, BN19RY, United Kingdom.
| |
Collapse
|
12
|
Abstract
The hypothesis that reactive oxygen species (ROS) can be not just associated with but causally implicated in disease was first made in 1956, but so far, the oxidative stress theory of disease has not led to major therapeutic breakthrough, and the use of antioxidant is now confined to the field of complementary medicine. This chapter reviews the lack of high-level clinical evidence for the effectiveness of antioxidants in preventing disease and the epistemological problems of the oxidative stress theory of disease. We conclude on possible ways forward to test this hypothesis with approaches that take into account personalized medicine. The previous oxidative stress model has helped neither to diagnose nor to treat possibly ROS-related or ROS-dependent diseases. The redox balance concept that low ROS levels are beneficial or tolerable and high levels are disease triggers and best reduced is apparently wrong. Physiological ROS signalling may become dysfunctional or a disease trigger by at least five mechanisms: a physiological source may appear at an unphysiological site, a physiological source may be underactivated (less common) or overactivated (more common), a new source may appear, a physiological source may be overactivated or underactivated, and a toxifying enzyme may convert an ROS signal molecule into a more reactive molecule. The latter three mechanisms may reach a physiological or nonphysiological target. All of these dysregulations may be the direct and essential cause of a disease (rarely the case) or just a secondary epiphenomenon, which will disappear once the non-ROS-related cause of the disease is cured (much more common). Importantly, these mechanisms are the same for almost every signalling system. Causal target validation (sources, toxifiers and targets) is essential in order to identify effective drugs and therapies for ROSopathies.
Collapse
Affiliation(s)
| | - Arshag D Mooradian
- Department of Medicine, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
13
|
Lorenzen I, Eble JA, Hanschmann EM. Thiol switches in membrane proteins - Extracellular redox regulation in cell biology. Biol Chem 2020; 402:253-269. [PMID: 33108336 DOI: 10.1515/hsz-2020-0266] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
Redox-mediated signal transduction depends on the enzymatic production of second messengers such as hydrogen peroxide, nitric oxide and hydrogen sulfite, as well as specific, reversible redox modifications of cysteine-residues in proteins. So-called thiol switches induce for instance conformational changes in specific proteins that regulate cellular pathways e.g., cell metabolism, proliferation, migration, gene expression and inflammation. Reduction, oxidation and disulfide isomerization are controlled by oxidoreductases of the thioredoxin family, including thioredoxins, glutaredoxins, peroxiredoxins and protein dsisulfide isomerases. These proteins are located in different cellular compartments, interact with substrates and catalyze specific reactions. Interestingly, some of these proteins are released by cells. Their extracellular functions and generally extracellular redox control have been widely underestimated. Here, we give an insight into extracellular redox signaling, extracellular thiol switches and their regulation by secreted oxidoreductases and thiol-isomerases, a topic whose importance has been scarcely studied so far, likely due to methodological limitations. We focus on the secreted redox proteins and characterized thiol switches in the ectodomains of membrane proteins, such as integrins and the metalloprotease ADAM17, which are among the best-characterized proteins and discuss their underlying mechanisms and biological implications.
Collapse
Affiliation(s)
- Inken Lorenzen
- Centre of Biochemistry and Molecular Biology, Structural Biology, Christian-Albrecht University of Kiel, Am Botanischen Garten 1-9, D-24118Kiel, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, D-48149Münster, Germany
| | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Life Science Center, Merowingerplatz 1a, D-40225Düsseldorf, Germany
| |
Collapse
|
14
|
Bergerhausen L, Grosche J, Meißner J, Hecker C, Caliandro MF, Westerhausen C, Kamenac A, Rezaei M, Mörgelin M, Poschmann G, Vestweber D, Hanschmann EM, Eble JA. Extracellular Redox Regulation of α7β Integrin-Mediated Cell Migration Is Signaled via a Dominant Thiol-Switch. Antioxidants (Basel) 2020; 9:antiox9030227. [PMID: 32164274 PMCID: PMC7139957 DOI: 10.3390/antiox9030227] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 12/21/2022] Open
Abstract
While adhering to extracellular matrix (ECM) proteins, such as laminin-111, cells temporarily produce hydrogen peroxide at adhesion sites. To study the redox regulation of α7β1 integrin-mediated cell adhesion to laminin-111, a conserved cysteine pair within the α-subunit hinge region was replaced for alanines. The molecular and cellular effects were analyzed by electron and atomic force microscopy, impedance-based migration assays, flow cytometry and live cell imaging. This cysteine pair constitutes a thiol-switch, which redox-dependently governs the equilibrium between an extended and a bent integrin conformation with high and low ligand binding activity, respectively. Hydrogen peroxide oxidizes the cysteines to a disulfide bond, increases ligand binding and promotes cell migration toward laminin-111. Inversely, extracellular thioredoxin-1 reduces the disulfide, thereby decreasing laminin binding. Mutation of this cysteine pair into the non-oxidizable hinge-mutant shows molecular and cellular effects similar to the reduced wild-type integrin, but lacks redox regulation. This proves the existence of a dominant thiol-switch within the α subunit hinge of α7β1 integrin, which is sufficient to implement activity regulation by extracellular redox agents in a redox-regulatory circuit. Our data reveal a novel and physiologically relevant thiol-based regulatory mechanism of integrin-mediated cell-ECM interactions, which employs short-lived hydrogen peroxide and extracellular thioredoxin-1 as signaling mediators.
Collapse
Affiliation(s)
- Lukas Bergerhausen
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany; (L.B.); (J.G.); (J.M.); (M.F.C.); (M.R.)
| | - Julius Grosche
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany; (L.B.); (J.G.); (J.M.); (M.F.C.); (M.R.)
| | - Juliane Meißner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany; (L.B.); (J.G.); (J.M.); (M.F.C.); (M.R.)
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.H.); (E.-M.H.)
| | - Michele F. Caliandro
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany; (L.B.); (J.G.); (J.M.); (M.F.C.); (M.R.)
| | - Christoph Westerhausen
- Biophysics Group, Department of Experimental Physics, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany (A.K.)
- Institute of Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
| | - Andrej Kamenac
- Biophysics Group, Department of Experimental Physics, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany (A.K.)
| | - Maryam Rezaei
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany; (L.B.); (J.G.); (J.M.); (M.F.C.); (M.R.)
| | | | - Gereon Poschmann
- Institute of Molecular Medicine I, Functional Redox Proteomics, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck-Institute of Molecular Biomedicine, 48149 Münster, Germany;
| | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.H.); (E.-M.H.)
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany; (L.B.); (J.G.); (J.M.); (M.F.C.); (M.R.)
- Correspondence: ; Tel.: +49-251-835-5591
| |
Collapse
|
15
|
Laurent Q, Berthet M, Cheng Y, Sakai N, Barluenga S, Winssinger N, Matile S. Probing for Thiol-Mediated Uptake into Bacteria. Chembiochem 2020; 21:69-73. [PMID: 31603284 DOI: 10.1002/cbic.201900378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Indexed: 01/02/2023]
Abstract
Cellular uptake mediated by cyclic oligochalcogenides (COCs) is emerging as a conceptually innovative method to penetrate mammalian cells. Their mode of action is based on dynamic covalent oligochalcogenide exchange with cellular thiols. To test thiol-mediated uptake in bacteria, five antibiotics have been equipped with up to three different COCs: One diselenolane and two dithiolanes. We found that the COCs do not activate antibiotics in Gram-negative bacteria. In Gram-positive bacteria, the COCs inactivate antibiotics that act in the cytoplasm and reduce the activity of antibiotics that act on the cell surface. These results indicate that thiol-mediated uptake operates in neither of the membranes of bacteria. COCs are likely to exchange with thiols on the inner, maybe also on the outer membrane, but do not move on. Concerning mammalian cells, the absence of a COC-mediated uptake into bacteria observed in this study disfavors trivial mechanisms, such as passive diffusion, and supports the existence of more sophisticated, so far poorly understood uptake pathways.
