1
|
Rafea R, Siragusa M, Fleming I. The Ever-Expanding Influence of the Endothelial Nitric Oxide Synthase. Basic Clin Pharmacol Toxicol 2025; 136:e70029. [PMID: 40150952 PMCID: PMC11950718 DOI: 10.1111/bcpt.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Nitric oxide (NO) generated by the endothelial NO synthase (eNOS) plays an essential role in the maintenance of vascular homeostasis and the prevention of vascular inflammation. There are a myriad of mechanisms that regulate the activity of the enzyme that may prove to represent interesting therapeutic opportunities. In this regard, the kinases that phosphorylate the enzyme and regulate its activity in situations linked to vascular disease seem to be particularly promising. Although the actions of NO were initially linked mainly to the activation of the guanylyl cyclase and the generation of cyclic GMP in vascular smooth muscle cells and platelets, it is now clear that NO elicits the majority of its actions via its ability to modify redox-activated cysteine residues in a process referred to as S-nitrosylation. The more wide spread use of mass spectrometry to detect S-nitrosylated proteins has helped to identify just how large the NO sphere of influence is and just how many cellular processes are affected. It may be an old target, but the sheer impact of eNOS on vascular health really justifies a revaluation of therapeutic options to maintain and protect its activity in situations associated with a high risk of developing cardiovascular disease.
Collapse
Affiliation(s)
- Riham Rafea
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurt am MainGermany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurt am MainGermany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurt am MainGermany
- Partner Site RheinMainGerman Center for Cardiovascular Research (DZHK)Frankfurt am MainGermany
| |
Collapse
|
2
|
Wei L, Liao W, Zhong Y, Tian Y, Wei S, Liu Y. NO-mediated protein S-nitrosylation under salt stress: Role and mechanism. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2024; 338:111927. [PMID: 37984610 DOI: 10.1016/j.plantsci.2023.111927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Salt stress is one of the major environmental stressors that remarkably hinders the processes of plant growth and development, thereby limiting crop productivity. An understanding of the molecular mechanisms underlying plant responses against salinity stimulus will help guide the rational design of crop plants to counter these challenges. Nitric oxide (NO) is a redox-related signaling molecule regulating diverse biological processes in plant. Accumulating evidences indicated NO exert its biological functions through posttranslational modification of proteins, notably via S-nitrosylation. During the past decade, the roles of S-nitrosylation as a regulator of plant and S-nitrosylated candidates have also been established and detected. Emerging evidence indicated that protein S-nitrosylation is ubiquitously involved in the regulation of plant response to salt stress. However, little is known about this pivotal molecular amendment in the regulation of salt stress response. Here, we describe current understanding on the regulatory mechanisms of protein S-nitrosylation in response to salt stress in plants and highlight key challenges in this field.
Collapse
Affiliation(s)
- Lijuan Wei
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou 434025, China
| | - Weibiao Liao
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District, Lanzhou 730070, China
| | - Yue Zhong
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou 434025, China
| | - Ye Tian
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou 434025, China
| | - Shouhui Wei
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou 434025, China.
| | - Yiqing Liu
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou 434025, China.
