1
|
Chakraborty S, Choudhuri A, Mishra A, Sengupta R. S-nitrosylation and S-glutathionylation: Lying at the forefront of redox dichotomy or a visible synergism? Biochem Biophys Res Commun 2025; 761:151734. [PMID: 40179738 DOI: 10.1016/j.bbrc.2025.151734] [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: 11/22/2024] [Revised: 03/06/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
The discovery of novel oxidoreductases and their specific functional revelations as cellular disulfide reductants, S-denitrosylases, or S-deglutathionylases, alongside the well-established major redoxins/antioxidant systems comprising thioredoxin and glutaredoxin, enlarges the spectrum of redox players in the intracellular milieu as well as pushes us to stand at the crossroads concerning the choice of antioxidants that can serve the benefit of catalyzing their cognate protein/non-protein substrates with better efficiencies than the rest. The complexity is extended to exploring the redundancy amongst the redoxin systems and identifying their overlapping or unique substrate preferences to intervene with oxidative or nitrosative stress-induced reversible protein posttranslational modifications such as S-nitrosylation and S-glutathionylation. Contrary to popular expectations of reiterating the theoretical and evidence-based existence of these modifications, the current review aims to take the first leap in delineating the logical reasons behind the competing susceptibility of reactive cysteine thiols toward either or both redox modifications and their subsequent extent of stability in the presence of cellular reductants (thioredoxin, glutaredoxin, thioredoxin-like mimetic or lipoic acid, dihydrolipoic acid, and glutathione), thus rebuilding the underpinnings of a 'redox-interactome' that can further pave the way for the global mapping of ideal substrates exhibiting stringencies or synergism in the context of translational redox research.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
2
|
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
|
3
|
Cui X, Gu J, Liu P, Lu R, Ren Z, Zhang Y, Wang F, Qi M, Liu Y, Li T. Genome-wide identification and characterization of the thioredoxin (TRX) gene family in tomato (Solanum lycopersicum) and a functional analysis of SlTRX2 under salt stress. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 220:109478. [PMID: 39826344 DOI: 10.1016/j.plaphy.2025.109478] [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: 10/25/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Thioredoxin is a multifunctional acidic protein widely presented in organisms that regulates intracellular redox processes, participating in a series of biochemical reactions in cells to affect the growth and development of plants. Although the thioredoxin (TRX) gene family has been widespread recognized across various plant species, and the tomato genome has been sequenced for years now, of tomato (Solanum lycopersicum) has remained largely uncharted in terms of identifying and unraveling the functional intricacies of is TRX genes. In this study, 53 SlTRX genes were identified, unevenly distributed across 11 of the 12 tomato chromosomes. These 53 SlTRX genes were categorized into 4 distinct subfamilies based on their evolutionary kinship and phylogenetic development. Expression profiling reveals that most of SlTRX genes exhibited distinct expression patterns across various tissues and developmental stages. In addition, the gene structure, conserved protein motifs and cis-elements of 53 SlTRX genes were analyzed simultaneously. In our rigorous in silico expression analysis, 8 SlTRX genes were meticulously selected for subsequent experiments. Subcellular localization indicated that these 8 SlTRX genes were localized in chloroplasts. Furthermore, these 8 SlTRX genes were responsive to abiotic stress (salt, drought and cold stress) under the qRT-PCR analysis, and their different expression patterns under diverse types of treatments indicated their possible roles in stress tolerance in tomato. Based on these results, SlTRX2, whose expression level continued to increase under salt stress, was selected for silencing to further investigate its function, and furthermore, silencing SlTRX2 inhibited plant growth and led to a significant reduction in photosynthesis under salt stress. Yeast two-hybrid and luciferase complementation imaging assays demonstrated that SlTRX2 may regulate tomato salt resistance by affecting related photosynthetic genes. Thus, our study establishes a valuable resource for further analysis on biological functions of SlTRX genes and will provide important insights in the mechanism of action under stress.
Collapse
Affiliation(s)
- Xiaoyu Cui
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Jiamao Gu
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Pengkun Liu
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Ruiqin Lu
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Zhen Ren
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Yueqi Zhang
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Feng Wang
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Mingfang Qi
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China
| | - Yufeng Liu
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China.
| | - Tianlai Li
- The Modern Facilities Horticultural Engineering Technology Center, Shenyang Agricultural University, 110866, Shenyang, Liaoning, China; The Key Laboratory of Protected Horticulture, Ministry of Education, 110866, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
Islam MI, Sultana S, Padmanabhan N, Rashid MU, Siddiqui TJ, Coombs KM, Vitiello PF, Karimi-Abdolrezaee S, Eftekharpour E. Thioredoxin-1 protein interactions in neuronal survival and neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167548. [PMID: 39454970 DOI: 10.1016/j.bbadis.2024.167548] [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: 07/03/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Neuronal cell death remains the principal pathophysiologic hallmark of neurodegenerative diseases and the main challenge for treatment strategies. Thioredoxin1 (Trx1) is a major cytoplasmic thiol oxidoreductase protein involved in redox signaling, hence a crucial player in maintaining neuronal health. Trx1 levels are notably reduced in neurodegenerative diseases including Alzheimer's and Parkinson's diseases, however, the impact of this decrease on neuronal physiology remains largely unexplored. This is mainly due to the nature of Trx1 redox regulatory role which is afforded by a rapid electron transfer to its oxidized protein substrates. During this reaction, Trx1 forms a transient bond with the oxidized disulfide bond in the substrate. This is a highly fast reaction which makes the identification of Trx1 substrates a technically challenging task. In this project, we utilized a transgenic mouse model expressing a Flag-tagged mutant form of Trx1 that can form stable disulfide bonds with its substrates, hence allowing identification of the Trx1 target proteins. Autophagy is a vital housekeeping process in neurons that is critical for degradation of damaged proteins under oxidative stress conditions and is interrupted in neurodegenerative diseases. Given Trx1's suggested involvement in autophagy, we aimed to identify potential Trx1 substrates following pharmacologic induction of autophagy in primary cortical neurons. Treatment with rapamycin, an autophagy inducer, significantly reduced neurite outgrowth and caused cytoskeletal alterations. Using immunoprecipitation and mass spectrometry, we have identified 77 Trx1 target proteins associated with a wide range of cellular functions including cytoskeletal organization and neurodegenerative diseases. Focusing on neuronal cytoskeleton organization, we identified a novel interaction between Trx1 and RhoB which was confirmed in genetic models of Trx1 downregulation in primary neuronal cultures and HT22 mouse immortalized hippocampal neurons. The applicability of these findings was also tested against the publicly available proteomic data from Alzheimer's patients. Our study uncovers a novel role for Trx1 in regulating neuronal cytoskeleton organization and provides a mechanistic explanation for its multifaceted role in the physiology and pathology of the nervous system, offering new insights into the molecular mechanisms underlying neurodegeneration.
Collapse
Affiliation(s)
- Md Imamul Islam
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | - Shakila Sultana
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | - Nirmala Padmanabhan
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | | | - Tabrez J Siddiqui
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | - Kevin M Coombs
- Department of Medical Microbiology, University of Manitoba, Canada
| | - Peter F Vitiello
- Department of Pediatrics, the University of Oklahoma Health Sciences Center, USA
| | | | | |
Collapse
|
5
|
Jiménez A, Martí MC, Sevilla F. Oxidative post-translational modifications of plant antioxidant systems under environmental stress. PHYSIOLOGIA PLANTARUM 2025; 177:e70118. [PMID: 39968905 PMCID: PMC11837463 DOI: 10.1111/ppl.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
Plants are often subject to environmental challenges posed by abiotic and biotic stresses, which are increasing under the current climate change conditions, provoking a loss in crop yield worldwide. Plants must cope with adverse situations such as increasing temperatures, air pollution or loss of agricultural land due to salinity, drought, contamination, and pathogen attacks, among others. Plants under stress conditions increase the production of reactive oxygen-, nitrogen-, and sulphur species (ROS/RNS/RSS), whose concentrations must be tightly regulated. The enzymatic antioxidant system and metabolites are in charge of their control to avoid their deleterious effects on cellular components, allowing their participation in signalling events. As signalling molecules, reactive species are involved in plant responses to the environment through post-translational modifications (PTMs) of proteins, which, in turn, may regulate the structure, function, and location of the antioxidant proteins by oxidative/nitrosative/persulfure modifications of different amino acid residues. In this review, we examine the different effects of these post-translational modifications, which are emerging as a fine-tuned point of control of the antioxidant systems involved in plant responses to climate change, a growing threat to crop production.
Collapse
Affiliation(s)
- Ana Jiménez
- Department of Stress Biology and Plant PathologyCEBAS‐CSICMurciaSpain
| | | | - Francisca Sevilla
- Department of Stress Biology and Plant PathologyCEBAS‐CSICMurciaSpain
| |
Collapse
|
6
|
Liang F, Wang M, Li J, Guo J. The evolution of S-nitrosylation detection methodology and the role of protein S-nitrosylation in various cancers. Cancer Cell Int 2024; 24:408. [PMID: 39702281 DOI: 10.1186/s12935-024-03568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024] Open
Abstract
S-nitrosylation (SNO) modification, a nitric oxide (NO)-mediated post-translational modification (PTM) of proteins, plays an important role in protein microstructure, degradation, activity, and stability. Due to the presence of reducing agents, the SNO modification process mediated by NO derivatives is often reversible and unstable. This reversible transformation between SNO modification and denitrification often influences the structure, activity, and function of proteins. The reversibility of SNO modifications also poses a challenge when verifying changes in the biological functions of proteins. Moreover, SNO modification of key signaling pathway proteins, such as caspase-3, NF-κB, and Bcl-2, can affect tumor proliferation, invasion, and apoptosis. The SNO-modified proteins play important roles in both promoting and inhibiting cancer, which indirectly confirms the duality and complexity of SNO modification functions. This article reviews the biological significance of various SNO-modified proteins in different cancers, providing a theoretical basis for determining whether the related changes of SNO-modified proteins are universal in cancers. Additionally, this review presents a comprehensive and detailed summary of the evolution of detection methods for SNO-modified proteins, providing a possible methodological basis for future research on SNO-modified proteins.