Collapse
Affiliation(s)
- Quentin Laurent
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Mathéo Berthet
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Yangyang Cheng
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Naomi Sakai
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Sofia Barluenga
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Nicolas Winssinger
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | - Stefan Matile
- National Centre of Competence in Research (NCCR), Chemical Biology, School of Chemistry and Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| |
Collapse
|
16
|
N-Acetyl-L-cysteine Protects Human Retinal Pigment Epithelial Cells from Oxidative Damage: Implications for Age-Related Macular Degeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5174957. [PMID: 31485293 PMCID: PMC6710748 DOI: 10.1155/2019/5174957] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/27/2019] [Indexed: 01/12/2023]
Abstract
Age-related macular degeneration (AMD) involves the loss of retinal pigment epithelium (RPE) and photoreceptors and is one of the leading causes of blindness in the elderly. Oxidative damage to proteins, lipids, and DNA has been associated with RPE dysfunction and AMD. In this study, we evaluated oxidative stress in AMD and the efficacy of antioxidant, N-acetyl-L-cysteine (NAC), in protecting RPE from oxidative damage. To test this idea, primary cultures of RPE from human donors with AMD (n = 32) or without AMD (No AMD, n = 21) were examined for expression of NADPH oxidase (NOX) genes, a source of reactive oxygen species (ROS). Additionally, the cells were pretreated with NAC for 2 hours and then treated with either hydrogen peroxide (H2O2) or tert-butyl hydroperoxide (t-BHP) to induce cellular oxidation. Twenty-four hours after treatment, ROS production, cell survival, the content of glutathione (GSH) and adenosine triphosphate (ATP), and cellular bioenergetics were measured. We found increased expression of p22phox, a NOX regulator, in AMD cells compared to No AMD cells (p = 0.02). In both AMD and No AMD cells, NAC pretreatment reduced t-BHP-induced ROS production and protected from H2O2-induced cell death and ATP depletion. In the absence of oxidation, NAC treatment improved mitochondrial function in both groups (p < 0.01). Conversely, the protective response exhibited by NAC was disease-dependent for some parameters. In the absence of oxidation, NAC significantly reduced ROS production (p < 0.001) and increased GSH content (p = 0.02) only in RPE from AMD donors. Additionally, NAC-mediated protection from H2O2-induced GSH depletion (p = 0.04) and mitochondrial dysfunction (p < 0.05) was more pronounced in AMD cells compared with No AMD cells. These results demonstrate the therapeutic benefit of NAC by mitigating oxidative damage in RPE. Additionally, the favorable outcomes observed for AMD RPE support NAC's relevance and the potential therapeutic value in treating AMD.
Collapse
|
17
|
Rosenberg N, Mor-Cohen R, Sheptovitsky VH, Romanenco O, Hess O, Lahav J. Integrin-mediated cell adhesion requires extracellular disulfide exchange regulated by protein disulfide isomerase. Exp Cell Res 2019; 381:77-85. [PMID: 31042499 DOI: 10.1016/j.yexcr.2019.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/26/2019] [Accepted: 04/14/2019] [Indexed: 01/28/2023]
Abstract
Cell adhesion to extracellular matrix, mediated by integrin receptors, is crucial for cell survival. Receptor-ligand interaction involves conformational changes in the integrin by a mechanism not fully elucidated. In addition to several direct evidence that there is disulfide re-arrangement of integrins, we previously demonstrated a role for extracellular thiols and protein disulfide isomerase (PDI) in integrin-mediated functions using platelets as model system. Exploring the possible generality of this mechanism, we now show, using three different nucleated cells which depend on adhesion for survival, that non-penetrating blockers of free thiols inhibit α2β1 and α5β1 integrin-mediated adhesion and that disulfide exchange takes place in that process. Inhibiting extracellular PDI mimics thiol blocking. Transfection with WT or enzymatically inactive PDI increased their membrane expression and enhanced cell adhesion, suggesting that PDI level is a limiting factor and that the chaperone activity of the enzyme contributes to adhesion. Exogenously added PDI also enhanced adhesion, further supporting the limiting factor of the enzyme. These data indicate that: a) Dependence on ecto-sulfhydryls for integrin-mediated adhesion is not exclusive to the platelet; b) PDI is involved in integrin-mediated adhesion, catalyzing disulfide bond exchange; c) PDI enhances cell adhesion by both its oxidoreductase activity and as a chaperone.
Collapse
Affiliation(s)
- Nurit Rosenberg
- The Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel-Hashomer and Dept. of Hematology, Sackler School of Medicine, Tel Aviv University, Israel.
| | - Ronit Mor-Cohen
- The Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center, Tel-Hashomer and Dept. of Hematology, Sackler School of Medicine, Tel Aviv University, Israel
| | - Vera Hazan Sheptovitsky
- Hemostasis Laboratory, Rabin Medical Center-Beilinson Hospital, Petah-Tiqva, and Dept. of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel
| | - Olga Romanenco
- Hemostasis Laboratory, Rabin Medical Center-Beilinson Hospital, Petah-Tiqva, and Dept. of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel
| | - Oded Hess
- Hemostasis Laboratory, Rabin Medical Center-Beilinson Hospital, Petah-Tiqva, and Dept. of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel
| | - Judith Lahav
- Hemostasis Laboratory, Rabin Medical Center-Beilinson Hospital, Petah-Tiqva, and Dept. of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel
| |
Collapse
|
18
|
Dendrimer-grafted bioreducible polycation/DNA multilayered films with low cytotoxicity and high transfection ability. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 98:737-745. [PMID: 30813078 DOI: 10.1016/j.msec.2018.12.111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 11/27/2018] [Accepted: 12/27/2018] [Indexed: 11/21/2022]
Abstract
Controlled release of incorporated foreign DNA from multilayered films plays an important role in surface-mediated gene delivery. Herein, multilayered polyelectrolyte complex thin films, composed of dendrimer-grafted bio-reducible cationic poly(disulfide amine) and plasmid DNA, were fabricated via layer-by-layer (LBL) assembly for in vitro localized gene delivery. The UV absorbance and thickness of the LBL films were found to have linear correlation with the numbers of poly(disulfide amine)/DNA bilayers. Although LBL films were stable in PBS buffer, their degradation could be triggered by reducing agents (i.e. glutathione, GSH). The degradation rate of the films is directly proportional to the GSH concentration, which in turn affected the corresponding gene expression. All poly(disulfide amine)/DNA films exhibited lower cytotoxicity and higher transfection activity in comparison with PEI/DNA multilayered films. Moreover, LBL films showed the highest transfection efficiency in the presence of 2.5 mM GSH when cultured with 293T cells, with ~36% GFP-positive 293T cells after 5-days of co-culture. These DNA-containing reducible films could potentially be useful in gene therapy and tissue engineering by controlling the release of incorporated DNA.
Collapse
|
19
|
You Y, Chen J, Zhu F, Xu Q, Han L, Gao X, Zhang X, Luo HR, Miao J, Sun X, Ren H, Du Y, Guo L, Wang X, Wang Y, Chen S, Huang N, Li J. Glutaredoxin 1 up-regulates deglutathionylation of α4 integrin and thereby restricts neutrophil mobilization from bone marrow. J Biol Chem 2018; 294:2616-2627. [PMID: 30598505 DOI: 10.1074/jbc.ra118.006096] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/27/2018] [Indexed: 12/31/2022] Open
Abstract
α4 integrin plays a crucial role in retention and release of neutrophils from bone marrow. Although α4 integrin is known to be a potential target of reactive oxygen species (ROS)-induced cysteine glutathionylation, the physiological significance and underlying regulatory mechanism of this event remain elusive. Here, using in vitro and in vivo biochemical and cell biology approaches, we show that physiological ROS-induced glutathionylation of α4 integrin in neutrophils increases the binding of neutrophil-associated α4 integrin to vascular cell adhesion molecule 1 (VCAM-1) on human endothelial cells. This enhanced binding was reversed by extracellular glutaredoxin 1 (Grx1), a thiol disulfide oxidoreductase promoting protein deglutathionylation. Furthermore, in a murine inflammation model, Grx1 disruption dramatically elevated α4 glutathionylation and subsequently enhanced neutrophil egress from the bone marrow. Corroborating this observation, intravenous injection of recombinant Grx1 into mice inhibited α4 glutathionylation and thereby suppressed inflammation-induced neutrophil mobilization from the bone marrow. Taken together, our results establish ROS-elicited glutathionylation and its modulation by Grx1 as pivotal regulatory mechanisms controlling α4 integrin affinity and neutrophil mobilization from the bone marrow under physiological conditions.