| |
Collapse
|
3
|
Lv D, Xu Z, Cheng P, Hu Z, Dong Y, Rong Y, Xu H, Wang Z, Cao X, Deng W, Tang B. S-Nitrosylation-mediated coupling of DJ-1 with PTEN induces PI3K/AKT/mTOR pathway-dependent keloid formation. BURNS & TRAUMA 2023; 11:tkad024. [PMID: 38116467 PMCID: PMC10729783 DOI: 10.1093/burnst/tkad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/15/2023] [Accepted: 04/03/2023] [Indexed: 12/21/2023]
Abstract
Background Keloids are aberrant dermal wound healing characterized by invasive growth, extracellular matrix deposition, cytokine overexpression and easy recurrence. Many factors have been implicated as pathological causes of keloids, particularly hyperactive inflammation, tension alignment and genetic predisposition. S-Nitrosylation (SNO), a unique form of protein modification, is associated with the local inflammatory response but its function in excessive fibrosis and keloid formation remains unknown. We aimed to discover the association between protein SNO and keloid formation. Methods Normal and keloid fibroblasts were isolated from collected normal skin and keloid tissues. The obtained fibroblasts were cultured in DMEM supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin. The effects of DJ-1 on cell proliferation, apoptosis, migration and invasion, and on the expression of proteins were assayed. TurboID-based proximity labelling and liquid chromatography-mass spectrometry were conducted to explore the potential targets of DJ-1. Biotin-switch assays and transnitrosylation reactions were used to detect protein SNO. Quantitative data were compared by two-tailed Student's t test. Results We found that DJ-1 served as an essential positive modulator to facilitate keloid cell proliferation, migration and invasion. A higher S-nitrosylated DJ-1 (SNO-DJ-1) level was observed in keloids, and the effect of DJ-1 on keloids was dependent on SNO of the Cys106 residue of the DJ-1 protein. SNO-DJ-1 was found to increase the level of phosphatase and tensin homolog (PTEN) S-nitrosylated at its Cys136 residue via transnitrosylation in keloids, thus diminishing the phosphatase activity of PTEN and activating the PI3K/AKT/mTOR pathway. Furthermore, Cys106-mutant DJ-1 is refractory to SNO and abrogates DJ-1-PTEN coupling and the SNO of the PTEN protein, thus repressing the PI3K/AKT/mTOR pathway and alleviating keloid formation. Importantly, the biological effect of DJ-1 in keloids is dependent on the SNO-DJ-1/SNO-PTEN/PI3K/AKT/mTOR axis. Conclusions For the first time, this study demonstrated the effect of transnitrosylation from DJ-1 to PTEN on promoting keloid formation via the PI3K/AKT/mTOR signaling pathway, suggesting that SNO of DJ-1 may be a novel therapeutic target for keloid treatment.
Collapse
Affiliation(s)
- Dongming Lv
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Zhongye Xu
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Pu Cheng
- Department of General Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen, China
| | - Zhicheng Hu
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Yunxian Dong
- Department of Plastic Surgery, Guangdong Second Provincial General Hospital, Southern Medical University, 466 Xingang Middle Road, Guangzhou, China
| | - Yanchao Rong
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Hailin Xu
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Zhiyong Wang
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Xiaoling Cao
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Bing Tang
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| |
Collapse
|
4
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Vascular nitric oxide resistance in type 2 diabetes. Cell Death Dis 2023; 14:410. [PMID: 37433795 PMCID: PMC10336063 DOI: 10.1038/s41419-023-05935-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/18/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023]
Abstract
Vascular nitric oxide (NO•) resistance, manifested by an impaired vasodilator function of NO• in both the macro- and microvessels, is a common state in type 2 diabetes (T2D) associated with developing cardiovascular events and death. Here, we summarize experimental and human evidence of vascular NO• resistance in T2D and discuss its underlying mechanisms. Human studies indicate a ~ 13-94% decrease in the endothelium (ET)-dependent vascular smooth muscle (VSM) relaxation and a 6-42% reduced response to NO• donors, i.e., sodium nitroprusside (SNP) and glyceryl trinitrate (GTN), in patients with T2D. A decreased vascular NO• production, NO• inactivation, and impaired responsiveness of VSM to NO• [occurred due to quenching NO• activity, desensitization of its receptor soluble guanylate cyclase (sGC), and/or impairment of its downstream pathway, cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG)] are the known mechanisms underlying the vascular NO• resistance in T2D. Hyperglycemia-induced overproduction of reactive oxygen species (ROS) and vascular insulin resistance are key players in this state. Therefore, upregulating vascular NO• availability, re-sensitizing or bypassing the non-responsive pathways to NO•, and targeting key vascular sources of ROS production may be clinically relevant pharmacological approaches to circumvent T2D-induced vascular NO• resistance.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Borisov VB, Forte E. Bioenergetics and Reactive Nitrogen Species in Bacteria. Int J Mol Sci 2022; 23:7321. [PMID: 35806323 PMCID: PMC9266656 DOI: 10.3390/ijms23137321] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
The production of reactive nitrogen species (RNS) by the innate immune system is part of the host's defense against invading pathogenic bacteria. In this review, we summarize recent studies on the molecular basis of the effects of nitric oxide and peroxynitrite on microbial respiration and energy conservation. We discuss possible molecular mechanisms underlying RNS resistance in bacteria mediated by unique respiratory oxygen reductases, the mycobacterial bcc-aa3 supercomplex, and bd-type cytochromes. A complete picture of the impact of RNS on microbial bioenergetics is not yet available. However, this research area is developing very rapidly, and the knowledge gained should help us develop new methods of treating infectious diseases.
