1
|
Grzelakowska A, Podsiadły R, Zielonka J. Phenyl Radical-Mediated Fluorogenic Cyclization for Specific Detection of Peroxynitrite. Anal Chem 2025. [PMID: 40146989 DOI: 10.1021/acs.analchem.4c06983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Peroxynitrite (ONOO-), a biological oxidizing and nitrating species responsible for post-translational modification of cellular proteins, has been implicated in numerous pathologies carrying an inflammatory component. Specific detection of ONOO- in biological systems remains a challenge, and boronates are regarded as the most promising class of probes for the detection and quantitation of ONOO-. Oxidation of boronate probes by ONOO- results in the formation of minor ONOO--specific products via a pathway involving a phenyl radical-type intermediate, in addition to the major phenolic product. Here, we report fluorogenic cyclization of the phenyl-type radical formed during oxidation of a boronate probe by ONOO-, with the production of a fluorescent product, and we propose a new approach for the specific detection of ONOO- based on this observation. We characterized the kinetics and stoichiometry of the reaction of benzophenone-2-boronic acid with ONOO- and identified 2-hydroxybenzophenone as the major product and fluorenone (FLN) and 2-nitrobenzophenone as the minor ONOO--specific products. Hydrogen peroxide neither alone nor in the presence of myeloperoxidase and nitrite produces FLN or 2-nitrobenzophenone. FLN can be selectively detected using fluorescence spectroscopy, providing a chemical principle for the development of next-generation probes for ONOO-, with noninvasive, fluorescence-based detection of ONOO--specific products. Fluorescence-based monitoring of FLN was successfully applied for the detection of ONOO- generated in situ from the decomposition of SIN-1, a thermal source of the superoxide radical anion and nitric oxide.
Collapse
Affiliation(s)
- Aleksandra Grzelakowska
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, Lodz 90-537, Poland
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Radosław Podsiadły
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, Lodz 90-537, Poland
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| |
Collapse
|
2
|
Kumar Rai R, Islam A, Shankar Pati R, Roy G. Cleavage of a Peroxide Bond via a Dual Attack by Functional Mimics of Glutathione Peroxidase. Chemistry 2025; 31:e202403483. [PMID: 39417606 DOI: 10.1002/chem.202403483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024]
Abstract
Nonmetal-containing peroxidase enzymes, including glutathione peroxidase (GPx), and peroxiredoxins, control cellular redox levels by catalyzing the reduction of H2O2. The remarkably higher reactivity of GPx enzyme as compared to the fully dissociated synthetic selenolate/thiolate molecule is probably due to the dual-attack on the peroxide bond (HO1-O2H) by the enzyme; The first one is a nucleophilic attack of the selenolate/thiolate moiety to O1 atom and the second attack at the O2 atom of the peroxide bond by the acidic "parked proton" from Trp or His residue present at the enzyme's active site, leading to the facile cleavage of O-O bond. Herein, we report two synthetic compounds (1 and 2), having a selenolate (Se-) and a proton donor (imidazolium or -COOH group) moieties, which showed excellent GPx-like activity via dual-attack on the peroxide bond. The combined effect of selenolate moiety that donates electrons to the antibonding (σ*) orbital of O1-O2 bond and the imidazolium or carboxylic acid moiety at the side chain that forms a strong H-bonding with the O2 atom facilitates O-O bond cleavage of H2O2 more efficiently. 1 and 2 exhibit remarkable ability in protecting Cu(I)-complex [TpmCu(CH3CN)]+ (9) against H2O2 by acting as a sacrificial antioxidant, thereby preventing metal-mediated ROS production.
Collapse
Affiliation(s)
- Rakesh Kumar Rai
- Department of Chemistry, Indian Institute of Technology Tirupati, Tirupati, A.P., 517619, India
| | - Amirul Islam
- Department of Chemistry, Indian Institute of Technology Tirupati, Tirupati, A.P., 517619, India
| | - Rudra Shankar Pati
- Department of Chemistry, Indian Institute of Technology Tirupati, Tirupati, A.P., 517619, India
| | - Gouriprasanna Roy
- Department of Chemistry, Indian Institute of Technology Tirupati, Tirupati, A.P., 517619, India
| |
Collapse
|
3
|
Kitajima N, Makihara K, Kurita H. On the Synergistic Effects of Cold Atmospheric Pressure Plasma Irradiation and Electroporation on Cytotoxicity of HeLa Cells. Int J Mol Sci 2025; 26:1093. [PMID: 39940861 PMCID: PMC11818767 DOI: 10.3390/ijms26031093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Cold atmospheric plasma (CAP) treatment induces cancer cell death through the generation of reactive oxygen and nitrogen species (RONS). However, the efficacy of RONS delivery into cells remains limited by membrane permeability. Here, we investigated whether combining CAP with pulsed electric fields (PEFs) could enhance cancer cell death through increased intracellular RONS uptake. HeLa cells were treated with argon atmospheric pressure plasma jet (Ar-APPJ), PEF, or their combination. The combined treatment showed significantly enhanced cell death compared to single treatments. While PEF treatment alone induced membrane permeabilization, the combination with Ar-APPJ resulted in more pronounced and sustained membrane disruption, as evidenced by increased calcein leakage. This enhanced effect was attributed to Ar-APPJ-induced lipid peroxidation interfering with membrane resealing after PEF-induced electroporation. We also demonstrated that PEF-induced membrane electroporation facilitates the intracellular uptake of CAP-generated RONS. These findings provide mechanistic insights into the synergistic effects of combined CAP and PEF treatments, suggesting enhanced cell death via multiple pathways.
Collapse
Affiliation(s)
| | | | - Hirofumi Kurita
- Department of Applied Chemistry and Life Science, Toyohashi University of Technology, Toyohashi 441-8580, Aichi, Japan
| |
Collapse
|
4
|
Babajani A, Eftekharinasab A, Bekeschus S, Mehdian H, Vakhshiteh F, Madjd Z. Reactive oxygen species from non-thermal gas plasma (CAP): implication for targeting cancer stem cells. Cancer Cell Int 2024; 24:344. [PMID: 39438918 PMCID: PMC11515683 DOI: 10.1186/s12935-024-03523-x] [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: 03/28/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
Cancer remains a major global health challenge, with the persistence of cancer stem cells (CSCs) contributing to treatment resistance and relapse. Despite advancements in cancer therapy, targeting CSCs presents a significant hurdle. Non-thermal gas plasma, also known as CAP, represents an innovative cancer treatment. It has recently gained attention for its often found to be selective, immunogenic, and potent anti-cancer properties. CAP is composed of a collection of transient, high-energy, and physically and chemically active entities, such as reactive oxygen species (ROS). It is acknowledged that the latter are responsible for a major portion of biomedical CAP effects. The dynamic interplay of CAP-derived ROS and other components contributes to the unique and versatile properties of CAP, enabling it to interact with biological systems and elicit various therapeutic effects, including its potential in cancer treatment. While CAP has shown promise in various cancer types, its application against CSCs is relatively unexplored. This review assesses the potential of CAP as a therapeutic strategy for targeting CSCs, focusing on its ability to regulate cellular states and achieve redox homeostasis. This is done by providing an overview of CSC characteristics and demonstrating recent findings on CAP's efficacy in targeting these cells. By contributing insights into the unique attributes of CSCs and the potential of CAP, this work contributes to an advanced understanding of innovative oncology strategies.
Collapse
Affiliation(s)
- Amirhesam Babajani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Hassan Mehdian
- Plasma Medicine Group, Plasma Research Institute, Kharazmi University, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
5
|
Dumitrel SI, Matichescu A, Dinu S, Buzatu R, Popovici R, Dinu DC, Bratu DC. New Insights Regarding the Use of Relevant Synthetic Compounds in Dentistry. Molecules 2024; 29:3802. [PMID: 39202881 PMCID: PMC11357206 DOI: 10.3390/molecules29163802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Worldwide, synthetic compounds are used for both in-office and at-home dental care. They are a valuable resource for both prophylactic and curative treatments for various dental problems, such as tooth decay, periodontal diseases, and many more. They are typically preferred due to their broad range of actions and ability to produce targeted, rapid, and long-lasting effects. Using a 0.12% chlorhexidine mouthwash is capable of reducing the plaque index from 47.69% to 2.37% and the bleeding index from 32.93% to 6.28% after just 2 weeks. Mouthwash with 0.1% OCT is also highly effective, as it significantly lowered the median plaque index and salivary bacterial counts in 152 patients in 5 days compared to a control group (p < 0.0001), while also reducing the gingival index (p < 0.001). When povidone-iodine was used as an irrigant during the surgical removal of mandibular third molars in 105 patients, it resulted in notably lower pain scores after 2 days compared to a control group (4.57 ± 0.60 vs. 5.71 ± 0.45). Sodium hypochlorite is excellent for root canal disinfection, as irrigating with 1% NaOCl completely eliminated the bacteria from canals in 65% patients. A 0.05% CPC mouthwash proved effective for perioperative patient care, significantly decreasing gingival bleeding (p < 0.001) and suppressing Streptococcus levels even one week post-surgery. Lastly, a 6% H2O2 paint-on varnish and 6% H2O2 tray formulations successfully bleached the teeth of 40 patients, maintaining a noticeably whiter appearance up to the 6-month follow-up, with significant color differences from the baseline (p < 0.005). Synthetic compounds have a large research base, which also provides a greater awareness of their mechanism of action and potential adverse effects. For a better understanding of how they work, several methods and assays are performed. These are protocolary techniques through which a compound's efficacy and toxicity are established.
Collapse
Affiliation(s)
- Stefania-Irina Dumitrel
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Anamaria Matichescu
- Department of Preventive, Community Dentistry and Oral Health, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 14A Tudor Vladimirescu Ave., 300173 Timisoara, Romania
- Translational and Experimental Clinical Research Centre in Oral Health, Victor Babes University of Medicine and Pharmacy, 14A Tudor Vladimirescu Ave., 300173 Timisoara, Romania
| | - Stefania Dinu
- Department of Pedodontics, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 9 No., Revolutiei 1989 Bv., 300041 Timisoara, Romania;
- Pediatric Dentistry Research Center, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 9 No., Revolutiei 1989 Bv., 300041 Timisoara, Romania
| | - Roxana Buzatu
- Department of Dental Aesthetics, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 9 No., Revolutiei 1989 Bv., 300041 Timisoara, Romania;
| | - Ramona Popovici
- Department of Management, Legislation and Communication in Dentistry, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 9 No., Revolutiei 1989 Bv., 300041 Timisoara, Romania;
| | - Dorin Cristian Dinu
- Family Dental Clinic, Private Practice, 24 Budapesta Street, 307160 Dumbravita, Romania;
| | - Dana Cristina Bratu
- Department of Orthodontics II, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy Timisoara, 9 No., Revolutiei 1989 Bv., 300041 Timisoara, Romania;
| |
Collapse
|
6
|
Li J, Lim JYS, Eu JQ, Chan AKMH, Goh BC, Wang L, Wong ALA. Reactive Oxygen Species Modulation in the Current Landscape of Anticancer Therapies. Antioxid Redox Signal 2024; 41:322-341. [PMID: 38445392 DOI: 10.1089/ars.2023.0445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Significance: Reactive oxygen species (ROS) are generated during mitochondrial oxidative metabolism, and are tightly controlled through homeostatic mechanisms to maintain intracellular redox, regulating growth and proliferation in healthy cells. However, ROS production is perturbed in cancers where abnormal accumulation of ROS leads to oxidative stress and genomic instability, triggering oncogenic signaling pathways on one hand, while increasing oxidative damage and triggering ROS-dependent death signaling on the other. Recent Advances: Our review illuminates how critical interactions between ROS and oncogenic signaling, the tumor microenvironment, and DNA damage response (DDR) pathways have led to interest in ROS modulation as a means of enhancing existing anticancer strategies and developing new therapeutic opportunities. Critical Issues: ROS equilibrium exists via a delicate balance of pro-oxidant and antioxidant species within cells. "Antioxidant" approaches have been explored mainly in the form of chemoprevention, but there is insufficient evidence to advocate its routine application. More progress has been made via the "pro-oxidant" approach of targeting cancer vulnerabilities and inducing oxidative stress. Various therapeutic modalities have employed this approach, including direct ROS-inducing agents, chemotherapy, targeted therapies, DDR therapies, radiotherapy, and immunotherapy. Finally, emerging delivery systems such as "nanosensitizers" as radiotherapy enhancers are currently in development. Future Directions: While approaches designed to induce ROS have shown considerable promise in selectively targeting cancer cells and dealing with resistance to conventional therapies, most are still in early phases of development and challenges remain. Further research should endeavor to refine treatment strategies, optimize drug combinations, and identify predictive biomarkers of ROS-based cancer therapies.