Collapse
Affiliation(s)
| | - Junli Chen
- From the Departments of Pathophysiology and
| | - Feimei Zhu
- From the Departments of Pathophysiology and
| | - Qian Xu
- From the Departments of Pathophysiology and
| | - Lu Han
- the State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xiang Gao
- the State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Zhang
- the State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Hongbo R Luo
- the Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115.,the Department of Lab Medicine, Children's Hospital Boston, Boston, Massachusetts 02115, and.,the Dana-Farber/Harvard Cancer Center, Boston, Massachusetts 02115
| | | | - Xiaodong Sun
- Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Hongyu Ren
- From the Departments of Pathophysiology and
| | - Yu Du
- From the Departments of Pathophysiology and
| | - Lijuan Guo
- From the Departments of Pathophysiology and
| | | | - Yi Wang
- From the Departments of Pathophysiology and
| | | | - Ning Huang
- From the Departments of Pathophysiology and
| | - Jingyu Li
- From the Departments of Pathophysiology and
| |
Collapse
|
20
|
Cha J, Kim H, Hwang NS, Kim P. Mild Reduction of the Cancer Cell Surface as an Anti-invasion Treatment. ACS APPLIED MATERIALS & INTERFACES 2018; 10:35676-35680. [PMID: 30288974 DOI: 10.1021/acsami.8b12566] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cancer cell invasion is the main reason for high mortality in patients with malignant cancers. There has been little improvement in cancer prognosis because of a high rate of infiltration. Therefore, successful treatment requires inhibition of cancer cell invasion. Here, we suggest a new approach to inhibit cancer cell invasion through mild reduction of cell surface proteins to expose free thiols. Through mild reduction, the cancer cell surfaces present free active thiols at the membranes, enhancing cell adhesion to extracellular matrix and decreasing motility. Collectively, we suggest cell surface modification as a new therapeutic approach to treat invading malignant cancers.
Collapse
Affiliation(s)
- Junghwa Cha
- Department of Bio and Brain Engineering , KAIST , Daejeon 34141 , Korea
| | - Hyunbum Kim
- Department of Chemical and Biological Engineering, Institute for Chemical Processes , Seoul National University , Seoul 08826 , Korea
| | - Nathaniel S Hwang
- Department of Chemical and Biological Engineering, Institute for Chemical Processes , Seoul National University , Seoul 08826 , Korea
- BioMAX Institute , Seoul National University , Seoul 08826 , Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering , KAIST , Daejeon 34141 , Korea
| |
Collapse
|
21
|
Cecchinato V, D'Agostino G, Raeli L, Nerviani A, Schiraldi M, Danelon G, Manzo A, Thelen M, Ciurea A, Bianchi ME, Rubartelli A, Pitzalis C, Uguccioni M. Redox-Mediated Mechanisms Fuel Monocyte Responses to CXCL12/HMGB1 in Active Rheumatoid Arthritis. Front Immunol 2018; 9:2118. [PMID: 30283452 PMCID: PMC6157448 DOI: 10.3389/fimmu.2018.02118] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/28/2018] [Indexed: 12/26/2022] Open
Abstract
Chemokine synergy-inducing molecules are emerging as regulating factors in cell migration. The alarmin HMGB1, in its reduced form, can complex with CXCL12 enhancing its activity on monocytes via the chemokine receptor CXCR4, while the form containing a disulfide bond, by binding to TLR2 or TLR4, initiates a cascade of events leading to production of cytokines and chemokines. So far, the possibility that the CXCL12/HMGB1 heterocomplex could be maintained in chronic inflammation was debated, due to the release of reactive oxygen species. Therefore, we have assessed if the heterocomplex could remain active in Rheumatoid Arthritis (RA) and its relevance in the disease assessment. Monocytes from RA patients with active disease require a low concentration of HMGB1 to enhance CXCL12-induced migration, in comparison to monocytes from patients in clinical remission or healthy donors. The activity of the heterocomplex depends on disease activity, on the COX2 and JAK/STAT pathways, and is determined by the redox potential of the microenvironment. In RA, the presence of an active thioredoxin system correlates with the enhanced cell migration, and with the presence of the heterocomplex in the synovial fluid. The present study highlights how, in an unbalanced microenvironment, the activity of the thioredoxin system plays a crucial role in sustaining inflammation. Prostaglandin E2 stimulation of monocytes from healthy donors is sufficient to recapitulate the response observed in patients with active RA. The activation of mechanisms counteracting the oxidative stress in the extracellular compartment preserves HMGB1 in its reduced form, and contributes to fuel the influx of inflammatory cells. Targeting the heterocomplex formation and its activity could thus be an additional tool for dampening the inflammation sustained by cell recruitment, for those patients with chronic inflammatory conditions who poorly respond to current therapies.
Collapse
Affiliation(s)
- Valentina Cecchinato
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Gianluca D'Agostino
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Lorenzo Raeli
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alessandra Nerviani
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Milena Schiraldi
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Gabriela Danelon
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Antonio Manzo
- Division of Rheumatology, Rheumatology and Translational Immunology Research Laboratories (LaRIT), IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Marcus Thelen
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Adrian Ciurea
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Marco E Bianchi
- San Raffaele University and Scientific Institute, Milan, Italy
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Costantino Pitzalis
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Mariagrazia Uguccioni
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
22
|
Buzás EI, Tóth EÁ, Sódar BW, Szabó-Taylor KÉ. Molecular interactions at the surface of extracellular vesicles. Semin Immunopathol 2018; 40:453-464. [PMID: 29663027 PMCID: PMC6208672 DOI: 10.1007/s00281-018-0682-0] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles such as exosomes, microvesicles, apoptotic bodies, and large oncosomes have been shown to participate in a wide variety of biological processes and are currently under intense investigation in many different fields of biomedicine. One of the key features of extracellular vesicles is that they have relatively large surface compared to their volume. Some extracellular vesicle surface molecules are shared with those of the plasma membrane of the releasing cell, while other molecules are characteristic for extracellular vesicular surfaces. Besides proteins, lipids, glycans, and nucleic acids are also players of extracellular vesicle surface interactions. Being secreted and present in high number in biological samples, collectively extracellular vesicles represent a uniquely large interactive surface area which can establish contacts both with cells and with molecules in the extracellular microenvironment. Here, we provide a brief overview of known components of the extracellular vesicle surface interactome and highlight some already established roles of the extracellular vesicle surface interactions in different biological processes in health and disease.
Collapse
Affiliation(s)
- Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
- MTA-SE Immune-Proteogenomics Research Group, Budapest, Hungary.
| | - Eszter Á Tóth
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Barbara W Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Katalin É Szabó-Taylor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
Echeverri-Ruiz N, Haynes T, Landers J, Woods J, Gemma MJ, Hughes M, Del Rio-Tsonis K. A biochemical basis for induction of retina regeneration by antioxidants. Dev Biol 2017; 433:394-403. [PMID: 29291983 PMCID: PMC5753421 DOI: 10.1016/j.ydbio.2017.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 12/19/2022]
Abstract
The use of antioxidants in tissue regeneration has been studied, but their mechanism of action is not well understood. Here, we analyze the role of the antioxidant N-acetylcysteine (NAC) in retina regeneration. Embryonic chicks are able to regenerate their retina after its complete removal from retinal stem/progenitor cells present in the ciliary margin (CM) of the eye only if a source of exogenous factors, such as FGF2, is present. This study shows that NAC modifies the redox status of the CM, initiates self-renewal of the stem/progenitor cells, and induces regeneration in the absence of FGF2. NAC works as an antioxidant by scavenging free radicals either independently or through the synthesis of glutathione (GSH), and/or by reducing oxidized proteins through a thiol disulfide exchange activity. We dissected the mechanism used by NAC to induce regeneration through the use of inhibitors of GSH synthesis and the use of other antioxidants with different biochemical structures and modes of action, and found that NAC induces regeneration through its thiol disulfide exchange activity. Thus, our results provide, for the first time, a biochemical basis for induction of retina regeneration. Furthermore, NAC induction was independent of FGF receptor signaling, but dependent on the MAPK (pErk1/2) pathway.