Collapse
Affiliation(s)
- Vitaliy B. Borisov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, 119991 Moscow, Russia
| | - Elena Forte
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
6
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
7
|
Mass spectrometry-based direct detection of multiple types of protein thiol modifications in pancreatic beta cells under endoplasmic reticulum stress. Redox Biol 2021; 46:102111. [PMID: 34425387 PMCID: PMC8379693 DOI: 10.1016/j.redox.2021.102111] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/26/2022] Open
Abstract
Thiol-based post-translational modifications (PTMs) play a key role in redox-dependent regulation and signaling. Functional cysteine (Cys) sites serve as redox switches, regulated through multiple types of PTMs. Herein, we aim to characterize the complexity of thiol PTMs at the proteome level through the establishment of a direct detection workflow. The LC-MS/MS based workflow allows for simultaneous quantification of protein abundances and multiple types of thiol PTMs. To demonstrate its utility, the workflow was applied to mouse pancreatic β-cells (β-TC-6) treated with thapsigargin to induce endoplasmic reticulum (ER) stress. This resulted in the quantification of >9000 proteins and multiple types of thiol PTMs, including intra-peptide disulfide (S–S), S-glutathionylation (SSG), S-sulfinylation (SO2H), S-sulfonylation (SO3H), S-persulfidation (SSH), and S-trisulfidation (SSSH). Proteins with significant changes in abundance were observed to be involved in canonical pathways such as autophagy, unfolded protein response, protein ubiquitination pathway, and EIF2 signaling. Moreover, ~500 Cys sites were observed with one or multiple types of PTMs with SSH and S–S as the predominant types of modifications. In many cases, significant changes in the levels of different PTMs were observed on various enzymes and their active sites, while their protein abundance exhibited little change. These results provide evidence of independent translational and post-translational regulation of enzyme activity. The observed complexity of thiol modifications on the same Cys residues illustrates the challenge in the characterization and interpretation of protein thiol modifications and their functional regulation. Simultaneous quantification of protein abundances and multiple types of thiol PTMs. Multiple types PTMs observed on the same Cys sites for redox-regulated proteins. Data revealed complexity of thiol PTMs and their regulation. Distinctive translational and post-translational regulation under ER stress in β-cells.
Collapse
|
8
|
Mikami R, Tsukagoshi S, Oda Y, Arai K. S-Denitrosylase-like Activity of Cyclic Diselenides Conjugated with Xaa-His Dipeptide: Role of Proline Spacer as a Key Activity Booster. Chembiochem 2021; 23:e202100394. [PMID: 34350692 DOI: 10.1002/cbic.202100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Indexed: 11/05/2022]
Abstract
This study developed dipeptide-conjugated 1,2-diselenan-4-amine (1), i.e., 1-Xaa-His, as a new class of S-denitrosylase mimic. The synthesized compounds, especially 1-Pro-His, remarkably promoted S-denitrosylation of nitrosothiols (RSNO) via a catalytic cycle involving the reversible redox reaction between the diselenide and its corresponding diselenol ([SeH,SeH]) form with coexisting reductant thiols (R'SH), during which the [SeH,SeH] form as a key reactive species reduces RSNO to the corresponding thiol (RSH). Structural analyses of 1-Pro-His suggested that the peptide backbone of [SeH,SeH] is rigidly bent to form a γ-turn, possibly including an NH···Se hydrogen bond between the imidazole ring of His and selenol group, thus stabilizing the [SeH,SeH] form thermodynamically, and dramatically enhancing the catalytic activity. Furthermore, the synthetic compounds were found to prohibit S-nitrosylation-induced protein misfolding in the presence of RSNO, eventually implying their potential as a drug seed for misfolding diseases caused by the dysregulation of the S-denitrosylation system.