Collapse
Affiliation(s)
- Feng Liang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Min Wang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jie Guo
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Oh CK, Nakamura T, Zhang X, Lipton SA. Redox regulation, protein S-nitrosylation, and synapse loss in Alzheimer's and related dementias. Neuron 2024; 112:3823-3850. [PMID: 39515322 PMCID: PMC11624102 DOI: 10.1016/j.neuron.2024.10.013] [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: 04/22/2024] [Revised: 09/12/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Redox-mediated posttranslational modification, as exemplified by protein S-nitrosylation, modulates protein activity and function in both health and disease. Here, we review recent findings that show how normal aging, infection/inflammation, trauma, environmental toxins, and diseases associated with protein aggregation can each trigger excessive nitrosative stress, resulting in aberrant protein S-nitrosylation and hence dysfunctional protein networks. These redox reactions contribute to the etiology of multiple neurodegenerative disorders as well as systemic diseases. In the CNS, aberrant S-nitrosylation reactions of single proteins or, in many cases, interconnected networks of proteins lead to dysfunctional pathways affecting endoplasmic reticulum (ER) stress, inflammatory signaling, autophagy/mitophagy, the ubiquitin-proteasome system, transcriptional and enzymatic machinery, and mitochondrial metabolism. Aberrant protein S-nitrosylation and transnitrosylation (transfer of nitric oxide [NO]-related species from one protein to another) trigger protein aggregation, neuronal bioenergetic compromise, and microglial phagocytosis, all of which contribute to the synapse loss that underlies cognitive decline in Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Chang-Ki Oh
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tomohiro Nakamura
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xu Zhang
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
8
|
Gřešková A, Petřivalský M. Thioredoxin System in Insects: Uncovering the Roles of Thioredoxins and Thioredoxin Reductase beyond the Antioxidant Defences. INSECTS 2024; 15:797. [PMID: 39452373 PMCID: PMC11508645 DOI: 10.3390/insects15100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Increased levels of reactive oxygen species (ROS) produced during aerobic metabolism in animals can negatively affect the intracellular redox status, cause oxidative stress and interfere with physiological processes in the cells. The antioxidant defence regulates ROS levels by interplaying diverse enzymes and non-enzymatic metabolites. The thioredoxin system, consisting of the enzyme thioredoxin reductase (TrxR), the redox-active protein thioredoxin (Trx) and NADPH, represent a crucial component of antioxidant defence. It is involved in the signalling and regulation of multiple developmental processes, such as cell proliferation or apoptotic death. Insects have evolved unique variations of TrxR, which resemble mammalian enzymes in overall structure and catalytic mechanisms, but the selenocysteine-cysteine pair in the active site is replaced by a cysteine-cysteine pair typical of bacteria. Moreover, the role of the thioredoxin system in insects is indispensable due to the absence of glutathione reductase, an essential enzyme of the glutathione system. However, the functions of the Trx system in insects are still poorly characterised. In the present review, we provide a critical overview of the current knowledge on the insect Trx system, focusing mainly on TrxR's role in the antioxidant and immune system of model insect species.
Collapse
Affiliation(s)
| | - Marek Petřivalský
- Department of Biochemistry, Faculty of Science, Palacký University in Olomouc, Šlechtitelů 27, 77900 Olomouc, Czech Republic
| |
Collapse
|
9
|
Bazbaz W, Kartawy M, Hamoudi W, Ojha SK, Khaliulin I, Amal H. The Role of Thioredoxin System in Shank3 Mouse Model of Autism. J Mol Neurosci 2024; 74:90. [PMID: 39347996 PMCID: PMC11457715 DOI: 10.1007/s12031-024-02270-y] [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: 08/23/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by difficulties in social interaction and communication, repetitive behaviors, and restricted interests. Unfortunately, the underlying molecular mechanism behind ASD remains unknown. It has been reported that oxidative and nitrosative stress are strongly linked to ASD. We have recently found that nitric oxide (NO•) and its products play an important role in this disorder. One of the key proteins associated with NO• is thioredoxin (Trx). We hypothesize that the Trx system is altered in the Shank3 KO mouse model of autism, which may lead to a decreased activity of the nuclear factor erythroid 2-related factor 2 (Nrf2), resulting in oxidative stress, and thus, contributing to ASD-related phenotypes. To test this hypothesis, we conducted in vivo behavioral studies and used primary cortical neurons derived from the Shank3 KO mice and human SH-SY5Y cells with SHANK3 mutation. We showed significant changes in the levels and activity of Trx redox proteins in the Shank3 KO mice. A Trx1 inhibitor PX-12 decreased Trx1 and Nrf2 expression in wild-type mice, causing abnormal alterations in the levels of synaptic proteins and neurotransmission markers, and an elevation of nitrosative stress. Trx inhibition resulted in an ASD-like behavioral phenotype, similar to that of Shank3 KO mice. Taken together, our findings confirm the strong link between the Trx system and ASD pathology, including the increased oxidative/nitrosative stress, and synaptic and behavioral deficits. The results of this study may pave the way for identifying novel drug targets for ASD.
Collapse
Affiliation(s)
- Wisam Bazbaz
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
10
|
Kumari R, Kapoor P, Mir BA, Singh M, Parrey ZA, Rakhra G, Parihar P, Khan MN, Rakhra G. Unlocking the versatility of nitric oxide in plants and insights into its molecular interplays under biotic and abiotic stress. Nitric Oxide 2024; 150:1-17. [PMID: 38972538 DOI: 10.1016/j.niox.2024.07.002] [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: 02/07/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
In plants, nitric oxide (NO) has become a versatile signaling molecule essential for mediating a wide range of physiological processes under various biotic and abiotic stress conditions. The fundamental function of NO under various stress scenarios has led to a paradigm shift in which NO is now seen as both a free radical liberated from the toxic product of oxidative metabolism and an agent that aids in plant sustenance. Numerous studies on NO biology have shown that NO is an important signal for germination, leaf senescence, photosynthesis, plant growth, pollen growth, and other processes. It is implicated in defense responses against pathogensas well as adaptation of plants in response to environmental cues like salinity, drought, and temperature extremes which demonstrates its multifaceted role. NO can carry out its biological action in a variety of ways, including interaction with protein kinases, modifying gene expression, and releasing secondary messengers. In addition to these signaling events, NO may also be in charge of the chromatin modifications, nitration, and S-nitrosylation-induced posttranslational modifications (PTM) of target proteins. Deciphering the molecular mechanism behind its essential function is essential to unravel the regulatory networks controlling the responses of plants to various environmental stimuli. Taking into consideration the versatile role of NO, an effort has been made to interpret its mode of action based on the post-translational modifications and to cover shreds of evidence for increased growth parameters along with an altered gene expression.
Collapse
Affiliation(s)
- Ritu Kumari
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Preedhi Kapoor
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Bilal Ahmad Mir
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Maninder Singh
- Department of Biotechnology and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Zubair Ahmad Parrey
- Plant Physiology and Biochemistry Section, Department of Botany, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Gurseen Rakhra
- Department of Nutrition & Dietetics, Faculty of Allied Health Sciences, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, 121004, India
| | - Parul Parihar
- Department of Biosciences and Biotechnology, Banasthali Vidyapith, Rajasthan, 304022, India
| | - M Nasir Khan
- Renewable Energy and Environmental Technology Center, University of Tabuk, Tabuk, 47913, Saudi Arabia
| | - Gurmeen Rakhra
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
11
|
Chakraborty S, Mishra A, Choudhuri A, Bhaumik T, Sengupta R. Leveraging the redundancy of S-denitrosylases in response to S-nitrosylation of caspases: Experimental strategies and beyond. Nitric Oxide 2024; 149:18-31. [PMID: 38823434 DOI: 10.1016/j.niox.2024.05.002] [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: 04/18/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Redox-based protein posttranslational modifications, such as S-nitrosylation of critical, active site cysteine thiols have garnered significant clinical attention and research interest, reasoning for one of the crucial biological implications of reactive messenger molecule, nitric oxide in the cellular repertoire. The stringency of the S-(de)nitrosylation-based redox switch governs the activity and contribution of several susceptible enzymes in signal transduction processes and diverse pathophysiological settings, thus establishing it as a transient yet reasonable, and regulated mechanism of NO adduction and release. Notably, endogenous proteases like cytosolic and mitochondrial caspases with a molecular weight ranging from 33 to 55 kDa are susceptible to performing this biochemistry in the presence of major oxidoreductases, which further unveils the enormous redox-mediated regulational control of caspases in the etiology of diseases. In addition to advancing the progress of the current state of understanding of 'redox biochemistry' in the field of medicine and enriching the existing dynamic S-nitrosoproteome, this review stands as a testament to an unprecedented shift in the underpinnings for redundancy and redox relay between the major redoxin/antioxidant systems, fine-tuning of which can command the apoptotic control of caspases at the face of nitro-oxidative stress. These intricate functional overlaps and cellular backups, supported rationally by kinetically favorable reaction mechanisms suggest the physiological relevance of identifying and involving such cognate substrates for cellular S-denitrosylases that can shed light on the bigger picture of extensively proposing targeted therapies and redox-based drug designing to potentially alleviate the side effects of NOx/ROS in disease pathogenesis.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Tamal Bhaumik
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
12
|
Assadsangabi A, Evans CA, Corfe BM, Lobo AJ. Exploring Predictive Biomarkers of Relapse in Ulcerative Colitis: A Proteomics Approach. Inflamm Bowel Dis 2024; 30:808-819. [PMID: 37889841 DOI: 10.1093/ibd/izad241] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Indexed: 10/29/2023]
Abstract
INTRODUCTION AND AIMS Risk stratification of subjects with a history of inflammatory bowel disease (IBD) into those likely to relapse and those who will remain quiescent continues to be a significant challenge. The aim of this study was to investigate whether certain proteomic signature profiles or biomarkers during remission are associated with future disease relapse in patients with ulcerative colitis (UC). METHODS Endoscopic rectal samples from patients with UC in clinical, endoscopic, and histological remission at index endoscopy were collected, as well as samplers from normal control individuals. The patients were stratified to early relapsers (ERs) if they developed clinical signs of UC flare within 6 months of index endoscopy or nonrelapsers (NRs) if there was no relapse after 36 months of follow-up. The pooled rectal samples from ERs, NRs, and control individuals were subjected to nano-liquid chromatography and tandem mass spectrometry as per standard iTRAQ (isobaric tags for relative and absolute quantitation) workflow methodology. Selected proteomics-yielded candidates were subjected to orthogonal validation via immunoblotting, in a biomarker discovery exercise. RESULTS Sixty-one patients were included, of whom 8 had clinical relapse within 6 months from the index endoscopy, and 43 patients had no clinical symptoms of relapse within the 36-month follow-up period. Ten patients who had clinical signs of relapse between 6 and 36 months were excluded. Seventeen control individuals were also included. Soluble proteomics analyses between ERs, NRs, and control individuals revealed a series of upregulated and downregulated proteins. Following orthogonal validation, upregulated TRX (P = .001) and IGHA1 (P = .001) were observed in ERs relative to NRs. CONCLUSIONS Several novel candidate tissue biomarkers have been identified in this study, which could discriminate patients with UC at risk of early relapse from those in long-term sustained remission. Our findings may pave the way for pre-emptive UC disease monitoring and therapeutic decision making.