Collapse
Affiliation(s)
- Jiaqi Li
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Jie Qing Eu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| |
Collapse
|
7
|
Pecchillo Cimmino T, Punziano C, Panico I, Petrone Z, Cassese M, Faraonio R, Barresi V, Esposito G, Ammendola R, Cattaneo F. Formyl-Peptide Receptor 2 Signaling Modulates SLC7A11/xCT Expression and Activity in Tumor Cells. Antioxidants (Basel) 2024; 13:552. [PMID: 38790657 PMCID: PMC11118824 DOI: 10.3390/antiox13050552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer cells exhibit high levels of oxidative stress and consequently require a high amount of cysteine for glutathione synthesis. Solute Carrier Family 7 Member 11 (SLC7A11), or xCT, mediates the cellular uptake of cystine in exchange for intracellular glutamate; imported extracellular cystine is reduced to cysteine in the cytosol through a NADPH-consuming reduction reaction. SLC7A11/xCT expression is under the control of stress-inducing conditions and of several transcription factors, such as NRF2 and ATF4. Formyl-peptide receptor 2 (FPR2) belongs to the FPR family, which transduces chemotactic signals mediating either inflammatory or anti-inflammatory responses according to the nature of its ligands and/or FPR2 binding with other FPR isoforms. The repertoire of FPR2 agonists with anti-inflammatory activities comprises WKYMVm peptide and Annexin A1 (ANXA1), and the downstream effects of the intracellular signaling cascades triggered by FPR2 include NADPH oxidase (NOX)-dependent generation of reactive oxygen species. Herein, we demonstrate that stimulation of CaLu-6 cells with either WKYMVm or ANXA1: (i) induces the redox-regulated activation of SLC7A11/xCT; (ii) promotes the synthesis of glutathione; (iii) prevents lipid peroxidation; and (iv) favors NRF2 nuclear translocation and activation. In conclusion, our overall results demonstrate that FPR2 agonists and NOX modulate SLC7A11/xCT expression and activity, thereby identifying a novel regulative pathway of the cystine/glutamate antiport that represents a new potential therapeutical target for the treatment of human cancers.
Collapse
Affiliation(s)
- Tiziana Pecchillo Cimmino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Carolina Punziano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Iolanda Panico
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Zeudi Petrone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Myrhiam Cassese
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Raffaella Faraonio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Vincenza Barresi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (T.P.C.); (C.P.); (I.P.); (Z.P.); (M.C.); (R.F.); (G.E.); (R.A.)
| |
Collapse
|
8
|
Berndt A, Lee J, Won W, Kimball K, Neiswanger C, Schattauer S, Wang Y, Yeboah F, Ruiz M, Evitts K, Rappleye M, Bremner S, Chun C, Smith N, Mack D, Young J, Lee CJ, Chavkin C. Ultra-fast genetically encoded sensor for precise real-time monitoring of physiological and pathophysiological peroxide dynamics. RESEARCH SQUARE 2024:rs.3.rs-4048855. [PMID: 38585715 PMCID: PMC10996778 DOI: 10.21203/rs.3.rs-4048855/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Hydrogen Peroxide (H2O2) is a central oxidant in redox biology due to its pleiotropic role in physiology and pathology. However, real-time monitoring of H2O2 in living cells and tissues remains a challenge. We address this gap with the development of an optogenetic hydRogen perOxide Sensor (oROS), leveraging the bacterial peroxide binding domain OxyR. Previously engineered OxyR-based fluorescent peroxide sensors lack the necessary sensitivity and response speed for effective real-time monitoring. By structurally redesigning the fusion of Escherichia coli (E. coli) ecOxyR with a circularly permutated green fluorescent protein (cpGFP), we created a novel, green-fluorescent peroxide sensor oROS-G. oROS-G exhibits high sensitivity and fast on-and-off kinetics, ideal for monitoring intracellular H2O2 dynamics. We successfully tracked real-time transient and steady-state H2O2 levels in diverse biological systems, including human stem cell-derived neurons and cardiomyocytes, primary neurons and astrocytes, and mouse brain ex vivo and in vivo. These applications demonstrate oROS's capabilities to monitor H2O2 as a secondary response to pharmacologically induced oxidative stress and when adapting to varying metabolic stress. We showcased the increased oxidative stress in astrocytes via Aβ-putriscine-MAOB axis, highlighting the sensor's relevance in validating neurodegenerative disease models. Lastly, we demonstrated acute opioid-induced generation of H2O2 signal in vivo which highlights redox-based mechanisms of GPCR regulation. oROS is a versatile tool, offering a window into the dynamic landscape of H2O2 signaling. This advancement paves the way for a deeper understanding of redox physiology, with significant implications for understanding diseases associated with oxidative stress, such as cancer, neurodegenerative, and cardiovascular diseases.
Collapse
|
9
|
Jain N, Sonawane PM, Roychaudhury A, Park SJ, An J, Kim CH, Nimse SB, Churchill DG. An indole-based near-infrared fluorescent "Turn-On" probe for H 2O 2: Selective detection and ultrasensitive imaging of zebrafish gallbladder. Talanta 2024; 269:125459. [PMID: 38011812 DOI: 10.1016/j.talanta.2023.125459] [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: 07/06/2023] [Revised: 09/25/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Fluorescent probes play essential roles in medical imaging, where the researchers can select one of many molecules to use to help monitor the status of living systems under investigation. To date, a few scaffolds that allow the in vivo detection of H2O2 are available only. Herein, we provide a highly sensitive and selective near-infrared fluorescent probe that detects H2O2 based on the ICT sensing mechanism. We report the first indole-incorporated fluorescent probe Indo-H2O2 that allows H2O2 detection with a LOD of 25.2 nM featuring a boronate group conjugated to an indole scaffold; the boronate cleaves upon reaction with H2O2. A 5-membered malononitrile derivative was incorporated; Indo-H2O2 has near-infrared (NIR) properties and the reaction time is low (∼25 min) compared to other related probes. Indo-H2O2 was successfully employed in both endogenous and exogenous imaging trials of H2O2 in living cells. Indo-H2O2 also allows the real-time monitoring of H2O2in vivo. It preferentially accesses the gallbladder of zebrafish. Our findings support Indo-H2O2 as a highly sensitive fluorescent NIR probe for detecting H2O2, and an idea to incorporate a central indole unit in future fluorescent probe designs.
Collapse
Affiliation(s)
- Neha Jain
- Department of Chemistry, Molecular Logic Gate Laboratory, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Prasad M Sonawane
- Department of Chemistry, Molecular Logic Gate Laboratory, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | | | - Su Jeong Park
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Jongkeol An
- Department of Chemistry, Molecular Logic Gate Laboratory, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Satish Balasaheb Nimse
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon, 24252, Republic of Korea.
| | - David G Churchill
- Department of Chemistry, Molecular Logic Gate Laboratory, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; KAIST Institute for Health Science and Technology (KIHST) (Therapeutic Bioengineering Section), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
10
|
Lee JD, Won W, Kimball K, Wang Y, Yeboah F, Evitts KM, Neiswanger C, Schattauer S, Rappleye M, Bremner SB, Chun C, Smith N, Mack DL, Young JE, Lee CJ, Chavkin C, Berndt A. Structure-guided engineering of a fast genetically encoded sensor for real-time H 2O 2 monitoring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578117. [PMID: 38352381 PMCID: PMC10862829 DOI: 10.1101/2024.01.31.578117] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Hydrogen Peroxide (H2O2) is a central oxidant in redox biology due to its pleiotropic role in physiology and pathology. However, real-time monitoring of H2O2 in living cells and tissues remains a challenge. We address this gap with the development of an optogenetic hydRogen perOxide Sensor (oROS), leveraging the bacterial peroxide binding domain OxyR. Previously engineered OxyR-based fluorescent peroxide sensors lack the necessary sensitivity or response speed for effective real-time monitoring. By structurally redesigning the fusion of Escherichia coli (E. coli) ecOxyR with a circularly permutated green fluorescent protein (cpGFP), we created a novel, green-fluorescent peroxide sensor oROS-G. oROS-G exhibits high sensitivity and fast on-and-off kinetics, ideal for monitoring intracellular H2O2 dynamics. We successfully tracked real-time transient and steady-state H2O2 levels in diverse biological systems, including human stem cell-derived neurons and cardiomyocytes, primary neurons and astrocytes, and mouse neurons and astrocytes in ex vivo brain slices. These applications demonstrate oROS's capabilities to monitor H2O2 as a secondary response to pharmacologically induced oxidative stress, G-protein coupled receptor (GPCR)-induced cell signaling, and when adapting to varying metabolic stress. We showcased the increased oxidative stress in astrocytes via Aβ-putriscine-MAOB axis, highlighting the sensor's relevance in validating neurodegenerative disease models. oROS is a versatile tool, offering a window into the dynamic landscape of H2O2 signaling. This advancement paves the way for a deeper understanding of redox physiology, with significant implications for diseases associated with oxidative stress, such as cancer, neurodegenerative disorders, and cardiovascular diseases.
Collapse
Affiliation(s)
- Justin Daho Lee
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Kandace Kimball
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Yihan Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Fred Yeboah
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kira M Evitts
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Carlie Neiswanger
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Selena Schattauer
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Michael Rappleye
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Samantha B Bremner
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Changho Chun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Netta Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - David L Mack
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Jessica E Young
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Charles Chavkin
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Andre Berndt
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Neuroscience of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| |
Collapse
|
11
|
Sørensen M, Pershagen G, Thacher JD, Lanki T, Wicki B, Röösli M, Vienneau D, Cantuaria ML, Schmidt JH, Aasvang GM, Al-Kindi S, Osborne MT, Wenzel P, Sastre J, Fleming I, Schulz R, Hahad O, Kuntic M, Zielonka J, Sies H, Grune T, Frenis K, Münzel T, Daiber A. Health position paper and redox perspectives - Disease burden by transportation noise. Redox Biol 2024; 69:102995. [PMID: 38142584 PMCID: PMC10788624 DOI: 10.1016/j.redox.2023.102995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023] Open
Abstract
Transportation noise is a ubiquitous urban exposure. In 2018, the World Health Organization concluded that chronic exposure to road traffic noise is a risk factor for ischemic heart disease. In contrast, they concluded that the quality of evidence for a link to other diseases was very low to moderate. Since then, several studies on the impact of noise on various diseases have been published. Also, studies investigating the mechanistic pathways underlying noise-induced health effects are emerging. We review the current evidence regarding effects of noise on health and the related disease-mechanisms. Several high-quality cohort studies consistently found road traffic noise to be associated with a higher risk of ischemic heart disease, heart failure, diabetes, and all-cause mortality. Furthermore, recent studies have indicated that road traffic and railway noise may increase the risk of diseases not commonly investigated in an environmental noise context, including breast cancer, dementia, and tinnitus. The harmful effects of noise are related to activation of a physiological stress response and nighttime sleep disturbance. Oxidative stress and inflammation downstream of stress hormone signaling and dysregulated circadian rhythms are identified as major disease-relevant pathomechanistic drivers. We discuss the role of reactive oxygen species and present results from antioxidant interventions. Lastly, we provide an overview of oxidative stress markers and adverse redox processes reported for noise-exposed animals and humans. This position paper summarizes all available epidemiological, clinical, and preclinical evidence of transportation noise as an important environmental risk factor for public health and discusses its implications on the population level.
