48 results on '"Benhar M"'
Search Results
2. Chapter Sixteen - Application of a Thioredoxin-Trapping Mutant for Analysis of the Cellular Nitrosoproteome
- Author
-
Benhar, M.
- Published
- 2017
- Full Text
- View/download PDF
3. The Use of Mechanical Valves in the Treatment of Valvular Heart Disease
- Author
-
D’Alessandro, Luigi C., Narducci, C., Pucci, A., Rabitti, G., Ragusa, B., Mamone, P., Benhar, M., Horstkotte, D., editor, and Loogen, F., editor
- Published
- 1986
- Full Text
- View/download PDF
4. Possible Risks of General Anesthesia in Patients with Intraventricular Conduction Disturbances.
- Author
-
Santini, M., Carrara, P., Benhar, M., Piovano, G., Rocchi, M., Di Mascolo, R., and Masini, V.
- Subjects
ANESTHESIA ,ANESTHESIOLOGY ,ELECTROPHYSIOLOGY ,NEUROLOGY ,PHYSIOLOGY ,HALOTHANE - Abstract
In order to assess the risk of complete AV block in patients, with intraventricular conduction disturbances who undergo general anesthesia, 20 patients with various conduction defects (7 LBBB, 1 RBBB and 1st degree AV block, 1 incomplete RBBB +9 RBBB+LAH and 2 RBBB+LPH) were studied by means of His bundle recording and corrected sinus node recovery time (CSNRT) before and after the subministration of thiopental (0.2 g. I.V.), succinylcholine (1 mg/kg 1.V.), Fluothane (1%) and Ethrane (1.6%). Nineteen patients displayed signs of dizziness or syncope; both the sinus rate and the CSNRT, did not undergo significant variations. A sight and not significant variation of intranodal conduction during sinus rhythm was observed after Fluothane administration (AH was prolonged by 8%). A less evident negative dromotropic action of thiopental and Ethrane was only revealed by atrial pacing. No significant variations were demonstrated in His-ventricular conduction after administration of the various drugs. The maximum average increase (1.5%) of the H-V interval was observed after administration of succinylcholine. Acute AV block distal to the His bundle appeared in three patients after succinylcholine administration. [ABSTRACT FROM AUTHOR]
- Published
- 1980
- Full Text
- View/download PDF
5. Possible risks of general anesthesia in patients with intraventricular conduction disturbances
- Author
-
V Masini, M Santini, R. Di Mascolo, Benhar M, Rocchi M, Carrara P, and G Piovano
- Subjects
Risk ,Heart block ,Bundle-Branch Block ,Succinylcholine ,Anesthesia, General ,Enflurane ,medicine ,Humans ,Sinus rhythm ,In patient ,Thiopental ,Bundle branch block ,business.industry ,Intraventricular conduction disturbances ,Cardiac Pacing, Artificial ,General Medicine ,medicine.disease ,Electrophysiology ,Heart Block ,Anesthesia ,Dromotropic ,Halothane ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
In order to assess the risk of complete AV block in patients with intraventricular conduction disturbances who undergo general anesthesia, 20 patients with various conduction defects (7 LBBB, 1 RBBB and 1st degree AV block, 1 incomplete RBBB, 9 RBBB + LAH and 2 RBBB + LPH) were studied by means of His bundle recording and corrected sinus node recovery time (CSNRT) before and after the subministration of thiopental (0.2 g I.V.), succinylcholine (1 mg/kg I.V.), Fluothane (1%) and Ethrane (1.6%). Nineteen patients displayed signs of dizziness or syncope; both the sinus rate and the CSNRT, did not undergo significant variations. A slight and not significant variation of intranodal conduction during sinus rhythm was observed after Fluothane administration (AH was prolonged by 8%). A less evident negative dromotropic action of thiopental and Ethrane was only revealed by atrial pacing. No significant variations were demonstrated in His-ventricular conduction after administration of the various drugs. The maximum average increase (1.5%) of the H-V interval was observed after administration of succinylcholine. Acute AV block distal to the His bundle appeared in three patients after succinylcholine administration.
- Published
- 1980
6. Endogenous histidine peptides are physiological antioxidants that prevent oligodendrocyte cell death and myelin loss in vivo.
- Author
-
Sajrawi C, Odeh M, Tiwari AK, Agranovich B, Abramovich I, Zubedat S, Saar G, Shaulov L, Avital A, Reznik D, Benhar M, Radzishevsky I, Engelender S, and Wolosker H
- Subjects
- Animals, Mice, Cuprizone toxicity, Oxidative Stress drug effects, Oxidative Stress physiology, Mice, Inbred C57BL, Histidine pharmacology, Histidine metabolism, Demyelinating Diseases metabolism, Demyelinating Diseases pathology, Demyelinating Diseases chemically induced, Glucosephosphate Dehydrogenase metabolism, Brain metabolism, Brain drug effects, Brain pathology, Male, Oligodendroglia metabolism, Oligodendroglia drug effects, Mice, Knockout, Antioxidants pharmacology, Antioxidants metabolism, Cell Death drug effects, Cell Death physiology, Myelin Sheath metabolism, Myelin Sheath drug effects, Myelin Sheath pathology
- Abstract
Histidine dipeptides (HDs) are synthesized in brain oligodendrocytes by carnosine synthase (carns1), but their role is unknown. Using metabolomics and in vivo experiments with both constitutive and oligodendrocyte-selective carns1-KO mouse models, we found that HDs are critical for oligodendrocyte survival and protect against oxidative stress. Carns1-KO mouse models had lower numbers of mature oligodendrocytes, increased lipid peroxidation, and behavioral changes. Cuprizone administration, which increases reactive oxygen species in vivo, resulted in higher oligodendrocyte death, demyelination, axonal alterations, and oxidative damage in the corpus callosum of carns1-KO mice. Gliosis and oxidative damage by cuprizone were prevented by pretreatment with the antioxidant N-acetylcysteine. NADPH levels were increased threefold in the brains of carns1-KO mice as an antioxidant response to oxidative stress through acceleration of the pentose phosphate pathway (PPP). This was due to overexpression of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Likewise, expression of NAD kinase, the biosynthetic enzyme for NADP+, and NAMPT, which replenishes the NAD+ pool, was higher in carns1-KO mice brains than in controls. Our observations suggest that HDs cell-autonomously protect oligodendrocytes from oxidative stress, with implications for demyelinating diseases., (© 2024 Wiley Periodicals LLC.)
- Published
- 2025
- Full Text
- View/download PDF
7. Widespread S-persulfidation in activated macrophages as a protective mechanism against oxidative-inflammatory stress.
- Author
-
Salti T, Braunstein I, Haimovich Y, Ziv T, and Benhar M
- Subjects
- Animals, Mice, Humans, Lipopolysaccharides, Inflammation metabolism, Cystathionine gamma-Lyase metabolism, Sulfides pharmacology, Interferon-gamma metabolism, Reactive Oxygen Species metabolism, Oxidation-Reduction, Proteomics methods, Oxidative Stress drug effects, Macrophages metabolism, Macrophages drug effects, Macrophages immunology, Hydrogen Sulfide metabolism, Hydrogen Sulfide pharmacology, Macrophage Activation drug effects
- Abstract
Acute inflammatory responses often involve the production of reactive oxygen and nitrogen species by innate immune cells, particularly macrophages. How activated macrophages protect themselves in the face of oxidative-inflammatory stress remains a long-standing question. Recent evidence implicates reactive sulfur species (RSS) in inflammatory responses; however, how endogenous RSS affect macrophage function and response to oxidative and inflammatory insults remains poorly understood. In this study, we investigated the endogenous pathways of RSS biogenesis and clearance in macrophages, with a particular focus on exploring how hydrogen sulfide (H
2 S)-mediated S-persulfidation influences macrophage responses to oxidative-inflammatory stress. We show that classical activation of mouse or human macrophages using lipopolysaccharide and interferon-γ (LPS/IFN-γ) triggers substantial production of H2 S/RSS, leading to widespread protein persulfidation. Biochemical and proteomic analyses revealed that this surge in cellular S-persulfidation engaged ∼2% of total thiols and modified over 800 functionally diverse proteins. S-persulfidation was found to be largely dependent on the cystine importer xCT and the H2 S-generating enzyme cystathionine γ-lyase and was independent of changes in the global proteome. We further investigated the role of the sulfide-oxidizing enzyme sulfide quinone oxidoreductase (SQOR), and found that it acts as a negative regulator of S-persulfidation. Elevated S-persulfidation following LPS/IFN-γ stimulation or SQOR inhibition was associated with increased resistance to oxidative stress. Upregulation of persulfides also inhibited the activation of the macrophage NLRP3 inflammasome and provided protection against inflammatory cell death. Collectively, our findings shed light on the metabolism and effects of RSS in macrophages and highlight the crucial role of persulfides in enabling macrophages to withstand and alleviate oxidative-inflammatory stress., Competing Interests: Declaration of competing interest All authors declare that they have no financial interests that may be relevant to the submitted work., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
8. Impact of Reactive Sulfur Species on Entamoeba histolytica : Modulating Viability, Motility, and Biofilm Degradation Capacity.
- Author
-
Ye J, Salti T, Zanditenas E, Trebicz-Geffen M, Benhar M, and Ankri S
- Abstract
Reactive sulfur species (RSS) like hydrogen sulfide (H
2 S) and cysteine persulfide (Cys-SSH) emerged as key signaling molecules with diverse physiological roles in the body, depending on their concentration and the cellular environment. While it is known that H2 S and Cys-SSH are produced by both colonocytes and by the gut microbiota through sulfur metabolism, it remains unknown how these RSS affect amebiasis caused by Entamoeba histolytica , a parasitic protozoan that can be present in the human gastrointestinal tract. This study investigates H2 S and Cys-SSH's impact on E. histolytica physiology and explores potential therapeutic implications. Exposing trophozoites to the H2 S donor, sodium sulfide (Na2 S), or to Cys-SSH led to rapid cytotoxicity. A proteomic analysis of Cys-SSH-challenged trophozoites resulted in the identification of >500 S-sulfurated proteins, which are involved in diverse cellular processes. Functional assessments revealed inhibited protein synthesis, altered cytoskeletal dynamics, and reduced motility in trophozoites treated with Cys-SSH. Notably, cysteine proteases (CPs) were significantly inhibited by S-sulfuration, affecting their bacterial biofilm degradation capacity. Immunofluorescence microscopy confirmed alterations in actin dynamics, corroborating the proteomic findings. Thus, our study reveals how RSS perturbs critical cellular functions in E. histolytica , potentially influencing its pathogenicity and interactions within the gut microbiota. Understanding these molecular mechanisms offers novel insights into amebiasis pathogenesis and unveils potential therapeutic avenues targeting RSS-mediated modifications in parasitic infections.- Published
- 2024
- Full Text
- View/download PDF
9. Global Thiol Proteome Analysis Provides Novel Insights into the Macrophage Inflammatory Response and Its Regulation by the Thioredoxin System.
