51
|
Hubbard D, Tutrow K, Gaston B. S-Nitroso-l-cysteine and ventilatory drive: A pediatric perspective. Pediatr Pulmonol 2022; 57:2291-2297. [PMID: 35785452 PMCID: PMC9489637 DOI: 10.1002/ppul.26036] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/09/2022] [Accepted: 05/29/2022] [Indexed: 01/01/2023]
Abstract
Though endogenous S-nitroso-l-cysteine (l-CSNO) signaling at the level of the carotid body increases minute ventilation (v̇E ), neither the background data nor the potential clinical relevance are well-understood by pulmonologists in general, or by pediatric pulmonologists in particular. Here, we first review how regulation of the synthesis, activation, transmembrane transport, target interaction, and degradation of l-CSNO can affect the ventilatory drive. In particular, we review l-CSNO formation by hemoglobin R to T conformational change and by nitric oxide (NO) synthases (NOS), and the downstream effects on v̇E through interaction with voltage-gated K+ (Kv) channel proteins and other targets in the peripheral and central nervous systems. We will review how these effects are independent of-and, in fact may be opposite to-those of NO. Next, we will review evidence that specific elements of this pathway may underlie disorders of respiratory control in childhood. Finally, we will review the potential clinical implications of this pathway in the development of respiratory stimulants, with a particular focus on potential pediatric applications.
Collapse
Affiliation(s)
- Dallin Hubbard
- Division of Pediatric PulmonologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kaylee Tutrow
- Division of Pediatric PulmonologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Benjamin Gaston
- Division of Pediatric PulmonologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
52
|
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
|
53
|
Jiao L, Su LY, Liu Q, Luo R, Qiao X, Xie T, Yang LX, Chen C, Yao YG. GSNOR deficiency attenuates MPTP-induced neurotoxicity and autophagy by facilitating CDK5 S-nitrosation in a mouse model of Parkinson's disease. Free Radic Biol Med 2022; 189:111-121. [PMID: 35918012 DOI: 10.1016/j.freeradbiomed.2022.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/14/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023]
Abstract
The S-nitrosoglutathione reductase (GSNOR) is a key denitrosating enzyme that regulates protein S-nitrosation, a process which has been found to be involved in the pathogenesis of Parkinson's disease (PD). However, the physiological function of GSNOR in PD remains unknown. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model, we found that GSNOR expression was significantly increased and accompanied by autophagy mediated by MPTP-induced cyclin dependent kinase 5 (CDK5), behavioral dyskinesias and dopaminergic neuron loss. Whereas, knockout of GSNOR, or treatment with the GSNOR inhibitor N6022, alleviated MPTP-induced PD-like pathology and neurotoxicity. Mechanistically, deficiency of GSNOR inhibited MPTP-induced CDK5 kinase activity and CDK5-mediated autophagy by increasing S-nitrosation of CDK5 at Cys83. Our study indicated that GSNOR is a key regulator of CDK5 S-nitrosation and is actively involved in CDK5-mediated autophagy induced by MPTP.
Collapse
Affiliation(s)
- Lijin Jiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ling-Yan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Rongcan Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ting Xie
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lu-Xiu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, and KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
54
|
Suppression of VEGFD expression by S-nitrosylation promotes the development of lung adenocarcinoma. J Exp Clin Cancer Res 2022; 41:239. [PMID: 35941690 PMCID: PMC9358865 DOI: 10.1186/s13046-022-02453-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background Vascular endothelial growth factor D (VEGFD), a member of the VEGF family, is implicated in angiogenesis and lymphangiogenesis, and is deemed to be expressed at a low level in cancers. S-nitrosylation, a NO (nitric oxide)-mediated post-translational modification has a critical role in angiogenesis. Here, we attempt to dissect the role and underlying mechanism of S-nitrosylation-mediated VEGFD suppression in lung adenocarcinoma (LUAD). Methods Messenger RNA and protein expression of VEGFD in LUAD were analyzed by TCGA and CPTAC database, respectively, and Assistant for Clinical Bioinformatics was performed for complex analysis. Mouse models with urethane (Ure)–induced LUAD or LUAD xenograft were established to investigate the role of S-nitrosylation in VEGFD expression and of VEGFD mutants in the oncogenesis of LUAD. Molecular, cellular, and biochemical approaches were applied to explore the underlying mechanism of S-nitrosylation-mediated VEGFD suppression. Tube formation and wound healing assays were used to examine the role of VEGFD on the angiogenesis and migration of LUAD cells, and the molecular modeling was applied to predict the protein stability of VEGFD mutant. Results VEGFD mRNA and protein levels were decreased to a different extent in multiple primary malignancies, especially in LUAD. Low VEGFD protein expression was closely related to the oncogenesis of LUAD and resultant from excessive NO-induced VEGFD S-nitrosylation at Cys277. Moreover, inhibition of S-nitrosoglutathione reductase consistently decreased the VEGFD denitrosylation at Cys277 and consequently promoted angiogenesis of LUAD. Finally, the VEGFDC277S mutant decreased the secretion of mature VEGFD by attenuating the PC7-dependent proteolysis and VEGFDC277S mutant thus reversed the effect of VEGFD on angiogenesis of LUAD. Conclusion Low-expression of VEGFD positively correlates with LUAD development. Aberrant S-nitrosylation of VEGFD negates itself to induce the tumorigenesis of LUAD, whereas normal S-nitrosylation of VEGFD is indispensable for its secretion and repression of angiogenesis of LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02453-8.