Collapse
Affiliation(s)
- Nancy Echeverri-Ruiz
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Tracy Haynes
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Joseph Landers
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Justin Woods
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Michael J Gemma
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Michael Hughes
- Department of Statistics and Statistical Consulting Center, Miami University, Oxford, OH 45056, USA
| | - Katia Del Rio-Tsonis
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| |
Collapse
|
24
|
Nosareva OL, Stepovaya EA, Ryazantseva NV, Shakhristova EV, Egorova MY, Novitsky VV. The Role of the Glutathione System in Oxidative Modification of Proteins and Dysregulation of Apoptosis in Jurkat Tumor Cells. Bull Exp Biol Med 2017; 164:199-202. [PMID: 29177874 DOI: 10.1007/s10517-017-3957-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Indexed: 12/17/2022]
Abstract
We compared changes in the redox status and intensity of oxidative modification of proteins in intact Jurkat tumor cells and cells cultured with buthionine sulfoximine, an inhibitor of the key enzyme of glutathione synthesis γ-glutamylcysteine synthetase. The glutathione system components play a role in modulation of the content of protein-bound glutathione, protein carbonyl derivatives, bityrosine, and oxidized tryptophan, and in dysregulation of apoptosis in Jurkat tumor cells. Inhibition of de novo synthesis of glutathione in Jurkat tumor cells was followed by accumulation of hydroxyl radical, a reduction in the level of protein-bound glutathione and oxidized tryptophan, and a rise in the concentration of protein carbonyl derivatives. These changes were accompanied by activation of programmed cell death.
Collapse
Affiliation(s)
- O L Nosareva
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - E A Stepovaya
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - N V Ryazantseva
- Siberian Federal University, Krasnoyarsk, Russia.,F. V. Voino-Yasentsky Krasnoyarsk State Medical University, Ministry of Health of the Russian Federation, Krasnoyarsk, Russia
| | - E V Shakhristova
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - M Yu Egorova
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - V V Novitsky
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| |
Collapse
|
25
|
Muralidharan P, Cserne Szappanos H, Ingley E, Hool LC. The cardiac L-type calcium channel alpha subunit is a target for direct redox modification during oxidative stress-the role of cysteine residues in the alpha interacting domain. Clin Exp Pharmacol Physiol 2017; 44 Suppl 1:46-54. [PMID: 28306174 DOI: 10.1111/1440-1681.12750] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/16/2017] [Accepted: 03/07/2017] [Indexed: 01/21/2023]
Abstract
Cardiovascular disease is the leading cause of death in the Western world. The incidence of cardiovascular disease is predicted to further rise with the increase in obesity and diabetes and with the aging population. Even though the survival rate from ischaemic heart disease has improved over the past 30 years, many patients progress to a chronic pathological condition, known as cardiac hypertrophy that is associated with an increase in morbidity and mortality. Reactive oxygen species (ROS) and calcium play an essential role in mediating cardiac hypertrophy. The L-type calcium channel is the main route for calcium influx into cardiac myocytes. There is now good evidence for a direct role for the L-type calcium channel in the development of cardiac hypertrophy. Cysteines on the channel are targets for redox modification and glutathionylation of the channel can modulate the function of the channel protein leading to the onset of pathology. The cysteine responsible for modification of L-type calcium channel function has now been identified. Detailed understanding of the role of cysteines as possible targets during oxidative stress may assist in designing therapy to prevent the development of hypertrophy and heart failure.
Collapse
Affiliation(s)
- Padmapriya Muralidharan
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, WA, Australia
| | - Henrietta Cserne Szappanos
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, WA, Australia
| | - Evan Ingley
- Harry Perkins Institute of Medical Research and Centre for Medical Research, University of Western Australia, Perth, WA, Australia.,School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA, Australia
| | - Livia C Hool
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Crawley, WA, Australia.,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| |
Collapse
|
26
|
Szabó-Taylor KÉ, Tóth EÁ, Balogh AM, Sódar BW, Kádár L, Pálóczi K, Fekete N, Németh A, Osteikoetxea X, Vukman KV, Holub M, Pállinger É, Nagy G, Winyard PG, Buzás EI. Monocyte activation drives preservation of membrane thiols by promoting release of oxidised membrane moieties via extracellular vesicles. Free Radic Biol Med 2017; 108:56-65. [PMID: 28323130 DOI: 10.1016/j.freeradbiomed.2017.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/06/2017] [Accepted: 03/14/2017] [Indexed: 12/15/2022]
Abstract
The redox state of cellular exofacial molecules is reflected by the amount of available thiols. Furthermore, surface thiols can be considered as indicators of immune cell activation. One group of thiol containing proteins, peroxiredoxins, in particular, have been associated with inflammation. In this study, we assessed surface thiols of the U937 and Thp1 monocyte cell lines and primary monocytes in vitro upon inflammatory stimulation by irreversibly labelling the cells with a fluorescent derivative of maleimide. We also investigated exofacial thiols on circulating blood mononuclear cells in patients with rheumatoid arthritis and healthy controls. When analysing extracellular vesicles, we combined thiol labelling with the use of antibodies to specific CD markers to exclude extracellular vesicle mimicking signals from thiol containing protein aggregates. Furthermore, differential detergent lysis was applied to confirm the vesicular nature of the detected extracellular events in blood plasma. We found an increase in exofacial thiols on monocytes upon in vitro stimulation by LPS or TNF, both in primary monocytes and monocytic cell lines (p<0.0005). At the same time, newly released extracellular vesicles showed a decrease in their exofacial thiols compared with those from unstimulated cells (p<0.05). We also found a significant elevation of surface thiols on circulating monocytes in rheumatoid arthritis patients (p<0.05) and newly released extracellular vesicles of isolated CD14+ cells from rheumatoid arthritis patients had decreased thiol levels compared with healthy subjects (p<0.01). Exofacial peroxiredoxin 1 was demonstrated on the surface of primary and cultured monocytes, and the number of peroxiredoxin 1 positive extracellular vesicles was increased in rheumatoid arthritis blood plasma (p<0.05). Furthermore, an overoxidised form of peroxiredoxin was detected in extracellular vesicle-enriched preparations from blood plasma. Our data show that cell surface thiols play a protective role and reflect oxidative stress resistance state in activated immune cells. Furthermore, they support a role of extracellular vesicles in the redox regulation of human monocytes, possibly representing an antioxidant mechanism.
Collapse
Affiliation(s)
- K É Szabó-Taylor
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary.
| | - E Á Tóth
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - A M Balogh
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - B W Sódar
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - L Kádár
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - K Pálóczi
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - N Fekete
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - A Németh
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - X Osteikoetxea
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - K V Vukman
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - M Holub
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - É Pállinger
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Gy Nagy
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary; Semmelweis University, Department of Rheumatology, 3rd Department of Internal Medicine, 1023 Budapest, Hungary
| | - P G Winyard
- University of Exeter Medical School, St Luke's Campus, Heavitree Road, Exeter EX1 2LU, United Kingdom
| | - E I Buzás
- Semmelweis University, Department of Genetics, Cell, and Immunobiology, Nagyvárad tér 4, 1089 Budapest, Hungary
| |
Collapse
|
27
|
Abstract
SIGNIFICANCE Secreted proteins are important both as signaling molecules and potential biomarkers. Recent Advances: Protein can undergo different types of oxidation, both in physiological conditions or under oxidative stress. Several redox proteomics techniques have been successfully applied to the identification of glutathionylated proteins, an oxidative post-translational modification consisting in the formation of a mixed disulfide between a protein cysteine and glutathione. Redox proteomics has also been used to study other forms of protein oxidation. CRITICAL ISSUES Because of the highest proportion of free cysteines in the cytosol, redox proteomics of protein thiols has focused, so far, on intracellular proteins. However, plasma proteins, such as transthyretin and albumin, have been described as glutathionylated or cysteinylated. The present review discusses the redox state of protein cysteines in relation to their cellular distribution. We describe the various approaches used to detect secreted glutathionylated proteins, the only thiol modification studied so far in secreted proteins, and the specific problems presented in the study of the secretome. FUTURE DIRECTIONS This review focusses on glutathionylated proteins secreted under inflammatory conditions and that may act as soluble mediators (cytokines). Future studies on the redox secretome (including other forms of oxidation) might identify new soluble mediators and biomarkers of oxidative stress. Antioxid. Redox Signal. 26, 299-312.