Collapse
Affiliation(s)
- Arash Assadsangabi
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
- Gastroenterology Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Gastroenterology Department, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Caroline A Evans
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
- Biological and Systems Engineering Group, ChELSI Institute, Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Bernard M Corfe
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Alan J Lobo
- Molecular Gastroenterology Research Group, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
- Gastroenterology Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom
| |
Collapse
|
13
|
Cecerska-Heryć E, Wiśniewska Z, Serwin N, Polikowska A, Goszka M, Engwert W, Michałów J, Pękała M, Budkowska M, Michalczyk A, Dołęgowska B. Can Compounds of Natural Origin Be Important in Chemoprevention? Anticancer Properties of Quercetin, Resveratrol, and Curcumin-A Comprehensive Review. Int J Mol Sci 2024; 25:4505. [PMID: 38674092 PMCID: PMC11050349 DOI: 10.3390/ijms25084505] [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: 02/15/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Malignant tumors are the second most common cause of death worldwide. More attention is being paid to the link between the body's impaired oxidoreductive balance and cancer incidence. Much attention is being paid to polyphenols derived from plants, as one of their properties is an antioxidant character: the ability to eliminate reactive oxygen and nitrogen species, chelate specific metal ions, modulate signaling pathways affecting inflammation, and raise the level and activity of antioxidant enzymes while lowering those with oxidative effects. The following three compounds, resveratrol, quercetin, and curcumin, are polyphenols modulating multiple molecular targets, or increasing pro-apoptotic protein expression levels and decreasing anti-apoptotic protein expression levels. Experiments conducted in vitro and in vivo on animals and humans suggest using them as chemopreventive agents based on antioxidant properties. The advantage of these natural polyphenols is low toxicity and weak adverse effects at higher doses. However, the compounds discussed are characterized by low bioavailability and solubility, which may make achieving the blood concentrations needed for the desired effect challenging. The solution may lie in derivatives of naturally occurring polyphenols subjected to structural modifications that enhance their beneficial effects or work on implementing new ways of delivering antioxidants that improve their solubility and bioavailability.
Collapse
Affiliation(s)
- Elżbieta Cecerska-Heryć
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Zofia Wiśniewska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Natalia Serwin
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Aleksandra Polikowska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Małgorzata Goszka
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Weronika Engwert
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Jaśmina Michałów
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Maja Pękała
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Anna Michalczyk
- Department of Psychiatry, Pomeranian Medical University of Szczecin, Broniewskiego 26, 71-460 Szczecin, Poland;
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (Z.W.); (N.S.); (A.P.); (M.G.); (W.E.); (J.M.); (M.P.); (B.D.)
| |
Collapse
|
14
|
Lambert L, de Carpentier F, André P, Marchand CH, Danon A. Type II metacaspase mediates light-dependent programmed cell death in Chlamydomonas reinhardtii. PLANT PHYSIOLOGY 2024; 194:2648-2662. [PMID: 37971939 PMCID: PMC10980519 DOI: 10.1093/plphys/kiad618] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/12/2023] [Accepted: 10/22/2023] [Indexed: 11/19/2023]
Abstract
Among the crucial processes that preside over the destiny of cells from any type of organism are those involving their self-destruction. This process is well characterized and conceptually logical to understand in multicellular organisms; however, the levels of knowledge and comprehension of its existence are still quite enigmatic in unicellular organisms. We use Chlamydomonas (Chlamydomonas reinhardtii) to lay the foundation for understanding the mechanisms of programmed cell death (PCD) in a unicellular photosynthetic organism. In this paper, we show that while PCD induces the death of a proportion of cells, it allows the survival of the remaining population. A quantitative proteomic analysis aiming at unveiling the proteome of PCD in Chlamydomonas allowed us to identify key proteins that led to the discovery of essential mechanisms. We show that in Chlamydomonas, PCD relies on the light dependence of a photosynthetic organism to generate reactive oxygen species and induce cell death. Finally, we obtained and characterized mutants for the 2 metacaspase genes in Chlamydomonas and showed that a type II metacaspase is essential for PCD execution.
Collapse
Affiliation(s)
- Lou Lambert
- Institut de Biologie Paris Seine, UMR 7238, CNRS, Sorbonne Université, Paris 75005, France
| | - Félix de Carpentier
- Institut de Biologie Paris Seine, UMR 7238, CNRS, Sorbonne Université, Paris 75005, France
- Doctoral School of Plant Sciences, Université Paris-Saclay, Saint-Aubin 91190, France
| | - Phuc André
- Institut de Biologie Paris Seine, UMR 7238, CNRS, Sorbonne Université, Paris 75005, France
| | - Christophe H Marchand
- Institut de Biologie Paris Seine, UMR 7238, CNRS, Sorbonne Université, Paris 75005, France
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique (CNRS), Paris F-75005, France
| | - Antoine Danon
- Institut de Biologie Paris Seine, UMR 7238, CNRS, Sorbonne Université, Paris 75005, France
| |
Collapse
|
15
|
Zhou HL, Grimmett ZW, Venetos NM, Stomberski CT, Qian Z, McLaughlin PJ, Bansal PK, Zhang R, Reynolds JD, Premont RT, Stamler JS. An enzyme that selectively S-nitrosylates proteins to regulate insulin signaling. Cell 2023; 186:5812-5825.e21. [PMID: 38056462 PMCID: PMC10794992 DOI: 10.1016/j.cell.2023.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/01/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Acyl-coenzyme A (acyl-CoA) species are cofactors for numerous enzymes that acylate thousands of proteins. Here, we describe an enzyme that uses S-nitroso-CoA (SNO-CoA) as its cofactor to S-nitrosylate multiple proteins (SNO-CoA-assisted nitrosylase, SCAN). Separate domains in SCAN mediate SNO-CoA and substrate binding, allowing SCAN to selectively catalyze SNO transfer from SNO-CoA to SCAN to multiple protein targets, including the insulin receptor (INSR) and insulin receptor substrate 1 (IRS1). Insulin-stimulated S-nitrosylation of INSR/IRS1 by SCAN reduces insulin signaling physiologically, whereas increased SCAN activity in obesity causes INSR/IRS1 hypernitrosylation and insulin resistance. SCAN-deficient mice are thus protected from diabetes. In human skeletal muscle and adipose tissue, SCAN expression increases with body mass index and correlates with INSR S-nitrosylation. S-nitrosylation by SCAN/SNO-CoA thus defines a new enzyme class, a unique mode of receptor tyrosine kinase regulation, and a revised paradigm for NO function in physiology and disease.
Collapse
Affiliation(s)
- Hua-Lin Zhou
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zachary W Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nicholas M Venetos
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Colin T Stomberski
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Zhaoxia Qian
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Precious J McLaughlin
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Puneet K Bansal
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rongli Zhang
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - James D Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Anesthesiology and Perioperative Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Richard T Premont
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Stamler
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
16
|
Chakraborty S, Choudhuri A, Mishra A, Bhattacharyya C, Billiar TR, Stoyanovsky DA, Sengupta R. Nitric oxide and thioredoxin modulate the activity of caspase 9 in HepG2 cells. Biochim Biophys Acta Gen Subj 2023; 1867:130452. [PMID: 37652366 PMCID: PMC10592080 DOI: 10.1016/j.bbagen.2023.130452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
The interdependent and finely tuned balance between the well-established redox-based modification, S-nitrosylation, and its counteractive mechanism of S-nitrosothiol degradation, i.e., S-denitrosylation of biological protein or non-protein thiols defines the cellular fate in the context of redox homeostasis. S-nitrosylation of cysteine residues by S-nitrosoglutathione, S-nitroso-L-cysteine-like physiological and S-nitroso-L-cysteine ethyl ester-like synthetic NO donors inactivate Caspase-3, 8, and 9, thereby hindering their apoptotic activity. However, spontaneous restoration of their activity upon S-denitrosylation of S-nitrosocaspases into their reduced, free thiol active states, aided by the members of the ubiquitous cellular redoxin (thioredoxin/ thioredoxin reductase/ NADPH) and low molecular weight dithiol (lipoic acid/ lipoamide dehydrogenase/ dihydrolipoic acid/ NADPH) systems imply a direct relevance to their proteolytic activities and further downstream signaling cascades. Additionally, our previous and current findings offer crucial insight into the concept of redundancy between thioredoxin and lipoic acid systems, and the redox-modulated control of the apoptotic and proteolytic activity of caspases, triggering their cyto- and neurotoxic effects in response to nitro-oxidative stress. Thus, this might lay the foundation for the exogenous introduction of precise and efficient NO or related donor drug delivery systems that can directly participate in catering to the S-(de)-nitrosylation-mediated functional outcomes of the cysteinyl proteases in pathophysiological settings.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology, Amity University, Kolkata 700135, West Bengal, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology, Amity University, Kolkata 700135, West Bengal, India
| | - Akansha Mishra
- Amity Institute of Biotechnology, Amity University, Kolkata 700135, West Bengal, India
| | - Camelia Bhattacharyya
- Amity Institute of Biotechnology, Amity University, Kolkata 700135, West Bengal, India
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Rajib Sengupta
- Amity Institute of Biotechnology, Amity University, Kolkata 700135, West Bengal, India; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
17
|
Dent MR, DeMartino AW. Nitric oxide and thiols: Chemical biology, signalling paradigms and vascular therapeutic potential. Br J Pharmacol 2023:10.1111/bph.16274. [PMID: 37908126 PMCID: PMC11058123 DOI: 10.1111/bph.16274] [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: 06/13/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023] Open
Abstract
Nitric oxide (• NO) interactions with biological thiols play crucial, but incompletely determined, roles in vascular signalling and other biological processes. Here, we highlight two recently proposed signalling paradigms: (1) the formation of a vasodilating labile nitrosyl ferrous haem (NO-ferrohaem) facilitated by thiols via thiyl radical generation and (2) polysulfides/persulfides and their interaction with • NO. We also describe the specific (bio)chemical routes in which • NO and thiols react to form S-nitrosothiols, a broad class of small molecules, and protein post-translational modifications that can influence protein function through catalytic site or allosteric structural changes. S-Nitrosothiol formation depends upon cellular conditions, but critically, an appropriate oxidant for either the thiol (yielding a thiyl radical) or • NO (yielding a nitrosonium [NO+ ]-donating species) is required. We examine the roles of these collective • NO/thiol species in vascular signalling and their cardiovascular therapeutic potential.