Collapse
|
55
|
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
|
56
|
Wengert LA, Backe SJ, Bourboulia D, Mollapour M, Woodford MR. TRAP1 Chaperones the Metabolic Switch in Cancer. Biomolecules 2022; 12:biom12060786. [PMID: 35740911 PMCID: PMC9221471 DOI: 10.3390/biom12060786] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial function is dependent on molecular chaperones, primarily due to their necessity in the formation of respiratory complexes and clearance of misfolded proteins. Heat shock proteins (Hsps) are a subset of molecular chaperones that function in all subcellular compartments, both constitutively and in response to stress. The Hsp90 chaperone TNF-receptor-associated protein-1 (TRAP1) is primarily localized to the mitochondria and controls both cellular metabolic reprogramming and mitochondrial apoptosis. TRAP1 upregulation facilitates the growth and progression of many cancers by promoting glycolytic metabolism and antagonizing the mitochondrial permeability transition that precedes multiple cell death pathways. TRAP1 attenuation induces apoptosis in cellular models of cancer, identifying TRAP1 as a potential therapeutic target in cancer. Similar to cytosolic Hsp90 proteins, TRAP1 is also subject to post-translational modifications (PTM) that regulate its function and mediate its impact on downstream effectors, or ‘clients’. However, few effectors have been identified to date. Here, we will discuss the consequence of TRAP1 deregulation in cancer and the impact of post-translational modification on the known functions of TRAP1.
Collapse
Affiliation(s)
- Laura A. Wengert
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sarah J. Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mark R. Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; (L.A.W.); (S.J.B.); (D.B.); (M.M.)
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Correspondence:
| |
Collapse
|
57
|
Veron D, Aggarwal PK, Li Q, Moeckel G, Kashgarian M, Tufro A. Podocyte VEGF-A Knockdown Induces Diffuse Glomerulosclerosis in Diabetic and in eNOS Knockout Mice. Front Pharmacol 2022; 12:788886. [PMID: 35280251 PMCID: PMC8906751 DOI: 10.3389/fphar.2021.788886] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/13/2021] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor-a (VEGF-A) and nitric oxide (NO) are essential for glomerular filtration barrier homeostasis, and are dysregulated in diabetic kidney disease (DKD). While NO availability is consistently low in diabetes, both high and low VEGF-A have been reported in patients with DKD. Here we examined the effect of inducible podocyte VEGF-A knockdown (VEGFKD) in diabetic mice and in endothelial nitric oxide synthase knockout mice (eNOS−/−). Diabetes was induced with streptozotocin using the Animal Models of Diabetic Complications Consortium (AMDCC) protocol. Induction of podocyte VEGFKD led to diffuse glomerulosclerosis, foot process effacement, and GBM thickening in both diabetic mice with intact eNOS and in non-diabetic eNOS−/−:VEGFKD mice. VEGFKD diabetic mice developed mild proteinuria and maintained normal glomerular filtration rate (GFR), associated with extremely high NO and thiol urinary excretion. In eNOS−/−:VEGFKD (+dox) mice severe diffuse glomerulosclerosis was associated with microaneurisms, arteriolar hyalinosis, massive proteinuria, and renal failure. Collectively, data indicate that combined podocyte VEGF-A and eNOS deficiency result in diffuse glomerulosclerosis in mice; compensatory NO and thiol generation prevents severe proteinuria and GFR loss in VEGFKD diabetic mice with intact eNOS, whereas VEGFKD induction in eNOS−/−:VEGFKD mice causes massive proteinuria and renal failure mimicking DKD in the absence of diabetes. Mechanistically, we identify VEGFKD-induced abnormal S-nitrosylation of specific proteins, including β3-integrin, laminin, and S-nitrosoglutathione reductase (GSNOR), as targetable molecular mechanisms involved in the development of advanced diffuse glomerulosclerosis and renal failure.
Collapse
Affiliation(s)
- Delma Veron
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States
| | - Pardeep K Aggarwal
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States
| | - Qi Li
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States.,Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Alda Tufro
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States.,Department of Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
58
|
Chen L, Sun S, Song CP, Zhou JM, Li J, Zuo J. Nitric oxide negatively regulates gibberellin signaling to coordinate growth and salt tolerance in Arabidopsis. J Genet Genomics 2022; 49:756-765. [PMID: 35276388 DOI: 10.1016/j.jgg.2022.02.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022]
Abstract
In response to dynamically altered environments, plants must finely coordinate the balance between growth and stress responses for their survival. However, the underpinning regulatory mechanisms remain largely elusive. The phytohormone gibberellin promotes growth via a derepression mechanism by proteasomal degradation of the DELLA transcription repressors. Conversely, the stress-induced burst of nitric oxide (NO) enhances stress tolerance, largely relaying on NO-mediated S-nitrosylation, a redox-based posttranslational modification. Here, we show that S-nitrosylation of Cys-374 in the Arabidopsis RGA protein, a key member of DELLAs, inhibits its interaction with the F-box protein SLY1, thereby preventing its proteasomal degradation under salinity condition. The accumulation of RGA consequently retards growth but enhances salt tolerance. We propose that NO negatively regulates gibberellin signaling via S-nitrosylation of RGA to coordinate the balance of growth and stress responses when challenged by adverse environments.
Collapse
Affiliation(s)
- Lichao Chen
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shuhao Sun
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Chun-Peng Song
- State Key Laboratory of Crop Stress Adaptation and Improvement, Collaborative Innovation Center of Crop Stress Biology, Henan University, Kaifeng, Henang 475001, China
| | - Jian-Min Zhou
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Hainan Yazhou Bay Seed Laboratory, Sanya, Hainan 572025, China
| | - Jiayang Li
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Hainan Yazhou Bay Seed Laboratory, Sanya, Hainan 572025, China
| | - Jianru Zuo
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Beijing 100101, China; Hainan Yazhou Bay Seed Laboratory, Sanya, Hainan 572025, China.