Collapse
Affiliation(s)
- Mette Sørensen
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Denmark.
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jesse Daniel Thacher
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Timo Lanki
- Department of Health Security, Finnish Institute for Health and Welfare, Kuopio, Finland; School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Benedikt Wicki
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Danielle Vienneau
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Manuella Lech Cantuaria
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Jesper Hvass Schmidt
- Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Gunn Marit Aasvang
- Department of Air Quality and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Sadeer Al-Kindi
- Department of Medicine, University Hospitals, Harrington Heart & Vascular Institute, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Michael T Osborne
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Wenzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt Am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35392, Gießen, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Katie Frenis
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
12
|
Xu H, Zeng Q, Zou K, Huang H, Chen J, Wang P, Yuan W, Xiao L, Tong P, Jin H. Glucocorticoid-induced activation of NOX/ROS/NF-κB signaling in MSCs contributes to the development of GONFH. Apoptosis 2023; 28:1332-1345. [PMID: 37306805 PMCID: PMC10258081 DOI: 10.1007/s10495-023-01860-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND This study aimed to investigate the pathogenic factors of glucocorticoids (GCs)-induced osteonecrosis of the femoral head (GONFH) and its underlying pathogenesis in vivo and in vitro. METHODS Radiographical (µCT) scanning, histopathological, immunohistochemical, reactive oxygen species (ROS) and tunel staining were conducted on GONFH patients and rats. ROS, tunel, flow cytometry, alkaline phosphatase, Oil red O staining, reverse transcription‑quantitative PCR and western blotting were applied to elucidate the exact pathogenesis mechanism. RESULTS Clinical and animal studies demonstrated increased levels of ROS, aggravated oxidative stress (OS) microenvironment, augmented apoptosis and imbalance in osteogenic/lipogenic in the GONFH group compared to the control group. The fate of mesenchymal stem cells (MSCs) directed by GCs is a crucial factor in determining GONFH. In vitro studies further revealed that GCs promote excessive ROS production through the expression of NOX family proteins, leading to a deterioration of the OS microenvironment in MSCs, ultimately resulting in apoptosis and imbalance in osteogenic/lipogenic differentiation. Furthermore, our results confirmed that the NOX inhibitor-diphenyleneiodonium chloride and the NF-κB inhibitor-BAY 11-7082 ameliorated apoptosis and osteogenic/lipogenic differentiation imbalance of MSCs induced by an excess of GCs. CONCLUSION We demonstrated for the first time that the aggravation of the OS microenvironment in MSCs caused by high doses of GCs leading to apoptosis and differentiation imbalance is a crucial factor in the pathogenesis of GONFH, mediated through activating the NOX/ROS/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Huihui Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Qinghe Zeng
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Kaiao Zou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Haipeng Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Jiali Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Pinger Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Wenhua Yuan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Luwei Xiao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Peijian Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| | - Hongting Jin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006 China
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053 China
- Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, Zhejiang 310053 China
| |
Collapse
|
13
|
Belmas T, Liesa M, Shum M. Quantifying mitochondrial redox and bilirubin content in intact primary hepatocytes of obese mice using fluorescent reporters. STAR Protoc 2023; 4:102408. [PMID: 37393613 PMCID: PMC10336327 DOI: 10.1016/j.xpro.2023.102408] [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: 03/20/2023] [Revised: 05/11/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023] Open
Abstract
Assessing the physiological role of H2O2 requires sensitive techniques to quantify H2O2 and antioxidants in live cells. Here, we present a protocol to assess the mitochondrial redox state and unconjugated bilirubin levels in intact live primary hepatocytes from obese mice. We described steps to quantify H2O2, GSSG/GSH, and bilirubin content in the mitochondrial matrix and the cytosol using the fluorescent reporters roGFP2-ORP1, GRX1-roGFP2, and UnaG, respectively. We detail hepatocyte isolation, plating, and transduction and live-cell imaging using a high-content imaging reader. For complete details on the use and execution of this protocol, please refer to Shum et al.1.
Collapse
Affiliation(s)
- Thomas Belmas
- Endocrinology - Nephrology Research Axis, CHU de Québec-Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Marc Liesa
- Institut de Biología Molecular de Barcelona, IBMB, CSIC, Barcelona, Catalonia, Spain; Department of Medicine, Endocrinology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Michaël Shum
- Endocrinology - Nephrology Research Axis, CHU de Québec-Université Laval Research Center, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
14
|
Wang K, Yao T, Xue J, Guo Y, Xu X. A Novel Fluorescent Probe for the Detection of Hydrogen Peroxide. BIOSENSORS 2023; 13:658. [PMID: 37367023 DOI: 10.3390/bios13060658] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023]
Abstract
Hydrogen peroxide (H2O2) is one of the important reactive oxygen species (ROS), which is closely related to many pathological and physiological processes in living organisms. Excessive H2O2 can lead to cancer, diabetes, cardiovascular diseases, and other diseases, so it is necessary to detect H2O2 in living cells. Since this work designed a novel fluorescent probe to detect the concentration of H2O2, the H2O2 reaction group arylboric acid was attached to the fluorescein 3-Acetyl-7-hydroxycoumarin as a specific recognition group for the selective detection of hydrogen peroxide. The experimental results show that the probe can effectively detect H2O2 with high selectivity and measure cellular ROS levels. Therefore, this novel fluorescent probe provides a potential monitoring tool for a variety of diseases caused by H2O2 excess.
Collapse
Affiliation(s)
- Kangkang Wang
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Tingting Yao
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Jiayu Xue
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yanqiu Guo
- Nanjing Luhe People's Hospital, Nanjing 210009, China
| | - Xiaowei Xu
- State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
15
|
Kulkarni VV, Wang Y, Pantaleon Garcia J, Evans SE. Redox-Dependent Activation of Lung Epithelial STAT3 Is Required for Inducible Protection against Bacterial Pneumonia. Am J Respir Cell Mol Biol 2023; 68:679-688. [PMID: 36826841 PMCID: PMC10257071 DOI: 10.1165/rcmb.2022-0342oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/24/2023] [Indexed: 02/25/2023] Open
Abstract
The lung epithelium is dynamic, capable of considerable structural and functional plasticity in response to pathogen challenges. Our laboratory has demonstrated that an inhaled combination of a Toll-like receptor (TLR) 2/6 agonist and a TLR9 agonist (Pam2ODN) results in robust protection against otherwise lethal pneumonias. We have previously shown that intact epithelial TLR signaling and generation of multisource epithelial reactive oxygen species (ROS) are required for inducible protection. Further investigating the mechanisms underlying this phenomenon of inducible resistance, reverse-phase protein array analysis demonstrated robust STAT3 (signal transducer and activator of transcription 3) phosphorylation following treatment of lung epithelial cells. We show here that Pam2ODN-induced STAT3 phosphorylation is IL-6-independent. We further found that therapeutic epithelial STAT3 activation is required for inducible protection against Pseudomonas aeruginosa pneumonia. Additional studies showed that inhibiting epithelial dual oxidases or scavenging ROS significantly reduced the Pam2ODN induction of STAT3 phosphorylation, suggesting a proximal role for ROS in inducible STAT3 activation. Dissecting these mechanisms, we analyzed the contributions of redox-sensitive kinases and found that Pam2ODN activated epithelial growth factor receptor in an ROS-dependent manner that is required for therapeutically inducible STAT3 activation. Taken together, we demonstrate that epithelial STAT3 is imperative for Pam2ODN's function and describe a novel redox-based mechanism for its activation. These key mechanistic insights may facilitate strategies to leverage inducible epithelial resistance to protect susceptible patients during periods of peak vulnerability.
Collapse
Affiliation(s)
- Vikram V. Kulkarni
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas; and
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yongxing Wang
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Scott E. Evans
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas; and
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
16
|
Yadav S, Waldeck-Weiermair M, Spyropoulos F, Bronson R, Pandey AK, Das AA, Sisti AC, Covington TA, Thulabandu V, Caplan S, Chutkow W, Steinhorn B, Michel T. Sensory ataxia and cardiac hypertrophy caused by neurovascular oxidative stress in chemogenetic transgenic mouse lines. Nat Commun 2023; 14:3094. [PMID: 37248315 PMCID: PMC10227029 DOI: 10.1038/s41467-023-38961-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/24/2023] [Indexed: 05/31/2023] Open
Abstract
Oxidative stress is associated with cardiovascular and neurodegenerative diseases. Here we report studies of neurovascular oxidative stress in chemogenetic transgenic mouse lines expressing yeast D-amino acid oxidase (DAAO) in neurons and vascular endothelium. When these transgenic mice are fed D-amino acids, DAAO generates hydrogen peroxide in target tissues. DAAO-TGCdh5 transgenic mice express DAAO under control of the putatively endothelial-specific Cdh5 promoter. When we provide these mice with D-alanine, they rapidly develop sensory ataxia caused by oxidative stress and mitochondrial dysfunction in neurons within dorsal root ganglia and nodose ganglia innervating the heart. DAAO-TGCdh5 mice also develop cardiac hypertrophy after chronic chemogenetic oxidative stress. This combination of ataxia, mitochondrial dysfunction, and cardiac hypertrophy is similar to findings in patients with Friedreich's ataxia. Our observations indicate that neurovascular oxidative stress is sufficient to cause sensory ataxia and cardiac hypertrophy. Studies of DAAO-TGCdh5 mice could provide mechanistic insights into Friedreich's ataxia.