Collapse
Affiliation(s)
- Matthew R. Dent
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony W. DeMartino
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Seth D, Stomberski CT, McLaughlin PJ, Premont RT, Lundberg K, Stamler JS. Comparison of the Nitric Oxide Synthase Interactomes and S-Nitroso-Proteomes: Furthering the Case for Enzymatic S-Nitrosylation. Antioxid Redox Signal 2023; 39:621-634. [PMID: 37053107 PMCID: PMC10619892 DOI: 10.1089/ars.2022.0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/13/2023] [Accepted: 04/08/2023] [Indexed: 04/14/2023]
Abstract
Aims: S-nitrosylation of proteins is the main mechanism through which nitric oxide (NO) regulates cellular function and likely represents the archetype redox-based signaling system across aerobic and anaerobic organisms. How NO generated by different nitric oxide synthase (NOS) isoforms leads to specificity of S-nitrosylation remains incompletely understood. This study aimed to identify proteins interacting with, and whose S-nitrosylation is mediated by, human NOS isoforms in the same cellular system, thereby illuminating the contribution of individual NOSs to specificity. Results: Of the hundreds of proteins interacting with each NOS, many were also S-nitrosylated. However, a large proportion of S-nitrosylated proteins (SNO-proteins) did not associate with NOS. Moreover, most NOS interactors and SNO-proteins were unique to each isoform. The amount of NO produced by each NOS isoform was unrelated to the numbers of SNO-proteins. Thus, NOSs promoted S-nitrosylation of largely distinct sets of target proteins. Different signaling pathways were enriched downstream of each NOS. Innovation and Conclusion: The interactomes and SNOomes of individual NOS isoforms were largely distinct. Only a small fraction of SNO-proteins interacted with their respective NOS. Amounts of S-nitrosylation were unrelated to the amount of NO generated by NOSs. These data argue against free diffusion of NO or NOS interactions as being necessary or sufficient for S-nitrosylation and favor roles for additional enzymes and/or regulatory elements in imparting SNO-protein specificity. Antioxid. Redox Signal. 39, 621-634.
Collapse
Affiliation(s)
- Divya Seth
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Colin T. Stomberski
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Precious J. McLaughlin
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Richard T. Premont
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Kathleen Lundberg
- Center for Proteomics and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jonathan S. Stamler
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
19
|
Liu F, Liu C, Chai Q, Zhao C, Meng H, Xue X, Yao TP, Zhang Y. Discovery of the First Irreversible HDAC6 Isoform Selective Inhibitor with Potent Anti-Multiple Myeloma Activity. J Med Chem 2023; 66:10080-10091. [PMID: 37463038 DOI: 10.1021/acs.jmedchem.3c00977] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
In our previous research, a series of phenylsulfonylfuroxan-based hydroxamates were developed, among which compound 1 exhibited remarkable in vitro and in vivo antitumor potency due to its histone deacetylase (HDAC) inhibitory and nitric oxide (NO)-donating activities. Herein, the in-depth study of compound 1 revealed that this HDAC inhibitor-NO donor hybrid could enduringly increase the intracellular levels of acetyl histones and acetyl α-tubulin, which could be ascribed to its irreversible inhibition toward class I HDACs and HDAC6. Structural modification of compound 1 led to a novel phenylsulfonylfuroxan-based hydroxamate 4, which exhibited considerable HDAC6 inhibitory activity and selectivity. Furthermore, compound 4 could inhibit intracellular HDAC6 both selectively and irreversibly. To the best of our knowledge, this is the first research reporting the irreversible inhibition of HDAC6. It was also demonstrated that compared with ACY-241 (a reversible HDAC6 inhibitor in clinical trials), the irreversible HDAC6 selective inhibitor 4 exhibited not only superior anti-multiple myeloma activity but also improved therapeutic index.
Collapse
Affiliation(s)
- Fengling Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chunxi Liu
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qipeng Chai
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chunlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hongwei Meng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Tso-Pang Yao
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - Yingjie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
20
|
Kalinina EV, Novichkova MD. S-Glutathionylation and S-Nitrosylation as Modulators of Redox-Dependent Processes in Cancer Cell. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:924-943. [PMID: 37751864 DOI: 10.1134/s0006297923070064] [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: 02/10/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 09/28/2023]
Abstract
Development of oxidative/nitrosative stress associated with the activation of oncogenic pathways results from the increase in the generation of reactive oxygen and nitrogen species (ROS/RNS) in tumor cells, where they can have a dual effect. At high concentrations, ROS/RNS cause cell death and limit tumor growth at certain phases of its development, while their low amounts promote oxidative/nitrosative modifications of key redox-dependent residues in regulatory proteins. The reversibility of such modifications as S-glutathionylation and S-nitrosylation that proceed through the electrophilic attack of ROS/RNS on nucleophilic Cys residues ensures the redox-dependent switch in the activity of signaling proteins, as well as the ability of these compounds to control cell proliferation and programmed cell death. The content of S-glutathionylated and S-nitrosylated proteins is controlled by the balance between S-glutathionylation/deglutathionylation and S-nitrosylation/denitrosylation, respectively, and depends on the cellular redox status. The extent of S-glutathionylation and S-nitrosylation of protein targets and their ratio largely determine the status and direction of signaling pathways in cancer cells. The review discusses the features of S-glutathionylation and S-nitrosylation reactions and systems that control them in cancer cells, as well as their relationship with redox-dependent processes and tumor growth.
Collapse
|
21
|
Kratzke M, Scaria G, Porter S, Kren B, Klein MA. Inhibition of Mitochondrial Antioxidant Defense and CDK4/6 in Mesothelioma. Molecules 2023; 28:molecules28114380. [PMID: 37298855 DOI: 10.3390/molecules28114380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
Advanced mesothelioma is considered an incurable disease and new treatment strategies are needed. Previous studies have demonstrated that mitochondrial antioxidant defense proteins and the cell cycle may contribute to mesothelioma growth, and that the inhibition of these pathways may be effective against this cancer. We demonstrated that the antioxidant defense inhibitor auranofin and the cyclin-dependent kinase 4/6 inhibitor palbociclib could decrease mesothelioma cell proliferation alone or in combination. In addition, we determined the effects of these compounds on colony growth, cell cycle progression, and the expression of key antioxidant defense and cell cycle proteins. Auranofin and palbociclib were effective in decreasing cell growth and inhibiting the above-described activity across all assays. Further study of this drug combination will elucidate the contribution of these pathways to mesothelioma activity and may reveal a new treatment strategy.
Collapse
Affiliation(s)
- Marian Kratzke
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
| | - George Scaria
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55417, USA
| | - Stephen Porter
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
| | - Betsy Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55417, USA
| | - Mark A Klein
- Hematology/Oncology Section, Primary Care Service Line, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55417, USA
| |
Collapse
|
22
|
Hobai IA. MECHANISMS OF CARDIAC DYSFUNCTION IN SEPSIS. Shock 2023; 59:515-539. [PMID: 36155956 DOI: 10.1097/shk.0000000000001997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Studies in animal models of sepsis have elucidated an intricate network of signaling pathways that lead to the dysregulation of myocardial Ca 2+ handling and subsequently to a decrease in cardiac contractile force, in a sex- and model-dependent manner. After challenge with a lethal dose of LPS, male animals show a decrease in cellular Ca 2+ transients (ΔCa i ), with intact myofilament function, whereas female animals show myofilament dysfunction, with intact ΔCa i . Male mice challenged with a low, nonlethal dose of LPS also develop myofilament desensitization, with intact ΔCa i . In the cecal ligation and puncture (CLP) model, the causative mechanisms seem similar to those in the LPS model in male mice and are unknown in female subjects. ΔCa i decrease in male mice is primarily due to redox-dependent inhibition of sarco/endoplasmic reticulum Ca 2+ ATP-ase (SERCA). Reactive oxygen species (ROS) are overproduced by dysregulated mitochondria and the enzymes NADPH/NADH oxidase, cyclooxygenase, and xanthine oxidase. In addition to inhibiting SERCA, ROS amplify cardiomyocyte cytokine production and mitochondrial dysfunction, making the process self-propagating. In contrast, female animals may exhibit a natural redox resilience. Myofilament dysfunction is due to hyperphosphorylation of troponin I, troponin T cleavage by caspase-3, and overproduction of cGMP by NO-activated soluble guanylate cyclase. Depleted, dysfunctional, or uncoupled mitochondria likely synthesize less ATP in both sexes, but the role of energy deficit is not clear. NO produced by NO synthase (NOS)-3 and mitochondrial NOSs, protein kinases and phosphatases, the processes of autophagy and sarco/endoplasmic reticulum stress, and β-adrenergic insensitivity may also play currently uncertain roles.
Collapse
Affiliation(s)
- Ion A Hobai
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
23
|
Abstract
Significance: Thioredoxin (Trx) is a powerful antioxidant that reduces protein disulfides to maintain redox stability in cells and is involved in regulating multiple redox-dependent signaling pathways. Recent Advance: The current accumulation of findings suggests that Trx participates in signaling pathways that interact with various proteins to manipulate their dynamic regulation of structure and function. These network pathways are critical for cancer pathogenesis and therapy. Promising clinical advances have been presented by most anticancer agents targeting such signaling pathways. Critical Issues: We herein link the signaling pathways regulated by the Trx system to potential cancer therapeutic opportunities, focusing on the coordination and strengths of the Trx signaling pathways in apoptosis, ferroptosis, immunomodulation, and drug resistance. We also provide a mechanistic network for the exploitation of therapeutic small molecules targeting the Trx signaling pathways. Future Directions: As research data accumulate, future complex networks of Trx-related signaling pathways will gain in detail. In-depth exploration and establishment of these signaling pathways, including Trx upstream and downstream regulatory proteins, will be critical to advancing novel cancer therapeutics. Antioxid. Redox Signal. 38, 403-424.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zhengjia Zhao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | | | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, China
| |
Collapse
|
24
|
Nagarajan N, Oka SI, Nah J, Wu C, Zhai P, Mukai R, Xu X, Kashyap S, Huang CY, Sung EA, Mizushima W, Titus AS, Takayama K, Mourad Y, Francisco J, Liu T, Chen T, Li H, Sadoshima J. Thioredoxin 1 promotes autophagy through transnitrosylation of Atg7 during myocardial ischemia. J Clin Invest 2023; 133:e162326. [PMID: 36480290 PMCID: PMC9888389 DOI: 10.1172/jci162326] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Modification of cysteine residues by oxidative and nitrosative stress affects structure and function of proteins, thereby contributing to the pathogenesis of cardiovascular disease. Although the major function of thioredoxin 1 (Trx1) is to reduce disulfide bonds, it can also act as either a denitrosylase or transnitrosylase in a context-dependent manner. Here we show that Trx1 transnitrosylates Atg7, an E1-like enzyme, thereby stimulating autophagy. During ischemia, Trx1 was oxidized at Cys32-Cys35 of the oxidoreductase catalytic center and S-nitrosylated at Cys73. Unexpectedly, Atg7 Cys545-Cys548 reduced the disulfide bond in Trx1 at Cys32-Cys35 through thiol-disulfide exchange and this then allowed NO to be released from Cys73 in Trx1 and transferred to Atg7 at Cys402. Experiments conducted with Atg7 C402S-knockin mice showed that S-nitrosylation of Atg7 at Cys402 promotes autophagy by stimulating E1-like activity, thereby protecting the heart against ischemia. These results suggest that the thiol-disulfide exchange and the NO transfer are functionally coupled, allowing oxidized Trx1 to mediate a salutary effect during myocardial ischemia through transnitrosylation of Atg7 and stimulation of autophagy.