Collapse
Affiliation(s)
- Pietro Ghezzi
- 1 Brighton & Sussex Medical School , Brighton, United Kingdom
| | - Philippe Chan
- 2 PISSARO Proteomic Platform, Institute for Research and Innovation in Biomedicine, University of Rouen , Rouen, France
| |
Collapse
|
28
|
Ghezzi P, Jaquet V, Marcucci F, Schmidt HHHW. The oxidative stress theory of disease: levels of evidence and epistemological aspects. Br J Pharmacol 2016; 174:1784-1796. [PMID: 27425643 DOI: 10.1111/bph.13544] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/26/2016] [Accepted: 06/22/2016] [Indexed: 12/12/2022] Open
Abstract
The theory that oxidative stress (OS) is at the root of several diseases is extremely popular. However, so far, no antioxidant has been recommended or offered by healthcare systems neither has any been approved as therapy by regulatory agencies that base their decisions on evidence-based medicine. This is simply because, so far, despite many preclinical and clinical studies indicating a beneficial effect of antioxidants in many disease conditions, randomised clinical trials have failed to provide the evidence of efficacy required for drug approval. In this review, we discuss the levels of evidence required to claim causality in preclinical research on OS, the weakness of the oversimplification associated with OS theory of disease and the importance of the narrative in its popularity. Finally, from a more translational perspective, we discuss the reasons why antioxidants acting by scavenging ROS might not only prevent their detrimental effects but also interfere with essential signalling roles. We propose that ROS have a complex metabolism and are generated by different enzymes at diverse sites and at different times. Aggregating this plurality of systems into a single theory of disease may not be the best way to develop new drugs, and future research may need to focus on specific oxygen-toxifying pathways rather than on non-specific ROS scavengers. Finally, similarly to what is nowadays required for clinical trials, we recommend making unpublished data available in repositories (open data), as this will allow big data approaches or meta-analyses, without the drawbacks of publication bias. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
| | - Vincent Jaquet
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Harald H H W Schmidt
- Maastricht University, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
29
|
Li Q, Guo R, Lin W. A Fluorescence Turn-On Probe for Thiols with a Tunable Dynamic Range. J Fluoresc 2016; 26:1077-81. [DOI: 10.1007/s10895-016-1796-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/27/2016] [Indexed: 10/22/2022]
|
30
|
Abstract
Disulfide bonds represent versatile posttranslational modifications whose roles encompass the structure, catalysis, and regulation of protein function. Due to the oxidizing nature of the extracellular environment, disulfide bonds found in secreted proteins were once believed to be inert. This notion has been challenged by the discovery of redox-sensitive disulfides that, once cleaved, can lead to changes in protein activity. These functional disulfides are twisted into unique configurations, leading to high strain and potential energy. In some cases, cleavage of these disulfides can lead to a gain of function in protein activity. Thus, these motifs can be referred to as switches. We describe the couples that control redox in the extracellular environment, examine several examples of proteins with switchable disulfides, and discuss the potential applications of disulfides in molecular biology.
Collapse
Affiliation(s)
- Michael C Yi
- Department of Chemical Engineering, Stanford University, Stanford, California 94305; ,
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, California 94305; , .,Department of Chemistry, Stanford University, Stanford, California 94305
| |
Collapse
|
31
|
|
32
|
Proteomic Identification of Oxidized Proteins in Entamoeba histolytica by Resin-Assisted Capture: Insights into the Role of Arginase in Resistance to Oxidative Stress. PLoS Negl Trop Dis 2016; 10:e0004340. [PMID: 26735309 PMCID: PMC4703340 DOI: 10.1371/journal.pntd.0004340] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/08/2015] [Indexed: 12/20/2022] Open
Abstract
Entamoeba histolytica is an obligate protozoan parasite of humans, and amebiasis, an infectious disease which targets the intestine and/or liver, is the second most common cause of human death due to a protozoan after malaria. Although amebiasis is usually asymptomatic, E. histolytica has potent pathogenic potential. During host infection, the parasite is exposed to reactive oxygen species that are produced and released by cells of the innate immune system at the site of infection. The ability of the parasite to survive oxidative stress (OS) is essential for a successful invasion of the host. Although the effects of OS on the regulation of gene expression in E. histolytica and the characterization of some proteins whose function in the parasite's defense against OS have been previously studied, our knowledge of oxidized proteins in E. histolytica is lacking. In order to fill this knowledge gap, we performed a large-scale identification and quantification of the oxidized proteins in oxidatively stressed E. histolytica trophozoites using resin-assisted capture coupled to mass spectrometry. We detected 154 oxidized proteins (OXs) and the functions of some of these proteins were associated with antioxidant activity, maintaining the parasite's cytoskeleton, translation, catalysis, and transport. We also found that oxidation of the Gal/GalNAc impairs its function and contributes to the inhibition of E. histolytica adherence to host cells. We also provide evidence that arginase, an enzyme which converts L-arginine into L-ornithine and urea, is involved in the protection of the parasite against OS. Collectively, these results emphasize the importance of OS as a critical regulator of E. histolytica's functions and indicate a new role for arginase in E. histolytica's resistance to OS. Reactive oxygen species are the most studied of environmental stresses generated by the host immune defense against pathogens. Although most of the studies that have investigated the effect of oxidative stress on an organism have focused on changes which occur at the protein level, only a few studies have investigated the oxidation status of these proteins. Infection with Entamoeba histolytica is known as amebiasis. This condition occurs worldwide, but is most associated with crowded living conditions and poor sanitation. The parasite is exposed inside the host to oxidative stress generated by cells of the host immune system. The nature of oxidized proteins in oxidatively stressed E. histolytica has never been studied. In this report, the authors present their quantitative results of a proteome-wide analysis of oxidized proteins in the oxidatively stressed parasite. They identified crucial redox-regulated proteins that are linked to the virulence of the parasite, such as the Gal/GalNAc lectin. They also discovered that arginase, a protein involved in ornithine synthesis, is also involved in the parasite's resistance to oxidative stress.
Collapse
|
33
|
Cort A, Ozben T, Saso L, De Luca C, Korkina L. Redox Control of Multidrug Resistance and Its Possible Modulation by Antioxidants. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4251912. [PMID: 26881027 PMCID: PMC4736404 DOI: 10.1155/2016/4251912] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/14/2015] [Accepted: 11/18/2015] [Indexed: 12/13/2022]
Abstract
Clinical efficacy of anticancer chemotherapies is dramatically hampered by multidrug resistance (MDR) dependent on inherited traits, acquired defence against toxins, and adaptive mechanisms mounting in tumours. There is overwhelming evidence that molecular events leading to MDR are regulated by redox mechanisms. For example, chemotherapeutics which overrun the first obstacle of redox-regulated cellular uptake channels (MDR1, MDR2, and MDR3) induce a concerted action of phase I/II metabolic enzymes with a temporal redox-regulated axis. This results in rapid metabolic transformation and elimination of a toxin. This metabolic axis is tightly interconnected with the inducible Nrf2-linked pathway, a key switch-on mechanism for upregulation of endogenous antioxidant enzymes and detoxifying systems. As a result, chemotherapeutics and cytotoxic by-products of their metabolism (ROS, hydroperoxides, and aldehydes) are inactivated and MDR occurs. On the other hand, tumour cells are capable of mounting an adaptive antioxidant response against ROS produced by chemotherapeutics and host immune cells. The multiple redox-dependent mechanisms involved in MDR prompted suggesting redox-active drugs (antioxidants and prooxidants) or inhibitors of inducible antioxidant defence as a novel approach to diminish MDR. Pitfalls and progress in this direction are discussed.