Collapse
Affiliation(s)
- Shambhu Yadav
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Markus Waldeck-Weiermair
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Fotios Spyropoulos
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Roderick Bronson
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Apabrita Ayan Das
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Alexander C Sisti
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Taylor A Covington
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Venkata Thulabandu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Shari Caplan
- Novartis Institutes for Biomedical Research, Cambridge, MA, 02139, USA
| | - William Chutkow
- Novartis Institutes for Biomedical Research, Cambridge, MA, 02139, USA
| | - Benjamin Steinhorn
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Thomas Michel
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Tang T, Gong Y, Gao Y, Pang X, Liu S, Xia Y, Liu D, Zhu L, Fan Q, Sun X. A pH-responsive liposomal nanoplatform for co-delivery of a Pt(IV) prodrug and cinnamaldehyde for effective tumor therapy. Front Bioeng Biotechnol 2023; 11:1191534. [PMID: 37214306 PMCID: PMC10196200 DOI: 10.3389/fbioe.2023.1191534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction: The tumor microenvironment (TME) is mainly characterized by abnormally elevated intracellular redox levels and excessive oxidative stress. However, the balance of the TME is also very fragile and susceptible to be disturbed by external factors. Therefore, several researchers are now focusing on intervening in redox processes as a therapeutic strategy to treat tumors. Here, we have developed a liposomal drug delivery platform that can load a Pt(IV) prodrug (DSCP) and cinnamaldehyde (CA) into a pH-responsive liposome to enrich more drugs in the tumor region for better therapeutic efficacy through enhanced permeability and retention effect. Methods: Using the glutathione-depleting properties of DSCP together with the ROS-generating properties of cisplatin and CA, we synergistically altered ROS levels in the tumor microenvironment to damage tumor cells and achieve anti-tumor effects in vitro. Results: A liposome loaded with DSCP and CA was successfully established, and this liposome effectively increased the level of ROS in the tumor microenvironment and achieved effective killing of tumor cells in vitro. Conclusion: In this study, novel liposomal nanodrugs loaded with DSCP and CA provided a synergistic strategy between conventional chemotherapy and disruption of TME redox homeostasis, leading to a significant increase in antitumor effects in vitro.
Collapse
Affiliation(s)
- Ting Tang
- Department of Dental Implantology, Hefei Stomatology Hospital, Clinical School of Anhui Medical University, Hefei, China
| | - Yufang Gong
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Gao
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinlong Pang
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuangqing Liu
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yulan Xia
- Department of Dental Implantology, Hefei Stomatology Hospital, Clinical School of Anhui Medical University, Hefei, China
| | - Dongsheng Liu
- Department of Dental Implantology, Hefei Stomatology Hospital, Clinical School of Anhui Medical University, Hefei, China
| | - Lin Zhu
- Department of Dental Implantology, Hefei Stomatology Hospital, Clinical School of Anhui Medical University, Hefei, China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao Sun
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
18
|
Wang YL, Chang CY, Hsu NS, Lo IW, Lin KH, Chen CL, Chang CF, Wang ZC, Ogasawara Y, Dairi T, Maruyama C, Hamano Y, Li TL. N-Formimidoylation/-iminoacetylation modification in aminoglycosides requires FAD-dependent and ligand-protein NOS bridge dual chemistry. Nat Commun 2023; 14:2528. [PMID: 37137912 PMCID: PMC10156733 DOI: 10.1038/s41467-023-38218-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/21/2023] [Indexed: 05/05/2023] Open
Abstract
Oxidized cysteine residues are highly reactive and can form functional covalent conjugates, of which the allosteric redox switch formed by the lysine-cysteine NOS bridge is an example. Here, we report a noncanonical FAD-dependent enzyme Orf1 that adds a glycine-derived N-formimidoyl group to glycinothricin to form the antibiotic BD-12. X-ray crystallography was used to investigate this complex enzymatic process, which showed Orf1 has two substrate-binding sites that sit 13.5 Å apart unlike canonical FAD-dependent oxidoreductases. One site could accommodate glycine and the other glycinothricin or glycylthricin. Moreover, an intermediate-enzyme adduct with a NOS-covalent linkage was observed in the later site, where it acts as a two-scissile-bond linkage facilitating nucleophilic addition and cofactor-free decarboxylation. The chain length of nucleophilic acceptors vies with bond cleavage sites at either N-O or O-S accounting for N-formimidoylation or N-iminoacetylation. The resultant product is no longer sensitive to aminoglycoside-modifying enzymes, a strategy that antibiotic-producing species employ to counter drug resistance in competing species.
Collapse
Affiliation(s)
- Yung-Lin Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Chin-Yuan Chang
- Department of Biology Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 30010, Taiwan
| | - Ning-Shian Hsu
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - I-Wen Lo
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Kuan-Hung Lin
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Chun-Liang Chen
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Chi-Fon Chang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Zhe-Chong Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yasushi Ogasawara
- Graduate School of Engineering, Hokkaido University, Kita-ku, Sapporo, Hokkaido, 060-8628, Japan
| | - Tohru Dairi
- Graduate School of Engineering, Hokkaido University, Kita-ku, Sapporo, Hokkaido, 060-8628, Japan
| | - Chitose Maruyama
- Graduate School of Bioscience and Biotechnology, Fukui Prefectural University, Eiheiji-cho, Fukui, 910-1195, Japan
- Fukui Bioincubation Center (FBIC), Fukui Prefectural University, Eiheiji-cho, Fukui, 910-1195, Japan
| | - Yoshimitsu Hamano
- Graduate School of Bioscience and Biotechnology, Fukui Prefectural University, Eiheiji-cho, Fukui, 910-1195, Japan.
- Fukui Bioincubation Center (FBIC), Fukui Prefectural University, Eiheiji-cho, Fukui, 910-1195, Japan.
| | - Tsung-Lin Li
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung City, 402, Taiwan.
| |
Collapse
|
19
|
Sui H, Xiao S, Jiang S, Wu S, Lin H, Cheng L, Ye L, Zhao Q, Yu Y, Tao L, Kong FM, Huang X, Cui R. Regorafenib induces NOX5-mediated endoplasmic reticulum stress and potentiates the anti-tumor activity of cisplatin in non-small cell lung cancer cells. Neoplasia 2023; 39:100897. [PMID: 36940556 PMCID: PMC10036942 DOI: 10.1016/j.neo.2023.100897] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Lung cancer is one of the most commonly diagnosed cancers worldwide. Although cisplatin-based chemotherapy regimens serve a pivotal role in non-small cell lung cancer (NSCLC) treatment, drug resistance and serious side effects limited its further clinical application. Regorafenib, a small-molecule multi-kinase inhibitor, was demonstrated to have promising anti-tumor activity in various solid tumors. In the present study, we found that regorafenib markedly enhanced cisplatin-induced cytotoxicity in lung cancer cells by activating reactive oxygen species (ROS)-mediated endoplasmic reticulum stress (ER Stress), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) signaling pathways. Regorafenib increased ROS generation by promoting NADPH oxidase 5 (NOX5) expression, and knocking down NOX5 attenuated ROS-mediated cytotoxicity of regorafenib in lung cancer cells. Additionally, mice xenograft model validated that synergistic anti-tumor effects of combined treatment with regorafenib and cisplatin. Our results suggested that combination therapy with regorafenib and cisplatin may serve as a potential therapeutic strategy for some NSCLC patients.
Collapse
Affiliation(s)
- Hehuan Sui
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China
| | - Sisi Xiao
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Suping Jiang
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China
| | - Siyuan Wu
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China
| | - Haizhen Lin
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Liyuan Cheng
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China
| | - Lihua Ye
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China
| | - Qi Zhao
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China
| | - Yun Yu
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lu Tao
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Feng-Ming Kong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xiaoying Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Ri Cui
- Cancer and Anticancer Drug Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Wenzhou University-Wenzhou Medical University Collaborative Innovation Center of Biomedical, Wenzhou, 325035, China.
| |
Collapse
|
20
|
Bhatt MR, Zondlo NJ. Synthesis and conformational preferences of peptides and proteins with cysteine sulfonic acid. Org Biomol Chem 2023; 21:2779-2800. [PMID: 36920119 DOI: 10.1039/d3ob00179b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Cysteine sulfonic acid (Cys-SO3H; cysteic acid) is an oxidative post-translational modification of cysteine, resulting from further oxidation from cysteine sulfinic acid (Cys-SO2H). Cysteine sulfonic acid is considered an irreversible post-translational modification, which serves as a biomarker of oxidative stress that has resulted in oxidative damage to proteins. Cysteine sulfonic acid is anionic, as a sulfonate (Cys-SO3-; cysteate), in the ionization state that is almost exclusively present at physiological pH (pKa ∼ -2). In order to understand protein structural changes that can occur upon oxidation to cysteine sulfonic acid, we analyzed its conformational preferences, using experimental methods, bioinformatics, and DFT-based computational analysis. Cysteine sulfonic acid was incorporated into model peptides for α-helix and polyproline II helix (PPII). Within peptides, oxidation of cysteine to the sulfonic acid proceeds rapidly and efficiently at room temperature in solution with methyltrioxorhenium (MeReO3) and H2O2. Peptides containing cysteine sulfonic acid were also generated on solid phase using trityl-protected cysteine and oxidation with MeReO3 and H2O2. Using methoxybenzyl (Mob)-protected cysteine, solid-phase oxidation with MeReO3 and H2O2 generated the Mob sulfone precursor to Cys-SO2- within fully synthesized peptides. These two solid-phase methods allow the synthesis of peptides containing either Cys-SO3- or Cys-SO2- in a practical manner, with no solution-phase synthesis required. Cys-SO3- had low PPII propensity for PPII propagation, despite promoting a relatively compact conformation in ϕ. In contrast, in a PPII initiation model system, Cys-SO3- promoted PPII relative to neutral Cys, with PPII initiation similar to Cys thiolate but less than Cys-SO2- or Ala. In an α-helix model system, Cys-SO3- promoted α-helix near the N-terminus, due to favorable helix dipole interactions and favorable α-helix capping via a sulfonate-amide side chain-main chain hydrogen bond. Across all peptides, the sulfonate side chain was significantly less ordered than that of the sulfinate. Analysis of Cys-SO3- in the PDB revealed a very strong propensity for local (i/i or i/i + 1) side chain-main chain sulfonate-amide hydrogen bonds for Cys-SO3-, with >80% of Cys-SO3- residues exhibiting these interactions. DFT calculations conducted to explore these conformational preferences indicated that side chain-main chain hydrogen bonds of the sulfonate with the intraresidue amide and/or with the i + 1 amide were favorable. However, hydrogen bonds to water or to amides, as well as interactions with oxophilic metals, were weaker for the sulfonate than the sulfinate, due to lower charge density on the oxygens in the sulfonate.
Collapse
Affiliation(s)
- Megh R Bhatt
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA.
| | - Neal J Zondlo
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
21
|
Pató A, Bölcskei K, Donkó Á, Kaszás D, Boros M, Bodrogi L, Várady G, Pape VFS, Roux BT, Enyedi B, Helyes Z, Watt FM, Sirokmány G, Geiszt M. Hydrogen peroxide production by epidermal dual oxidase 1 regulates nociceptive sensory signals. Redox Biol 2023; 62:102670. [PMID: 36958249 PMCID: PMC10038790 DOI: 10.1016/j.redox.2023.102670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/22/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Keratinocytes of the mammalian skin provide not only mechanical protection for the tissues, but also transmit mechanical, chemical, and thermal stimuli from the external environment to the sensory nerve terminals. Sensory nerve fibers penetrate the epidermal basement membrane and function in the tight intercellular space among keratinocytes. Here we show that epidermal keratinocytes produce hydrogen peroxide upon the activation of the NADPH oxidase dual oxidase 1 (DUOX1). This enzyme can be activated by increasing cytosolic calcium levels. Using DUOX1 knockout animals as a model system we found an increased sensitivity towards certain noxious stimuli in DUOX1-deficient animals, which is not due to structural changes in the skin as evidenced by detailed immunohistochemical and electron-microscopic analysis of epidermal tissue. We show that DUOX1 is expressed in keratinocytes but not in the neural sensory pathway. The release of hydrogen peroxide by activated DUOX1 alters both the activity of neuronal TRPA1 and redox-sensitive potassium channels expressed in dorsal root ganglia primary sensory neurons. We describe hydrogen peroxide, produced by DUOX1 as a paracrine mediator of nociceptive signal transmission. Our results indicate that a novel, hitherto unknown redox mechanism modulates noxious sensory signals.