| |
Collapse
|
59
|
Sengupta S, Nath R, Bhattacharjee A. Characterizing the effect of S-nitrosoglutathione on Saccharomyces cerevisiae: Upregulation of alcohol dehydrogenase and inactivation of aconitase. Process Biochem 2022. [DOI: 10.1016/j.procbio.2021.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
60
|
Georas SN, Wright RJ, Ivanova A, Israel E, LaVange LM, Akuthota P, Carr TF, Denlinger LC, Fajt ML, Kumar R, O'Neal WK, Phipatanakul W, Szefler SJ, Aronica MA, Bacharier LB, Burbank AJ, Castro M, Crotty Alexander L, Bamdad J, Cardet JC, Comhair SAA, Covar RA, DiMango EA, Erwin K, Erzurum SC, Fahy JV, Gaffin JM, Gaston B, Gerald LB, Hoffman EA, Holguin F, Jackson DJ, James J, Jarjour NN, Kenyon NJ, Khatri S, Kirwan JP, Kraft M, Krishnan JA, Liu AH, Liu MC, Marquis MA, Martinez F, Mey J, Moore WC, Moy JN, Ortega VE, Peden DB, Pennington E, Peters MC, Ross K, Sanchez M, Smith LJ, Sorkness RL, Wechsler ME, Wenzel SE, White SR, Zein J, Zeki AA, Noel P. The Precision Interventions for Severe and/or Exacerbation-Prone (PrecISE) Asthma Network: An overview of Network organization, procedures, and interventions. J Allergy Clin Immunol 2022; 149:488-516.e9. [PMID: 34848210 PMCID: PMC8821377 DOI: 10.1016/j.jaci.2021.10.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
Asthma is a heterogeneous disease, with multiple underlying inflammatory pathways and structural airway abnormalities that impact disease persistence and severity. Recent progress has been made in developing targeted asthma therapeutics, especially for subjects with eosinophilic asthma. However, there is an unmet need for new approaches to treat patients with severe and exacerbation-prone asthma, who contribute disproportionately to disease burden. Extensive deep phenotyping has revealed the heterogeneous nature of severe asthma and identified distinct disease subtypes. A current challenge in the field is to translate new and emerging knowledge about different pathobiologic mechanisms in asthma into patient-specific therapies, with the ultimate goal of modifying the natural history of disease. Here, we describe the Precision Interventions for Severe and/or Exacerbation-Prone Asthma (PrecISE) Network, a groundbreaking collaborative effort of asthma researchers and biostatisticians from around the United States. The PrecISE Network was designed to conduct phase II/proof-of-concept clinical trials of precision interventions in the population with severe asthma, and is supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health. Using an innovative adaptive platform trial design, the PrecISE Network will evaluate up to 6 interventions simultaneously in biomarker-defined subgroups of subjects. We review the development and organizational structure of the PrecISE Network, and choice of interventions being studied. We hope that the PrecISE Network will enhance our understanding of asthma subtypes and accelerate the development of therapeutics for severe asthma.
Collapse
Affiliation(s)
- Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY.
| | | | - Anastasia Ivanova
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Elliot Israel
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Lisa M LaVange
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Praveen Akuthota
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Tara F Carr
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Merritt L Fajt
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | | | - Wanda K O'Neal
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | | | - Stanley J Szefler
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Mark A Aronica
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Allison J Burbank
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, Mo
| | - Laura Crotty Alexander
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Julie Bamdad
- Division of Lung Diseases, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Md
| | | | | | | | | | - Kim Erwin
- Institute for Healthcare Delivery Design, University of Illinois at Chicago, Chicago, Ill
| | | | - John V Fahy
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | | | - Benjamin Gaston
- Wells Center for Pediatric Research, Indiana University, Indianapolis, Ind
| | - Lynn B Gerald
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | | | - Daniel J Jackson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - John James
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Sumita Khatri
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - John P Kirwan
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, La
| | - Monica Kraft
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Ill
| | - Andrew H Liu
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Mark C Liu
- Pulmonary and Critical Care Medicine, Department of Medicine, the Johns Hopkins University, Baltimore, Md
| | - M Alison Marquis
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Fernando Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jacob Mey
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, La
| | - Wendy C Moore
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - James N Moy
- Rush University Medical Center, Chicago, Ill
| | - Victor E Ortega
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - David B Peden
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | | | - Michael C Peters
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | - Kristie Ross
- The Cleveland Clinic, Cleveland, Ohio; UH Rainbow Babies and Children's Hospitals, Cleveland, Ohio
| | - Maria Sanchez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | | | - Ronald L Sorkness
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Michael E Wechsler
- Children's Hospital Colorado, Aurora, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Sally E Wenzel
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Steven R White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Joe Zein
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amir A Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Patricia Noel
- Division of Lung Diseases, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Md
| |
Collapse
|
61
|
Hydropersulfides (RSSH) and Nitric Oxide (NO) Signaling: Possible Effects on S-Nitrosothiols (RS-NO). Antioxidants (Basel) 2022; 11:antiox11010169. [PMID: 35052673 PMCID: PMC8773330 DOI: 10.3390/antiox11010169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 01/05/2023] Open
Abstract
S-Nitrosothiol (RS-NO) formation in proteins and peptides have been implicated as factors in the etiology of many diseases and as possible regulators of thiol protein function. They have also been proposed as possible storage forms of nitric oxide (NO). However, despite their proposed functions/roles, there appears to be little consensus regarding the physiological mechanisms of RS-NO formation and degradation. Hydropersulfides (RSSH) have recently been discovered as endogenously generated species with unique reactivity. One important reaction of RSSH is with RS-NO, which leads to the degradation of RS-NO as well as the release of NO. Thus, it can be speculated that RSSH can be a factor in the regulation of steady-state RS-NO levels, and therefore may be important in RS-NO (patho)physiology. Moreover, RSSH-mediated NO release from RS-NO may be a possible mechanism allowing RS-NO to serve as a storage form of NO.