Collapse
Affiliation(s)
- Aysegul Cort
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Sanko University, İncili Pınar, Gazi Muhtar Paşa Bulvarı, Sehitkamil, 27090 Gaziantep, Turkey
| | - Tomris Ozben
- Department of Biochemistry, Akdeniz University Medical Faculty, Campus, Dumlupınar Street, 07070 Antalya, Turkey
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Chiara De Luca
- Evidence-Based Well-Being (EB-WB) Ltd., 31 Alt-Stralau, 10245 Berlin, Germany
| | - Liudmila Korkina
- Centre of Innovative Biotechnological Investigations Nanolab, 197 Vernadskogo Prospekt, Moscow 119571, Russia
| |
Collapse
|
34
|
Endothelial function does not improve with high-intensity continuous exercise training in SHR: implications of eNOS uncoupling. Hypertens Res 2015; 39:70-8. [PMID: 26537830 DOI: 10.1038/hr.2015.114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/30/2015] [Accepted: 08/20/2015] [Indexed: 12/28/2022]
Abstract
Exercise training is a well-recognized way to improve vascular endothelial function by increasing nitric oxide (NO) bioavailability. However, in hypertensive subjects, unlike low- and moderate-intensity exercise training, the beneficial effects of continuous high-intensity exercise on endothelial function are not clear, and the underlying mechanisms remain unknown. The aim of this study was to investigate the impact of high-intensity exercise on vascular function, especially on the NO pathway, in spontaneous hypertensive rats (SHR). These effects were studied on WKY, sedentary SHR and SHR that exercised at moderate (SHR-MOD) and high intensity (SHR-HI) on a treadmill (1 h per day; 5 days per week for 6 weeks at 55% and 80% of their maximal aerobic velocity, respectively). Endothelial function and specific NO contributions to acetylcholine-mediated relaxation were evaluated by measuring the aortic ring isometric forces. Endothelial nitric oxide synthase (eNOS) expression and phosphorylation (ser1177) were evaluated by western blotting. The total aortic and eNOS-dependent reactive oxygen species (ROS) production was assessed using electron paramagnetic resonance in aortic tissue. Although the aortas of SHR-HI had increased eNOS levels without alteration of eNOS phosphorylation, high-intensity exercise had no beneficial effect on endothelium-dependent vasorelaxation, unlike moderate exercise. This result was associated with increased eNOS-dependent ROS production in the aortas of SHR-HI. Notably, the use of the recoupling agent BH4 or a thiol-reducing agent blunted eNOS-dependent ROS production in the aortas of SHR-HI. In conclusion, the lack of a positive effect of high-intensity exercise on endothelial function in SHR was mainly explained by redox-dependent eNOS uncoupling, resulting in a switch from NO to O2(-) generation.
Collapse
|
35
|
The flavo-oxidase QSOX1 supports vascular smooth muscle cell migration and proliferation: Evidence for a role in neointima growth. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1334-46. [DOI: 10.1016/j.bbadis.2015.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/13/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022]
|
36
|
Checconi P, Salzano S, Bowler L, Mullen L, Mengozzi M, Hanschmann EM, Lillig CH, Sgarbanti R, Panella S, Nencioni L, Palamara AT, Ghezzi P. Redox proteomics of the inflammatory secretome identifies a common set of redoxins and other glutathionylated proteins released in inflammation, influenza virus infection and oxidative stress. PLoS One 2015; 10:e0127086. [PMID: 25985305 PMCID: PMC4436175 DOI: 10.1371/journal.pone.0127086] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 04/11/2015] [Indexed: 01/06/2023] Open
Abstract
Protein cysteines can form transient disulfides with glutathione (GSH), resulting in the production of glutathionylated proteins, and this process is regarded as a mechanism by which the redox state of the cell can regulate protein function. Most studies on redox regulation of immunity have focused on intracellular proteins. In this study we have used redox proteomics to identify those proteins released in glutathionylated form by macrophages stimulated with lipopolysaccharide (LPS) after pre-loading the cells with biotinylated GSH. Of the several proteins identified in the redox secretome, we have selected a number for validation. Proteomic analysis indicated that LPS stimulated the release of peroxiredoxin (PRDX) 1, PRDX2, vimentin (VIM), profilin1 (PFN1) and thioredoxin 1 (TXN1). For PRDX1 and TXN1, we were able to confirm that the released protein is glutathionylated. PRDX1, PRDX2 and TXN1 were also released by the human pulmonary epithelial cell line, A549, infected with influenza virus. The release of the proteins identified was inhibited by the anti-inflammatory glucocorticoid, dexamethasone (DEX), which also inhibited tumor necrosis factor (TNF)-α release, and by thiol antioxidants (N-butanoyl GSH derivative, GSH-C4, and N-acetylcysteine (NAC), which did not affect TNF-α production. The proteins identified could be useful as biomarkers of oxidative stress associated with inflammation, and further studies will be required to investigate if the extracellular forms of these proteins has immunoregulatory functions.
Collapse
Affiliation(s)
- Paola Checconi
- Institute Pasteur, Cenci-Bolognetti Foundation, "Sapienza" University of Rome, Rome, Italy
- Brighton & Sussex Medical School, Falmer, Brighton, United Kingdom
| | - Sonia Salzano
- Brighton & Sussex Medical School, Falmer, Brighton, United Kingdom
| | - Lucas Bowler
- University of Brighton, Pharmacy and Biomolecular Sciences, Moulsecoomb, Brighton, United Kingdom
| | - Lisa Mullen
- Brighton & Sussex Medical School, Falmer, Brighton, United Kingdom
| | - Manuela Mengozzi
- Brighton & Sussex Medical School, Falmer, Brighton, United Kingdom
| | - Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| | | | - Simona Panella
- IRCSS San Raffaele Pisana, Telematic University, Rome, Italy
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci-Bolognetti Foundation, "Sapienza" University of Rome, Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci-Bolognetti Foundation, "Sapienza" University of Rome, Rome, Italy
| | - Anna Teresa Palamara
- IRCSS San Raffaele Pisana, Telematic University, Rome, Italy
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci-Bolognetti Foundation, "Sapienza" University of Rome, Rome, Italy
| | - Pietro Ghezzi
- Brighton & Sussex Medical School, Falmer, Brighton, United Kingdom
- * E-mail:
| |
Collapse
|
37
|
Zou Y, Xie L, Carroll S, Muniz M, Gibson H, Wei WZ, Liu H, Mao G. Layer-by-layer films with bioreducible and nonbioreducible polycations for sequential DNA release. Biomacromolecules 2014; 15:3965-75. [PMID: 25360688 DOI: 10.1021/bm5010433] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Layer-by-layer (LbL) films containing cationic polyelectrolytes and anionic bioactive molecules such as DNA are promising biomaterials for controlled and localized gene delivery for a number of biomedical applications including cancer DNA vaccine delivery. Bioreducible LbL films made of disulfide-containing poly(amido amine)s (PAAs) and plasmid DNA can be degraded by redox-active membrane proteins through the thiol-disulfide exchange reaction to release DNA exclusively into the extracellular microenvironment adjacent to the film. In order to better understand the film degradation mechanism and nature of the released species, the bioreducible film degradation is studied by atomic force microscopy, fluorescence, and dynamic light scattering in solutions containing a reducing agent. The PAA/DNA LbL film undergoes fast bulk degradation with micrometer-sized pieces breaking off from the substrate. This bulk degradation behavior is arrested by periodic insertions of a nonbioreducible poly(ethylenimine) (PEI) layer. The LbL films containing PAA/DNA and PEI/DNA bilayers display sequential film disassembly and are capable of continuously releasing DNA nanoparticles over a prolonged time. Insertion of the PEI layer enables the bioreducible LbL films to transfect human embryonic kidney 293 cells. The data conclude that the PEI layer is effective as a barrier layer against interlayer diffusion during LbL film assembly and more importantly during film disassembly. Without the barrier layer, the high mobility of cleaved PAA fragments is responsible for bulk degradation of bioreducible LbL films, which may prevent their ultimate gene-delivery applications. This work establishes a direct link among film internal structure, disassembly mechanism, and transfection efficiency. It provides a simple method to design bioreducible LbL films for sequential and long-time DNA release.