Collapse
Affiliation(s)
- Narayani Nagarajan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Changgong Wu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Koichiro Takayama
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Tong Chen
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
25
|
Chakraborty S, Sircar E, Bhattacharyya C, Choudhuri A, Mishra A, Dutta S, Bhatta S, Sachin K, Sengupta R. S-Denitrosylation: A Crosstalk between Glutathione and Redoxin Systems. Antioxidants (Basel) 2022; 11:1921. [PMID: 36290644 PMCID: PMC9598160 DOI: 10.3390/antiox11101921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 08/27/2023] Open
Abstract
S-nitrosylation of proteins occurs as a consequence of the derivatization of cysteine thiols with nitric oxide (NO) and is often associated with diseases and protein malfunction. Aberrant S-nitrosylation, in addition to other genetic and epigenetic factors, has gained rapid importance as a prime cause of various metabolic, respiratory, and cardiac disorders, with a major emphasis on cancer and neurodegeneration. The S-nitrosoproteome, a term used to collectively refer to the diverse and dynamic repertoire of S-nitrosylated proteins, is relatively less explored in the field of redox biochemistry, in contrast to other covalently modified versions of the same set of proteins. Advancing research is gradually unveiling the enormous clinical importance of S-nitrosylation in the etiology of diseases and is opening up new avenues of prompt diagnosis that harness this phenomenon. Ever since the discovery of the two robust and highly conserved S-nitrosoglutathione reductase and thioredoxin systems as candidate denitrosylases, years of rampant speculation centered around the identification of specific substrates and other candidate denitrosylases, subcellular localization of both substrates and denitrosylases, the position of susceptible thiols, mechanisms of S-denitrosylation under basal and stimulus-dependent conditions, impact on protein conformation and function, and extrapolating these findings towards the understanding of diseases, aging and the development of novel therapeutic strategies. However, newer insights in the ever-expanding field of redox biology reveal distinct gaps in exploring the crucial crosstalk between the redoxins/major denitrosylase systems. Clarifying the importance of the functional overlap of the glutaredoxin, glutathione, and thioredoxin systems and examining their complementary functions as denitrosylases and antioxidant enzymatic defense systems are essential prerequisites for devising a rationale that could aid in predicting the extent of cell survival under high oxidative/nitrosative stress while taking into account the existence of the alternative and compensatory regulatory mechanisms. This review thus attempts to highlight major gaps in our understanding of the robust cellular redox regulation system, which is upheld by the concerted efforts of various denitrosylases and antioxidants.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Esha Sircar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, Uttarakhand, India
| | - Camelia Bhattacharyya
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Sreejita Dutta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Sneha Bhatta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Kumar Sachin
- Department of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| |
Collapse
|
26
|
Lundberg JO, Weitzberg E. Nitric oxide signaling in health and disease. Cell 2022; 185:2853-2878. [DOI: 10.1016/j.cell.2022.06.010] [Citation(s) in RCA: 346] [Impact Index Per Article: 115.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 10/16/2022]
|
27
|
Focus on Nitric Oxide Homeostasis: Direct and Indirect Enzymatic Regulation of Protein Denitrosation Reactions in Plants. Antioxidants (Basel) 2022; 11:antiox11071411. [PMID: 35883902 PMCID: PMC9311986 DOI: 10.3390/antiox11071411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Protein cysteines (Cys) undergo a multitude of different reactive oxygen species (ROS), reactive sulfur species (RSS), and/or reactive nitrogen species (RNS)-derived modifications. S-nitrosation (also referred to as nitrosylation), the addition of a nitric oxide (NO) group to reactive Cys thiols, can alter protein stability and activity and can result in changes of protein subcellular localization. Although it is clear that this nitrosative posttranslational modification (PTM) regulates multiple signal transduction pathways in plants, the enzymatic systems that catalyze the reverse S-denitrosation reaction are poorly understood. This review provides an overview of the biochemistry and regulation of nitro-oxidative modifications of protein Cys residues with a focus on NO production and S-nitrosation. In addition, the importance and recent advances in defining enzymatic systems proposed to be involved in regulating S-denitrosation are addressed, specifically cytosolic thioredoxins (TRX) and the newly identified aldo-keto reductases (AKR).
Collapse
|
28
|
Chakraborty S, Mukherjee P, Sengupta R. Ribonucleotide reductase: Implications of thiol S-nitrosylation and tyrosine nitration for different subunits. Nitric Oxide 2022; 127:26-43. [PMID: 35850377 DOI: 10.1016/j.niox.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/20/2022] [Accepted: 07/08/2022] [Indexed: 11/20/2022]
Abstract
Ribonucleotide reductase (RNR) is a multi-subunit enzyme responsible for catalyzing the rate-limiting step in the production of deoxyribonucleotides essential for DNA synthesis and repair. The active RNR complex is composed of multimeric R1 and R2 subunits. The RNR catalysis involves the formation of tyrosyl radicals in R2 subunits and thiyl radicals in R1 subunits. Despite the quaternary structure and cofactor diversity, all the three classes of RNR have a conserved cysteine residue at the active site which is converted into a thiyl radical that initiates the substrate turnover, suggesting that the catalytic mechanism is somewhat similar for all three classes of the RNR enzyme. Increased RNR activity has been associated with malignant transformation, cancer cell growth, and tumorigenesis. Efforts concerning the understanding of RNR inhibition in designing potent RNR inhibitors/drugs as well as developing novel approaches for antibacterial, antiviral treatments, and cancer therapeutics with improved radiosensitization have been made in clinical research. This review highlights the precise and potent roles of NO in RNR inhibition by targeting both the subunits. Under nitrosative stress, the thiols of the R1 subunits have been found to be modified by S-nitrosylation and the tyrosyl radicals of the R2 subunits have been modified by nitration. In view of the recent advances and progresses in the field of nitrosative modifications and its fundamental role in signaling with implications in health and diseases, the present article focuses on the regulations of RNR activity by S-nitrosylation of thiols (R1 subunits) and nitration of tyrosyl residues (R2 subunits) which will further help in designing new drugs and therapies.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology, Amity University, Kolkata, 700135, West Bengal, India
| | - Prerona Mukherjee
- Amity Institute of Biotechnology, Amity University, Kolkata, 700135, West Bengal, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology, Amity University, Kolkata, 700135, West Bengal, India.
| |
Collapse
|
29
|
Almeida VS, Miller LL, Delia JPG, Magalhães AV, Caruso IP, Iqbal A, Almeida FCL. Deciphering the Path of S-nitrosation of Human Thioredoxin: Evidence of an Internal NO Transfer and Implication for the Cellular Responses to NO. Antioxidants (Basel) 2022; 11:antiox11071236. [PMID: 35883729 PMCID: PMC9311519 DOI: 10.3390/antiox11071236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 01/27/2023] Open
Abstract
Nitric oxide (NO) is a free radical with a signaling capacity. Its cellular functions are achieved mainly through S-nitrosation where thioredoxin (hTrx) is pivotal in the S-transnitrosation to specific cellular targets. In this study, we use NMR spectroscopy and mass spectrometry to follow the mechanism of S-(trans)nitrosation of hTrx. We describe a site-specific path for S-nitrosation by measuring the reactivity of each of the 5 cysteines of hTrx using cysteine mutants. We showed the interdependence of the three cysteines in the nitrosative site. C73 is the most reactive and is responsible for all S-transnitrosation to other cellular targets. We observed NO internal transfers leading to C62 S-nitrosation, which serves as a storage site for NO. C69-SNO only forms under nitrosative stress, leading to hTrx nuclear translocation.
Collapse
Affiliation(s)
- Vitor S. Almeida
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil
- Institute of Chemistry, Rural Federal University of Rio de Janeiro (UFRRJ), Seropédica 23897-000, Brazil
| | - Lara L. Miller
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
| | - João P. G. Delia
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
| | - Augusto V. Magalhães
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
| | - Icaro P. Caruso
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
- Institute of Chemistry, Rural Federal University of Rio de Janeiro (UFRRJ), Seropédica 23897-000, Brazil
- Multiuser Center for Biomolecular Innovation (CMIB), Department of Physics, Institute of Biosciences, Letters and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
| | - Anwar Iqbal
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
- Department of Chemical Sciences, University of Lakki Marwat, Lakki Marwat 28420, Pakistan
| | - Fabio C. L. Almeida
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil; (V.S.A.); (L.L.M.); (J.P.G.D.); (A.V.M.); (I.P.C.); (A.I.)
- Correspondence:
| |
Collapse
|
30
|
Charles R, Eaton P. Redox Regulation of Soluble Epoxide Hydrolase-Implications for Cardiovascular Health and Disease. Cells 2022; 11:cells11121932. [PMID: 35741062 PMCID: PMC9221603 DOI: 10.3390/cells11121932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/25/2022] Open
Abstract
Cell responses to changes in their redox state are significantly mediated by reversible oxido-reductive post-translational modifications of proteins, potentially altering their activities or interactions. These modifications are important for the homeostatic responses of cells to environmental changes that alter their redox state. Such redox regulatory mechanisms not only operate to maintain health, but can become dysregulated and contribute to pathophysiology. In this review, we focus on the redox control of soluble epoxide hydrolase (sEH), which is widely expressed, including in blood vessels and cardiomyocytes. We review the different types of oxidative modifications that regulate sEH and how they may alter cardiovascular physiology and affect disease progression during stress.