Collapse
Affiliation(s)
- Rumi Mikami
- Tokai University - Shonan Campus: Tokai Daigaku, Chemistry, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| | - Shunsuke Tsukagoshi
- Tokai University - Shonan Campus: Tokai Daigaku, Chemistry, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| | - Yoshiki Oda
- Tokai University - Shonan Campus: Tokai Daigaku, Technology Joint Management Office, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| | - Kenta Arai
- Tokai University, Chemistry, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| |
Collapse
|
9
|
Marletta MA. Revisiting Nitric Oxide Signaling: Where Was It, and Where Is It Going? Biochemistry 2021; 60:3491-3496. [PMID: 34096266 DOI: 10.1021/acs.biochem.1c00276] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nitric oxide (NO) has long been known to be an intermediate in bacterial pathways of denitrification. Only in the middle to late 1980s was it found to play a central role in a much broader biological context. For example, it is now well established that NO acts as a signaling agent in metazoans, including humans, yet NO is toxic and very reactive under biological conditions. How is the biology in which NO plays a role controlled? How is NO used and the inherent toxicity avoided? Looking back at the initial discovery time, to the present, and on to the future provides many answers to questions such as those listed above.
Collapse
Affiliation(s)
- Michael A Marletta
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, California 94720-3220, United States
| |
Collapse
|
10
|
Gao W, Huang M, Chen X, Chen J, Zou Z, Li L, Ji K, Nie Z, Yang B, Wei Z, Xu P, Jia J, Zhang Q, Shen H, Wang Q, Li K, Zhu L, Wang M, Ye S, Zeng S, Lin Y, Rong Z, Xu Y, Zhu P, Zhang H, Hao B, Liu Q. The role of S-nitrosylation of PFKM in regulation of glycolysis in ovarian cancer cells. Cell Death Dis 2021; 12:408. [PMID: 33859186 PMCID: PMC8050300 DOI: 10.1038/s41419-021-03681-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/02/2023]
Abstract
One of the malignant transformation hallmarks is metabolism reprogramming, which plays a critical role in the biosynthetic needs of unchecked proliferation, abrogating cell death programs, and immunologic escape. However, the mechanism of the metabolic switch is not fully understood. Here, we found that the S-nitrosoproteomic profile of endogenous nitrogen oxide in ovarian cancer cells targeted multiple components in metabolism processes. Phosphofructokinase (PFKM), one of the most important regulatory enzymes of glycolysis, was S-nitrosylated by nitric oxide synthase NOS1 at Cys351. S-nitrosylation at Cys351 stabilized the tetramer of PFKM, leading to resist negative feedback of downstream metabolic intermediates. The PFKM-C351S mutation decreased the proliferation rate of cultured cancer cells, and reduced tumor growth and metastasis in the mouse xenograft model. These findings indicated that S-nitrosylation at Cys351 of PFKM by NOS1 contributes to the metabolic reprogramming of ovarian cancer cells, highlighting a critical role of endogenous nitrogen oxide on metabolism regulations in tumor progression.
Collapse
Affiliation(s)
- Wenwen Gao
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mengqiu Huang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xi Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jianping Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhiwei Zou
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Southern Hospital Zengcheng Branch, Southern Medical University, Guangzhou, 528308, China
| | - Linlin Li
- First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, 450001, China
| | - Kaiyuan Ji
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhirui Nie
- Guangzhou Panyu Central Hospital, Guangzhou, 511400, China
| | - Bingsheng Yang
- Pearl River Hospital, Southern Medical University, Guangzhou, 528308, China
| | - Zibo Wei
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Pengfei Xu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junshuang Jia
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qianbing Zhang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hongfen Shen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qianli Wang
- Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province, 450003, China
| | - Keyi Li
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lingqun Zhu
- Guangzhou Concord Cancer Center, Guangzhou, 528308, China
| | - Meng Wang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuangyan Ye
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Sisi Zeng
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying Lin
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhili Rong
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yang Xu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Peng Zhu
- Central Lab of Shenzhen Pingshan People's Hospital, Shenzhen, 518118, P. R. China
| | - Hui Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China.