- Author
-
Abu Hariri H, Braunstein I, Salti T, Glaser F, Gefen T, Geva-Zatorsky N, Ziv T, and Benhar M
- Subjects
- Humans, Proteome metabolism, Proteomics, Lipopolysaccharides pharmacology, Thioredoxins metabolism, Oxidation-Reduction, Macrophages metabolism, Sulfhydryl Compounds metabolism, Cysteine metabolism
- Abstract
Aims: Oxidative modifications of cysteine (Cys) thiols regulate various physiological processes, including inflammatory responses. The thioredoxin (Trx) system plays a key role in thiol redox control. The aim of this study was to characterize the dynamic cysteine proteome of human macrophages upon activation by the prototypical proinflammatory agent, bacterial lipopolysaccharide (LPS), and/or perturbation of the Trx system. Results: In this study, we profiled the cellular and redox proteome of human THP-1-derived macrophages during the early phase of LPS activation and/or inhibition of Trx system activity by auranofin (AF) by employing a peptide-centric, resin-assisted capture, redox proteomic workflow. Among 4200 identified cysteines, oxidation of nearly 10% was selectively affected by LPS or AF treatments. Notably, the proteomic analysis uncovered a subset of ∼100 thiols, mapped to proteins involved in diverse processes, whose oxidation is antagonistically regulated by LPS and Trx. Compared with the redox proteome, the cellular proteome was largely unchanged, highlighting the importance of redox modification as a mechanism that allows for rapid modulation of macrophage activities in response to a proinflammatory or pro-oxidant insult. Structural-functional analyses provided mechanistic insights into redox regulation of selected proteins, including the glutathione-synthesizing enzyme, glutamate-cysteine ligase, and the autophagy adaptor, SQSTM1/p62, suggesting mechanisms by which macrophages adapt and fine-tune their responses according to a changing inflammatory and redox environment. Innovation: This study provides a rich resource for further characterization of redox mechanisms that regulate macrophage inflammatory activities. Conclusion: The dynamic thiol redox proteome allows macrophages to efficiently respond and adapt to redox and inflammatory challenges. Antioxid. Redox Signal. 38, 388-402.
- Published
- 2023
- Full Text
- View/download PDF
10. S-nitrosocysteine and glutathione depletion synergize to induce cell death in human tumor cells: Insights into the redox and cytotoxic mechanisms.
- Author
-
Knany A, Engelman R, Hariri HA, Biswal S, Wolfenson H, and Benhar M
- Subjects
- Buthionine Sulfoximine pharmacology, Cell Death, Cysteine analogs & derivatives, Humans, Oxidation-Reduction, Glutathione metabolism, S-Nitrosothiols
- Abstract
Nitric oxide (NO)-dependent signaling and cytotoxic effects are mediated in part via protein S-nitrosylation. The magnitude and duration of S-nitrosylation are governed by the two main thiol reducing systems, the glutathione (GSH) and thioredoxin (Trx) antioxidant systems. In recent years, approaches have been developed to harness the cytotoxic potential of NO/nitrosylation to inhibit tumor cell growth. However, progress in this area has been hindered by insufficient understanding of the balance and interplay between cellular nitrosylation, other oxidative processes and the GSH/Trx systems. In addition, the mechanistic relationship between thiol redox imbalance and cancer cell death is not fully understood. Herein, we explored the redox and cellular effects induced by the S-nitrosylating agent, S-nitrosocysteine (CysNO), in GSH-sufficient and -deficient human tumor cells. We used l-buthionine-sulfoximine (BSO) to induce GSH deficiency, and employed redox, biochemical and cellular assays to interrogate molecular mechanisms. We found that, under GSH-sufficient conditions, a CysNO challenge (100-500 μM) results in a marked yet reversible increase in protein S-nitrosylation in the absence of appreciable S-oxidation. In contrast, under GSH-deficient conditions, CysNO induces elevated and sustained levels of both S-nitrosylation and S-oxidation. Experiments in various cancer cell lines showed that administration of CysNO or BSO alone commonly induce minimal cytotoxicity whereas BSO/CysNO combination therapy leads to extensive cell death. Studies in HeLa cancer cells revealed that treatment with BSO/CysNO results in dual inhibition of the GSH and Trx systems, thereby amplifying redox stress and causing cellular dysfunction. In particular, BSO/CysNO induced rapid oxidation and collapse of the actin cytoskeletal network, followed by loss of mitochondrial function, leading to profound and irreversible decrease in ATP levels. Further observations indicated that BSO/CysNO-induced cell death occurs via a caspase-independent mechanism that involves multiple stress-induced pathways. The present findings provide new insights into the relationship between cellular nitrosylation/oxidation, thiol antioxidant defenses and cell death. These results may aid future efforts to develop NO/redox-based anticancer approaches., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Oxidants, Antioxidants and Thiol Redox Switches in the Control of Regulated Cell Death Pathways.
- Author
-
Benhar M
- Abstract
It is well appreciated that biological reactive oxygen and nitrogen species such as hydrogen peroxide, superoxide and nitric oxide, as well as endogenous antioxidant systems, are important modulators of cell survival and death in diverse organisms and cell types. In addition, oxidative stress, nitrosative stress and dysregulated cell death are implicated in a wide variety of pathological conditions, including cancer, cardiovascular and neurological diseases. Therefore, much effort is devoted to elucidate the molecular mechanisms linking oxidant/antioxidant systems and cell death pathways. This review is focused on thiol redox modifications as a major mechanism by which oxidants and antioxidants influence specific regulated cell death pathways in mammalian cells. Growing evidence indicates that redox modifications of cysteine residues in proteins are involved in the regulation of multiple cell death modalities, including apoptosis, necroptosis and pyroptosis. In addition, recent research suggests that thiol redox switches play a role in the crosstalk between apoptotic and necrotic forms of regulated cell death. Thus, thiol-based redox circuits provide an additional layer of control that determines when and how cells die.
- Published
- 2020
- Full Text
- View/download PDF
12. Opposing effects of polysulfides and thioredoxin on apoptosis through caspase persulfidation.
- Author
-
Braunstein I, Engelman R, Yitzhaki O, Ziv T, Galardon E, and Benhar M
- Subjects
- Caspase 3 metabolism, Caspase 9 metabolism, Caspase Inhibitors pharmacology, Enzyme Activation drug effects, HeLa Cells, Humans, Signal Transduction drug effects, Thioredoxin-Disulfide Reductase metabolism, Apoptosis drug effects, Caspases metabolism, Sulfides metabolism, Sulfides pharmacology, Thioredoxins pharmacology
- Abstract
Hydrogen sulfide has been implicated in a large number of physiological processes including cell survival and death, encouraging research into its mechanisms of action and therapeutic potential. Results from recent studies suggest that the cellular effects of hydrogen sulfide are mediated in part by sulfane sulfur species, including persulfides and polysulfides. In the present study, we investigated the apoptosis-modulating effects of polysulfides, especially on the caspase cascade, which mediates the intrinsic apoptotic pathway. Biochemical analyses revealed that organic or synthetic polysulfides strongly and rapidly inhibit the enzymatic activity of caspase-3, a major effector protease in apoptosis. We attributed the caspase-3 inhibition to persulfidation of its catalytic cysteine. In apoptotically stimulated HeLa cells, short-term exposure to polysulfides triggered the persulfidation and deactivation of cleaved caspase-3. These effects were antagonized by the thioredoxin/thioredoxin reductase system (Trx/TrxR). Trx/TrxR restored the activity of polysulfide-inactivated caspase-3 in vitro , and TrxR inhibition potentiated polysulfide-mediated suppression of caspase-3 activity in situ We further found that under conditions of low TrxR activity, early cell exposure to polysulfides leads to enhanced persulfidation of initiator caspase-9 and decreases apoptosis. Notably, we show that the proenzymes procaspase-3 and -9 are basally persulfidated in resting (unstimulated) cells and become depersulfidated during their processing and activation. Inhibition of TrxR attenuated the depersulfidation and activation of caspase-9. Taken together, our results reveal that polysulfides target the caspase-9/3 cascade and thereby suppress cancer cell apoptosis, and highlight the role of Trx/TrxR-mediated depersulfidation in enabling caspase activation., (© 2020 Braunstein et al.)
- Published
- 2020
- Full Text
- View/download PDF
13. Selective Persulfide Detection Reveals Evolutionarily Conserved Antiaging Effects of S-Sulfhydration.
- Author
-
Zivanovic J, Kouroussis E, Kohl JB, Adhikari B, Bursac B, Schott-Roux S, Petrovic D, Miljkovic JL, Thomas-Lopez D, Jung Y, Miler M, Mitchell S, Milosevic V, Gomes JE, Benhar M, Gonzalez-Zorn B, Ivanovic-Burmazovic I, Torregrossa R, Mitchell JR, Whiteman M, Schwarz G, Snyder SH, Paul BD, Carroll KS, and Filipovic MR
- Published
- 2020
- Full Text
- View/download PDF
14. Selective Persulfide Detection Reveals Evolutionarily Conserved Antiaging Effects of S-Sulfhydration.
- Author
-
Zivanovic J, Kouroussis E, Kohl JB, Adhikari B, Bursac B, Schott-Roux S, Petrovic D, Miljkovic JL, Thomas-Lopez D, Jung Y, Miler M, Mitchell S, Milosevic V, Gomes JE, Benhar M, Gonzalez-Zorn B, Ivanovic-Burmazovic I, Torregrossa R, Mitchell JR, Whiteman M, Schwarz G, Snyder SH, Paul BD, Carroll KS, and Filipovic MR
- Subjects
- Animals, Caenorhabditis elegans, Cell Line, Cyclohexanones chemistry, Cysteine chemistry, Cysteine metabolism, Drosophila melanogaster, Escherichia coli, Fibroblasts, Humans, Male, Mice, Mice, Inbred C57BL, Oxidative Stress physiology, Rats, Rats, Wistar, Saccharomyces cerevisiae, Staining and Labeling, Aging metabolism, Hydrogen Sulfide metabolism, Protein Processing, Post-Translational physiology, Sulfides metabolism
- Abstract
Life on Earth emerged in a hydrogen sulfide (H
2 S)-rich environment eons ago and with it protein persulfidation mediated by H2 S evolved as a signaling mechanism. Protein persulfidation (S-sulfhydration) is a post-translational modification of reactive cysteine residues, which modulate protein structure and/or function. Persulfides are difficult to label and study due to their reactivity and similarity with cysteine. Here, we report a facile strategy for chemoselective persulfide bioconjugation using dimedone-based probes, to achieve highly selective, rapid, and robust persulfide labeling in biological samples with broad utility. Using this method, we show persulfidation is an evolutionarily conserved modification and waves of persulfidation are employed by cells to resolve sulfenylation and prevent irreversible cysteine overoxidation preserving protein function. We report an age-associated decline in persulfidation that is conserved across evolutionary boundaries. Accordingly, dietary or pharmacological interventions to increase persulfidation associate with increased longevity and improved capacity to cope with stress stimuli., (Crown Copyright © 2019. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
15. S-Nitrosylation of α1-Antitrypsin Triggers Macrophages Toward Inflammatory Phenotype and Enhances Intra-Cellular Bacteria Elimination.