Collapse
|
31
|
TRX2/Rab35 Interaction Impairs Exosome Secretion by Inducing Rab35 Degradation. Int J Mol Sci 2022; 23:ijms23126557. [PMID: 35743001 PMCID: PMC9224307 DOI: 10.3390/ijms23126557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Given that exosomes mediate intercellular communication by delivering cellular components to recipient cells or tissue, they have the potential to be engineered to deliver therapeutic payloads. However, the regulatory mechanism of exosome secretion is poorly understood. In addition, mitochondrial components have been found in exosomes, suggesting communication between mitochondria and exosomes. However, the molecular mechanism of the mitochondria and vesicle interaction remains unclear. Here, we showed that mitochondrial thioredoxin 2 (TRX2) decreased exosome concentrations and inhibited HCT116 cell migration. Coimmunoprecipitation/mass spectrometry (Co-IP/MS) showed that TRX2 interacted with Rab35. TRX2 and Rab35 bound to each other at their N-terminal motifs and colocalized on mitochondria. Furthermore, TRX2 induced Rab35 degradation, resulting in impaired exosome secretion. Additionally, Rab35 mediated the suppressive effects of TRX2 on cell migration, and TRX2 suppressed cell migration through exosomes. Taken together, this study first found an interaction between TRX2 and Rab35. These results revealed a new role for TRX2 in the regulation of exosome secretion and cell migration and explained the upstream regulatory mechanism of Rab35. Furthermore, these findings also provide new molecular evidence for communication between mitochondria and vesicles.
Collapse
|
32
|
TXN inhibitor impedes radioresistance of colorectal cancer cells with decreased ALDH1L2 expression via TXN/NF-κB signaling pathway. Br J Cancer 2022; 127:637-648. [PMID: 35597868 PMCID: PMC9381770 DOI: 10.1038/s41416-022-01835-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Background Colorectal cancer (CRC) is prevalent worldwide and is often challenged by treatment failure and recurrence due to resistance to radiotherapy. Here, we aimed to identify the elusive underlying molecular mechanisms of radioresistance in CRC. Methods Weighted gene co-expression network analysis was used to identify potential radiation-related genes. Colony formation and comet assays and multi-target single-hit survival and xenograft animal models were used to validate the results obtained from the bioinformatic analysis. Immunohistochemistry was performed to examine the clinical characteristics of ALDH1L2. Co-immunoprecipitation, immunofluorescence and flow cytometry were used to understand the molecular mechanisms underlying radioresistance. Results Bioinformatic analysis, in vitro, and in vivo experiments revealed that ALDH1L2 is a radiation-related gene, and a decrease in its expression induces radioresistance in CRC cells by inhibiting ROS-mediated apoptosis. Patients with low ALDH1L2 expression exhibit resistance to radiotherapy. Mechanistically, ALDH1L2 interacts with thioredoxin (TXN) and regulates the downstream NF-κB signaling pathway. PX-12, the TXN inhibitor, overcomes radioresistance due to decreased ALDH1L2. Conclusions Our results provide valuable insights into the potential role of ALDH1L2 in CRC radiotherapy. We propose that the simultaneous application of TXN inhibitors and radiotherapy would significantly ameliorate the clinical outcomes of patients with CRC having low ALDH1L2. ![]()
Collapse
|
33
|
Awan MUN, Yan F, Mahmood F, Bai L, Liu J, Bai J. The Functions of Thioredoxin 1 in Neurodegeneration. Antioxid Redox Signal 2022; 36:1023-1036. [PMID: 34465198 DOI: 10.1089/ars.2021.0186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Significance: Thioredoxin 1 (Trx1) is a ubiquitous protein that is found in organisms ranging from prokaryotes to eukaryotes. Trx1 acts as reductases in redox regulation and protects proteins from oxidative aggregation and inactivation. Trx1 helps the cells to cope with various environmental stresses and inhibits programmed cell death. It is beneficial to neuroregeneration and resistance against oxidative stress-associated neuron damage. Trx1 also plays important roles in suppressing neurodegenerative disorders. Recent Advances: Trx1 is a redox regulating protein involved in neuronal protection. According to a previous study, Trx1 expression is increased by nerve growth factor (NGF) and necessary for neurite outgrowth of PC12 cells. Trx1 has been shown to promote the growth of neurons. Trx1 knockout or knockdown has the worse impact on cell viability and survival. Critical Issues: Trx1 has functions in central nervous system. Trx1 plays the defensive roles against oxidative stress in neurodegenerative diseases. Future Directions: In this review, we focus on the structure of Trx1 and basic functions of Trx1. Trx1 plays a neuroprotective role by suppressing endoplasmic reticulum stress in Parkinson's disease. Neurodegenerative diseases have no cure and carry a high cost to the health care system and patient's families. Trx1 may be taken as a new target for neurodegenerative disorder therapy. Further studies of the Trx1 roles and mechanisms on neurodegenerative diseases are needed. Antioxid. Redox Signal. 36, 1023-1036.
Collapse
Affiliation(s)
- Maher Un Nisa Awan
- Laboratory of Molecular Neurobiology, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.,Laboratory of Molecular Neurobiology, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Fang Yan
- Laboratory of Molecular Neurobiology, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Faisal Mahmood
- Laboratory of Molecular Neurobiology, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Liping Bai
- Laboratory of Molecular Neurobiology, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jingyu Liu
- Laboratory of Molecular Neurobiology, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Laboratory of Molecular Neurobiology, Medical School, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
34
|
A new look at the role of nitric oxide in preeclampsia: protein S-nitrosylation. Pregnancy Hypertens 2022; 29:14-20. [DOI: 10.1016/j.preghy.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022]
|
35
|
Ogata FT, Simões Sato AY, Coppo L, Arai RJ, Stern AI, Pequeno Monteiro H. Thiol-Based Antioxidants and the Epithelial/Mesenchymal Transition in Cancer. Antioxid Redox Signal 2022; 36:1037-1050. [PMID: 34541904 DOI: 10.1089/ars.2021.0199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The epithelial/mesenchymal transition (EMT) is commonly associated with tumor metastasis. Oxidative and nitrosative stress is maintained in cancer cells and is involved in the EMT. Cancer cells are endowed with high levels of enzymatic and nonenzymatic antioxidants, which counteract the effects of oxidative and nitrosative stress. Thiol-based antioxidant systems such as the thioredoxin/thioredoxin reductase (Trx/TrxR) and glutathione/glutaredoxin (GSH/Grx) are continually active in cancer cells, while the thioredoxin-interacting protein (Txnip), the negative regulator of the Trx/TrxR system, is downregulated. Recent Advances: Trx/TrxR and GSH/Grx systems play a major role in maintaining EMT signaling and cancer cell progression. Critical Issues: Enhanced stress conditions stimulated in cancer cells inhibit EMT signaling. The elevated expression levels of the Trx/TrxR and GSH/Grx systems in these cells provide the antioxidant protection necessary to guarantee the occurrence of the EMT. Future Directions: Elevation of the intracellular reactive oxygen species and nitric oxide concentrations in cancer cells has been viewed as a promising strategy for elimination of these cells. The development of inhibitors of GSH synthesis and of the Trx/TrxR system together with genetic-based strategies to enhance Txnip levels may provide the necessary means to achieve this goal. Antioxid. Redox Signal. 36, 1037-1050.
Collapse
Affiliation(s)
- Fernando Toshio Ogata
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMol, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alex Yuri Simões Sato
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMol, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lucia Coppo
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Roberto Jun Arai
- Department of Oncology and Radiology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina-Universidade de São Paulo, São Paulo, Brazil
| | - Arnold Ira Stern
- Grossman School of Medicine, New York University, New York, New York, USA
| | - Hugo Pequeno Monteiro
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMol, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Ye H, Wu J, Liang Z, Zhang Y, Huang Z. Protein S-Nitrosation: Biochemistry, Identification, Molecular Mechanisms, and Therapeutic Applications. J Med Chem 2022; 65:5902-5925. [PMID: 35412827 DOI: 10.1021/acs.jmedchem.1c02194] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein S-nitrosation (SNO), a posttranslational modification (PTM) of cysteine (Cys) residues elicited by nitric oxide (NO), regulates a wide range of protein functions. As a crucial form of redox-based signaling by NO, SNO contributes significantly to the modulation of physiological functions, and SNO imbalance is closely linked to pathophysiological processes. Site-specific identification of the SNO protein is critical for understanding the underlying molecular mechanisms of protein function regulation. Although careful verification is needed, SNO modification data containing numerous functional proteins are a potential research direction for druggable target identification and drug discovery. Undoubtedly, SNO-related research is meaningful not only for the development of NO donor drugs but also for classic target-based drug design. Herein, we provide a comprehensive summary of SNO, including its origin and transport, identification, function, and potential contribution to drug discovery. Importantly, we propose new views to develop novel therapies based on potential protein SNO-sourced targets.
Collapse
Affiliation(s)
- Hui Ye
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Jianbing Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Zhuangzhuang Liang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| |
Collapse
|
37
|
Li X, Yang G, Zhang W, Qin B, Ye Z, Shi H, Zhao X, Chen Y, Song B, Mei Z, Zhao Q, Wang F. USP13: Multiple Functions and Target Inhibition. Front Cell Dev Biol 2022; 10:875124. [PMID: 35445009 PMCID: PMC9014248 DOI: 10.3389/fcell.2022.875124] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
As a deubiquitination (DUB) enzyme, ubiquitin-specific protease 13 (USP13) is involved in a myriad of cellular processes, such as mitochondrial energy metabolism, autophagy, DNA damage response, and endoplasmic reticulum-associated degradation (ERAD), by regulating the deubiquitination of diverse key substrate proteins. Thus, dysregulation of USP13 can give rise to the occurrence and development of plenty of diseases, in particular malignant tumors. Given its implications in the stabilization of disease-related proteins and oncology targets, considerable efforts have been committed to the discovery of inhibitors targeting USP13. Here, we summarize an overview of the recent advances of the structure, function of USP13, and its relations to diseases, as well as discovery and development of inhibitors, aiming to provide the theoretical basis for investigation of the molecular mechanism of USP13 action and further development of more potent druggable inhibitors.