- Platform of Metabolomics, Center for Precision Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China.
| | - Bingtao Hao
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Medical Genetic Institute of Henan Province, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genoics, Henan Provincial People's Hospital Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, 450003, China.
| | - Qiuzhen Liu
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Pingshan General Hospital of Southern Medical University, Southern Medical University, Shenzhen, 518118, China.
| |
Collapse
|
11
|
Richards LA, Schonhoff CM. Nitric oxide and sex differences in dendritic branching and arborization. J Neurosci Res 2021; 99:1390-1400. [PMID: 33538046 DOI: 10.1002/jnr.24789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is an important signaling molecule with many functions in the nervous system. Derived from the enzymatic conversion of arginine by several nitric oxide synthases (NOS), NO plays significant roles in neuronal developmental events such as the establishment of dendritic branching or arbors. A brief summary of the discovery, molecular biology, and chemistry of NO, and a description of important NO-mediated signal transduction pathways with emphasis on the role for NO in the development of dendritic branching during neurodevelopment are presented. Important sex differences in neuronal nitric oxide synthase expression during neuronal development are considered. Finally, a survey of endogenous and exogenous substances that disrupt dendritic patterning is presented with particular emphasis on how these molecules may drive NO-mediated sex differences in dendritic branching.
Collapse
Affiliation(s)
- Laura A Richards
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA
| | - Christopher M Schonhoff
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA.,Department of Biomedical Sciences, Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA, USA
| |
Collapse
|
12
|
Zhang C, Xu Y, Wang G, Fang C, Bao H, Zhang Y, Lu H. FluoroTRAQ: Quantitative Analysis of Protein S-Nitrosylation through Fluorous Solid-Phase Extraction Combining with iTRAQ by Mass Spectrometry. Anal Chem 2020; 92:15317-15322. [PMID: 33174720 DOI: 10.1021/acs.analchem.0c01706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
S-Nitrosylation is an important post-translational modification that occurs on cysteine amino acid and regulates signal transduction in diverse cell processes. Dysregulation of protein nitrosylation has shown close association with cardiovascular and neurological diseases, thus demanding further precise and in-depth understanding. Mass spectrometry-based proteomics has been the method of choice for analyzing S-nitrosylated (SNO-) proteins. However, due to their extremely low expression level and rapid turnover rate, quantitative analysis of the S-nitrosylation at the proteomic level remains challenging. Herein, we developed a novel approach termed FluoroTRAQ, which combined the fluorous solid-phase extraction of SNO-peptides and iTRAQ labeling for the quantitative analysis of the SNO-proteome with high sensitivity and specificity. This new analytical strategy was subsequently applied to examine the dynamic SNO-proteome changes of human umbilical vein endothelial cells upon in vitro S-nitrosoglutathione induction. Our data identified a number of novel SNO-proteins and revealed their temporal modulation as validated by biotin switch assay. Our study offered a practical approach for quantitative analysis of protein S-nitrosylation.
Collapse
Affiliation(s)
- Cheng Zhang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Yaoyao Xu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Guoli Wang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Caiyun Fang
- Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032 P. R. China
| | - Huimin Bao
- Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032 P. R. China
| | - Ying Zhang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Haojie Lu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China.,Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032 P. R. China
| |
Collapse
|
13
|
Rhee SG, Woo HA. Multiple functions of 2-Cys peroxiredoxins, I and II, and their regulations via post-translational modifications. Free Radic Biol Med 2020; 152:107-115. [PMID: 32151745 DOI: 10.1016/j.freeradbiomed.2020.02.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/14/2022]
Abstract
Peroxiredoxins (Prxs) are an unusual family of thiol-specific peroxidases that possess a binding site for H2O2 and rely on a conserved cysteine residue for rapid reaction with H2O2. Among 6 mammalian isoforms (Prx I to VI), Prx I and Prx II are mainly found in the cytosol and nucleus. Prx I and Prx II function as antioxidant enzymes and protein chaperone under oxidative distress conditions. Under oxidative eustress conditions, Prx I and Prx II regulate the levels of H2O2 at specific area of the cells as well as sense and transduce H2O2 signaling to target proteins. Prx I and Prx II are known to be covalently modified on multiple sites: Prx I is hyperoxidized on Cys52; phosphorylated on Ser32, Thr90, and Tyr194; acetylated on Lys7, Lys16, Lys27, Lys35, and Lys197; glutathionylated on Cys52, Cys83, and Cys173; and nitrosylated on Cys52 and Cys83, whereas Prx II is hyperoxidized on Cys51; phosphorylated on Thr89, Ser112, and Thr182; acetylated on Ala2 and Lys196; glutathionylated on Cys51 and Cys172; and nitrosylated on Cys51 and Cys172. In this review, we describe how these post-translational modifications affect various functions of Prx I and Prx II.