Collapse
Affiliation(s)
- Anna Pató
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624, Pécs, Hungary
| | - Ágnes Donkó
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary
| | - Diána Kaszás
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary; MTA-SE Lendület Tissue Damage Research Group, Hungarian Academy of Sciences and Semmelweis University, H-1094, Budapest, Hungary; HCEMM-SE Inflammatory Signaling Research Group, Department of Physiology, Semmelweis University, H-1094, Budapest, Hungary
| | - Melinda Boros
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624, Pécs, Hungary
| | - Lilla Bodrogi
- Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, H-2100, Gödöllő, Hungary
| | - György Várady
- Research Centre for Natural Sciences, Institute of Enzymology, H-1117, Budapest, Hungary
| | - Veronika F S Pape
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary
| | - Benoit T Roux
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary; MTA-SE Lendület Tissue Damage Research Group, Hungarian Academy of Sciences and Semmelweis University, H-1094, Budapest, Hungary; HCEMM-SE Inflammatory Signaling Research Group, Department of Physiology, Semmelweis University, H-1094, Budapest, Hungary
| | - Balázs Enyedi
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary; MTA-SE Lendület Tissue Damage Research Group, Hungarian Academy of Sciences and Semmelweis University, H-1094, Budapest, Hungary; HCEMM-SE Inflammatory Signaling Research Group, Department of Physiology, Semmelweis University, H-1094, Budapest, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624, Pécs, Hungary; Eötvös Lorand Research Network, Chronic Pain Research Group, University of Pécs, H-7624, Pécs, Hungary; National Laboratory for Drug Research and Development, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - Fiona M Watt
- European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Gábor Sirokmány
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary.
| | - Miklós Geiszt
- Department of Physiology, Semmelweis University, Faculty of Medicine, H-1094, Budapest, Hungary.
| |
Collapse
|
22
|
Xiao C, Izquierdo-Roca V, Rivera-Gil P. Real Time and Spatiotemporal Quantification of pH and H 2O 2 Imbalances with a Multiplex Surface-Enhanced Raman Spectroscopy Nanosensor. ACS MATERIALS AU 2023; 3:164-175. [PMID: 38089722 PMCID: PMC9999477 DOI: 10.1021/acsmaterialsau.2c00069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 06/03/2024]
Abstract
Oxidative stress is involved in many aging-related pathological disorders and is the result of defective cellular management of redox reactions. Particularly, hydrogen peroxide (H2O2), is a major byproduct and a common oxidative stress biomarker. Monitoring its dynamics and a direct correlation to diseases remains a challenge due to the complexity of redox reactions. Sensitivity and specificity are major drawbacks for H2O2 sensors regardless of their readout. Luminiscent boronate-based probes such as 3-mercaptophenylboronic acid (3-MPBA) are emerging as the most effective quantitation tool due to their specificity and sensitivity. Problems associated with these probes are limited intracellular sensing, water solubility, selectivity, and quenching. We have synthesized a boronate-based nanosensor with a surface-enhanced Raman spectroscopy (SERS) readout to solve these challenges. Furthermore, we found out that environmental pH gradients, as found in biological samples, affect the sensitivity of boronate-based sensors. When the sensor is in an alkaline environment, the oxidation of 3-MPBA by H2O2 is more favored than in an acidic environment. This leads to different H2O2 measurements depending on pH. To solve this issue, we synthesized a multiplex nanosensor capable of concomitantly quantifying pH and H2O2. Our nanosensor first measures the local pH and based on this value, provides the amount of H2O2. It seems that this pH-dependent sensitivity effect applies to all boronic acid based probes. We tested the multiplexing ability by quantitatively measuring intra- and extracellular pH and H2O2 dynamics under physiological and pathological conditions on healthy cells and cells in which H+ and/or H2O2 homeostasis has been altered.
Collapse
Affiliation(s)
- Can Xiao
- Department
of Medicine and Life Sciences, Universitat
Pompeu Fabra, Carrer Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Victor Izquierdo-Roca
- Catalonia
Institute for Energy Research (IREC), Jardins de les Dones de Negre 1, 08930 Sant Adrià del Besòs-Barcelona, Spain
| | - Pilar Rivera-Gil
- Department
of Medicine and Life Sciences, Universitat
Pompeu Fabra, Carrer Doctor Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
23
|
Puente-Cobacho B, Varela-López A, Quiles JL, Vera-Ramirez L. Involvement of redox signalling in tumour cell dormancy and metastasis. Cancer Metastasis Rev 2023; 42:49-85. [PMID: 36701089 PMCID: PMC10014738 DOI: 10.1007/s10555-022-10077-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/27/2022] [Indexed: 01/27/2023]
Abstract
Decades of research on oncogene-driven carcinogenesis and gene-expression regulatory networks only started to unveil the complexity of tumour cellular and molecular biology. This knowledge has been successfully implemented in the clinical practice to treat primary tumours. In contrast, much less progress has been made in the development of new therapies against metastasis, which are the main cause of cancer-related deaths. More recently, the role of epigenetic and microenviromental factors has been shown to play a key role in tumour progression. Free radicals are known to communicate the intracellular and extracellular compartments, acting as second messengers and exerting a decisive modulatory effect on tumour cell signalling. Depending on the cellular and molecular context, as well as the intracellular concentration of free radicals and the activation status of the antioxidant system of the cell, the signalling equilibrium can be tilted either towards tumour cell survival and progression or cell death. In this regard, recent advances in tumour cell biology and metastasis indicate that redox signalling is at the base of many cell-intrinsic and microenvironmental mechanisms that control disseminated tumour cell fate and metastasis. In this manuscript, we will review the current knowledge about redox signalling along the different phases of the metastatic cascade, including tumour cell dormancy, making emphasis on metabolism and the establishment of supportive microenvironmental connections, from a redox perspective.
Collapse
Affiliation(s)
- Beatriz Puente-Cobacho
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Laura Vera-Ramirez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain. .,Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.
| |
Collapse
|
24
|
Zhang H, Wang A, Li G, Zhai Q, Huang Z, Wang X, Cao Z, Liu L, Liu G, Chen B, Zhu K, Xu Y, Xu Y. Osteoporotic bone loss from excess iron accumulation is driven by NOX4-triggered ferroptosis in osteoblasts. Free Radic Biol Med 2023; 198:123-136. [PMID: 36738798 DOI: 10.1016/j.freeradbiomed.2023.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/08/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Excess iron accumulation is a risk factor for osteopenia and osteoporosis, and ferroptosis is becoming well understood as iron-dependent form of cell death resulting from lipid peroxide accumulation. However, any pathological impacts of ferroptosis on osteoporosis remain unknown. Here, we show that ferroptosis is involved in excess-iron-induced bone loss and demonstrate that osteoporotic mice and humans have elevated skeletal accumulation of the NADPH oxidase 4 (NOX4) enzyme. Mechanistically, we found that the NOX4 locus contains iron-response element-like (IRE-like) sequences that are normally bound (and repressed) by the iron regulatory protein 1 (IRP1) protein. Binding with iron induces dissociation of IRP1 from the IRE-like sequences and thereby activates NOX4 transcription. Elevated NOX4 increases lipid peroxide accumulation and causes obvious dysregulation of mitochondrial morphology and function in osteoblasts. Excitingly, the osteoporotic bone loss which we initially observed in an excessive-iron accumulating mouse line (Hepc1-/-) was blocked upon treatment with the ferroptosis-inhibitor ferrostatin-1 (Ferr-1) and with the iron chelator deferoxamine (DFO), suggesting a potential therapeutic strategy for preventing osteoporotic bone loss based on disruption of ferroptosis.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Aifei Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Institute of Osteoporosis Diagnosis and Treatments of Soochow University, Suzhou, 215004, China
| | - Guangfei Li
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Qiaocheng Zhai
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhengyun Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xiao Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zihou Cao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Institute of Osteoporosis Diagnosis and Treatments of Soochow University, Suzhou, 215004, China
| | - Lulin Liu
- Institute of Osteoporosis Diagnosis and Treatments of Soochow University, Suzhou, 215004, China
| | - Gongwen Liu
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215004, China
| | - Bin Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Keyu Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Institute of Osteoporosis Diagnosis and Treatments of Soochow University, Suzhou, 215004, China
| | - Ying Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Institute of Osteoporosis Diagnosis and Treatments of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
25
|
Messina MS, Quargnali G, Chang CJ. Activity-Based Sensing for Chemistry-Enabled Biology: Illuminating Principles, Probes, and Prospects for Boronate Reagents for Studying Hydrogen Peroxide. ACS BIO & MED CHEM AU 2022; 2:548-564. [PMID: 36573097 PMCID: PMC9782337 DOI: 10.1021/acsbiomedchemau.2c00052] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
Activity-based sensing (ABS) offers a general approach that exploits chemical reactivity as a method for selective detection and manipulation of biological analytes. Here, we illustrate the value of this chemical platform to enable new biological discovery through a case study in the design and application of ABS reagents for studying hydrogen peroxide (H2O2), a major type of reactive oxygen species (ROS) that regulates a diverse array of vital cellular signaling processes to sustain life. Specifically, we summarize advances in the use of activity-based boronate probes for the detection of H2O2 featuring high molecular selectivity over other ROS, with an emphasis on tailoring designs in chemical structure to promote new biological principles of redox signaling.
Collapse
Affiliation(s)
- Marco S. Messina
- Department
of Chemistry and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark, Delaware 19716, United States
| | - Gianluca Quargnali
- Department
of Chemistry and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Christopher J. Chang
- Department
of Chemistry and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
26
|
Mu K, Kitts DD. Application of a HyPer-3 sensor to monitor intracellular H 2O 2 generation induced by phenolic acids in differentiated Caco-2 cells. Anal Biochem 2022; 659:114934. [PMID: 36206845 DOI: 10.1016/j.ab.2022.114934] [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: 08/23/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 12/14/2022]
Abstract
Intestinal epithelial cells (IECs) are an important point of contact between dietary food components consumed and subsequent whole-body utilization for body maintenance and growth. Selective bioactive phenolic acids, widely present in fruits, vegetables and beverages can generate hydrogen peroxide (H2O2) and contribute to the cellular redox balance, hence influencing well-known cellular antioxidant and pro-oxidant mechanisms. Our findings have showed that increasing extracellular H2O2 resulted in associated changes in intracellular H2O2 levels in Caco-2 cells (p < 0.05) which was facilitated by activity of a family of water channel membrane proteins, termed aquaporins (AQPs). To demonstrate this, a HyPer-3 genetically encoded fluorescent H2O2 sensitive indicator was used to enable fluorescent real-time imaging of intracellular H2O2 levels as a measure of changes occurring in extracellular H2O2 in differentiated Caco-2 cells exposed to different phenolic acids. The use of confocal microscopy and flow cytometry, respectively, captured visualization and quantification of H2O2 uptake in differentiated Caco-2 cells. DFP00173, an aquaporin 3 (AQP3) inhibitor was effective at inhibiting the intracellular uptake of H2O2 and was sensitive to varied levels of H2O2 generated when different phenolic acids were added to the culture media. In summary, HyPer-3 was shown to be an effective technique to demonstrate relative capabilities of structurally different dietary phenolic acids that have potential to alter intestinal redox balance by changing intracellular H2O2, and either antioxidant or pro-oxidant activity, respectively.