Collapse
Affiliation(s)
- Yi Zou
- Department of Chemical Engineering and Materials Science, Wayne State University , 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Oupický D, Li J. Bioreducible polycations in nucleic acid delivery: past, present, and future trends. Macromol Biosci 2014; 14:908-22. [PMID: 24678057 PMCID: PMC4410047 DOI: 10.1002/mabi.201400061] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/19/2014] [Indexed: 12/16/2022]
Abstract
Polycations that are degradable by reduction of disulfide bonds are developed for applications in delivery of nucleic acids. This Feature Article surveys methods of synthesis of bioreducible polycations and discusses current understanding of the mechanism of action of bioreducible polyplexes. Emphasis is placed on the relationship between the biological redox environment and toxicity, trafficking, transfection activity, and in vivo behavior of bioreducible polycations and polyplexes.
Collapse
Affiliation(s)
- David Oupický
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Durham Research Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| | | |
Collapse
|
39
|
Eble JA, de Rezende FF. Redox-relevant aspects of the extracellular matrix and its cellular contacts via integrins. Antioxid Redox Signal 2014; 20:1977-93. [PMID: 24040997 PMCID: PMC3993061 DOI: 10.1089/ars.2013.5294] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/29/2013] [Accepted: 09/16/2013] [Indexed: 12/30/2022]
Abstract
SIGNIFICANCE The extracellular matrix (ECM) fulfills essential functions in multicellular organisms. It provides the mechanical scaffold and environmental cues to cells. Upon cell attachment, the ECM signals into the cells. In this process, reactive oxygen species (ROS) are physiologically used as signalizing molecules. RECENT ADVANCES ECM attachment influences the ROS-production of cells. In turn, ROS affect the production, assembly and turnover of the ECM during wound healing and matrix remodeling. Pathological changes of ROS levels lead to excess ECM production and increased tissue contraction in fibrotic disorders and desmoplastic tumors. Integrins are cell adhesion molecules which mediate cell adhesion and force transmission between cells and the ECM. They have been identified as a target of redox-regulation by ROS. Cysteine-based redox-modifications, together with structural data, highlighted particular regions within integrin heterodimers that may be subject to redox-dependent conformational changes along with an alteration of integrin binding activity. CRITICAL ISSUES In a molecular model, a long-range disulfide-bridge within the integrin β-subunit and disulfide bridges within the genu and calf-2 domains of the integrin α-subunit may control the transition between the bent/inactive and upright/active conformation of the integrin ectodomain. These thiol-based intramolecular cross-linkages occur in the stalk domain of both integrin subunits, whereas the ligand-binding integrin headpiece is apparently unaffected by redox-regulation. FUTURE DIRECTIONS Redox-regulation of the integrin activation state may explain the effect of ROS in physiological processes. A deeper understanding of the underlying mechanism may open new prospects for the treatment of fibrotic disorders.
Collapse
Affiliation(s)
- Johannes A. Eble
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
- Excellence Cluster Cardio-Pulmonary System, Center for Molecular Medicine, Vascular Matrix Biology, Frankfurt University Hospital, Frankfurt/Main, Germany
| | - Flávia Figueiredo de Rezende
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
- Excellence Cluster Cardio-Pulmonary System, Center for Molecular Medicine, Vascular Matrix Biology, Frankfurt University Hospital, Frankfurt/Main, Germany
| |
Collapse
|
40
|
Murphy DD, Reddy EC, Moran N, O'Neill S. Regulation of platelet activity in a changing redox environment. Antioxid Redox Signal 2014; 20:2074-89. [PMID: 24206201 DOI: 10.1089/ars.2013.5698] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SIGNIFICANCE The regulation of platelet function is finely tuned by a balance between the vasculature's redox environment and the oxidative processes that occur in it. The activation of platelets at sites of vascular damage is essential for the maintenance of normal hemostasis. In the extracellular milieu, a normal redox environment is maintained by thiol/disulfide redox couples, which include reduced and oxidized glutathione (GSH/GSSG) and cysteine (Cys/CySS). Oxidative changes in either of the plasma redox potentials are directly linked with risk factors for cardiovascular disease. RECENT ADVANCES Many proteins found on the surface of platelets contain cysteine residues that are targets for oxidation. These include platelet-specific integrins and thiol isomerase enzymes that respond to changes in the extracellular redox environment, thus influencing normal platelet responses. CRITICAL ISSUES The post-translational modification of critical cysteine thiol groups is linked to alterations in redox potentials and occurs both intracellularly and extracellularly in normal platelet activation. Platelet integrins, in particular, are prime targets for redox modification due to their high cysteine content. Although the role of thiol/disulfide bond exchange in platelet activation is established, the effects of a changing redox environment on platelet reactivity are unclear. FUTURE DIRECTIONS A thorough understanding of these mechanisms and how they interact with other platelet signaling events is of the utmost importance for the development of novel therapeutic targets so that we can protect against inappropriate thrombus formation.
Collapse
Affiliation(s)
- Desmond D Murphy
- 1 Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland , Dublin, Ireland
| | | | | | | |
Collapse
|
41
|
Layani-Bazar A, Skornick I, Berrebi A, Pauker MH, Noy E, Silberman A, Albeck M, Longo DL, Kalechman Y, Sredni B. Redox Modulation of Adjacent Thiols in VLA-4 by AS101 Converts Myeloid Leukemia Cells from a Drug-Resistant to Drug-Sensitive State. Cancer Res 2014; 74:3092-103. [DOI: 10.1158/0008-5472.can-13-2159] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Abstract
Most proteins in nature are chemically modified after they are made to control how, when, and where they function. The 3 core features of proteins are posttranslationally modified: amino acid side chains can be modified, peptide bonds can be cleaved or isomerized, and disulfide bonds can be cleaved. Cleavage of peptide bonds is a major mechanism of protein control in the circulation, as exemplified by activation of the blood coagulation and complement zymogens. Cleavage of disulfide bonds is emerging as another important mechanism of protein control in the circulation. Recent advances in our understanding of control of soluble blood proteins and blood cell receptors by functional disulfide bonds is discussed as is how these bonds are being identified and studied.
Collapse
|
43
|
Mermelekas G, Makridakis M, Koeck T, Vlahou A. Redox proteomics: from residue modifications to putative biomarker identification by gel- and LC-MS-based approaches. Expert Rev Proteomics 2014; 10:537-49. [DOI: 10.1586/14789450.2013.855611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
44
|
Pellom ST, Michalek RD, Crump KE, Langston PK, Juneau DG, Grayson JM. Increased cell surface free thiols identify effector CD8+ T cells undergoing T cell receptor stimulation. PLoS One 2013; 8:e81134. [PMID: 24236211 PMCID: PMC3827480 DOI: 10.1371/journal.pone.0081134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 10/18/2013] [Indexed: 11/18/2022] Open
Abstract
Recognition of peptide Major Histocompatibility Complexes (MHC) by the T cell receptor causes rapid production of reactive oxygen intermediates (ROI) in naïve CD8+ T cells. Because ROI such as H2O2 are membrane permeable, mechanisms must exist to prevent overoxidation of surface proteins. In this study we used fluorescently labeled conjugates of maleimide to measure the level of cell surface free thiols (CSFT) during the development, activation and differentiation of CD8+ T cells. We found that during development CSFT were higher on CD8 SP compared to CD4 SP or CD4CD8 DP T cells. After activation CSFT became elevated prior to division but once proliferation started levels continued to rise. During acute viral infection CSFT levels were elevated on antigen-specific effector cells compared to memory cells. Additionally, the CSFT level was always higher on antigen-specific CD8+ T cells in lymphoid compared to nonlymphoid organs. During chronic viral infection, CSFT levels were elevated for extended periods on antigen-specific effector CD8+ T cells. Finally, CSFT levels on effector CD8+ T cells, regardless of infection, identified cells undergoing TCR stimulation. Taken together these data suggest that CD8+ T cells upregulate CSFT following receptor ligation and ROI production during infection to prevent overoxidation of surface proteins.