Collapse
Affiliation(s)
- Sue Goo Rhee
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 120-749, South Korea; The Biochemistry and Biophysics Center, NHLBI, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Hyun Ae Woo
- College of Pharmacy and College of Natural Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| |
Collapse
|
14
|
Ramesh M, Gopinath P, Govindaraju T. Role of Post-translational Modifications in Alzheimer's Disease. Chembiochem 2020; 21:1052-1079. [PMID: 31863723 DOI: 10.1002/cbic.201900573] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/19/2019] [Indexed: 12/22/2022]
Abstract
The global burden of Alzheimer's disease (AD) is growing. Valiant efforts to develop clinical candidates for treatment have continuously met with failure. Currently available palliative treatments are temporary and there is a constant need to search for reliable disease pathways, biomarkers and drug targets for developing diagnostic and therapeutic tools to address the unmet medical needs of AD. Challenges in drug-discovery efforts raise further questions about the strategies of current conventional diagnosis; drug design; and understanding of disease pathways, biomarkers and targets. In this context, post-translational modifications (PTMs) regulate protein trafficking, function and degradation, and their in-depth study plays a significant role in the identification of novel biomarkers and drug targets. Aberrant PTMs of disease-relevant proteins could trigger pathological pathways, leading to disease progression. Advancements in proteomics enable the generation of patterns or signatures of such modifications, and thus, provide a versatile platform to develop biomarkers based on PTMs. In addition, understanding and targeting the aberrant PTMs of various proteins provide viable avenues for addressing AD drug-discovery challenges. This review highlights numerous PTMs of proteins relevant to AD and provides an overview of their adverse effects on the protein structure, function and aggregation propensity that contribute to the disease pathology. A critical discussion offers suggestions of methods to develop PTM signatures and interfere with aberrant PTMs to develop viable diagnostic and therapeutic interventions in AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - Pushparathinam Gopinath
- Department of Chemistry, SRM-Institute of Science and Technology, Kattankulathur, 603203, Chennai, Tamilnadu, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru, 560064, Karnataka, India
| |
Collapse
|
15
|
Glutathione and Nitric Oxide: Key Team Players in Use and Disuse of Skeletal Muscle. Nutrients 2019; 11:nu11102318. [PMID: 31575008 PMCID: PMC6836164 DOI: 10.3390/nu11102318] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Glutathione (GSH) is the main non-enzymatic antioxidant playing an important role in detoxification, signal transduction by modulation of protein thiols redox status and direct scavenging of radicals. The latter function is not only performed against reactive oxygen species (ROS) but GSH also has a fundamental role in buffering nitric oxide (NO), a physiologically-produced molecule having-multifaceted functions. The efficient rate of GSH synthesis and high levels of GSH-dependent enzymes are characteristic features of healthy skeletal muscle where, besides the canonical functions, it is also involved in muscle contraction regulation. Moreover, NO production in skeletal muscle is a direct consequence of contractile activity and influences several metabolic myocyte pathways under both physiological and pathological conditions. In this review, we will consider the homeostasis and intersection of GSH with NO and then we will restrict the discussion on their role in processes related to skeletal muscle function and degeneration.