Collapse
Affiliation(s)
- Kaiwen Mu
- Food Science, Food Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, 2205 East Mall, Vancouver, B.C, V6T 1Z4, Canada.
| | - David D Kitts
- Food Science, Food Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, 2205 East Mall, Vancouver, B.C, V6T 1Z4, Canada.
| |
Collapse
|
27
|
Wang Y, Liu X, Huang W, Liang J, Chen Y. The intricate interplay between HIFs, ROS, and the ubiquitin system in the tumor hypoxic microenvironment. Pharmacol Ther 2022; 240:108303. [PMID: 36328089 DOI: 10.1016/j.pharmthera.2022.108303] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/16/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alterations in protein ubiquitination and hypoxia-inducible factor (HIF) signaling both contribute to tumorigenesis and tumor progression. Ubiquitination is a dynamic process that is coordinately regulated by E3 ligases and deubiquitinases (DUBs), which have emerged as attractive therapeutic targets. HIF expression and transcriptional activity are usually increased in tumors, leading to poor clinical outcomes. Reactive oxygen species (ROS) are upregulated in tumors and have multiple effects on HIF signaling and the ubiquitin system. A growing body of evidence has shown that multiple E3 ligases and UBDs function synergistically to control the expression and activity of HIF, thereby allowing cancer cells to cope with the hypoxic microenvironment. Conversely, several E3 ligases and DUBs are regulated by hypoxia and/or HIF signaling. Hypoxia also induces ROS production, which in turn modulates the stability or activity of HIF, E3 ligases, and DUBs. Understanding the complex networks between E3 ligase, DUBs, ROS, and HIF will provide insights into the fundamental mechanism of the cellular response to hypoxia and help identify novel molecular targets for cancer treatment. We review the current knowledge on the comprehensive relationship between E3 ligase, DUBs, ROS, and HIF signaling, with a particular focus on the use of E3 ligase or DUB inhibitors in cancer.
Collapse
Affiliation(s)
- Yijie Wang
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China
| | - Xiong Liu
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Weixiao Huang
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Junjie Liang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China.
| | - Yan Chen
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China; School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
28
|
Yang YZ, Qiu WX, Xu ZY, Sun Z, Qing M, Li NB, Luo HQ. Rational design of a fluorescent probe for specific sensing of hydrogen peroxide/glucose and intracellular imaging applications. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 277:121254. [PMID: 35452901 DOI: 10.1016/j.saa.2022.121254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 06/14/2023]
Abstract
A new type of dye with advantages of high selectivity and sensitivity is formed by using the strategy of hybridization between the luminescent unit and recognition unit. Based on this strategy, we exploit a novel dye bonding the benzopyrylium salt as a luminescent unit and phenylboronate group as a response site, which is served as a fluorescent probe 1 for specific recognition of hydrogen peroxide in biological application. Probe 1 employs a unique recognition switch, phenylboronate unit, to"turn-on"a highly specific and rapid fluorescence response toward hydrogen peroxide combined with the 1,6-rearrangement elimination reaction strategy. Meanwhile, probe 1 has the ability to glucose assay by taking advantage of glucose oxidase/glucose enzymatic reaction. What's more, the probe 1 is capable of tracking endogenous hydrogen peroxide in living cells and intracellular imaging. Therefore, the newly developed bioprobe 1 is expected to be used to monitor hydrogen peroxide and glucose levels in complex organisms.
Collapse
Affiliation(s)
- Yu Zhu Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China; Department of Basic Teaching, Zunyi Medical and Pharmaceutical College, Zunyi 563006, PR China
| | - Wan Xiang Qiu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Zi Yi Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Zhe Sun
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Min Qing
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Nian Bing Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China.
| | - Hong Qun Luo
- Key Laboratory of Luminescence Analysis and Molecular Sensing, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
29
|
A NIR fluorescent probe for the selective detection of hydrogen peroxide by acetyl-hydrolyzing in cells. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
Yang T, Lin X, Li H, Zhou X, Fan F, Yang J, Luo Y, Liu X. Acetyl-11-Keto-Beta Boswellic Acid (AKBA) Protects Lens Epithelial Cells Against H2O2-Induced Oxidative Injury and Attenuates Cataract Progression by Activating Keap1/Nrf2/HO-1 Signaling. Front Pharmacol 2022; 13:927871. [PMID: 35899124 PMCID: PMC9310784 DOI: 10.3389/fphar.2022.927871] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 11/14/2022] Open
Abstract
Age-related cataract (ARC) is one of the leading blinding eye diseases worldwide. Chronic oxidative stress and the apoptosis of human lens epithelial cells (HLECs) have been suggested to be the mechanism underlying cataract formation. Acetyl-11-keto-β-boswellic acid (AKBA) is a pentacyclic triterpene with antioxidative and antiapoptotic effects. In this study, we investigated the potential effects of AKBA on oxidative-induced HLECs injury and cataract formation. H2O2 was used to simulate HLECs oxidative injury in vitro, and Na2SeO3 was applied to establish an in vivo cataract model. In our current study, a cell counting kit-8 (CCK-8) assay was performed to evaluate the effects of H2O2 and AKBA on cell viability in vitro. Intracellular reactive oxygen species (ROS) levels were measured with the ROS assay to verify the antioxidant capacity of AKBA. Apoptotic cells were detected and measured by TUNEL staining and flow cytometry, and quantitative real-time (qRT)-PCR and Western blotting were applied to examine the transcription and expression of apoptosis-related proteins. Furthermore, immunofluorescence staining was performed to locate factor-erythroid 2-related factor 2 (Nrf2), and the protein levels of Nrf2, kelch-like ECH-associated protein 1 (Keap1) and heme oxygenase-1 (HO-1) were determined by Western blotting. Finally, we observed the degree of lens opacity and performed hematoxylin-eosin (H&E) staining to assess the protective effect of AKBA on cataract formation in vivo. AKBA increased HLECs viability under H2O2 stimulation, decreased intracellular ROS levels and alleviated the cell apoptosis rate in vitro. AKBA significantly decreased the expression of caspase-3 and Bax and increased the content of Bcl-2. The results of immunofluorescence and immunohistochemical staining proved that the expression and nuclear translocation of Nrf2 were activated with AKBA treatment in vivo and in vitro. Moreover, computational docking results showed that AKBA could bind specifically to the predicted Keap1/Nrf2 binding sites. After AKBA activation, Nrf2 dissociates from the Nrf2/Keap1 complex, translocates into the nucleus, and subsequently promotes HO-1 expression. In addition, AKBA attenuated lens opacity in selenite-induced cataracts. Overall, these findings indicated that AKBA alleviated oxidative injury and cataract formation by activating the Keap1/Nrf2/HO-1 cascade. Therefore, our current study highlights that AKBA may serve as a promising treatment for ARC progression.
Collapse
Affiliation(s)
- Tianke Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiaolei Lin
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Hongzhe Li
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiyue Zhou
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Fan Fan
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jianing Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yi Luo
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- *Correspondence: Yi Luo, ; Xin Liu,
| | - Xin Liu
- Department of Ophthalmology, Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- *Correspondence: Yi Luo, ; Xin Liu,
| |
Collapse
|
31
|
Tawa M, Okamura T. Factors influencing the soluble guanylate cyclase heme redox state in blood vessels. Vascul Pharmacol 2022; 145:107023. [PMID: 35718342 DOI: 10.1016/j.vph.2022.107023] [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: 03/11/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 11/15/2022]
Abstract
Soluble guanylate cyclase (sGC) plays an important role in maintaining vascular homeostasis, as an acceptor for the biological messenger nitric oxide (NO). However, only reduced sGC (with a ferrous heme) can be activated by NO; oxidized (ferric heme) and apo (absent heme) sGC cannot. In addition, the proportions of reduced, oxidized, and apo sGC change under pathological conditions. Although diseased blood vessels often show decreased NO bioavailability in the vascular wall, a shift of sGC heme redox balance in favor of the oxidized/apo forms can also occur. Therefore, sGC is of growing interest as a drug target for various cardiovascular diseases. Notably, the balance between NO-sensitive reduced sGC and NO-insensitive oxidized/apo sGC in the body is regulated in a reversible manner by various biological molecules and proteins. Many studies have attempted to identify endogenous factors and determinants that influence this redox state. For example, various reactive nitrogen and oxygen species are capable of inducing the oxidation of sGC heme. Conversely, a heme reductase and some antioxidants reduce the ferric heme in sGC to the ferrous state. This review summarizes the factors and mechanisms identified by these studies that operate to regulate the sGC heme redox state.
Collapse
Affiliation(s)
- Masashi Tawa
- Department of Pathological and Molecular Pharmacology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-1094, Japan.
| | - Tomio Okamura
- Emeritus Professor, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
32
|
Murphy MP, Bayir H, Belousov V, Chang CJ, Davies KJA, Davies MJ, Dick TP, Finkel T, Forman HJ, Janssen-Heininger Y, Gems D, Kagan VE, Kalyanaraman B, Larsson NG, Milne GL, Nyström T, Poulsen HE, Radi R, Van Remmen H, Schumacker PT, Thornalley PJ, Toyokuni S, Winterbourn CC, Yin H, Halliwell B. Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat Metab 2022; 4:651-662. [PMID: 35760871 PMCID: PMC9711940 DOI: 10.1038/s42255-022-00591-z] [Citation(s) in RCA: 576] [Impact Index Per Article: 192.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/19/2022] [Indexed: 01/14/2023]
Abstract
Multiple roles of reactive oxygen species (ROS) and their consequences for health and disease are emerging throughout biological sciences. This development has led researchers unfamiliar with the complexities of ROS and their reactions to employ commercial kits and probes to measure ROS and oxidative damage inappropriately, treating ROS (a generic abbreviation) as if it were a discrete molecular entity. Unfortunately, the application and interpretation of these measurements are fraught with challenges and limitations. This can lead to misleading claims entering the literature and impeding progress, despite a well-established body of knowledge on how best to assess individual ROS, their reactions, role as signalling molecules and the oxidative damage that they can cause. In this consensus statement we illuminate problems that can arise with many commonly used approaches for measurement of ROS and oxidative damage, and propose guidelines for best practice. We hope that these strategies will be useful to those who find their research requiring assessment of ROS, oxidative damage and redox signalling in cells and in vivo.
Collapse
Affiliation(s)
- Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| | - Hülya Bayir
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vsevolod Belousov
- Federal Center of Brain Research and Neurotechnologies, Moscow, Russian Federation
| | | | - Kelvin J A Davies
- Gerontology, Molecular & Computational Biology, and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Tobias P Dick
- German Cancer Research Center, DKFZ-ZMBH Alliance and Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | - Henry J Forman
- Gerontology, Molecular & Computational Biology, and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, CA, USA
- School of Natural Sciences, University of California, Merced, Merced, CA, USA
| | - Yvonne Janssen-Heininger
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - David Gems
- University of Vermont, Burlington, VT, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Nils-Göran Larsson
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Rafael Radi
- Universidad de la República, Montevideo, Uruguay
| | | | | | - Paul J Thornalley
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Shinya Toyokuni
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Christine C Winterbourn
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Huiyong Yin
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Barry Halliwell
- Department of Biochemistry and Life Sciences Institute Neurobiogy Programme, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
33
|
A puromycin-dependent activity-based sensing probe for histochemical staining of hydrogen peroxide in cells and animal tissues. Nat Protoc 2022; 17:1691-1710. [PMID: 35562423 DOI: 10.1038/s41596-022-00694-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/01/2022] [Indexed: 12/13/2022]
Abstract
Hydrogen peroxide (H2O2) is a key member of the reactive oxygen species family of transient small molecules that has broad contributions to oxidative stress and redox signaling. The development of selective and sensitive chemical probes can enable the study of H2O2 biology in cell, tissue and animal models. Peroxymycin-1 is a histochemical activity-based sensing probe that responds to H2O2 via chemoselective boronate oxidation to release puromycin, which is then covalently incorporated into nascent proteins by the ribosome and can be detected by antibody staining. Here, we describe an optimized two-step, one-pot protocol for synthesizing Peroxymycin-1 with improved yields over our originally reported procedure. We also present detailed procedures for applying Peroxymycin-1 to a broad range of biological samples spanning cells to animal tissues for profiling H2O2 levels through histochemical detection by using commercially available anti-puromycin antibodies. The preparation of Peroxymycin-1 takes 9 h, the confocal imaging experiments of endogenous H2O2 levels across different cancer cell lines take 1 d, the dot blot analysis of mouse liver tissues takes 1 d and the confocal imaging of mouse liver tissues takes 3-4 d.