- Author
-
Kaner Z, Engelman R, Schuster R, Rider P, Greenberg D, Av-Gay Y, Benhar M, and Lewis EC
- Subjects
- Animals, Female, Macrophages, Peritoneal microbiology, Mice, alpha 1-Antitrypsin genetics, Immunity, Innate, Macrophages, Peritoneal immunology, Nitric Oxide immunology, Salmonella typhi immunology, alpha 1-Antitrypsin immunology
- Abstract
Background: Human α1-antitrypsin (hAAT) is a circulating anti-inflammatory serine-protease inhibitor that rises during acute phase responses. in vivo , hAAT reduces bacterial load, without directly inhibiting bacterial growth. In conditions of excess nitric-oxide (NO), hAAT undergoes S-nitrosylation (S-NO-hAAT) and gains antibacterial capacity. The impact of S-NO-hAAT on immune cells has yet to be explored. Aim: Study the effects of S-NO-hAAT on immune cells during bacterial infection. Methods: Clinical-grade hAAT was S-nitrosylated and then compared to unmodified hAAT, functionally, and structurally. Intracellular bacterial clearance by THP-1 macrophages was assessed using live Salmonella typhi . Murine peritoneal macrophages were examined, and signaling pathways were evaluated. S-NO-hAAT was also investigated after blocking free mambranal cysteine residues on cells. Results: S-NO-hAAT (27.5 uM) enhances intracellular bacteria elimination by immunocytes (up to 1-log reduction). S-NO-hAAT causes resting macrophages to exhibit a pro-inflammatory and antibacterial phenotype, including release of inflammatory cytokines and induction of inducible nitric oxide synthase (iNOS) and TLR2. These pro-inflammatory effects are dependent upon cell surface thiols and activation of MAPK pathways. Conclusions: hAAT duality appears to be context-specific, involving S-nitrosylation in a nitric oxide rich environment. Our results suggest that S-nitrosylation facilitates the antibacterial activity of hAAT by promoting its ability to activate innate immune cells. This pro-inflammatory effect may involve transferring of nitric oxide from S-NO-hAAT to a free cysteine residue on cellular targets.
- Published
- 2019
- Full Text
- View/download PDF
16. Roles of mammalian glutathione peroxidase and thioredoxin reductase enzymes in the cellular response to nitrosative stress.
- Author
-
Benhar M
- Subjects
- Animals, Humans, Glutathione Peroxidase metabolism, Nitrosative Stress physiology, Selenoproteins metabolism, Thioredoxin-Disulfide Reductase metabolism
- Abstract
Mammalian cells employ elaborate antioxidant systems to effectively handle reactive oxygen and nitrogen species (ROS and RNS). At the heart of these systems operate two selenoprotein families consisting of glutathione peroxidase (GPx) and thioredoxin reductase (TrxR) enzymes. Although mostly studied in the context of oxidative stress, considerable evidence has amassed to indicate that these selenoenzymes also play important roles in nitrosative stress responses. GPx and TrxR, together with their redox partners, metabolize nitrosothiols and peroxynitrite, two major RNS. As such, these enzymes play active roles in the cellular defense against nitrosative stress. However, under certain conditions, these enzymes are inactivated by nitrosothiols or peroxynitrite, which may exacerbate oxidative and nitrosative stress in cells. The selenol groups in the active sites of GPx and TrxR enzymes are critically involved in these beneficial and detrimental processes. Further elucidation of the biochemical interactions between distinct RNS and GPx/TrxR will lead to a better understanding of the roles of these selenoenzymes in cellular homeostasis and disease., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. Correction: Nitrosothiol-Trapping-Based Proteomic Analysis of S-Nitrosylation in Human Lung Carcinoma Cells.
- Author
-
Ben-Lulu S, Ziv T, Weisman-Shomer P, and Benhar M
- Abstract
[This corrects the article DOI: 10.1371/journal.pone.0169862.].
- Published
- 2017
- Full Text
- View/download PDF
18. Nitrosothiol-Trapping-Based Proteomic Analysis of S-Nitrosylation in Human Lung Carcinoma Cells.
- Author
-
Ben-Lulu S, Ziv T, Weisman-Shomer P, and Benhar M
- Subjects
- A549 Cells, Cell Cycle drug effects, Cysteine chemistry, Cysteine pharmacokinetics, Cysteine pharmacology, Cytokines pharmacology, Humans, Inflammation metabolism, Inflammation pathology, Lung Neoplasms pathology, Signal Transduction drug effects, Cysteine analogs & derivatives, Lung Neoplasms metabolism, Neoplasm Proteins biosynthesis, Proteomics methods, S-Nitrosothiols chemistry, S-Nitrosothiols pharmacokinetics, S-Nitrosothiols pharmacology, Staining and Labeling methods
- Abstract
Nitrosylation of cysteines residues (S-nitrosylation) mediates many of the cellular effects of nitric oxide in normal and diseased cells. Recent research indicates that S-nitrosylation of certain proteins could play a role in tumor progression and responsiveness to therapy. However, the protein targets of S-nitrosylation in cancer cells remain largely unidentified. In this study, we used our recently developed nitrosothiol trapping approach to explore the nitrosoproteome of human A549 lung carcinoma cells treated with S-nitrosocysteine or pro-inflammatory cytokines. Using this approach, we identified about 300 putative nitrosylation targets in S-nitrosocysteine-treated A549 cells and approximately 400 targets in cytokine-stimulated cells. Among the more than 500 proteins identified in the two screens, the majority represent novel targets of S-nitrosylation, as revealed by comparison with publicly available nitrosoproteomic data. By coupling the trapping procedure with differential thiol labeling, we identified nearly 300 potential nitrosylation sites in about 150 proteins. The proteomic results were validated for several proteins by an independent approach. Bioinformatic analysis highlighted important cellular pathways that are targeted by S-nitrosylation, notably, cell cycle and inflammatory signaling. Taken together, our results identify new molecular targets of nitric oxide in lung cancer cells and suggest that S-nitrosylation may regulate signaling pathways that are critically involved in lung cancer progression., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2017
- Full Text
- View/download PDF
19. Inhibitory nitrosylation of mammalian thioredoxin reductase 1: Molecular characterization and evidence for its functional role in cellular nitroso-redox imbalance.
- Author
-
Engelman R, Ziv T, Arnér ESJ, and Benhar M
- Subjects
- Amino Acid Sequence, Animals, Catalytic Domain, Cysteine analogs & derivatives, Cysteine chemistry, Cysteine pharmacology, Glutathione metabolism, HeLa Cells, Humans, NADP chemistry, Nitric Oxide Donors chemistry, Nitric Oxide Donors pharmacology, Oxidation-Reduction, Protein Processing, Post-Translational, Rats, S-Nitrosothiols chemistry, S-Nitrosothiols pharmacology, Selenocysteine chemistry, Thioredoxin Reductase 1 antagonists & inhibitors, Thioredoxin Reductase 1 chemistry, Thioredoxin Reductase 1 metabolism
- Abstract
Mammalian thioredoxin 1 (Trx1) and the selenoprotein Trx reductase 1 (TrxR1) are key cellular enzymes that function coordinately in thiol-based redox regulation and signaling. Recent studies have revealed that the Trx1/TrxR1 system has an S-nitrosothiol reductase (denitrosylase) activity through which it can regulate nitric oxide-related cellular processes. In this study we revealed that TrxR1 is itself susceptible to nitrosylation, characterized the underlying mechanism, and explored its functional significance. We found that nitrosothiol or nitric oxide donating agents rapidly and effectively inhibited the activity of recombinant or endogenous TrxR1. In particular, the NADPH-reduced TrxR1 was partially and reversibly inhibited upon exposure to low concentrations (<10μM) of S-nitrosocysteine (CysNO) and markedly and continuously inhibited at higher doses. Concurrently, TrxR1 very efficiently reduced low, but not high, levels of CysNO. Biochemical and mass spectrometric analyses indicated that its active site selenocysteine residue renders TrxR1 highly susceptible to nitrosylation-mediated inhibition, and revealed both thiol and selenol modifications at the two redox active centers of the enzyme. Studies in HeLa cancer cells demonstrated that endogenous TrxR1 is sensitive to nitrosylation-dependent inactivation and pointed to an important role for glutathione in reversing or preventing this process. Notably, depletion of cellular glutathione with l-buthionine-sulfoximine synergized with nitrosating agents in promoting sustained nitrosylation and inactivation of TrxR1, events that were accompanied by significant oxidation of Trx1 and extensive cell death. Collectively, these findings expand our knowledge of the role and regulation of the mammalian Trx system in relation to cellular nitroso-redox imbalance. The observations raise the possibility of exploiting the nitrosylation susceptibility of TrxR1 for killing tumor cells., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
20. Dual targeting of the thioredoxin and glutathione systems in cancer and HIV.
- Author
-
Benhar M, Shytaj IL, Stamler JS, and Savarino A
- Subjects
- Acquired Immunodeficiency Syndrome therapy, Animals, Humans, Mice, Neoplasms pathology, Neoplasms therapy, Oxidation-Reduction, Acquired Immunodeficiency Syndrome immunology, Glutathione immunology, HIV-1 immunology, Neoplasms immunology, Thioredoxins immunology
- Abstract
Although the use of antioxidants for the treatment of cancer and HIV/AIDS has been proposed for decades, new insights gained from redox research have suggested a very different scenario. These new data show that the major cellular antioxidant systems, the thioredoxin (Trx) and glutathione (GSH) systems, actually promote cancer growth and HIV infection, while suppressing an effective immune response. Mechanistically, these systems control both the redox- and NO-based pathways (nitroso-redox homeostasis), which subserve innate and cellular immune defenses. Dual inhibition of the Trx and GSH systems synergistically kills neoplastic cells in vitro and in mice and decreases resistance to anticancer therapy. Similarly, the population of HIV reservoir cells that constitutes the major barrier to a cure for AIDS is exquisitely redox sensitive and could be selectively targeted by Trx and GSH inhibitors. Trx and GSH inhibition may lead to a reprogramming of the immune response, tilting the balance between the immune system and cancer or HIV in favor of the former, allowing elimination of diseased cells. Thus, therapies based on silencing of the Trx and GSH pathways represent a promising approach for the cure of both cancer and AIDS and warrant further investigation.