Collapse
|
16
|
Gong B, Shi Q. Identifying S-nitrosylated proteins and unraveling S-nitrosoglutathione reductase-modulated sodic alkaline stress tolerance in Solanum lycopersicum L. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2019; 142:84-93. [PMID: 31277045 DOI: 10.1016/j.plaphy.2019.06.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 05/13/2023]
Abstract
S-nitrosylation, regulated by S-nitrosoglutathione reductase (GSNOR), is considered as an important route for nitric oxide (NO)-modulated stress tolerance in plants. However, genetic evidence for the GSNOR-mediated integrated regulation of S-nitrosylation and plant stress response remains elusive until now. In the present study, we used a site-specific nitrosoproteomic approach to identify 334 endogenously S-nitrosylated proteins with 425 S-nitrosylated sites from the wild type (WT) and GSNOR-knockdown (G) tomato plants under both control (C) and sodic alkaline stress (S) conditions. In detail, the results revealed 68, 92, 54 and 56 up-regulated, as well as 10, 36, 14 and 10 down-regulated S-nitrosylated proteins in G-C/WT-C, G-S/WT-S, WT-S/WT-C, and G-S/G-C, which is the first dataset for S-nitrosylated proteins in Solanaceae. These S-nitrosylated proteins are involved in a wide range of various metabolic, cellular and catalytic processes. Based on this data, proteins involving in NO homeostasis control, signaling of Ca2+, ethylene and MAPK, reactive oxygen species (ROS) scavenging, osmotic regulation, as well as energy support pathway have been identified and selected as the key and sensitive targets that were regulated by GSNOR-modulated S-nitrosylation in response to sodic alkaline stress. Taken together, GSNOR is actively involved in the regulation of sodic alkaline stress tolerance by S-nitrosylation. And the present study provided valuable resources and new clues for the study of S-nitrosylation-regulated metabolism in tomato plants.
Collapse
Affiliation(s)
- Biao Gong
- State Key Laboratory of Crop Biology, Shandong Agricultural University, Taian 271018, PR China; Collaborative Innovation Center of Fruit & Vegetable Quality and Efficient Production in Shandong, Key Laboratory of Biology and Genetic Improvement of Horticultural Crops in Huanghuai Region, Ministry of Agriculture and Rural Affairs, PR China
| | - Qinghua Shi
- State Key Laboratory of Crop Biology, Shandong Agricultural University, Taian 271018, PR China; Collaborative Innovation Center of Fruit & Vegetable Quality and Efficient Production in Shandong, Key Laboratory of Biology and Genetic Improvement of Horticultural Crops in Huanghuai Region, Ministry of Agriculture and Rural Affairs, PR China.
| |
Collapse
|
17
|
Picciano AL, Crane BR. A nitric oxide synthase-like protein from Synechococcus produces NO/NO 3- from l-arginine and NADPH in a tetrahydrobiopterin- and Ca 2+-dependent manner. J Biol Chem 2019; 294:10708-10719. [PMID: 31113865 PMCID: PMC6615690 DOI: 10.1074/jbc.ra119.008399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/17/2019] [Indexed: 01/01/2023] Open
Abstract
Nitric oxide synthases (NOSs) are heme-based monooxygenases that convert l-Arg to l-citrulline and nitric oxide (NO), a key signaling molecule and cytotoxic agent in mammals. Bacteria also contain NOS proteins, but the role of NO production within these organisms, where understood, differs considerably from that of mammals. For example, a NOS protein in the marine cyanobacterium Synechococcus sp. PCC 7335 (syNOS) has recently been proposed to function in nitrogen assimilation from l-Arg. syNOS retains the oxygenase (NOSox) and reductase (NOSred) domains present in mammalian NOS enzymes (mNOSs), but also contains an N-terminal globin domain (NOSg) homologous to bacterial flavohemoglobin proteins. Herein, we show that syNOS functions as a dimer and produces NO from l-Arg and NADPH in a tetrahydrobiopterin (H4B)-dependent manner at levels similar to those produced by other NOSs but does not require Ca2+-calmodulin, which regulates NOSred-mediated NOSox reduction in mNOSs. Unlike other bacterial NOSs, syNOS cannot function with tetrahydrofolate and requires high Ca2+ levels (>200 μm) for its activation. NOSg converts NO to NO3- in the presence of O2 and NADPH; however, NOSg did not protect Escherichia coli strains against nitrosative stress, even in a mutant devoid of NO-protective flavohemoglobin. We also found that syNOS does not have NOS activity in E. coli (which lacks H4B) and that the recombinant protein does not confer growth advantages on l-Arg as a nitrogen source. Our findings indicate that syNOS has both NOS and NO oxygenase activities, requires H4B, and may play a role in Ca2+-mediated signaling.