Collapse
Affiliation(s)
- Xiaolong Li
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ge Yang
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Wenyao Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Biying Qin
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zifan Ye
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Huijing Shi
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xinmeng Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yihang Chen
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Bowei Song
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ziqing Mei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | | | - Feng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
- *Correspondence: Feng Wang,
| |
Collapse
|
38
|
Retamal MA, Altenberg GA. Role and Posttranslational Regulation of Cx46 Hemichannels and Gap Junction Channels in the Eye Lens. Front Physiol 2022; 13:864948. [PMID: 35431975 PMCID: PMC9006113 DOI: 10.3389/fphys.2022.864948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/14/2022] [Indexed: 12/31/2022] Open
Abstract
Connexins are a family of proteins that can form two distinct types of channels: hemichannels and gap junction channels. Hemichannels are composed of six connexin subunits and when open allow for exchanges between the cytoplasm and the extracellular milieu. Gap junction channels are formed by head-to-head docking of two hemichannels in series, each one from one of two adjacent cells. These channels allow for exchanges between the cytoplasms of contacting cells. The lens is a transparent structure located in the eye that focuses light on the retina. The transparency of the lens depends on its lack of blood irrigation and the absence of organelles in its cells. To survive such complex metabolic scenario, lens cells express Cx43, Cx46 and Cx50, three connexins isoforms that form hemichannels and gap junction channels that allow for metabolic cooperation between lens cells. This review focuses on the roles of Cx46 hemichannels and gap junction channels in the lens under physiological conditions and in the formation of cataracts, with emphasis on the modulation by posttranslational modifications.
Collapse
Affiliation(s)
- Mauricio A. Retamal
- Universidad del Desarrollo, Centro de Fisiología Celular e Integrativa, Clínica Alemana Facultad de Medicina, Santiago, Chile
- Universidad del Desarrollo, Programa de Comunicación Celular en Cáncer, Clínica Alemana Facultad de Medicina, Santiago, Chile
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- *Correspondence: Mauricio A. Retamal, ; Guillermo A. Altenberg,
| | - Guillermo A. Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- *Correspondence: Mauricio A. Retamal, ; Guillermo A. Altenberg,
| |
Collapse
|
39
|
Gupta KJ, Kaladhar VC, Fitzpatrick TB, Fernie AR, Møller IM, Loake GJ. Nitric oxide regulation of plant metabolism. MOLECULAR PLANT 2022; 15:228-242. [PMID: 34971792 DOI: 10.1016/j.molp.2021.12.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 10/31/2021] [Accepted: 12/23/2021] [Indexed: 05/17/2023]
Abstract
Nitric oxide (NO) has emerged as an important signal molecule in plants, having myriad roles in plant development. In addition, NO also orchestrates both biotic and abiotic stress responses, during which intensive cellular metabolic reprogramming occurs. Integral to these responses is the location of NO biosynthetic and scavenging pathways in diverse cellular compartments, enabling plants to effectively organize signal transduction pathways. NO regulates plant metabolism and, in turn, metabolic pathways reciprocally regulate NO accumulation and function. Thus, these diverse cellular processes are inextricably linked. This review addresses the numerous redox pathways, located in the various subcellular compartments that produce NO, in addition to the mechanisms underpinning NO scavenging. We focus on how this molecular dance is integrated into the metabolic state of the cell. Within this context, a reciprocal relationship between NO accumulation and metabolite production is often apparent. We also showcase cellular pathways, including those associated with nitrate reduction, that provide evidence for this integration of NO function and metabolism. Finally, we discuss the potential importance of the biochemical reactions governing NO levels in determining plant responses to a changing environment.
Collapse
Affiliation(s)
- Kapuganti Jagadis Gupta
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, Delhi 110067 India.
| | - Vemula Chandra Kaladhar
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, Delhi 110067 India
| | - Teresa B Fitzpatrick
- Vitamins and Environmental Stress Responses in Plants, Department of Botany and Plant Biology, University of Geneva, Geneva 1211 Switzerland
| | - Alisdair R Fernie
- Max-Planck-Institute of Molecular Plant Physiology, Am Mühlenberg 1, Potsdam-Golm 14476 Germany
| | - Ian Max Møller
- Department of Molecular Biology and Genetics, Aarhus University, Forsøgsvej 1, 4200 Slagelse, Denmark
| | - Gary J Loake
- Institute of Molecular Plant Sciences, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK.
| |
Collapse
|
40
|
Liu Y, Xue N, Zhang B, Lv H, Li S. Role of Thioredoxin-1 and its inducers in human health and diseases. Eur J Pharmacol 2022; 919:174756. [PMID: 35032486 DOI: 10.1016/j.ejphar.2022.174756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 12/27/2022]
Abstract
Thioredoxin-1 (Trx-1) is a small redox-active protein normally found in mammalian cells that responds to the changing redox environment by contributing electrons or regulating related proteins. There is growing evidence that Trx-1 has multiple functions, including cytoprotective, anti-apoptotic, antioxidant and anti-inflammatory effects. To date, researchers have found that Trx-1 deficiency leads to severe damage in various disease models, such as atherosclerosis, cerebral ischemia, diabetes and tumors. Conversely, activation of Trx-1 has a protective effect against these diseases. Accordingly, a variety of Trx-1 inducers have been widely used in the clinic with significant therapeutic value. In this paper, we summarize the pathogenesis of Trx-1 involvement in the above-mentioned diseases and describe the protective effects of Trx-1 inducers on them.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China
| | - Nianyu Xue
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China
| | - Boxi Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China
| | - Hongming Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China.
| | - Shize Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China.
| |
Collapse
|
41
|
Pande A, Mun BG, Rahim W, Khan M, Lee DS, Lee GM, Al Azzawi TNI, Hussain A, Kim CK, Yun BW. Phytohormonal Regulation Through Protein S-Nitrosylation Under Stress. FRONTIERS IN PLANT SCIENCE 2022; 13:865542. [PMID: 35401598 PMCID: PMC8988057 DOI: 10.3389/fpls.2022.865542] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/18/2022] [Indexed: 05/18/2023]
Abstract
The liaison between Nitric oxide (NO) and phytohormones regulates a myriad of physiological processes at the cellular level. The interaction between NO and phytohormones is mainly influenced by NO-mediated post-translational modifications (PTMs) under basal as well as induced conditions. Protein S-nitrosylation is the most prominent and widely studied PTM among others. It is the selective but reversible redox-based covalent addition of a NO moiety to the sulfhydryl group of cysteine (Cys) molecule(s) on a target protein to form S-nitrosothiols. This process may involve either direct S-nitrosylation or indirect S-nitrosylation followed by transfer of NO group from one thiol to another (transnitrosylation). During S-nitrosylation, NO can directly target Cys residue (s) of key genes involved in hormone signaling thereby regulating their function. The phytohormones regulated by NO in this manner includes abscisic acid, auxin, gibberellic acid, cytokinin, ethylene, salicylic acid, jasmonic acid, brassinosteroid, and strigolactone during various metabolic and physiological conditions and environmental stress responses. S-nitrosylation of key proteins involved in the phytohormonal network occurs during their synthesis, degradation, or signaling roles depending upon the response required to maintain cellular homeostasis. This review presents the interaction between NO and phytohormones and the role of the canonical NO-mediated post-translational modification particularly, S-nitrosylation of key proteins involved in the phytohormonal networks under biotic and abiotic stresses.
Collapse
Affiliation(s)
- Anjali Pande
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Bong Gyu Mun
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Waqas Rahim
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Murtaza Khan
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Da Sol Lee
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Geun Mo Lee
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Tiba Nazar Ibrahim Al Azzawi
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
| | - Adil Hussain
- Laboratory of Cell Biology, Department of Entomology, Abdul Wali Khan University, Mardan, Pakistan
| | - Chang Kil Kim
- Department of Horticultural Sciences, Kyungpook National University, Daegu, South Korea
- *Correspondence: Chang Kil Kim,
| | - Byung Wook Yun
- Laboratory of Plant Molecular Pathology and Functional Genomics, Department of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu, South Korea
- Byung Wook Yun,
| |
Collapse
|
42
|
Lee HM, Choi JW, Choi MS. Role of Nitric Oxide and Protein S-Nitrosylation in Ischemia-Reperfusion Injury. Antioxidants (Basel) 2021; 11:57. [PMID: 35052559 PMCID: PMC8772765 DOI: 10.3390/antiox11010057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/06/2021] [Accepted: 12/24/2021] [Indexed: 12/19/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a process in which damage is induced in hypoxic tissue when oxygen supply is resumed after ischemia. During IRI, restoration of reduced nitric oxide (NO) levels may alleviate reperfusion injury in ischemic organs. The protective mechanism of NO is due to anti-inflammatory effects, antioxidant effects, and the regulation of cell signaling pathways. On the other hand, it is generally known that S-nitrosylation (SNO) mediates the detrimental or protective effect of NO depending on the action of the nitrosylated target protein, and this is also applied in the IRI process. In this review, the effect of each change of NO and SNO during the IRI process was investigated.
Collapse
Affiliation(s)
- Hyang-Mi Lee
- College of Pharmacy, Dongduk Women’s University, Seoul 02748, Korea;
| | - Ji Woong Choi
- College of Pharmacy, Gachon University, Incheon 21936, Korea
| | - Min Sik Choi
- Laboratory of Pharmacology, College of Pharmacy, Dongduk Women’s University, Seoul 02748, Korea
| |
Collapse
|
43
|
He Q, Qu M, Xu C, Shi W, Hussain M, Jin G, Zhu H, Zeng LH, Wu X. The emerging roles of nitric oxide in ferroptosis and pyroptosis of tumor cells. Life Sci 2021; 290:120257. [PMID: 34952041 DOI: 10.1016/j.lfs.2021.120257] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/06/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Tumor cells can develop resistance to cell death which is divided into necrosis and programmed cell death (PCD). PCD, including apoptosis, autophagy, ferroptosis, pyroptosis, and necroptosis. Ferroptosis and pyroptosis, two new forms of cell death, have gradually been of interest to researchers. Boosting ferroptosis and pyroptosis of tumor cells could be a potential cancer therapy. Nitric oxide (NO) is a ubiquitous, lipophilic, highly diffusible, free-radical signaling molecule that plays various roles in tumorigenesis. In addition, NO also has regulatory mechanisms through S-nitrosylation that do not depend on the classic NO/sGC/cGMP signaling. The current tumor treatment strategy for NO is to promote cell death through promoting S-nitrosylation-induced apoptosis while multiple drawbacks dampen this tumor therapy. However, numerous studies have suggested that suppression of NO is perceived to active ferroptosis and pyroptosis, which could be a better anti-tumor treatment. In this review, ferroptosis and pyroptosis are described in detail. We summarize that NO influences ferroptosis and pyroptosis and infer that S-nitrosylation mediates ferroptosis- and pyroptosis-related signaling pathways. It could be a potential cancer therapy different from NO-induced apoptosis of tumor cells. Finally, the information shows the drugs that manipulate endogenous production and exogenous delivery of NO to modulate the levels of S-nitrosylation.