- Published
- 2016
- Full Text
- View/download PDF
21. Emerging Roles of Protein S-Nitrosylation in Macrophages and Cancer Cells.
- Author
-
Benhar M
- Subjects
- Alcohol Dehydrogenase genetics, Alcohol Dehydrogenase metabolism, Animals, Glutathione metabolism, Humans, Macrophages immunology, Neoplasms metabolism, Nitric Oxide metabolism, Protein Processing, Post-Translational, Receptor Protein-Tyrosine Kinases metabolism, Thioredoxins chemistry, Macrophages metabolism, Neoplasms pathology, Thioredoxins metabolism
- Abstract
Despite long and intensive investigation, the mechanisms by which nitric oxide (NO) regulates immune function and carcinogenesis remain incompletely understood. Protein S-nitrosylation, the covalent attachment of a nitroso group to a cysteine thiol, has emerged as a central mechanism of NO-dependent cellular regulation. In particular, recent research has revealed important roles for S-nitrosylation/denitrosylation in modulating the activity of macrophage and tumor cell proteins, implicating Snitrosylation in the regulation of macrophage function as well as in tumor development and response to therapy. This review summarizes recent progress in the identification and characterization of S-nitrosylated proteins in macrophages and cancer cells. The review highlights key findings and insights obtained from functional and proteomic studies about the roles of S-nitrosylation in signaling, transcription, apoptosis and other cellular processes relevant to macrophage function and cancer progression. Some of the implications of recent discoveries for the development of novel anticancer approaches are also discussed.
- Published
- 2016
- Full Text
- View/download PDF
22. Nitric oxide and the thioredoxin system: a complex interplay in redox regulation.
- Author
-
Benhar M
- Subjects
- Animals, Humans, Oxidation-Reduction, Nitric Oxide physiology, Thioredoxins physiology
- Abstract
Background: The free radical nitric oxide (NO) and the thiol oxidoreductase thioredoxin (Trx) play essential roles in cellular redox regulation. Recent biochemical and cellular studies have revealed a complex thiol-dependent crosstalk between NO and Trx that modulates multiple redox-dependent pathways., Scope of Review: This review aims to discuss recent progress, as well as the remaining questions, regarding the interaction and cross regulation between NO and Trx in cellular function and dysfunction., Major Conclusions: The importance and ubiquity of NO-mediated S-nitrosylation of protein thiols as a signaling mechanism is increasingly recognized as is the central role of Trx in regulating S-nitrosylation processes. By denitrosylating diverse protein substrates, Trx plays an active role in attenuating NO signaling as well as in ameliorating nitrosative stress. Yet, at the same time, Trx can also support the activity of NO synthases, thus promoting NO production and its downstream effects. Finally, NO can reciprocally modulate the redox activity of Trx and Trx reductase., General Significance: Further elucidation of the crosstalk between NO and Trx will be important for an improved understanding of the effects of reactive oxygen and nitrogen species on cellular signaling and function., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
23. Blocking IL1β Pathway Following Paclitaxel Chemotherapy Slightly Inhibits Primary Tumor Growth but Promotes Spontaneous Metastasis.
- Author
-
Voloshin T, Alishekevitz D, Kaneti L, Miller V, Isakov E, Kaplanov I, Voronov E, Fremder E, Benhar M, Machluf M, Apte RN, and Shaked Y
- Subjects
- Animals, Cell Line, Tumor, Cells, Cultured, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Gene Expression Regulation, Neoplastic drug effects, Humans, Interleukin 1 Receptor Antagonist Protein administration & dosage, Interleukin-1beta blood, Interleukin-1beta metabolism, Macrophages drug effects, Macrophages metabolism, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Neoplasm Metastasis, Neoplasms, Experimental blood supply, Neoplasms, Experimental genetics, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic prevention & control, Paclitaxel administration & dosage, Receptors, Interleukin-1 antagonists & inhibitors, Receptors, Interleukin-1 metabolism, Reverse Transcriptase Polymerase Chain Reaction, Tumor Burden drug effects, Antineoplastic Combined Chemotherapy Protocols pharmacology, Interleukin-1beta genetics, Neoplasms, Experimental drug therapy, Signal Transduction drug effects
- Abstract
Acquired resistance to therapy is a major obstacle in clinical oncology, and little is known about the contributing mechanisms of the host response to therapy. Here, we show that the proinflammatory cytokine IL1β is overexpressed in response to paclitaxel chemotherapy in macrophages, subsequently promoting the invasive properties of malignant cells. In accordance, blocking IL1β, or its receptor, using either genetic or pharmacologic approach, results in slight retardation of primary tumor growth; however, it accelerates metastasis spread. Tumors from mice treated with combined therapy of paclitaxel and the IL1 receptor antagonist anakinra exhibit increased number of M2 macrophages and vessel leakiness when compared with paclitaxel monotherapy-treated mice, indicating a prometastatic role of M2 macrophages in the IL1β-deprived microenvironment. Taken together, these findings demonstrate the dual effects of blocking the IL1 pathway on tumor growth. Accordingly, treatments using "add-on" drugs to conventional therapy should be investigated in appropriate tumor models consisting of primary tumors and their metastases., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
24. Thioredoxin-mimetic peptides as catalysts of S-denitrosylation and anti-nitrosative stress agents.
- Author
-
Kronenfeld G, Engelman R, Weisman-Shomer P, Atlas D, and Benhar M
- Subjects
- Catalysis, Cell Line, Humans, Molecular Weight, Thioredoxins chemistry, Molecular Mimicry, Nitrosation, Thioredoxins metabolism
- Abstract
S-nitrosylation, the coupling of a nitric oxide moiety to a reactive cysteine residue to form an S-nitrosothiol (SNO), is an important posttranslational mechanism for regulating protein activity. Growing evidence indicates that hyper-S-nitrosylation may contribute to cellular dysfunction associated with various human diseases. It is also increasingly appreciated that thioredoxin and thioredoxin reductase play significant roles in the cellular catabolism of SNO and protection from nitrosative stress. Here, we investigated the SNO reductase activity and protective effects of thioredoxin-mimetic peptides (TXMs), Ac-Cys-Pro-Cys-amide (CB3) and Ac-Cys-Gly-Pro-Cys-amide (CB4), both under cell-free conditions and in nitrosatively stressed cultured cells. In vitro biochemical analyses revealed that the TXM peptides reduced small-molecule SNO compounds, such as S-nitrosoglutathione (GSNO), and acted as general and efficient protein-denitrosylating agents. In particular, CB3 was found to be a highly potent SNO-metabolizing agent. Notably, CB3 mimicked the activity of thioredoxin by coupling with thioredoxin reductase to enhance GSNO reduction. Moreover, in a cell-free lysate system, both CB3 and CB4 synergized with an NADPH-dependent activity to denitrosylate proteins. Further investigation revealed that the TXM peptides protect the peroxiredoxin-thioredoxin system from SNO-dependent inhibition. Indeed, SNO-inhibited Prx1 was efficiently denitrosylated and reactivated by CB3 or CB4. In addition, CB3 protected thioredoxin reductase from SNO-mediated inactivation both in vitro and in intact cells. Finally, CB3 and CB4 partially rescued human neuroblastoma SH-SY5Y cells and rat insulinoma INS-1 832/13 cells from GSNO-induced growth inhibition. Collectively, the present findings indicate the efficient denitrosylation activity and protective effects of TXM peptides and suggest their potential therapeutic value in treating pathological conditions related to nitrosative stress., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. A substrate trapping approach identifies proteins regulated by reversible S-nitrosylation.
- Author
-
Ben-Lulu S, Ziv T, Admon A, Weisman-Shomer P, and Benhar M
- Subjects
- Animals, Cell Line, Cysteine analogs & derivatives, Cysteine pharmacology, HEK293 Cells, Humans, Interferon-gamma pharmacology, Macrophages cytology, Macrophages drug effects, Mice, Monocytes cytology, Monocytes drug effects, Mutation, Protein Processing, Post-Translational, Proteins chemistry, S-Nitrosothiols pharmacology, Signal Transduction drug effects, Thioredoxins genetics, Thioredoxins metabolism, Macrophages metabolism, Monocytes metabolism, Nitric Oxide pharmacology, Proteins metabolism, Proteomics methods
- Abstract
Protein S-nitrosylation, the nitric oxide-mediated posttranslational modification of cysteine residues, has emerged as an important regulatory mechanism in diverse cellular processes. Yet, knowledge about the S-nitrosoproteome in different cell types and cellular contexts is still limited and many questions remain regarding the precise roles of protein S-nitrosylation and denitrosylation. Here we present a novel strategy to identify reversibly nitrosylated proteins. Our approach is based on nitrosothiol capture and enrichment using a thioredoxin trap mutant, followed by protein identification by mass spectrometry. Employing this approach, we identified more than 400 putative nitroso-proteins in S-nitrosocysteine-treated human monocytes and about 200 nitrosylation substrates in endotoxin and cytokine-stimulated mouse macrophages. The large majority of these represent novel nitrosylation targets and they include many proteins with key functions in cellular homeostasis and signaling. Biochemical and functional experiments in vitro and in cells validated the proteomic results and further suggested a role for thioredoxin in the denitrosylation and activation of inducible nitric oxide synthase and the protein kinase MEK1. Our findings contribute to a better understanding of the macrophage S-nitrosoproteome and the role of thioredoxin-mediated denitrosylation in nitric oxide signaling. The approach described here may prove generally useful for the identification and exploration of nitroso-proteomes under various physiological and pathophysiological conditions., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
26. Suppression of the pro-inflammatory NLRP3/interleukin-1β pathway in macrophages by the thioredoxin reductase inhibitor auranofin.