Collapse
|
62
|
GSNOR regulates ganoderic acid content in Ganoderma lucidum under heat stress through S-nitrosylation of catalase. Commun Biol 2022; 5:32. [PMID: 35017648 PMCID: PMC8752759 DOI: 10.1038/s42003-021-02988-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022] Open
Abstract
As a master regulator of the balance between NO signaling and protein S-nitrosylation, S-nitrosoglutathione (GSNO) reductase (GSNOR) is involved in various developmental processes and stress responses. However, the proteins and specific sites that can be S-nitrosylated, especially in microorganisms, and the physiological functions of S-nitrosylated proteins remain unclear. Herein, we show that the ganoderic acid (GA) content in GSNOR-silenced (GSNORi) strains is significantly lower (by 25%) than in wild type (WT) under heat stress (HS). Additionally, silencing GSNOR results in an 80% increase in catalase (CAT) activity, which consequently decreases GA accumulation via inhibition of ROS signaling. The mechanism of GSNOR-mediated control of CAT activity may be via protein S-nitrosylation. In support of this possibility, we show that CAT is S-nitrosylated (as shown via recombinant protein in vitro and via GSNORi strains in vivo). Additionally, Cys (cysteine) 401, Cys642 and Cys653 in CAT are S-nitrosylation sites (assayed via mass spectrometry analysis), and Cys401 may play a pivotal role in CAT activity. These findings indicate a mechanism by which GSNOR responds to stress and regulates secondary metabolite content through protein S-nitrosylation. Our results also define a new S-nitrosylation site and the function of an S-nitrosylated protein regulated by GSNOR in microorganisms. Liu et al. identify catalase as a target of S-nitrosylation by GSNOR and the specific sites of S-nitrosylation critical for its role in regulating secondary metabolite production in Ganoderma lucidum under heat stress. This study suggests that GSNOR regulates other metabolic pathways in microorganisms through S-nitrosylation of target proteins in response to environmental changes.
Collapse
|
63
|
OUP accepted manuscript. FEMS Microbiol Lett 2022; 369:6544667. [DOI: 10.1093/femsle/fnab162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
|
64
|
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
|
65
|
Zhao S, Song TY, Wang ZY, Gao J, Cao JW, Hu LL, Huang ZR, Xie LP, Ji Y. S-nitrosylation of Hsp90 promotes cardiac hypertrophy in mice through GSK3β signaling. Acta Pharmacol Sin 2021; 43:1979-1988. [PMID: 34934196 PMCID: PMC9343375 DOI: 10.1038/s41401-021-00828-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022]
Abstract
Cardiac hypertrophy, as one of the major predisposing factors for chronic heart failure, lacks effective interventions. Exploring the pathogenesis of cardiac hypertrophy will reveal potential therapeutic targets. S-nitrosylation is a kind of posttranslational modification that occurs at active cysteines of proteins to mediate various cellular processes. We here identified heat shock protein 90 (Hsp90) as a highly S-nitrosylated target in the hearts of rodents with hypertrophy, and the role of Hsp90 in cardiac hypertrophy remains undefined. The S-nitrosylation of Hsp90 (SNO-Hsp90) levels were elevated in angiotensin II (Ang II)- or phenylephrine (PE)-treated neonatal rat cardiomyocytes (NRCMs) in vitro as well as in cardiomyocytes isolated from mice subjected to transverse aortic constriction (TAC) in vivo. We demonstrated that the elevated SNO-Hsp90 levels were mediated by decreased S-nitrosoglutathione reductase (GSNOR) expression during cardiac hypertrophy, and delivery of GSNOR adeno-associated virus expression vectors (AAV9-GSNOR) decreased the SNO-Hsp90 levels to attenuate cardiac hypertrophy. Mass spectrometry analysis revealed that cysteine 589 (Cys589) might be the S-nitrosylation site of Hsp90. Delivery of the mutated AAV9-Hsp90-C589A inhibited SNO-Hsp90 levels and attenuated cardiac hypertrophy. We further revealed that SNO-Hsp90 led to increased interaction of glycogen synthase kinase 3β (GSK3β) and Hsp90, leading to elevated GSK3β phosphorylation and decreased eIF2Bε phosphorylation, thereby aggravating cardiac hypertrophy. Application of GSK3β inhibitor TWS119 abolished the protective effect of Hsp90-C589A mutation in Ang II-treated NRCMs. In conclusion, this study demonstrates a critical role of SNO-Hsp90 in cardiac hypertrophy, which may be of a therapeutic target for cardiac hypertrophy treatment.
Collapse
|
66
|
Pillars and Gaps of S-Nitrosylation-Dependent Epigenetic Regulation in Physiology and Cancer. Life (Basel) 2021; 11:life11121424. [PMID: 34947954 PMCID: PMC8704633 DOI: 10.3390/life11121424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Nitric oxide (NO) is a diffusible signaling molecule produced by three isoforms of nitric oxide synthase, which release NO during the metabolism of the amino acid arginine. NO participates in pathophysiological responses of many different tissues, inducing concentration-dependent effect. Indeed, while low NO levels generally have protective effects, higher NO concentrations induce cytotoxic/cytostatic actions. In recent years, evidences have been accumulated unveiling S-nitrosylation as a major NO-dependent post-translational mechanism ruling gene expression. S-nitrosylation is a reversible, highly regulated phenomenon in which NO reacts with one or few specific cysteine residues of target proteins generating S-nitrosothiols. By inducing this chemical modification, NO might exert epigenetic regulation through direct effects on both DNA and histones as well as through indirect actions affecting the functions of transcription factors and transcriptional co-regulators. In this light, S-nitrosylation may also impact on cancer cell gene expression programs. Indeed, it affects different cell pathways and functions ranging from the impairment of DNA damage repair to the modulation of the activity of signal transduction molecules, oncogenes, tumor suppressors, and chromatin remodelers. Nitrosylation is therefore a versatile tool by which NO might control gene expression programs in health and disease.