Collapse
Affiliation(s)
- Qiangqiang He
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China
| | - Meiyu Qu
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Shi
- Department of Biology and Genetics, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Musaddique Hussain
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Guojian Jin
- Department of Internal Medicine, Shaoxing Central Hospital Anchang Branch, Shaoxing City 312080, China
| | - Haibin Zhu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310015, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
44
|
Kalous KS, Wynia-Smith SL, Smith BC. Sirtuin Oxidative Post-translational Modifications. Front Physiol 2021; 12:763417. [PMID: 34899389 PMCID: PMC8652059 DOI: 10.3389/fphys.2021.763417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Increased sirtuin deacylase activity is correlated with increased lifespan and healthspan in eukaryotes. Conversely, decreased sirtuin deacylase activity is correlated with increased susceptibility to aging-related diseases. However, the mechanisms leading to decreased sirtuin activity during aging are poorly understood. Recent work has shown that oxidative post-translational modification by reactive oxygen (ROS) or nitrogen (RNS) species results in inhibition of sirtuin deacylase activity through cysteine nitrosation, glutathionylation, sulfenylation, and sulfhydration as well as tyrosine nitration. The prevalence of ROS/RNS (e.g., nitric oxide, S-nitrosoglutathione, hydrogen peroxide, oxidized glutathione, and peroxynitrite) is increased during inflammation and as a result of electron transport chain dysfunction. With age, cellular production of ROS/RNS increases; thus, cellular oxidants may serve as a causal link between loss of sirtuin activity and aging-related disease development. Therefore, the prevention of inhibitory oxidative modification may represent a novel means to increase sirtuin activity during aging. In this review, we explore the role of cellular oxidants in inhibiting individual sirtuin human isoform deacylase activity and clarify the relevance of ROS/RNS as regulatory molecules of sirtuin deacylase activity in the context of health and disease.
Collapse
Affiliation(s)
- Kelsey S Kalous
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sarah L Wynia-Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
45
|
Doulias PT, Tenopoulou M, Zakopoulos I, Ischiropoulos H. Organic mercury solid phase chemoselective capture for proteomic identification of S-nitrosated proteins and peptides. Nitric Oxide 2021; 117:1-6. [PMID: 34536587 DOI: 10.1016/j.niox.2021.09.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 12/26/2022]
Abstract
Cysteine S-nitrosation mediates NO signaling and protein function under pathophysiological conditions. Herein, we provide a detailed protocol regarding the organic mercury chemoselective enrichment of S-nitrosated proteins and peptides. We discuss key aspects of the enrichment strategy and provide technical tips for the best performance of the experimental protocol.
Collapse
Affiliation(s)
- Paschalis-Thomas Doulias
- Children's Hospital of Philadelphia Research Institute, 3517 Civic Center Boulevard, Philadelphia, PA, 19104-4318, USA; Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, 45110, Greece.
| | - Margarita Tenopoulou
- Children's Hospital of Philadelphia Research Institute, 3517 Civic Center Boulevard, Philadelphia, PA, 19104-4318, USA; Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, 45110, Greece
| | - Iordanis Zakopoulos
- Children's Hospital of Philadelphia Research Institute, 3517 Civic Center Boulevard, Philadelphia, PA, 19104-4318, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, 3517 Civic Center Boulevard, Philadelphia, PA, 19104-4318, USA.
| |
Collapse
|
46
|
Atlas D. Emerging therapeutic opportunities of novel thiol-amides, NAC-amide (AD4/NACA) and thioredoxin mimetics (TXM-Peptides) for neurodegenerative-related disorders. Free Radic Biol Med 2021; 176:120-141. [PMID: 34481041 DOI: 10.1016/j.freeradbiomed.2021.08.239] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/17/2021] [Accepted: 08/29/2021] [Indexed: 12/23/2022]
Abstract
Understanding neurodegenerative diseases have challenged scientists for decades. It has become apparent that a decrease in life span is often correlated with the development of neurodegenerative disorders. Oxidative stress and the subsequent inflammatory damages appear to contribute to the different molecular and biochemical mechanisms associated with neurodegeneration. In this review, I examine the protective properties of novel amino acid based compounds, comprising the AD series (AD1-AD7) in particular N-acetylcysteine amide, AD4, also called NACA, and the series of thioredoxin mimetic (TXM) peptides, TXM-CB3-TXM-CB16. Designed to cross the blood-brain-barrier (BBB) and permeate the cell membrane, these antioxidant/anti-inflammatory compounds may enable effective treatment of neurodegenerative related disorders. The review addresses the molecular mechanism of cellular protection exhibited by these new reagents, focusing on the reversal of oxidative stress, mitochondrial stress, inflammatory damages, and prevention of premature cell death. In addition, it will cover the outlook of the clinical prospects of AD4/NACA and the thioredoxin-mimetic peptides, which are currently in development.
Collapse
Affiliation(s)
- Daphne Atlas
- Professor of Neurochemistry, Dept. of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel.
| |
Collapse
|
47
|
Zimmer A, Teixeira RB, Constantin RL, Fernandes-Piedras TRG, Campos-Carraro C, Türck P, Visioli F, Baldo G, Schenkel PC, Araujo AS, Belló-Klein A. Thioredoxin system activation is associated with the progression of experimental pulmonary arterial hypertension. Life Sci 2021; 284:119917. [PMID: 34478759 DOI: 10.1016/j.lfs.2021.119917] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/19/2021] [Accepted: 08/22/2021] [Indexed: 11/21/2022]
Abstract
In addition to being an antioxidant, thioredoxin (Trx) is known to stimulate signaling pathways involved in cell proliferation and to inhibit apoptosis. The aim of this study was to explore the role of Trx in some of these pathways along the progression of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH). Male rats were first divided into two groups: monocrotaline (MCT - 60 mg/kg i.p.) and control (received saline), that were further divided into three groups: 1, 2, and 3 weeks. Animals were submitted to echocardiographic analysis. Right and left ventricles were used for the measurement of hypertrophy, through morphometric and histological analysis. The lung was prepared for biochemical and molecular analysis. One week after MCT injection, there was an increase in thioredoxin reductase (TrxR) activity, a reduction in glutathione reductase (GR) activity, and an increase in Trx-1 and vitamin D3 up-regulated protein-1 (VDUP-1) expression. Two weeks after MCT injection, there was an increase in VDUP-1, Akt and cleaved caspase-3 activation, and a decrease in Trx-1 and Nrf2 expression. PAH-induced by MCT promoted a reduction in Nrf2 and Trx-1 expression as well as an increase in Akt and VDUP-1 expression after three weeks. The increase in pulmonary vascular resistance was accompanied by increased TrxR activity, suggesting an association between the Trx system and functional changes in the progression of PAH. It seems that Trx-1 activation was an adaptive response to MCT administration to cope with pulmonary remodeling and disease progression, suggesting a potential new target for PAH therapeutics.
Collapse
Affiliation(s)
- Alexsandra Zimmer
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Rayane Brinck Teixeira
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Rosalia Lempk Constantin
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Tânia Regina Gatelli Fernandes-Piedras
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristina Campos-Carraro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Patrick Türck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Visioli
- Laboratory of Oral Pathology, Post-Graduation Program in Dentistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Guilherme Baldo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Paulo Cavalheiro Schenkel
- Laboratory of Cardiovascular Physiology, Department of Physiology and Pharmacology, Biology Institute, Universidade Federal de Pelotas (UFPel), Pelotas, Rio Grande do Sul, Brazil.
| | - Alex Sander Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
48
|
Chibani K, Pucker B, Dietz KJ, Cavanagh A. Genome-wide analysis and transcriptional regulation of the typical and atypical thioredoxins in Arabidopsis thaliana. FEBS Lett 2021; 595:2715-2730. [PMID: 34561866 DOI: 10.1002/1873-3468.14197] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
Thioredoxins (TRXs), a large subclass of ubiquitous oxidoreductases, are involved in thiol redox regulation. Here, we performed a comprehensive analysis of TRXs in the Arabidopsis thaliana genome, revealing 41 genes encoding 18 typical and 23 atypical TRXs, and 6 genes encoding thioredoxin reductases (TRs). The high number of atypical TRXs indicates special functions in plants that mostly await elucidation. We identified an atypical class of thioredoxins called TRX-c in the genomes of photosynthetic eukaryotes. Localized to the chloroplast, TRX-c displays atypical CPLC, CHLC and CNLC motifs in the active sites. In silico analysis of the transcriptional regulations of TRXs revealed high expression of TRX-c in leaves and strong regulation under cold, osmotic, salinity and metal ion stresses.
Collapse
Affiliation(s)
- Kamel Chibani
- School of Life Sciences, University of Essex, Colchester, UK
- Department of Biochemistry and Physiology of Plants, Faculty of Biology, University of Bielefeld, Germany
| | - Boas Pucker
- Department of Sciences, University of Cambridge, UK
| | - Karl-Josef Dietz
- Department of Biochemistry and Physiology of Plants, Faculty of Biology, University of Bielefeld, Germany
| | - Amanda Cavanagh
- School of Life Sciences, University of Essex, Colchester, UK
| |
Collapse
|
49
|
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: 20] [Impact Index Per Article: 5.0] [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
|
50
|
Endogenous S-nitrosocysteine proteomic inventories identify a core of proteins in heart metabolic pathways. Redox Biol 2021; 47:102153. [PMID: 34610554 PMCID: PMC8497991 DOI: 10.1016/j.redox.2021.102153] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Protein cysteine residues are essential for protein folding, participate in enzymatic catalysis, and coordinate the binding of metal ions to proteins. Enzymatically catalyzed and redox-dependent post-translational modifications of cysteine residues are also critical for signal transduction and regulation of protein function and localization. S-nitrosylation, the addition of a nitric oxide equivalent to a cysteine residue, is a redox-dependent modification. In this study, we curated and analyzed four different studies that employed various chemoselective platforms coupled to mass spectrometry to precisely identify S-nitrosocysteine residues in mouse heart proteins. Collectively 1974 S-nitrosocysteine residues in 761 proteins were identified and 33.4% were identified in two or more studies. A core of 75 S-nitrosocysteine residues in 44 proteins were identified in all four studies. Bioinformatic analysis of each study indicated a significant enrichment of mitochondrial proteins participating in metabolism. Regulatory proteins in glycolysis, TCA cycle, oxidative phosphorylation and ATP production, long chain fatty acid β-oxidation, and ketone and amino acid metabolism constitute the major functional pathways impacted by protein S-nitrosylation. In the cardiovascular system, nitric oxide signaling regulates vasodilation and cardiac muscle contractility. The meta-analysis of the proteomic data supports the hypothesis that nitric oxide signaling via protein S-nitrosylation is also a regulator of cardiomyocyte metabolism that coordinates fuel utilization to maximize ATP production. As such, protein cysteine S-nitrosylation represents a third functional dimension of nitric oxide signaling in the cardiovascular system to ensure optimal cardiac function.
Collapse
|