- Author
-
Isakov E, Weisman-Shomer P, and Benhar M
- Subjects
- Animals, Cell Line, Gene Expression Regulation drug effects, Inflammasomes metabolism, Lipopolysaccharides pharmacology, Mice, NLR Family, Pyrin Domain-Containing 3 Protein, Thioredoxin-Disulfide Reductase metabolism, Toll-Like Receptor 4 agonists, Toll-Like Receptor 4 metabolism, Transcription, Genetic drug effects, Antirheumatic Agents pharmacology, Auranofin pharmacology, Carrier Proteins metabolism, Interleukin-1beta metabolism, Macrophages, Peritoneal metabolism, Signal Transduction drug effects, Thioredoxin-Disulfide Reductase antagonists & inhibitors
- Abstract
Background: The thioredoxin/thioredoxin reductase system, which is best known for its essential role in antioxidant defense and redox homeostasis, is increasingly implicated in the regulation of multiple cellular signaling pathways. In the present study, we asked if the thioredoxin system in macrophages might regulate toll-like receptor 4 (TLR4)-dependent gene expression and consequent responses., Methods: Using microarray analysis we analyzed the effect of auranofin, a highly potent and specific inhibitor of thioredoxin reductase, on the transcriptional program activated in J774 macrophages by the TLR4 agonist, lipopolysaccharide (LPS). We used quantitative real-time PCR (qPCR), Western blotting, ELISA and cytotoxicity assays to confirm and extend the microarray results., Results: Global transcriptional profiling revealed that macrophage treatment with auranofin exerted a selective effect on LPS-induced gene expression, suppressing the induction of a small number of genes. Interestingly, among these suppressed genes were three members of the interleukin-1 (IL-1) family of genes, among which IL-1β was most affected. qPCR analyses confirmed the repressive effects of auranofin on IL-1 genes. In addition, qPCR and Western blot analyses showed that auranofin impaired TLR4-dependent induction of the inflammasome receptor NLRP3, which plays a critical role in IL-1β processing. Consistent with these findings, inflammasome-dependent release of IL-1β from stimulated macrophages was suppressed by auranofin as was inflammasome-mediated cell death., Conclusions: Our findings suggest a regulatory role for the thioredoxin system in macrophage inflammatory signaling. Inhibition of the thioredoxin system in macrophages exerts an anti-inflammatory effect by repressing the activation of the NLRP3/IL-1β pathway., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
27. Proteomic identification of S-nitrosylated proteins in the parasite Entamoeba histolytica by resin-assisted capture: insights into the regulation of the Gal/GalNAc lectin by nitric oxide.
- Author
-
Hertz R, Ben Lulu S, Shahi P, Trebicz-Geffen M, Benhar M, and Ankri S
- Subjects
- Carbohydrates chemistry, Cell Adhesion, Chromatography, Affinity, Cysteine chemistry, Entamoebiasis immunology, Entamoebiasis parasitology, Glycolysis, HeLa Cells, Humans, Protein Transport, Proteome, Proteomics, Protozoan Proteins genetics, Cysteine analogs & derivatives, Entamoeba histolytica chemistry, Lectins chemistry, Nitric Oxide chemistry, Nitrogen chemistry, S-Nitrosothiols chemistry
- Abstract
Entamoeba histolytica is a gastrointestinal protozoan parasite that causes amebiasis, a disease which has a worldwide distribution with substantial morbidity and mortality. Nitrosative stress, which is generated by innate immune cells, is one of the various environmental challenges that E. histolytica encounters during its life cycle. Although the effects of nitric oxide (NO) on the regulation of gene expression in this parasite have been previously investigated, our knowledge on S-nitrosylated proteins in E.histolytica is lacking. In order to fill this knowledge gap, we performed a large-scale detection of S-nitrosylated (SNO) proteins in E.histolytica trophozoites that were treated with the NO donor, S-nitrosocysteine by resin-assisted capture (RAC). We found that proteins involved in glycolysis, gluconeogenesis, translation, protein transport, and adherence to target cells such as the heavy subunit of Gal/GalNac lectin are among the S-nitrosylated proteins that were enriched by SNO-RAC. We also found that the S-nitrosylated cysteine residues in the carbohydrate recognition domain (CRD) of Gal/GalNAc lectin impairs its function and contributes to the inhibition of E.histolytica adherence to host cells. Collectively, these results advance our understanding of the mechanism of reduced E.histolytica adherence to mammalian cells by NO and emphasize the importance of NO as a regulator of key physiological functions in E.histolytica.
- Published
- 2014
- Full Text
- View/download PDF
28. Multilevel regulation of 2-Cys peroxiredoxin reaction cycle by S-nitrosylation.
- Author
-
Engelman R, Weisman-Shomer P, Ziv T, Xu J, Arnér ES, and Benhar M
- Subjects
- Animals, Catalysis, Cysteine chemistry, Cysteine genetics, Cysteine metabolism, Disulfides chemistry, Disulfides metabolism, HeLa Cells, Homeodomain Proteins genetics, Humans, Mice, Nitric Oxide genetics, Oxidation-Reduction, Thioredoxin-Disulfide Reductase chemistry, Thioredoxin-Disulfide Reductase genetics, Thioredoxin-Disulfide Reductase metabolism, Homeodomain Proteins chemistry, Homeodomain Proteins metabolism, Nitric Oxide chemistry, Nitric Oxide metabolism
- Abstract
S-nitrosothiols (SNOs), formed by nitric oxide (NO)-mediated S-nitrosylation, and hydrogen peroxide (H2O2), a prominent reactive oxygen species, are implicated in diverse physiological and pathological processes. Recent research has shown that the cellular action and metabolism of SNOs and H2O2 involve overlapping, thiol-based mechanisms, but how these reactive species may affect each other's fate and function is not well understood. In this study we investigated how NO/SNO may affect the redox cycle of mammalian peroxiredoxin-1 (Prx1), a representative of the 2-Cys Prxs, a group of thioredoxin (Trx)-dependent peroxidases. We found that, both in a cell-free system and in cells, NO/SNO donors such as S-nitrosocysteine and S-nitrosoglutathione readily induced the S-nitrosylation of Prx1, causing structural and functional alterations. In particular, nitrosylation promoted disulfide formation involving the pair of catalytic cysteines (Cys-52 and Cys-173) and disrupted the oligomeric structure of Prx1, leading to loss of peroxidase activity. A highly potent inhibition of the peroxidase catalytic reaction by NO/SNO was seen in assays employing the coupled Prx-Trx system. In this setting, S-nitrosocysteine (10 μM) effectively blocked the Trx-mediated regeneration of oxidized Prx1. This effect appeared to be due to both competition between S-nitrosocysteine and Prx1 for the Trx system and direct modulation by S-nitrosocysteine of Trx reductase activity. Our findings that NO/SNO target both Prx and Trx reductase may have implications for understanding the impact of nitrosylation on cellular redox homeostasis.
- Published
- 2013
- Full Text
- View/download PDF
29. Thioredoxin-mimetic peptides (TXM) reverse auranofin induced apoptosis and restore insulin secretion in insulinoma cells.
- Author
-
Cohen-Kutner M, Khomsky L, Trus M, Aisner Y, Niv MY, Benhar M, and Atlas D
- Subjects
- Animals, Caspase 3 metabolism, Catalytic Domain, Cell Line, Tumor, Free Radicals metabolism, Insulin Secretion, Insulinoma, Janus Kinases metabolism, MAP Kinase Kinase Kinase 5 metabolism, Molecular Dynamics Simulation, Oligopeptides chemistry, Oxidative Stress, Peptidomimetics chemistry, Phosphorylation, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerases metabolism, Rats, Structure-Activity Relationship, Thioredoxin-Disulfide Reductase antagonists & inhibitors, Thioredoxins chemistry, p38 Mitogen-Activated Protein Kinases metabolism, Apoptosis drug effects, Auranofin pharmacology, Insulin metabolism, Oligopeptides pharmacology, Peptidomimetics pharmacology, Thioredoxins metabolism
- Abstract
The thioredoxin reductase/thioredoxin system (TrxR/Trx1) plays a major role in protecting cells from oxidative stress. Disruption of the TrxR-Trx1 system keeps Trx1 in the oxidized state leading to cell death through activation of the ASK1-Trx1 apoptotic pathway. The potential mechanism and ability of tri- and tetra-oligopeptides derived from the canonical -CxxC- motif of the Trx1-active site to mimic and enhance Trx1 cellular activity was examined. The Trx mimetics peptides (TXM) protected insulinoma INS 832/13 cells from oxidative stress induced by selectively inhibiting TrxR with auranofin (AuF). TXM reversed the AuF-effects preventing apoptosis, and increasing cell-viability. The TXM peptides were effective in inhibiting AuF-induced MAPK, JNK and p38(MAPK) phosphorylation, in correlation with preventing caspase-3 cleavage and thereby PARP-1 dissociation. The ability to form a disulfide-bridge-like conformation was estimated from molecular dynamics simulations. The TXM peptides restored insulin secretion and displayed Trx1 denitrosylase activity. Their potency was 10-100-fold higher than redox reagents like NAC, AD4, or ascorbic acid. Unable to reverse ERK1/2 phosphorylation, TXM-CB3 (NAc-Cys-Pro-Cys amide) appeared to function in part, through inhibiting ASK1-Trx dissociation. These highly effective anti-apoptotic effects of Trx1 mimetic peptides exhibited in INS 832/13 cells could become valuable in treating adverse oxidative-stress related disorders such as diabetes., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
30. Increased adipocyte S-nitrosylation targets anti-lipolytic action of insulin: relevance to adipose tissue dysfunction in obesity.