Collapse
|
34
|
Liu Q, Liu R, Zhou Y, Wang W, Wu G, Yang N. Phospholipase Dδ and H 2S increase the production of NADPH oxidase-dependent H 2O 2 to respond to osmotic stress-induced stomatal closure in Arabidopsis thaliana. JOURNAL OF PLANT PHYSIOLOGY 2022; 270:153617. [PMID: 35042010 DOI: 10.1016/j.jplph.2022.153617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
Osmotic stress is one of the main stresses that seriously affects the survival of plants, destroying normal cell activities, and potentially leading to plant death. Phospholipase D (PLD), a major lipid hydrolase, hydrolyzes membrane phospholipids to produce phosphatidic acid (PA) and responds to many abiotic stresses. Hydrogen sulfide (H2S) emerges as the third gaseous signaling molecule involved in the complex network of signaling events. Hydrogen peroxide (H2O2) plays a crucial role as a signaling molecule in plant development and growth, and responds to various abiotic and biotic stresses. In this study, the functions and the relationship of PLDδ, H2S, and H2O2 in osmotic stress-induced stomatal closure were explored. By using the seedlings of ecotype (WT), PLDδ-deficient mutant (pldδ), l-cysteine desulfhydrase (LCD)-deficient mutant (lcd), and pldδlcd double mutant, atrbohD, and atrbohF mutant as materials, and the stomatal aperture were analyzed. The relative water loss of pldδ, lcd, and pldδlcd was higher than that of WT. Exogenous PA and NaHS could partially alleviate the leaf wilting and yellowing phenotypes of pldδ, lcd, and pldδlcd under osmotic stress, but the mutants could not be restored to the same phenotype as WT. The fluorescence intensity of H2O2 in guard cells of pldδ, lcd, and pldδlcd was lower than that of WT, indicating that PLDδ and LCD were involved in the production of H2O2 in guard cells. Exogenous application of H2O2 to WT, pldδ, lcd, and pldδlcd significantly induced stomatal closure under osmotic stress. Exogenous NaHS induced stomatal closure of WT, but could not induce stomatal closure of atrbohD and atrbohF under osmotic stress. These results suggest that the accumulation of H2O2 was essential to induce stomatal closure under osmotic stress, and PLDδ and LCD acted upstream of H2O2.
Collapse
Affiliation(s)
- Qin Liu
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Ruirui Liu
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Yaping Zhou
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Wei Wang
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Guofan Wu
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China
| | - Ning Yang
- College of Life Science, Northwest Normal University, Lanzhou, 730070, China.
| |
Collapse
|
35
|
KAMINSKY R, YANCHYSHYN A, BELEMETS N, KURYK O, SAMBORSKA I, DZEVULSKA I, PELLICANO R. Hyperhomocysteinemia in the pathogenesis of cardiovascular and endocrine diseases: translational messages. MINERVA BIOTECHNOLOGY AND BIOMOLECULAR RESEARCH 2022; 34. [DOI: 10.23736/s2724-542x.21.02805-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
|
36
|
Singh R, Manna PP. Reactive oxygen species in cancer progression and its role in therapeutics. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The redox status in pathogenesis is critically regulated by careful balance between the generation of reactive oxygen species (ROS) and their elimination. Increased ROS level above the cellular tolerability threshold results in apoptotic or necrotic cell death. ROS belongs to a group of highly reactive compounds that have evolved to play key roles in cellular signaling pathways. It’s widely assumed that a reasonable amount of ROS is essential for a variety of biological processes. Elevated levels of ROS are known to cause various pathologic conditions like neurological disorders, cardiovascular conditions, inflammation, autoimmunity, and cancer. ROS is well known to initiate and assist in progression of tumor by promoting proliferation and survival of cancer cells and thus facilitates pro-tumorigenic signaling in tumor microenvironment. As cancer cells become more resilient to the effects of ROS manipulating drugs, increased antioxidant capacity attenuates their susceptibility to cancer treatment. Excessive environmental stress, on the other hand, can cause cancer cells to die. This review summarizes various molecular mechanisms including the role of checkpoint inhibitors that can be harnessed to develop effective therapeutic strategies for targeting ROS
related signaling in cancer.
Collapse
Affiliation(s)
- Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
37
|
Bekhet OH, Eid ME. The interplay between reactive oxygen species and antioxidants in cancer progression and therapy: a narrative review. Transl Cancer Res 2022; 10:4196-4206. [PMID: 35116715 PMCID: PMC8799102 DOI: 10.21037/tcr-21-629] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022]
Abstract
Objective To unveil the role of reactive oxygen species (ROS) and antioxidants in signaling and involvement in cancer progression and therapy. Background Cancer is considered one of the main causes of mortality in developed countries and expected to be more in developing countries as well. Although some cancers may develop at young age, yet almost all types of cancers are an accumulation of genetic and epigenetic cell damages. Cancer is considered a diverse collection of diseases on a cellular level rather than a single disease; and each disease has a different cause as well. ROS have been seen as harmful toxic molecules; however, they are recognized for cellular signaling capabilities. Elevated levels of ROS have protumorigenic activities; they induce cancer cell proliferation, and adaptation to hypoxia in addition to other effects like DNA damage and genetic instability. They are produced excessively by cancer cells to hyperactivate cellular transformation meanwhile increasing antioxidant capacity to avoid cell death. Methods We discussed peer reviewed published research work from 1987 to 2021. In this paper, we review the role of antioxidants as defensive barrier against excessive ROS levels for maintaining oxidation-reduction (redox) balance; however, antioxidant can also strive in tumor cells with their scavenging capacities and maintain protumorigenic signaling and resist the cancer cell oxidative stress and apoptosis. High doses of antioxidant compounds could be toxic to cells as they are capable of reacting with the physiological concentrations of ROS present for normal cellular processes and signaling. Conclusions Maintaining cellular redox homeostasis is vital for healthy biological system. Therefore, therapeutic modalities for cancer including antioxidants and ROS management should be used at certain doses to target specific redox pathways involved in cancer progression without disrupting the overall redox balance in normal cells.
Collapse
Affiliation(s)
- Osama Hussein Bekhet
- Pole of Endocrinology, Diabetes and Nutrition, Catholic University of Louvain, Woluwe-Saint-Lambert, Belgium
| | - Mohamed Elsayed Eid
- Laboratory of Natural Products Chemistry, Mediterranean Agronomic Institute of Chania, Crete, Greece
| |
Collapse
|
38
|
Recent advance in dual-functional luminescent probes for reactive species and common biological ions. Anal Bioanal Chem 2022; 414:5087-5103. [DOI: 10.1007/s00216-021-03792-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Indexed: 01/17/2023]
|
39
|
Long MJC, Huang KT, Aye Y. The not so identical twins: (dis)similarities between reactive electrophile and oxidant sensing and signaling. Chem Soc Rev 2021; 50:12269-12291. [PMID: 34779447 DOI: 10.1039/d1cs00467k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this tutorial review, we compare and contrast the chemical mechanisms of electrophile/oxidant sensing, and the molecular mechanisms of signal propagation. We critically analyze biological systems in which these different pathways are believed to be manifest and what the data really mean. Finally, we discuss applications of this knowledge to disease treatment and drug development.
Collapse
Affiliation(s)
| | - Kuan-Ting Huang
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
40
|
Li J, Ma X, Yang W, Guo C, Zhai J, Xie X. Enhanced Sulfite-Selective Sensing and Cell Imaging with Fluorescent Nanoreactors Containing a Ratiometric Lipid Peroxidation Sensor. Anal Chem 2021; 93:11758-11764. [PMID: 34410685 DOI: 10.1021/acs.analchem.1c02167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The detection of SO2 and its derivatives is indispensable for monitoring atmospheric, water quality, and biological fluctuation of oxidative stress and metabolism of biothiols within native cellular contexts. In this article, the brush copolymer nanoreactors containing amine-terminated PDMS were used to encapsulate the fluorescent indicator C11-BDP, forming sulfite-sensitive nanoreactors (ssNRs). Surprisingly, the ssNRs were found to be highly selective to sulfite over a range of reactive oxygen/nitrogen/sulfur species and anions, which was not observed with freely dissolved indicators. The ssNRs showed a rapid response (t95 = 65 s), an excellent detection limit (0.7 μM), and a very high sensitivity (ca. 1000-fold ratiometric intensity change) to sulfite. For cellular studies, the ssNRs exhibited negligible toxicity and could be endocytosed into endosomes and lysosomes. Finally, the ssNRs allowed us to visualize the different responses of three different types of cells (pre-adipocytes, RAW264.7, and HeLa cells) to external stimuli in the culture media with sulfites and lipopolysaccharides.
Collapse
Affiliation(s)
- Jing Li
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xueqing Ma
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China.,Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Yang
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chao Guo
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jingying Zhai
- Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaojiang Xie
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
41
|
Walczak A, Radek M, Majsterek I. The Role of ER Stress-Related Phenomena in the Biology of Malignant Peripheral Nerve Sheath Tumors. Int J Mol Sci 2021; 22:ijms22179405. [PMID: 34502310 PMCID: PMC8430526 DOI: 10.3390/ijms22179405] [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: 06/29/2021] [Revised: 08/11/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are rare but one of the most aggressive types of cancer. Currently, there are no effective chemotherapy strategies for these malignancies. The inactivation of the neurofibromatosis type I (NF1) gene, followed by loss of TP53, is an early stage in MPNST carcinogenesis. NF1 is a negative regulator of the Ras proteins family, which are key factors in regulating cell growth, homeostasis and survival. Cell cycle dysregulation induces a stress phenotype, such as proteotoxic stress, metabolic stress, and oxidative stress, which should result in cell death. However, in the case of neoplastic cells, we observe not only the avoidance of apoptosis, but also the impact of stress factors on the treatment effectiveness. This review focuses on the pathomechanisms underlying MPNST cells physiology, and discusses the possible ways to develop a successful treatment based on the molecular background of the disease.
Collapse
Affiliation(s)
- Anna Walczak
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-647 Lodz, Poland;
| | - Maciej Radek
- Department of Neurosurgery and Peripheral Nerve Surgery, Medical University of Lodz, 90-647 Lodz, Poland;
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-647 Lodz, Poland;
- Correspondence:
| |
Collapse
|
42
|
A tandem activity-based sensing and labeling strategy enables imaging of transcellular hydrogen peroxide signaling. Proc Natl Acad Sci U S A 2021; 118:2018513118. [PMID: 33622793 DOI: 10.1073/pnas.2018513118] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) like hydrogen peroxide (H2O2) are transient species that have broad actions in signaling and stress, but spatioanatomical understanding of their biology remains insufficient. Here, we report a tandem activity-based sensing and labeling strategy for H2O2 imaging that enables capture and permanent recording of localized H2O2 fluxes. Peroxy Green-1 Fluoromethyl (PG1-FM) is a diffusible small-molecule probe that senses H2O2 by a boronate oxidation reaction to trigger dual release and covalent labeling of a fluorescent product, thus preserving spatial information on local H2O2 changes. This unique reagent enables visualization of transcellular redox signaling in a microglia-neuron coculture cell model, where selective activation of microglia for ROS production increases H2O2 in nearby neurons. In addition to identifying ROS-mediated cell-to-cell communication, this work provides a starting point for the design of chemical probes that can achieve high spatial fidelity by combining activity-based sensing and labeling strategies.