Collapse
Affiliation(s)
- Samuel Troy Pellom
- Laboratory of Lymphocyte Function, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Ryan D. Michalek
- Metabolon Corporation, Durham, North Carolina, United States of America
| | - Katie E. Crump
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - P. Kent Langston
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Daniel G. Juneau
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jason M. Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
45
|
Cheng H, Lee SH, Wu S. Effects of N-acetyl-L-cysteine on adhesive strength between breast cancer cell and extracellular matrix proteins after ionizing radiation. Life Sci 2013; 93:798-803. [PMID: 24113073 DOI: 10.1016/j.lfs.2013.09.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/19/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
Abstract
AIMS To evaluate the effect of N-acetyl-L-cysteine (LNAC), a common ROS scavenger, on the adhesive affinity between MDA-MB-231 breast cancer cells and extracellular matrix (ECM) proteins after IR. MAIN METHODS Using static cell adhesion assays to determine the effect of various times and duration of LNAC (10mM) treatment on IR (20Gy)-altered adhesive affinity between MDA-MB-231 breast cancer cells and ECM, especially fibronectin; using fluorescence dye carboxy- 2,7-dichlorodihydrofluorescein diacetate to determine intracellular levels of ROS; using flow cytometry to determine cell surface integrin β1; and using Western blot analysis to determine vimentin expression. KEY FINDINGS Our results indicated that continuously treating the breast cancer cells with LNAC for 24h, starting immediately after IR, could inhibit IR-induced cell adhesion to ECM proteins at 24h post-IR. The reduction of cell adhesive affinity was correlated with a down-regulation of IR-induced ROS production and surface expression of activated integrin β1. When the cells were pretreated for 1h, the inhibitory effects of LNAC were found to be either reduced or completely abrogated followed by 24h or 2h treatments, respectively. In addition to cell adhesion, treatment with LNAC inhibited IR-induced expression of vimentin, an epithelial-mesenchymal transition marker (EMT). SIGNIFICANCE The benefits of administering antioxidants during radiation therapy have been the subject of much controversy. Our results suggest that if antioxidant treatment is to be combined with IR therapy, time of administration and treatment duration are important variables to consider.
Collapse
Affiliation(s)
- Huiwen Cheng
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA
| | | | | |
Collapse
|
46
|
Rodríguez-Lara V, Morales-Rivero A, Rivera-Cambas AM, Fortoul TI. Vanadium inhalation induces actin changes in mice testicular cells. Toxicol Ind Health 2013; 32:367-74. [PMID: 24097359 DOI: 10.1177/0748233713501364] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Infertility is becoming a health problem, which has increased mainly in megacities, and several studies have shown its association with environmental pollution. Air pollution has been linked to alterations in sperm parameters, both in humans and animal models. In male humans, it has been associated with reduced semen quality and DNA alterations. Vanadium is a transition element that has increased in recent decades as a component of air suspended matter and has been associated with reprotoxic effects in animal models. Few are the mechanisms described by which the vanadium produces these effects, and cytoskeleton interaction is a possibility. We reported immunohistochemical changes in actin testicular cytoskeleton in a vanadium inhalation experimental mice model. Our findings show that exposure to vanadium pentoxide (0.02 M) results in actin decrease in testicular cells from 3-12 weeks exposure time; this effect was statistically significant and exposure time dependent. Actin cytoskeleton damage is a mechanism that could explain vanadium reprotoxic effects and its association with impaired fertility.
Collapse
Affiliation(s)
- Vianey Rodríguez-Lara
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, México
| | - Alonso Morales-Rivero
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, México
| | - Angelica Muñiz Rivera-Cambas
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, México
| | - Teresa I Fortoul
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, México
| |
Collapse
|
47
|
Abstract
The redox proteome consists of reversible and irreversible covalent modifications that link redox metabolism to biologic structure and function. These modifications, especially of Cys, function at the molecular level in protein folding and maturation, catalytic activity, signaling, and macromolecular interactions and at the macroscopic level in control of secretion and cell shape. Interaction of the redox proteome with redox-active chemicals is central to macromolecular structure, regulation, and signaling during the life cycle and has a central role in the tolerance and adaptability to diet and environmental challenges.
Collapse
Affiliation(s)
- Young-Mi Go
- From the Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322
| | | |
Collapse
|
48
|
Metcalfe C, Cresswell P, Barclay AN. Interleukin-2 signalling is modulated by a labile disulfide bond in the CD132 chain of its receptor. Open Biol 2013; 2:110036. [PMID: 22645657 PMCID: PMC3352089 DOI: 10.1098/rsob.110036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 12/21/2011] [Indexed: 12/22/2022] Open
Abstract
Certain disulfide bonds present in leucocyte membrane proteins are labile and can be reduced in inflammation. This can cause structural changes that result in downstream functional effects, for example, in integrin activation. Recent studies have shown that a wide range of membrane proteins have labile disulfide bonds including CD132, the common gamma chain of the receptors for several cytokines including interleukin-2 and interleukin-4 (IL-2 and IL-4). The Cys(183)-Cys(232) disulfide bond in mouse CD132 is susceptible to reduction by enzymes such as thioredoxin (TRX), gamma interferon-inducible lysosomal thiolreductase and protein disulfide isomerase, which are commonly secreted during immune activation. The Cys(183)-Cys(232) disulfide bond is also reduced in an in vivo lipopolysaccharide (LPS)-induced acute model of inflammation. Conditions that lead to the reduction of the Cys(183)-Cys(232) disulfide bond in CD132 inhibit proliferation of an IL-2-dependent T cell clone and concomitant inhibition of the STAT-5 signalling pathway. The same reducing conditions had no effect on the proliferation of an IL-2-independent T cell clone, nor did they reduce disulfide bonds in IL-2 itself. We postulate that reduction of the Cys(183)-Cys(232) disulfide in CD132 inhibits IL-2 binding to the receptor complex. Published data show that the Cys(183)-Cys(232) disulfide bond is exposed at the surface of CD132 and in close contact with IL-2 and IL-4 in their respective receptor complexes. In addition, mutants in these Cys residues in human CD132 lead to immunodeficiency and loss of IL-2 binding. These results have wider implications for the regulation of cytokine receptors in general, as their activity can be modulated by a 'redox regulator' mechanism caused by the changes in the redox environment that occur during inflammation and activation of the immune system.
Collapse
Affiliation(s)
- Clive Metcalfe
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | | |
Collapse
|
49
|
Lacroix S, Egrise D, Van Simaeys G, Doumont G, Monclus M, Sherer F, Herbaux T, Leroy D, Goldman S. [18F]-FBEM, a tracer targeting cell-surface protein thiols for cell trafficking imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:409-16. [DOI: 10.1002/cmmi.1540] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 03/02/2013] [Accepted: 03/10/2013] [Indexed: 11/06/2022]
|
50
|
Abstract
Protein action in nature is largely controlled by the level of expression and by post-translational modifications. Post-translational modifications result in a proteome that is at least two orders of magnitude more diverse than the genome. There are three basic types of post-translational modifications: covalent modification of an amino acid side chain, hydrolytic cleavage or isomerization of a peptide bond, and reductive cleavage of a disulfide bond. This review addresses the modification of disulfide bonds. Protein disulfide bonds perform either a structural or a functional role, and there are two types of functional disulfide: the catalytic and allosteric bonds. The allosteric disulfide bonds control the function of the mature protein in which they reside by triggering a change when they are cleaved. The change can be in ligand binding, substrate hydrolysis, proteolysis, or oligomer formation. The allosteric disulfides are cleaved by oxidoreductases or by thiol/disulfide exchange, and the configurations of the disulfides and the secondary structures that they link share some recurring features. How these bonds are being identified using bioinformatics and experimental screens and what the future holds for this field of research are also discussed.
Collapse
Affiliation(s)
- Kristina M Cook
- Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney NSW2052, Australia
| | | |
Collapse
|