- Author
-
Ovadia H, Haim Y, Nov O, Almog O, Kovsan J, Bashan N, Benhar M, and Rudich A
- Subjects
- Animals, Cysteine chemistry, Female, Humans, Insulin Resistance physiology, Lipolysis, Male, Mice, Mice, Inbred C57BL, Mutagenesis, Site-Directed, Oxidative Stress, Phosphorylation, Adipocytes cytology, Adipose Tissue metabolism, Insulin metabolism, Nitrogen chemistry, Obesity metabolism
- Abstract
Protein S-nitrosylation is a reversible protein modification implicated in both physiological and pathophysiological regulation of protein function. In obesity, skeletal muscle insulin resistance is associated with increased S-nitrosylation of insulin-signaling proteins. However, whether adipose tissue is similarly affected in obesity and, if so, what are the causes and functional consequences of increased S-nitrosylation in this tissue are unknown. Total protein S-nitrosylation was increased in intra-abdominal adipose tissue of obese humans and in high fat-fed or leptin-deficient ob/ob mice. Both the insulin receptor β-subunit and Akt were S-nitrosylated, correlating with body weight. Elevated protein and mRNA expression of inducible NO synthase and decreased protein levels of thioredoxin reductase were associated with increased adipose tissue S-nitrosylation. Cultured differentiated pre-adipocyte cell lines exposed to the NO donors S-nitrosoglutathione (GSNO) or S-nitroso-N-acetylpenicillamine exhibited diminished insulin-stimulated phosphorylation of Akt but not of GSK3 nor of insulin-stimulated glucose uptake. Yet the anti-lipolytic action of insulin was markedly impaired in both cultured adipocytes and in mice injected with GSNO prior to administration of insulin. In cells, impaired ability of insulin to diminish phosphorylated PKA substrates in response to isoproterenol suggested impaired insulin-induced activation of PDE3B. Consistently, increased S-nitrosylation of PDE3B was detected in adipose tissue of high fat-fed obese mice. Site-directed mutagenesis revealed that Cys-768 and Cys-1040, two putative sites for S-nitrosylation adjacent to the substrate-binding site of PDE3B, accounted for ∼50% of its GSNO-induced S-nitrosylation. Collectively, PDE3B and the anti-lipolytic action of insulin may constitute novel targets for increased S-nitrosylation of adipose tissue in obesity.
- Published
- 2011
- Full Text
- View/download PDF
31. Analysis of protein S-nitrosylation.
- Author
-
Schonhoff CM and Benhar M
- Subjects
- Nitric Oxide metabolism, Proteins chemistry, S-Nitrosothiols metabolism, Biochemistry methods, Protein Processing, Post-Translational, Proteins metabolism, S-Nitrosothiols analysis
- Abstract
S-Nitrosylation, the redox-based modification of cysteine thiol side chains by nitric oxide, is a dynamic and reversible post-translational modification of proteins that subserves many important cellular functions. Analysis of protein S-nitrosylation is often challenging due to methodological limitations and the effects of various chemical and physical parameters. Despite these technical challenges, a growing number of useful methods are now available to analyze protein S-nitrosylation. In this unit, several important methods to measure protein S-nitrosylation and denitrosylation are discussed and evaluated. Recommendations are given regarding the potential and the applicability of the methods discussed.
- Published
- 2011
- Full Text
- View/download PDF
32. Identification of S-nitrosylated targets of thioredoxin using a quantitative proteomic approach.
- Author
-
Benhar M, Thompson JW, Moseley MA, and Stamler JS
- Subjects
- Amino Acids metabolism, Animals, Cell Line, Chromatography, Liquid, Humans, Isotope Labeling, Jurkat Cells, Mice, Models, Theoretical, S-Nitrosothiols metabolism, Tandem Mass Spectrometry, Proteomics, Thioredoxins metabolism
- Abstract
Reversible protein cysteine nitrosylation (S-nitrosylation) is a common mechanism utilized in signal transduction and other diverse cellular processes. Protein denitrosylation is largely mediated by cysteine denitrosylases, but the functional scope and significance of these enzymes are incompletely defined, in part due to limited information on their cognate substrates. Here, using Jurkat cells, we employed stable isotope labeling by amino acids in cell culture (SILAC), coupled to the biotin switch technique and mass spectrometry, to identify 46 new substrates of one denitrosylase, thioredoxin 1. These substrates are involved in a wide range of cellular functions including cytoskeletal organization, cellular metabolism, signal transduction, and redox homeostasis. We also identified multiple S-nitrosylated proteins that are not substrates of thioredoxin 1. A verification of our principal findings was made in a second cell type (RAW264.7 cells). Our results point to thioredoxin 1 as a major protein denitrosylase in mammalian cells and demonstrate the utility of quantitative proteomics for large-scale identification of denitrosylase substrates.
- Published
- 2010
- Full Text
- View/download PDF
33. Thioredoxin-interacting protein (Txnip) is a feedback regulator of S-nitrosylation.
- Author
-
Forrester MT, Seth D, Hausladen A, Eyler CE, Foster MW, Matsumoto A, Benhar M, Marshall HE, and Stamler JS
- Subjects
- Carrier Proteins genetics, Cell Line, Cell Survival physiology, Humans, Nitric Oxide genetics, Oxidation-Reduction, Thioredoxin-Disulfide Reductase antagonists & inhibitors, Thioredoxin-Disulfide Reductase genetics, Thioredoxin-Disulfide Reductase metabolism, Thioredoxins genetics, Thioredoxins metabolism, Carrier Proteins metabolism, Nitric Oxide metabolism, Oxidative Stress physiology
- Abstract
Nitric oxide exerts a plethora of biological effects via protein S-nitrosylation, a redox-based reaction that converts a protein Cys thiol to a S-nitrosothiol. However, although the regulation of protein S-nitrosylation has been the subject of extensive study, much less is known about the systems governing protein denitrosylation. Most recently, thioredoxin/thioredoxin reductases were shown to mediate both basal and stimulus-coupled protein denitrosylation. We now demonstrate that protein denitrosylation by thioredoxin is regulated dynamically by thioredoxin-interacting protein (Txnip), a thioredoxin inhibitor. Endogenously synthesized nitric oxide represses Txnip, thereby facilitating thioredoxin-mediated denitrosylation. Autoregulation of denitrosylation thus allows cells to survive nitrosative stress. Our findings reveal that denitrosylation of proteins is dynamically regulated, establish a physiological role for thioredoxin in protection from nitrosative stress, and suggest new approaches to manipulate cellular S-nitrosylation.
- Published
- 2009
- Full Text
- View/download PDF
34. Protein denitrosylation: enzymatic mechanisms and cellular functions.
- Author
-
Benhar M, Forrester MT, and Stamler JS
- Subjects
- Humans, Nitric Oxide metabolism, Nitrosation, Oxidation-Reduction, S-Nitrosoglutathione metabolism, S-Nitrosothiols metabolism, Signal Transduction, Aldehyde Oxidoreductases metabolism, Proteins metabolism, Thioredoxins metabolism
- Abstract
S-Nitrosylation, the redox-based modification of Cys thiol side chains by nitric oxide, is a common mechanism in signal transduction. Dysregulated S-nitrosylation contributes to a range of human pathologies. New roles for protein denitrosylation in regulating S-nitrosylation are being revealed. Recently, several denitrosylases - the enzymes that mediate Cys denitrosylation - have been discovered, of which two enzyme systems in particular, the S-nitrosoglutathione reductase and thioredoxin systems, have been shown to be physiologically relevant. These highly conserved enzymes regulate signalling through multiple classes of receptors and influence diverse cellular responses. In addition, they protect from nitrosative stress in microorganisms, mammals and plants, thereby exerting profound effects on host-microbe interactions and innate immunity.
- Published
- 2009
- Full Text
- View/download PDF
35. Detection of protein S-nitrosylation with the biotin-switch technique.
- Author
-
Forrester MT, Foster MW, Benhar M, and Stamler JS
- Subjects
- Animals, Biotin metabolism, Cell Extracts chemistry, Chemistry Techniques, Analytical instrumentation, Chemistry Techniques, Analytical trends, Cysteine chemistry, Cysteine metabolism, Humans, Oxidation-Reduction, Signal Transduction, Sulfhydryl Compounds chemistry, Biotin chemistry, Chemistry Techniques, Analytical methods, Nitric Oxide metabolism, Protein Processing, Post-Translational physiology, S-Nitrosothiols chemistry
- Abstract
Protein S-nitrosylation, the posttranslational modification of cysteine thiols to form S-nitrosothiols, is a principle mechanism of nitric oxide-based signaling. Studies have demonstrated myriad roles for S-nitrosylation in organisms from bacteria to humans, and recent efforts have greatly advanced our scientific understanding of how this redox-based modification is dynamically regulated during physiological and pathophysiological conditions. The focus of this review is the biotin-switch technique (BST), which has become a mainstay assay for detecting S-nitrosylated proteins in complex biological systems. Potential pitfalls and modern adaptations of the BST are discussed, as are future directions for this assay in the burgeoning field of protein S-nitrosylation.
- Published
- 2009
- Full Text
- View/download PDF
36. Regulated protein denitrosylation by cytosolic and mitochondrial thioredoxins.
- Author
-
Benhar M, Forrester MT, Hess DT, and Stamler JS
- Subjects
- Animals, Apoptosis, Auranofin pharmacology, Binding Sites, Caspase 3 metabolism, Caspase Inhibitors, Cell Line, Dinitrochlorobenzene pharmacology, HeLa Cells, Humans, Jurkat Cells, Macrophages metabolism, Mitochondria enzymology, Rats, Recombinant Proteins metabolism, T-Lymphocytes metabolism, fas Receptor metabolism, Cytosol metabolism, Mitochondria metabolism, Mitochondrial Proteins metabolism, Nitric Oxide metabolism, S-Nitrosothiols metabolism, Thioredoxin-Disulfide Reductase metabolism, Thioredoxins metabolism
- Abstract
Nitric oxide acts substantially in cellular signal transduction through stimulus-coupled S-nitrosylation of cysteine residues. The mechanisms that might subserve protein denitrosylation in cellular signaling remain uncharacterized. Our search for denitrosylase activities focused on caspase-3, an exemplar of stimulus-dependent denitrosylation, and identified thioredoxin and thioredoxin reductase in a biochemical screen. In resting human lymphocytes, thioredoxin-1 actively denitrosylated cytosolic caspase-3 and thereby maintained a low steady-state amount of S-nitrosylation. Upon stimulation of Fas, thioredoxin-2 mediated denitrosylation of mitochondria-associated caspase-3, a process required for caspase-3 activation, and promoted apoptosis. Inhibition of thioredoxin-thioredoxin reductases enabled identification of additional substrates subject to endogenous S-nitrosylation. Thus, specific enzymatic mechanisms may regulate basal and stimulus-induced denitrosylation in mammalian cells.