Collapse
Affiliation(s)
- Angela L Picciano
- From the Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | - Brian R Crane
- From the Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| |
Collapse
|
18
|
Ghosh S, Roy P, Prasad S, Mugesh G. Crystal-facet-dependent denitrosylation: modulation of NO release from S-nitrosothiols by Cu 2O polymorphs. Chem Sci 2019; 10:5308-5318. [PMID: 31191887 PMCID: PMC6540961 DOI: 10.1039/c9sc01374a] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/24/2019] [Indexed: 01/07/2023] Open
Abstract
Nitric oxide (NO), a gaseous small molecule generated by the nitric oxide synthase (NOS) enzymes, plays key roles in signal transduction. The thiol groups present in many proteins and small molecules undergo nitrosylation to form the corresponding S-nitrosothiols. The release of NO from S-nitrosothiols is a key strategy to maintain the NO levels in biological systems. However, the controlled release of NO from the nitrosylated compounds at physiological pH remains a challenge. In this paper, we describe the synthesis and NO releasing ability of Cu2O nanomaterials and provide the first experimental evidence that the nanocrystals having different crystal facets within the same crystal system exhibit different activities toward S-nitrosothiols. We used various imaging techniques and time-dependent spectroscopic measurements to understand the nature of catalytically active species involved in the surface reactions. The denitrosylation reactions by Cu2O can be carried out multiple times without affecting the catalytic activity.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India .
| | - Punarbasu Roy
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India .
| | - Sanjay Prasad
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India .
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India .
| |
Collapse
|
19
|
Zhang J, Huang D, Wang C, Wang B, Fang H, Huo J, Liao W. Recent Progress in Protein S-Nitrosylation in Phytohormone Signaling. PLANT & CELL PHYSIOLOGY 2019; 60:494-502. [PMID: 30668813 DOI: 10.1093/pcp/pcz012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/15/2019] [Indexed: 06/09/2023]
Abstract
The free radical nitric oxide (NO) is a critical regulator in modulation of wide range of growth and developmental processes as well as environmental responses in plants. In most cases, NO interacts with plant hormones to regulate these processes. It is clear that NO might work through either transcriptional or post-translational level. The redox-based post-translational modification S-nitrosylation has been recognized as a NO-dependent regulatory mechanism in recent years. In general, S-nitrosylation can be understood as a product of reversible reaction where NO moiety group covalently attaches to thiol of cysteine residue resulting in the formation of S-nitrosothiol in target proteins. Recently, the crosstalk between S-nitrosylation and phytohormones has been emerging. Furthermore, several proteins involved in plant hormone signaling have been reported to undergo S-nitrosylation, which might subsequently mediate plant growth and defense response. In this review, we focus on the recent processes in protein S-nitrosylation in phytohormone signaling. In addition, both importance and challenges of future work on protein S-nitrosylation in plant hormone network are also highlighted.
Collapse
Affiliation(s)
- Jing Zhang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| | - Dengjing Huang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| | - Chunlei Wang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| | - Bo Wang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| | - Hua Fang
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| | - Jianqiang Huo
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| | - Weibiao Liao
- College of Horticulture, Gansu Agricultural University, 1 Yinmen Village, Anning District Lanzhou, P.R. China
| |
Collapse
|
20
|
Hasan MM, Manavalan B, Khatun MS, Kurata H. Prediction of S-nitrosylation sites by integrating support vector machines and random forest. Mol Omics 2019; 15:451-458. [DOI: 10.1039/c9mo00098d] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cysteine S-nitrosylation is a type of reversible post-translational modification of proteins, which controls diverse biological processes.
Collapse
Affiliation(s)
- Md. Mehedi Hasan
- Department of Bioscience and Bioinformatics
- Kyushu Institute of Technology
- Iizuka
- Japan
- Japan Society for the Promotion of Science
| | | | - Mst. Shamima Khatun
- Department of Bioscience and Bioinformatics
- Kyushu Institute of Technology
- Iizuka
- Japan
| | - Hiroyuki Kurata
- Department of Bioscience and Bioinformatics
- Kyushu Institute of Technology
- Iizuka
- Japan
- Biomedical Informatics R&D Center
| |
Collapse
|