Collapse
|
43
|
A microtubule-localizing activity-based sensing fluorescent probe for imaging hydrogen peroxide in living cells. Bioorg Med Chem Lett 2021; 48:128252. [PMID: 34245851 DOI: 10.1016/j.bmcl.2021.128252] [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: 05/27/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 11/21/2022]
Abstract
Hydrogen peroxide (H2O2) is a major reactive oxygen species (ROS) in living systems with broad roles spanning both oxidative stress and redox signaling. Indeed, owing to its potent redox activity, regulating local sites of H2O2 generation and trafficking is critical to determining downstream physiological and/or pathological consequences. We now report the design, synthesis, and biological evaluation of Microtubule Peroxy Yellow 1 (MT-PY1), an activity-based sensing fluorescent probe bearing a microtubule-targeting moiety for detection of H2O2 in living cells. MT-PY1 utilizes a boronate trigger to show a selective and robust turn-on response to H2O2 in aqueous solution and in living cells. Live-cell microscopy experiments establish that the probe co-localizes with microtubules and retains its localization after responding to changes in levels of H2O2, including detection of endogenous H2O2 fluxes produced upon growth factor stimulation. This work adds to the arsenal of activity-based sensing probes for biological analytes that enable selective molecular imaging with subcellular resolution.
Collapse
|
44
|
Smolyarova DD, Podgorny OV, Bilan DS, Belousov VV. A guide to genetically encoded tools for the study of H 2 O 2. FEBS J 2021; 289:5382-5395. [PMID: 34173331 DOI: 10.1111/febs.16088] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/23/2021] [Accepted: 06/24/2021] [Indexed: 01/09/2023]
Abstract
Cell metabolism heavily relies on the redox reactions that inevitably generate reactive oxygen species (ROS). It is now well established that ROS fluctuations near basal levels coordinate numerous physiological processes in living organisms, thus exhibiting regulatory functions. Hydrogen peroxide, the most long-lived ROS, is a key contributor to ROS-dependent signal transduction in the cell. H2 O2 is known to impact various targets in the cell; therefore, the question of how H2 O2 modulates physiological processes in a highly specific manner is central in redox biology. To resolve this question, novel genetic tools have recently been created for detecting H2 O2 and emulating its generation in living organisms with unmatched spatiotemporal resolution. Here, we review H2 O2 -sensitive genetically encoded fluorescent sensors and opto- and chemogenetic tools for controlled H2 O2 generation.
Collapse
Affiliation(s)
- Daria D Smolyarova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Russia
| | - Oleg V Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Dmitry S Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod V Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, Moscow, Russia.,Institute for Cardiovascular Physiology, Georg August University Göttingen, Germany
| |
Collapse
|
45
|
Yang N, Sun H, Xue Y, Zhang W, Wang H, Tao H, Liang X, Li M, Xu Y, Chen L, Zhang L, Huang L, Geng D. Inhibition of MAGL activates the Keap1/Nrf2 pathway to attenuate glucocorticoid-induced osteonecrosis of the femoral head. Clin Transl Med 2021; 11:e447. [PMID: 34185425 PMCID: PMC8167863 DOI: 10.1002/ctm2.447] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 01/12/2023] Open
Abstract
Glucocorticoids (GCs) are used in treating viral infections, acute spinal cord injury, autoimmune diseases, and shock. Several patients develop GC-induced osteonecrosis of the femoral head (ONFH). However, the pathogenic mechanisms underlying GC-induced ONFH remain poorly understood. GC-directed bone marrow mesenchymal stem cells (BMSCs) fate is an important factor that determines GC-induced ONFH. At high concentrations, GCs induce BMSC apoptosis by promoting oxidative stress. In the present study, we aimed to elucidate the molecular mechanisms that relieve GC-induced oxidative stress in BMSCs, which would be vital for treating ONFH. The endocannabinoid system regulates oxidative stress in multiple organs. Here, we found that monoacylglycerol lipase (MAGL), a key molecule in the endocannabinoid system, was significantly upregulated during GC treatment in osteoblasts both in vitro and in vivo. MAGL expression was positively correlated with expression of the NADPH oxidase family and apoptosis-related proteins. Functional analysis showed that MAGL inhibition markedly reduced oxidative stress and partially rescued BMSC apoptosis. Additionally, in vivo studies indicated that MAGL inhibition effectively attenuated GC-induced ONFH. Pathway analysis showed that MAGL inhibition regulated oxidative stress in BMSCs via the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. The expression of Nrf2, a major regulator of intracellular antioxidants, was upregulated by inhibiting MAGL. Nrf2 activation can mimic the effect of MAGL inhibition and significantly reduce GC-induced oxidative damage in BMSCs. The beneficial effects of MAGL inhibition were attenuated after the blockade of the Keap1/Nrf2 antioxidant signaling pathway. Notably, pharmacological blockade of MAGL conferred femoral head protection in GC-induced ONFH, even after oxidative stress responses were initiated. Therefore, MAGL may represent a novel target for the prevention and treatment of GC-induced ONFH.
Collapse
Affiliation(s)
- Ning Yang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Houyi Sun
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Yi Xue
- Department of OrthopaedicsChangshu Hospital Affiliated to Nanjing University of Traditional Chinese MedicineChangshuChina
| | - Weicheng Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Hongzhi Wang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Huaqiang Tao
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Xiaolong Liang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Meng Li
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Yaozeng Xu
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Liang Chen
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Liang Zhang
- Department of Orthopaedics, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Lixin Huang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| | - Dechun Geng
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouChina
| |
Collapse
|
46
|
Fong D, Swager TM. Trace Detection of Hydrogen Peroxide via Dynamic Double Emulsions. J Am Chem Soc 2021; 143:4397-4404. [PMID: 33724029 DOI: 10.1021/jacs.1c00683] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hydrogen peroxide is a dynamic signaling molecule in biological systems. We report herein a versatile double emulsion sensor that can detect femtomolar quantities of aqueous hydrogen peroxide. The mechanism responsible for this sensitivity is a peroxide induced change in double emulsion structure, which results in a modified directional emission from dyes dissolved in the high index organic phase. The morphology (structure) of the double emulsion is controlled via interfacial tensions and a methyltrioxorhenium catalyzed sulfide oxidation results in an enhancement of the surfactant effectiveness. The incipient polar sulfoxide induced decrease of the interfacial tension at the organic-water (O-W) interface results in an increased interfacial area between the organic phase and water and a diminished emission perpendicular to the supporting substrate. The modularity of our sensory system is demonstrated through cascade catalysis between methyltrioxorhenium and oxidase enzymes, with the latter producing hydrogen peroxide as a byproduct to enable for the selective and sensitive detection of molecular and ionic enzymatic substrates.
Collapse
Affiliation(s)
- Darryl Fong
- Department of Chemistry and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Timothy M Swager
- Department of Chemistry and Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
47
|
A high-performance genetically encoded fluorescent biosensor for imaging physiological peroxynitrite. Cell Chem Biol 2021; 28:1542-1553.e5. [PMID: 33581056 DOI: 10.1016/j.chembiol.2021.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/08/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
Peroxynitrite is a reactive nitrogen species (RNS) that plays critical roles in signal transduction, stress response, and numerous human diseases. Advanced molecular tools that permit the selective, sensitive, and noninvasive detection of peroxynitrite are essential for understanding its pathophysiological functions. Here, we present pnGFP-Ultra, a high-performance, reaction-based, genetically encodable biosensor for imaging peroxynitrite in live cells. pnGFP-Ultra features a p-boronophenylalanine-modified chromophore as the sensing moiety and exhibits a remarkable ~110-fold fluorescence turn-on response toward peroxynitrite while displaying virtually no cross-reaction with other reactive oxygen/nitrogen species. To facilitate the expression of pnGFP-Ultra in mammalian cells, we engineered an efficient noncanonical amino acid (ncAA) expression system that is broadly applicable to the mammalian expression of ncAA-containing proteins. pnGFP-Ultra robustly detected peroxynitrite production in activated macrophages and primary glial cells. pnGFP-Ultra fills an important technical gap and represents a valuable addition to the molecular toolbox for probing RNS biology.
Collapse
|
48
|
Nan Y, Gu Y, Liu Z, Zhou Q, Zhao W, Xu W. Versatile quantitative biopsy: an approach for cost-effective detection of hydrogen peroxide in tissue specimens. NEW J CHEM 2021. [DOI: 10.1039/d0nj05569g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the presence of H2O2, the amylose released by the micelles is converted into glucose by glucoamylase and detected by a PGM.
Collapse
Affiliation(s)
- Yanxia Nan
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - Yu Gu
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - Zhen Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - Qiulan Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - Wenjie Zhao
- College of Chemistry and Chemical Engineering
- Inner Mongolia University
- Hohhot
- China
| | - Weijian Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| |
Collapse
|
49
|
Freire Boullosa L, Van Loenhout J, Deben C. Endogenous antioxidants in the prognosis and treatment of lung cancer. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
50
|
Chintala S, Maness PF, Petroff JT, Throgmorton JC, Zhang M, Omlid SM, McCulla RD. Photo-oxidation and Thermal Oxidations of Triptycene Thiols by Aryl Chalcogen Oxides. ACS OMEGA 2020; 5:32349-32356. [PMID: 33376871 PMCID: PMC7758903 DOI: 10.1021/acsomega.0c04293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/02/2020] [Indexed: 06/12/2023]
Abstract
Oxidation of thiols yield sulfenic acids, which are very unstable intermediates. As sulfenic acids are reactive, they form disulfides in the presence of thiols. However, sulfenic acids also oxidize to sulfinic acids (-SO2H) and sulfonic acids (-SO3H) at higher concentrations of oxidants. Hydrogen peroxide is a commonly used oxidant for the oxidation of thiols to yield sulfenic acids. However, hydrogen peroxide also oxidizes other reactive functional groups present in a molecule. In this work, the reaction intermediates arising from the oxidation of sterically hindered thiols by aryl chalcogen oxides, dibenzothiophene S-oxide (DBTO), dibenzoselenophene Se-oxide (DBSeO), and dibenzotellurophene Te-oxide (DBTeO), were investigated. Photodeoxygenation of DBTO produces triplet atomic oxygen [O(3P)], which has previously shown to preferentially react with thiols over other functional groups. Similarly, aryl selenoxides have also shown that they can thermally react selectively with thiols at room temperature to yield disulfides. Conversely, aryl telluroxides have been reported to oxidize thiols to disulfides thermally with no selectivity toward thiols. The results from this study demonstrate that sulfenic acids are an intermediate in the oxidation of thiols by DBTeO and by photodeoxygenation of DBTO. The results also showed that the oxidation of thiols by DBSeO yields sulfonic acids. Triptycene-9-thiol and 9-fluorotriptycene-10-thiol were for the thiols used in this oxidation reaction. This work expands the list of oxidants that can be used to oxidize thiols to obtain sulfenic acids.
Collapse
|