- Published
- 2008
- Full Text
- View/download PDF
37. Regulation of beta-adrenergic receptor signaling by S-nitrosylation of G-protein-coupled receptor kinase 2.
- Author
-
Whalen EJ, Foster MW, Matsumoto A, Ozawa K, Violin JD, Que LG, Nelson CD, Benhar M, Keys JR, Rockman HA, Koch WJ, Daaka Y, Lefkowitz RJ, and Stamler JS
- Subjects
- Animals, Arrestins metabolism, Cell Line, Cell Line, Tumor, Cysteine metabolism, G-Protein-Coupled Receptor Kinase 2, Homeostasis, Humans, Lung metabolism, Mice, Myocardium metabolism, Nitric Acid metabolism, Phosphorylation, Signal Transduction, beta-Adrenergic Receptor Kinases chemistry, beta-Arrestins, Receptors, Adrenergic, beta metabolism, S-Nitrosothiols metabolism, beta-Adrenergic Receptor Kinases metabolism
- Abstract
beta-adrenergic receptors (beta-ARs), prototypic G-protein-coupled receptors (GPCRs), play a critical role in regulating numerous physiological processes. The GPCR kinases (GRKs) curtail G-protein signaling and target receptors for internalization. Nitric oxide (NO) and/or S-nitrosothiols (SNOs) can prevent the loss of beta-AR signaling in vivo, but the molecular details are unknown. Here we show in mice that SNOs increase beta-AR expression and prevent agonist-stimulated receptor downregulation; and in cells, SNOs decrease GRK2-mediated beta-AR phosphorylation and subsequent recruitment of beta-arrestin to the receptor, resulting in the attenuation of receptor desensitization and internalization. In both cells and tissues, GRK2 is S-nitrosylated by SNOs as well as by NO synthases, and GRK2 S-nitrosylation increases following stimulation of multiple GPCRs with agonists. Cys340 of GRK2 is identified as a principal locus of inhibition by S-nitrosylation. Our studies thus reveal a central molecular mechanism through which GPCR signaling is regulated.
- Published
- 2007
- Full Text
- View/download PDF
38. Nitrosative stress in the ER: a new role for S-nitrosylation in neurodegenerative diseases.
- Author
-
Benhar M, Forrester MT, and Stamler JS
- Subjects
- Animals, Endoplasmic Reticulum chemistry, Humans, Nitrosation, Protein Folding, Endoplasmic Reticulum metabolism, Neurodegenerative Diseases metabolism, Nitric Oxide chemistry, Nitric Oxide physiology
- Abstract
S-Nitrosylation, the covalent addition of a nitrogen monoxide group to a cysteine thiol, has been shown to modify the function of a broad spectrum of mammalian, plant, and microbial proteins and thereby to convey the ubiquitous influence of nitric oxide on cellular signal transduction and host defense. Accumulating evidence indicates that dysregulated, diminished, or excessive S-nitrosylation may be implicated in a wide range of pathophysiological conditions. A recent study establishes a functional relationship between inhibitory S-nitrosylation of the redox enzyme protein disulfide isomerase (PDI), defects in regulation of protein folding within the endoplasmic reticulum (ER), and neurodegeneration. Further, an examination of human brains afflicted with Parkinson's or Alzheimer's disease supports a causal role for the S-nitrosylation of PDI and consequent ER stress in these prevalent neurodegenerative disorders.
- Published
- 2006
- Full Text
- View/download PDF
39. A central role for S-nitrosylation in apoptosis.
- Author
-
Benhar M and Stamler JS
- Subjects
- Animals, Cysteine metabolism, Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) metabolism, Humans, Macrophages metabolism, Macrophages physiology, Mice, Mice, Knockout, Models, Biological, Neurons metabolism, Neurons physiology, Nitric Oxide metabolism, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism, Nuclear Proteins metabolism, Ubiquitin-Protein Ligases, Seven in Absentia Proteins, Apoptosis physiology, S-Nitrosothiols metabolism, Signal Transduction physiology
- Published
- 2005
- Full Text
- View/download PDF
40. A low molecular weight copper chelator crosses the blood-brain barrier and attenuates experimental autoimmune encephalomyelitis.
- Author
-
Offen D, Gilgun-Sherki Y, Barhum Y, Benhar M, Grinberg L, Reich R, Melamed E, and Atlas D
- Subjects
- Acetylcysteine analogs & derivatives, Administration, Oral, Animals, Antimetabolites, Antineoplastic pharmacology, Biological Transport physiology, Blood-Brain Barrier metabolism, Cell Line, Cell Membrane Permeability, Disease Models, Animal, Drug Evaluation, Preclinical, Encephalomyelitis, Autoimmune, Experimental metabolism, Erythrocytes drug effects, Erythrocytes metabolism, Female, Humans, JNK Mitogen-Activated Protein Kinases, Matrix Metalloproteinase 9 metabolism, Mice, Mice, Inbred C3H, Mitogen-Activated Protein Kinases metabolism, Molecular Weight, Phosphorylation drug effects, Reactive Oxygen Species metabolism, Sulfhydryl Compounds metabolism, Treatment Outcome, p38 Mitogen-Activated Protein Kinases, Acetylcysteine metabolism, Acetylcysteine pharmacology, Chelating Agents metabolism, Chelating Agents pharmacology, Copper metabolism, Encephalomyelitis, Autoimmune, Experimental drug therapy
- Abstract
Increasing evidence suggests that enhanced production of reactive oxygen species (ROS) activates the MAP kinases, c-Jun N-terminal protein kinase (JNK) and mitogen-activated protein kinase MAPK (p38). These phosphorylated intermediates at the stress-activated pathway induce expression of matrix metalloproteinases (MMPs), leading to inflammatory responses and pathological damages involved in the etiology of multiple sclerosis (MS). Here we report that N-acetylcysteine amide (AD4) crosses the blood-brain barrier (BBB), chelates Cu(2+), which catalyzes free radical formation, and prevents ROS-induced activation of JNK, p38 and MMP-9. In the myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, oral administration of AD4 drastically reduced the clinical signs, inflammation, MMP-9 activity, and protected axons from demylination damages. In agreement with the in vitro studies, we propose that ROS scavenging by AD4 in MOG-treated animals prevented MMP's induction and subsequent damages through inhibition of MAPK pathway. The low toxicity of AD4 coupled with BBB penetration makes this compound an excellent potential candidate for the therapy of MS and other neurodegenerative disorders.
- Published
- 2004
- Full Text
- View/download PDF
41. Cisplatin-induced activation of the EGF receptor.
- Author
-
Benhar M, Engelberg D, and Levitzki A
- Subjects
- 3T3 Cells, Animals, Glioma metabolism, Glioma pathology, Humans, Ligands, Mice, Tumor Cells, Cultured, src-Family Kinases metabolism, Cisplatin pharmacology, ErbB Receptors metabolism
- Abstract
Cisplatin (CDDP) is an efficient DNA-damaging antitumor agent employed for the treatment of various human cancers. CDDP activates nuclear as well as cytoplasmatic signaling pathways involved in regulation of the cell cycle, damage repair and programmed cell death. Here we report that CDDP also activates a membrane-integrated protein, the epidermal growth factor receptor (EGFR). We show that EGFR is activated in response to CDDP in various types of cells that overexpress the receptor, including transformed human glioma cells and human breast tumor cells. CDDP-induced EGFR activation requires its kinase activity, as it can be blocked by an EGFR kinase inhibitor or by expression of a kinase dead receptor. We also show that CDDP-induced EGFR activation is independent of receptor ligand. CDDP induces the activation of c-Src, and EGFR activation is blocked by Src-family inhibitor PP1, suggesting that Src kinases mediate CDDP-induced EGFR activation. We propose that EGFR activation in response to CDDP is a survival response, since inhibition of EGFR activation enhances CDDP-induced death. These findings show that signals generated by DNA damage can modulate EGFR activity, and argue that interfering with CDDP-induced EGFR activation in tumor cells might be a useful approach to sensitize these cells to genotoxic agents.
- Published
- 2002
- Full Text
- View/download PDF
42. Toward a PKB inhibitor: modification of a selective PKA inhibitor by rational design.
- Author
-
Reuveni H, Livnah N, Geiger T, Klein S, Ohne O, Cohen I, Benhar M, Gellerman G, and Levitzki A
- Subjects
- 3T3 Cells, Adenosine Triphosphate chemistry, Animals, Apoptosis, Binding, Competitive, Calcium-Calmodulin-Dependent Protein Kinases antagonists & inhibitors, Calcium-Calmodulin-Dependent Protein Kinases chemistry, Cell Line, Combinatorial Chemistry Techniques methods, Cyclic AMP-Dependent Protein Kinases chemistry, Enzyme Inhibitors chemistry, Glycogen Synthase Kinase 3, Humans, Isoquinolines chemistry, Mice, Phosphorylation, Protein Serine-Threonine Kinases chemistry, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins c-akt, Structure-Activity Relationship, Substrate Specificity, Tumor Cells, Cultured enzymology, Tumor Cells, Cultured pathology, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Enzyme Inhibitors chemical synthesis, Isoquinolines chemical synthesis, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Sulfonamides
- Abstract
Protein kinase B/Akt (PKB) is an anti-apoptotic protein kinase that has strongly elevated activity in human malignancies. We therefore initiated a program to develop PKB inhibitors, "Aktstatins". We screened about 500 compounds for PKB inhibitors, using a radioactive assay and an ELISA assay that we established for this purpose. These compounds were produced as combinatorial libraries, designed using the structure of the selective PKA inhibitor H-89 as a starting point. We have identified a successful lead compound, which inhibits PKB activity in vitro and in cells overexpressing active PKB. The new compound shows reversed selectivity to H-89: In contrast to H-89, which inhibits PKA 70 times better than PKB, the new compound, NL-71-101, inhibits PKB 2.4-fold better than PKA. The new compound, but not H-89, induces apoptosis in tumor cells in which PKB is amplified. We have identified structural features in NL-71-101 that are significant for the specificity and that can be used for future development and optimization of PKB inhibitors.