Collapse
|
67
|
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
|
68
|
Treffon P, Rossi J, Gabellini G, Trost P, Zaffagnini M, Vierling E. Quantitative Proteome Profiling of a S-Nitrosoglutathione Reductase (GSNOR) Null Mutant Reveals a New Class of Enzymes Involved in Nitric Oxide Homeostasis in Plants. FRONTIERS IN PLANT SCIENCE 2021; 12:787435. [PMID: 34956283 PMCID: PMC8695856 DOI: 10.3389/fpls.2021.787435] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 06/14/2023]
Abstract
Nitric oxide (NO) is a short-lived radical gas that acts as a signaling molecule in all higher organisms, and that is involved in multiple plant processes, including germination, root growth, and fertility. Regulation of NO-levels is predominantly achieved by reaction of oxidation products of NO with glutathione to form S-nitrosoglutathione (GSNO), the principal bioactive form of NO. The enzyme S-nitrosoglutathione reductase (GSNOR) is a major route of NADH-dependent GSNO catabolism and is critical to NO homeostasis. Here, we performed a proteomic analysis examining changes in the total leaf proteome of an Arabidopsis thaliana GSNOR null mutant (hot5-2/gsnor1-3). Significant increases or decreases in proteins associated with chlorophyll metabolism and with redox and stress metabolism provide insight into phenotypes observed in hot5-2/gsnor1-3 plants. Importantly, we identified a significant increase in proteins that belong to the aldo-keto reductase (AKR) protein superfamily, AKR4C8 and 9. Because specific AKRs have been linked to NO metabolism in mammals, we expressed and purified A. thaliana AKR4C8 and 9 and close homologs AKR4C10 and 11 and determined that they have NADPH-dependent activity in GSNO and S-nitroso-coenzyme A (SNO-CoA) reduction. Further, we found an increase of NADPH-dependent GSNO reduction activity in hot5-2/gsnor1-3 mutant plants. These data uncover a new, NADPH-dependent component of NO metabolism that may be integrated with NADH-dependent GSNOR activity to control NO homeostasis in plants.
Collapse
Affiliation(s)
- Patrick Treffon
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Jacopo Rossi
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | - Giuseppe Gabellini
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | - Paolo Trost
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | - Mirko Zaffagnini
- Department of Pharmacy and Biotechnologies, University of Bologna, Bologna, Italy
| | - Elizabeth Vierling
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
69
|
Liu M, Zaman R, Sawczak V, Periasamy A, Sun F, Zaman K. S-nitrosothiols signaling in cystic fibrosis airways. J Biosci 2021. [DOI: 10.1007/s12038-021-00223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
70
|
Sebag SC, Zhang Z, Qian Q, Li M, Zhu Z, Harata M, Li W, Zingman LV, Liu L, Lira VA, Potthoff MJ, Bartelt A, Yang L. ADH5-mediated NO bioactivity maintains metabolic homeostasis in brown adipose tissue. Cell Rep 2021; 37:110003. [PMID: 34788615 PMCID: PMC8640996 DOI: 10.1016/j.celrep.2021.110003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/23/2021] [Accepted: 10/22/2021] [Indexed: 01/21/2023] Open
Abstract
Brown adipose tissue (BAT) thermogenic activity is tightly regulated by cellular redox status, but the underlying molecular mechanisms are incompletely understood. Protein S-nitrosylation, the nitric-oxide-mediated cysteine thiol protein modification, plays important roles in cellular redox regulation. Here we show that diet-induced obesity (DIO) and acute cold exposure elevate BAT protein S-nitrosylation, including UCP1. This thermogenic-induced nitric oxide bioactivity is regulated by S-nitrosoglutathione reductase (GSNOR; alcohol dehydrogenase 5 [ADH5]), a denitrosylase that balances the intracellular nitroso-redox status. Loss of ADH5 in BAT impairs cold-induced UCP1-dependent thermogenesis and worsens obesity-associated metabolic dysfunction. Mechanistically, we demonstrate that Adh5 expression is induced by the transcription factor heat shock factor 1 (HSF1), and administration of an HSF1 activator to BAT of DIO mice increases Adh5 expression and significantly improves UCP1-mediated respiration. Together, these data indicate that ADH5 controls BAT nitroso-redox homeostasis to regulate adipose thermogenesis, which may be therapeutically targeted to improve metabolic health.
Collapse
Affiliation(s)
- Sara C. Sebag
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,These authors contributed equally
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,These authors contributed equally
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Zhiyong Zhu
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mikako Harata
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Wenxian Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Leonid V. Zingman
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Limin Liu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vitor A. Lira
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J. Potthoff
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention, Ludwig Maximilians University Munich Pettenkoferstr. 9, 80336 Munich, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Technische Universität München, Biedersteiner Str. 29, 80802 München, Germany,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Lead contact,Correspondence:
| |
Collapse
|
71
|
Zeng M, He Y, Gao X, Wang Y, Deng S, Ye T, Wang X, Xue H. Characteristics and functions of glyceraldehyde 3-phosphate dehydrogenase S-nitrosylation during controlled aging of elm and Arabidopsis seeds. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:7020-7034. [PMID: 34244712 DOI: 10.1093/jxb/erab322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/08/2021] [Indexed: 06/13/2023]
Abstract
Seed aging is the gradual decline in seed vigor, during which programmed cell death (PCD) occurs. The functions of nitric oxide (NO) are exerted through protein S-nitrosylation, a reversible post-translational modification. During seed aging, more than 80 proteins are S-nitrosylated, but the particular role of individual proteins is unknown. Here, we showed that the S-nitrosylation level of glyceraldehyde 3-phosphate dehydrogenase (UpGAPDH) in elm (Ulmus pumila L.) seeds increased after controlled deterioration treatment. UpGAPDH was S-nitrosylated at Cys154 during S-nitrosoglutathione (GSNO) treatment, and its oligomerization was triggered both in vitro and in elm seeds. Interestingly, UpGAPDH interacted with the mitochondrial voltage-dependent anion channel in an S-nitrosylation-dependent way. Some UpGAPDH-green fluorescent protein in Arabidopsis protoplasts co-localized with mitochondria during the GSNO treatment, while the S-nitrosylation-defective UpGAPDH C154S-GFP protein did not. Seeds of oxUpGAPDH lines showed cell death and lost seed vigor rapidly during controlled deterioration treatment-triggered seed aging, while those overexpressing S-nitrosylation-defective UpGAPDH-Cys154 did not. Our results suggest that S-nitrosylation of UpGAPDH may accelerate cell death and seed deterioration during controlled deterioration treatment. These results provide new insights into the effects of UpGAPDH S-nitrosylation on protein interactions and seed aging.