- Published
- 2002
- Full Text
- View/download PDF
43. ROS, stress-activated kinases and stress signaling in cancer.
- Author
-
Benhar M, Engelberg D, and Levitzki A
- Subjects
- Animals, Apoptosis, Humans, Neoplasms enzymology, Oxidative Stress, Signal Transduction, Mitogen-Activated Protein Kinases metabolism, Neoplasms metabolism, Reactive Oxygen Species metabolism
- Abstract
Anticancer therapy is frequently efficient in early stages of the disease, whereas advanced tumors are usually resistant to the same treatments. The molecular basis for this change is not entirely understood. Many anticancer agents are DNA- or cytoskeleton-damaging drugs that show some specificity towards dividing cells. However, recent studies show that these agents also activate stress-signaling cascades that may play a role in eliciting the observed therapeutic effects. We discuss recent findings that suggest that induction of stress signaling in oncogenically transformed cells is integrated into apoptotic pathways. Reactive oxygen species (ROS) and stress-activated protein kinases (SAPKs), which are potentiated in recently transformed cells, emerge as key effectors of cell death. In advanced tumors, however, these agents are downregulated and, consequently, death signaling is suppressed. Such changes in ROS and SAPK activity levels during the course of tumor development may underlie the changes in responsiveness to anticancer therapy.
- Published
- 2002
- Full Text
- View/download PDF
44. Enhanced ROS production in oncogenically transformed cells potentiates c-Jun N-terminal kinase and p38 mitogen-activated protein kinase activation and sensitization to genotoxic stress.
- Author
-
Benhar M, Dalyot I, Engelberg D, and Levitzki A
- Subjects
- 3T3 Cells, Animals, Anisomycin pharmacology, Antineoplastic Agents pharmacology, Apoptosis, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Cell Line, Transformed, Cell Survival, Cisplatin pharmacology, Dose-Response Relationship, Drug, Enzyme Activation, ErbB Receptors metabolism, Flow Cytometry, Fluoresceins pharmacology, Fluorescent Dyes pharmacology, Humans, Immunoblotting, In Situ Nick-End Labeling, JNK Mitogen-Activated Protein Kinases, MAP Kinase Kinase 3, MAP Kinase Kinase 6, Mice, Mitogen-Activated Protein Kinase Kinases metabolism, Plasmids metabolism, Protein Synthesis Inhibitors pharmacology, Protein-Tyrosine Kinases metabolism, Stress, Physiological, Time Factors, Transfection, p38 Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinases metabolism, Reactive Oxygen Species
- Abstract
Many primary tumors as well as transformed cell lines display high sensitivity to chemotherapeutic drugs and radiation. The molecular mechanisms that underlie this sensitivity are largely unknown. Here we show that the sensitization of transformed cells to stress stimuli is due to the potentiation of the c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase pathways. Activation of these pathways by the antitumor drug cis-platin (CDDP) and by other stress agents is markedly enhanced and is induced by lower stress doses in NIH 3T3 cells overexpressing epidermal growth factor receptor, HER1-2 kinase, or oncogenic Ras than in nontransformed NIH 3T3 cells. Inhibition of stress kinase activity by specific inhibitors reduces CDDP-mediated cell death in transformed cells, whereas overactivation of stress kinase pathways augments cells death. Potentiation of stress kinases is a common feature of cells transformed by different oncogenes, including cells derived from human tumors, and is shown here to be independent of the activity of the particular transforming oncoprotein. We further show that the mechanism that underlies potentiation of stress kinases in transformed cells involves reactive oxygen species (ROS), whose production is elevated in these cells. JNK/p38 activation is inhibited by antioxidants and in particular by inhibitors of the mitochondrial respiratory chain and NADPH oxidase. Conversely, by artificially elevating ROS levels in nontransformed NIH 3T3 cells we were able to induce potentiation of JNK/p38 activation. Taken together, our findings suggest that ROS-dependent potentiation of stress kinase pathways accounts for the sensitization of transformed cells to stress and anticancer drugs.
- Published
- 2001
- Full Text
- View/download PDF
45. Differential expression pattern of Rab-GDI isoforms during the parotid gland secretion cycle.
- Author
-
Benhar M, Boschwitz H, and Linial M
- Subjects
- Adrenergic beta-Agonists pharmacology, Animals, Brain metabolism, Cloning, Molecular, Exocytosis, GTP-Binding Proteins genetics, Gene Expression, Isoproterenol pharmacology, Male, Periodicity, RNA, Messenger analysis, Rats, Subcellular Fractions chemistry, rab2 GTP-Binding Protein, rab4 GTP-Binding Proteins, Cell Degranulation physiology, GTP-Binding Proteins biosynthesis, Guanine Nucleotide Dissociation Inhibitors, Parotid Gland physiology
- Abstract
Rab GDP dissociation inhibitor (GDI) plays an important role in regulating the GDP/GTP cycle of small GTP binding proteins of the Rab family. It also regulates their association to membranes. The small family of Rab-GDI consists of several closely related isoforms, the functional differences between which are still unknown. Here we show that multiple GDI isoforms are expressed in rat parotid gland and that the individual GDI isoforms have a characteristic expression both at the RNA and at the protein level, during the parotid secretory cycle. GDIalpha, the major isoform in brain, is expressed throughout the secretory process and is equally distributed between cytoplasmic and membranous fractions. In contrast, an isoform related to, but different from GDIbeta is found predominantly in the cytoplasmic fraction and its expression is detected only after beta-adrenergic stimulation of the gland, at the end of the secretion phase, when exocytosis is already completed. The induction of such a GDI isoform at the beginning of the recovery stage correlates with the expression pattern of Rab1 and Rab5, but not Rab2 and Rab4. Our results suggest different functional roles for multiple GDI isoforms along the secretion and recovery phases in rat parotid gland.
- Published
- 1997
- Full Text
- View/download PDF
46. [Follow-up of patients undergoing surgery for aortic dissection: evaluation with transesophageal echocardiography].
- Author
-
Di Segni M, Minardi G, Pucci E, Boccardi L, Mamone P, Pucci A, Benhar M, D'Alessandro LC, and Giovannini E
- Subjects
- Adult, Aged, Aortic Dissection classification, Aortic Dissection diagnostic imaging, Aortic Aneurysm classification, Aortic Aneurysm diagnostic imaging, Female, Follow-Up Studies, Humans, Male, Middle Aged, Aortic Dissection surgery, Aortic Aneurysm surgery, Echocardiography methods
- Abstract
Background: Transesophageal echocardiography (TEE) is a useful means in the diagnosis of acute aortic dissection (AD), owing to its very high sensibility and specificity. In this study, TEE was performed to assess post-surgical evolution., Patients: Between 1982 and 1991, 119 pts. were operated on in our institution for AD (De Bakey I and II type): 87 pts. underwent replacement of the ascending aorta with a composite tubular graft bearing a mechanical valve; 26 had a simple tubular graft and 6 had aortic reconstruction. Sixty-eight of 72 discharged pts. were followed for up to 9.5 years (mean 4.5 +/- 2.6). Nine years after surgery actuarial survival of discharged pts. was 75%. Seven pts. died after a mean period of 3.4 years from surgery: only one died from postoperative complication (dehiscence of proximal anastomosis), none for aortic rupture distal to the graft. TEE was performed in 32 of these pts. and in other two operated on elsewhere, after 4.4 +/- 2.7 years from surgery; before the operation, type I AD was diagnosed in 23 pts. and type II in 11 pts., Results: In 10/11 pts. with type II AD the aortic arch and the descending aorta looked normal; in one patient a localized intimal flap was found up to the arch. The descending aorta diameter was somewhat higher than in normal subjects (25.2 +/- 2.8 vs 21.9 +/- 3.7 mm), but in only one case was it beyond 2DS (32 mm). In all type I pts. an intimal flap persisted distal to the graft, along the whole thoracic aorta. Within the false lumen a flow was detected by color-Doppler in 14/23 pts. (61%), and spontaneous echo-contrast was noted in 14 pts. (61%). A thrombus was observed in 7 pts. (30%) and it was generally localized; in only one case it was extensive with total obliteration of the false lumen. In 16 pts. (70%) communications between the two lumina were found. The descending aorta diameter ranged from 25 to 53 mm, and mean value was higher than in normal subjects (34.2 +/- 6.2 vs 21.9 +/- 3.7 mm)., Conclusions: In most pts. with type II AD, surgery can be a definitive treatment, as the remaining aorta keeps to normal size and appearance. In type I AD, operation is only palliative, as the dissection persists: the false lumen is often perfused through one or more communications with the true lumen and seldom its obliteration is noted. The persistence of dissection does not necessarily seem to be an ominous finding, as the survival of the study population was high and no patient died from aortic rupture. Nevertheless, long-term prognosis can be affected by aorta dilation that often (but not always) follows the persistence of wall dissection. For its high reliability, easy feasibility and low cost TEE is a very useful method for following up patients operated on for AD and for detecting those who are at higher risk of aortic rupture because of lumen dilation.
- Published
- 1992
47. [Creatine phosphate: an additive to cardioplegic solutions. Clinical study].
- Author
-
D'Alessandro LC, Cini R, Stazi G, Pucci A, De Benedictis F, Rabitti G, Benhar M, Giacopino F, Pezza E, and Cioli G
- Subjects
- Adenosine Triphosphate metabolism, Heart Diseases surgery, Humans, Myocardial Contraction, Random Allocation, Ventricular Fibrillation drug therapy, Heart drug effects, Heart Arrest, Induced, Myocardium metabolism, Phosphocreatine pharmacology
- Published
- 1987
48. [General anesthesia in patients with intraventricular conduction abnormalities (author's transl)].
- Author
-
Santini M, Carrara P, Benhar M, Lazzari G, Rocchi M, Dini P, Di Mascolo R, Alliegro A, Messina G, and Masini V
- Subjects
- Enflurane pharmacology, Halothane pharmacology, Heart Block chemically induced, Heart Conduction System drug effects, Humans, Succinylcholine adverse effects, Succinylcholine pharmacology, Zygomatic Fractures surgery, Anesthesia, General adverse effects, Bundle-Branch Block complications
- Abstract
This study was undertaken to assess the risk of developing complete heart block during general anesthesia in patients with bundle branch block. His bundle electrograms were recorded during sinus rhythm and after atrail pacing in 11 patients (5 LBBB, 2 RBBB, 3 RBBB and LAH, 1 RBBB and LBH) before and after administration of Pentothal 0.20 g e.v., Succinylcholine 1 mg/kg e.v., and breathing of fluothane 1%or Ethrane. Minimal effects on sinus functions and A-V node conduction was observed during anesthesia; Fluothane only increased slightly AH intervals (+11%). Both Fluothane and Ethrane effects on HV conduction was insignificant. In 9 patients HV intervals increased of 5% after Succinylcholine; 2 patients developed a complete heart block distal to his after the drug. Possible causes of the complete heart block are discussed and a direct effect of Succinylcholine is hypothesized.
- Published
- 1977
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.