Collapse
Affiliation(s)
- MeiYan Zeng
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| | - YuQi He
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| | - Xue Gao
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| | - Yu Wang
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| | - ShiMing Deng
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
- College of Forestry and Horticulture, Hubei Minzu University, Enshi, China
| | - TianTian Ye
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| | - XiaoFeng Wang
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| | - Hua Xue
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, National Engineering Laboratory for Tree Breeding, College of Biological Sciences and Biotechnology, Beijing Forestry University, 35 Tsinghua East Road, Beijing, 100083, China
| |
Collapse
|
72
|
Liu Q, Gu T, Su LY, Jiao L, Qiao X, Xu M, Xie T, Yang LX, Yu D, Xu L, Chen C, Yao YG. GSNOR facilitates antiviral innate immunity by restricting TBK1 cysteine S-nitrosation. Redox Biol 2021; 47:102172. [PMID: 34678655 PMCID: PMC8577438 DOI: 10.1016/j.redox.2021.102172] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/17/2021] [Indexed: 11/16/2022] Open
Abstract
Innate immunity is the first line of host defense against pathogens. This process is modulated by multiple antiviral protein modifications, such as phosphorylation and ubiquitination. Here, we showed that cellular S-nitrosoglutathione reductase (GSNOR) is actively involved in innate immunity activation. GSNOR deficiency in mouse embryo fibroblasts (MEFs) and RAW264.7 macrophages reduced the antiviral innate immune response and facilitated herpes simplex virus-1 (HSV-1) and vesicular stomatitis virus (VSV) replication. Concordantly, HSV-1 infection in Gsnor-/- mice and wild-type mice with GSNOR being inhibited by N6022 resulted in higher mortality relative to the respective controls, together with severe infiltration of immune cells in the lungs. Mechanistically, GSNOR deficiency enhanced cellular TANK-binding kinase 1 (TBK1) protein S-nitrosation at the Cys423 site and inhibited TBK1 kinase activity, resulting in reduced interferon production for antiviral responses. Our study indicated that GSNOR is a critical regulator of antiviral responses and S-nitrosation is actively involved in innate immunity.
Collapse
Affiliation(s)
- Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Tianle Gu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ling-Yan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Lijin Jiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Beijing, 100101, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China
| | - Ting Xie
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Beijing, 100101, China
| | - Lu-Xiu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China
| | - Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China
| | - Chang Chen
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Beijing, 100101, China.
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
73
|
Pophal M, Grimmett ZW, Chu C, Margevicius S, Raffay T, Ross K, Jafri A, Giddings O, Stamler JS, Gaston B, Reynolds JD. Airway Thiol-NO Adducts as Determinants of Exhaled NO. Antioxidants (Basel) 2021; 10:antiox10101527. [PMID: 34679661 PMCID: PMC8532745 DOI: 10.3390/antiox10101527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022] Open
Abstract
Thiol-NO adducts such as S-nitrosoglutathione (GSNO) are endogenous bronchodilators in human airways. Decreased airway S-nitrosothiol concentrations are associated with asthma. Nitric oxide (NO), a breakdown product of GSNO, is measured in exhaled breath as a biomarker in asthma; an elevated fraction of expired NO (FENO) is associated with asthmatic airway inflammation. We hypothesized that FENO could reflect airway S-nitrosothiol concentrations. To test this hypothesis, we first studied the relationship between mixed expired NO and airway S-nitrosothiols in patients endotracheally intubated for respiratory failure. The inverse (Lineweaver-Burke type) relationship suggested that expired NO could reflect the rate of pulmonary S-nitrosothiol breakdown. We thus studied NO evolution from the lungs of mice (GSNO reductase −/−) unable reductively to catabolize GSNO. More NO was produced from GSNO in the −/− compared to wild type lungs. Finally, we formally tested the hypothesis that airway GSNO increases FENO using an inhalational challenge model in normal human subjects. FENO increased in all subjects tested, with a median t1/2 of 32.0 min. Taken together, these data demonstrate that FENO reports, at least in part, GSNO breakdown in the lungs. Unlike GSNO, NO is not present in the lungs in physiologically relevant concentrations. However, FENO following a GSNO challenge could be a non-invasive test for airway GSNO catabolism.
Collapse
Affiliation(s)
- Megan Pophal
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
| | - Zachary W. Grimmett
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
| | - Clara Chu
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
| | - Seunghee Margevicius
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Thomas Raffay
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Kristie Ross
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Anjum Jafri
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Olivia Giddings
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (T.R.); (K.R.); (A.J.); (O.G.)
| | - Jonathan S. Stamler
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
- Division of Cardiology, Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Benjamin Gaston
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-274-8899
| | - James D. Reynolds
- Institute for Transformative Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (M.P.); (Z.W.G.); (C.C.); (J.S.S.); (J.D.R.)
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Department of Anesthesiology & Perioperative Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
74
|
Ageeva-Kieferle A, Georgii E, Winkler B, Ghirardo A, Albert A, Hüther P, Mengel A, Becker C, Schnitzler JP, Durner J, Lindermayr C. Nitric oxide coordinates growth, development, and stress response via histone modification and gene expression. PLANT PHYSIOLOGY 2021; 187:336-360. [PMID: 34003928 PMCID: PMC8418403 DOI: 10.1093/plphys/kiab222] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/18/2021] [Indexed: 05/02/2023]
Abstract
Nitric oxide (NO) is a signaling molecule with multiple regulatory functions in plant physiology and stress response. In addition to direct effects on transcriptional machinery, NO executes its signaling function via epigenetic mechanisms. We report that light intensity-dependent changes in NO correspond to changes in global histone acetylation (H3, H3K9, and H3K9/K14) in Arabidopsis (Arabidopsis thaliana) wild-type leaves, and that this relationship depends on S-nitrosoglutathione reductase and histone deacetylase 6 (HDA6). The activity of HDA6 was sensitive to NO, demonstrating that NO participates in regulation of histone acetylation. Chromatin immunoprecipitation sequencing and RNA-seq analyses revealed that NO participates in the metabolic switch from growth and development to stress response. This coordinating function of NO might be particularly important in plant ability to adapt to a changing environment, and is therefore a promising foundation for mitigating the negative effects of climate change on plant productivity.
Collapse
Affiliation(s)
| | - Elisabeth Georgii
- Institute of Biochemical Plant Pathology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Barbro Winkler
- Research Unit Environmental Simulation, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Andrea Ghirardo
- Research Unit Environmental Simulation, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Andreas Albert
- Research Unit Environmental Simulation, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Patrick Hüther
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna BioCenter (VBC), Vienna 1030, Austria
| | - Alexander Mengel
- Institute of Biochemical Plant Pathology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Claude Becker
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna BioCenter (VBC), Vienna 1030, Austria
- Faculty of Biology, Ludwig-Maximilians-University Munich, LMU Biocenter, Martinsried 82152, Germany
| | - Jörg-Peter Schnitzler
- Research Unit Environmental Simulation, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Jörg Durner
- Institute of Biochemical Plant Pathology, Helmholtz Zentrum München, Neuherberg 85764, Germany
- Chair of Biochemical Plant Pathology, Technische Universität München, Freising 85354, Germany
| | - Christian Lindermayr
- Institute of Biochemical Plant Pathology, Helmholtz Zentrum München, Neuherberg 85764, Germany
- Author for communication:
| |
Collapse
|
75
|
Chatterji A, Banerjee D, Billiar TR, Sengupta R. Understanding the role of S-nitrosylation/nitrosative stress in inflammation and the role of cellular denitrosylases in inflammation modulation: Implications in health and diseases. Free Radic Biol Med 2021; 172:604-621. [PMID: 34245859 DOI: 10.1016/j.freeradbiomed.2021.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/22/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022]
Abstract
S-nitrosylation is a very fundamental post-translational modification of protein and non-protein thiols due the involvement of it in a variety of cellular processes including activation/inhibition of several ion channels such as ryanodine receptor in the cardiovascular system; blood vessel dilation; cGMP signaling and neurotransmission. S-nitrosothiol homeostasis in the cell is tightly regulated and perturbations in homeostasis result in an altered redox state leading to a plethora of disease conditions. However, the exact role of S-nitrosylated proteins and nitrosative stress metabolites in inflammation and in inflammation modulation is not well-reviewed. The cell utilizes its intricate defense mechanisms i.e. cellular denitrosylases such as Thioredoxin (Trx) and S-nitrosoglutathione reductase (GSNOR) systems to combat nitric oxide (NO) pathology which has also gained current attraction as novel anti-inflammatory molecules. This review attempts to provide state-of-the-art knowledge from past and present research on the mechanistic role of nitrosative stress intermediates (RNS, OONO-, PSNO) in pulmonary and autoimmune diseases and how cellular denitrosylases particularly GSNOR and Trx via imparting opposing effects can modulate and reduce inflammation in several health and disease conditions. This review would also bring into notice the existing gaps in current research where denitrosylases can be utilized for ameliorating inflammation that would leave avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- Ajanta Chatterji
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Debasmita Banerjee
- Department of Molecular Biology and Biotechnology, University of Kalyani, Block C, Nadia, Kalyani, West Bengal, 741235, India
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 5213, USA
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
76
|
Zimmermann J, Oestreicher J, Geissel F, Deponte M, Morgan B. An intracellular assay for activity screening and characterization of glutathione-dependent oxidoreductases. Free Radic Biol Med 2021; 172:340-349. [PMID: 34146665 DOI: 10.1016/j.freeradbiomed.2021.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022]
Abstract
The thioredoxin fold superfamily is highly diverse and contains many enzymatically active glutathione-dependent thiol-disulfide oxidoreductases, for example glutaredoxins and protein disulfide isomerases. However, many thioredoxin fold proteins remain completely uncharacterized, their cellular function is unknown, and it is unclear if they have a redox-dependent enzymatic activity with glutathione or not. Investigation of enzymatic activity traditionally involved time-consuming in vitro characterization of recombinant proteins, limiting the capacity to study novel mechanisms and structure-function relationships. To accelerate our investigation of glutathione-dependent oxidoreductases, we have developed a high-throughput and semi-quantitative assay in yeast. We combined overexpression of the glutathione transporter OPT1 with genetic fusion constructs between glutathione-dependent oxidoreductases and redox-sensitive green fluorescent protein 2 (roGFP2) to allow the rapid characterization of enzymatic activity with physiological substrates. We show that the kinetics of roGFP2 oxidation by glutathione disulfide correlate well with the in vitro-determined activity of the genetically fused glutaredoxins or mutants thereof. Our assay thus allows direct screening of glutaredoxin activity and rapid investigation of structure-function relationships. We also demonstrate that our assay can be used to monitor roGFP2 oxidation by S-nitrosoglutathione (GSNO). We show that glutaredoxins efficiently catalyze oxidation of roGFP2 by GSNO in both live yeast cells and in vitro. In summary, we have established a novel assay for activity screening and characterization of glutathione-dependent oxidoreductases.
Collapse
Affiliation(s)
- Jannik Zimmermann
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123 Saarbrücken, Germany
| | - Julian Oestreicher
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123 Saarbrücken, Germany
| | - Fabian Geissel
- Faculty of Chemistry, Department of Biochemistry, University of Kaiserslautern, Kaiserslautern, Germany
| | - Marcel Deponte
- Faculty of Chemistry, Department of Biochemistry, University of Kaiserslautern, Kaiserslautern, Germany.
| | - Bruce Morgan
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123 Saarbrücken, Germany.
| |
Collapse
|
77
|
Mikami R, Tsukagoshi S, Oda Y, Arai K. S-Denitrosylase-like Activity of Cyclic Diselenides Conjugated with Xaa-His Dipeptide: Role of Proline Spacer as a Key Activity Booster. Chembiochem 2021; 23:e202100394. [PMID: 34350692 DOI: 10.1002/cbic.202100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Indexed: 11/05/2022]
Abstract
This study developed dipeptide-conjugated 1,2-diselenan-4-amine (1), i.e., 1-Xaa-His, as a new class of S-denitrosylase mimic. The synthesized compounds, especially 1-Pro-His, remarkably promoted S-denitrosylation of nitrosothiols (RSNO) via a catalytic cycle involving the reversible redox reaction between the diselenide and its corresponding diselenol ([SeH,SeH]) form with coexisting reductant thiols (R'SH), during which the [SeH,SeH] form as a key reactive species reduces RSNO to the corresponding thiol (RSH). Structural analyses of 1-Pro-His suggested that the peptide backbone of [SeH,SeH] is rigidly bent to form a γ-turn, possibly including an NH···Se hydrogen bond between the imidazole ring of His and selenol group, thus stabilizing the [SeH,SeH] form thermodynamically, and dramatically enhancing the catalytic activity. Furthermore, the synthetic compounds were found to prohibit S-nitrosylation-induced protein misfolding in the presence of RSNO, eventually implying their potential as a drug seed for misfolding diseases caused by the dysregulation of the S-denitrosylation system.
Collapse
Affiliation(s)
- Rumi Mikami
- Tokai University - Shonan Campus: Tokai Daigaku, Chemistry, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| | - Shunsuke Tsukagoshi
- Tokai University - Shonan Campus: Tokai Daigaku, Chemistry, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| | - Yoshiki Oda
- Tokai University - Shonan Campus: Tokai Daigaku, Technology Joint Management Office, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| | - Kenta Arai
- Tokai University, Chemistry, Kitakaname, 259-1292, Hiratsuka-shi, JAPAN
| |
Collapse
|
78
|
Yoon H, Park S, Lim M. Dynamics of photodissociation of nitric oxide from S-nitrosylated cysteine and N-acetylated cysteine derivatives in water. Phys Chem Chem Phys 2021; 23:13512-13525. [PMID: 34124727 DOI: 10.1039/d1cp01743h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cysteine and N-acetylated cysteine derivatives are ubiquitous in biological systems; they have thiol groups that bind NO to form S-nitrosothiols (RSNOs) such as S-nitrosocysteine (CySNO), S-nitroso-N-acetylcysteine (NacSNO), and S-nitroso-N-acetylpenicillamine (NapSNO). Although they have been utilised as thermally or catalytically decomposing NO donors, their photochemical applications are yet to be fully explored owing to the lack of photodissociation dynamics. To this end, the photoexcitation dynamics of these RSNOs in water at 330 nm were investigated using femtosecond time-resolved infrared (TRIR) spectroscopy over a broad time range encompassing the entire reaction, which includes the primary reaction, secondary reactions of the reaction intermediates, and product formation. We discovered that the acetate and amide groups in these RSNOs have strong vibrational bands sensitive to the bondage of NO and the electronic state of the compound, which facilitates the identification of reaction intermediates involved in photoexcitation. The simplest thiol available with the acetate group-thioglycolic acid-was nitrosylated; it produced S-nitrosothioglycolic acid (TgSNO) and was comparatively investigated. Transient absorption bands in the TRIR spectra of the RSNOs were assigned using quantum chemical calculations. Photoexcited cysteine-related RSNOs either decompose into RS and NO within 0.3 ps after excitation at 330 nm with a primary quantum yield (Φ1) of 0.46-1 or relax into an electronically excited intermediate state lying at 42 ± 3 kcal mol-1 above the ground state, which relaxes into the ground state with a time constant of 460-520 ps. A majority (62-80%) of the RS radical geminately rebinds with NO at a time constant of 3-7 ps. The remaining RS reacts with the neighbouring RSNO, which produces additional NO and RSSR with a (nearly) diffusion-limited rate constant that doubles the amount of NO produced; further, it remarkably extends the time window for the dissociated NO to react with the target compound. The final fraction of NO produced from these RSNOs at 330 nm was 0.32-0.58, and it depends on the geminate rebinding yield and Φ1. The detailed dynamics of the photoexcited RSNO can be utilised in the quantitative application of these RSNOs in practical use and in the synthesis of more efficient photoactivated NO precursors.
Collapse
Affiliation(s)
- Hojeong Yoon
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Korea
| | - Seongchul Park
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Korea
| | - Manho Lim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan 46241, Korea
| |
Collapse
|
79
|
Yan Y, Shi Q, Gong B. S-nitrosoglutathione Reductase-Mediated Nitric Oxide Affects Axillary Buds Outgrowth of Solanum lycopersicum L. by Regulating Auxin and Cytokinin Signaling. PLANT & CELL PHYSIOLOGY 2021; 62:458-471. [PMID: 33493306 DOI: 10.1093/pcp/pcab002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/07/2021] [Indexed: 06/12/2023]
Abstract
Auxin and cytokinin are two kinds of important phytohormones that mediate outgrowth of axillary buds in plants. How nitric oxide and its regulator of S-nitrosoglutathione reductase (GSNOR) take part in auxin and cytokinin signaling for controlling axillary buds outgrowth remains elusive. We investigated the roles of GSNOR during tomato axillary bud outgrowth by using physiological, biochemical and genetic approaches. GSNOR negatively regulated NO homeostasis. Suppression of GSNOR promoted axillary bud outgrowth by inhibiting the expression of FZY in both apical and axillary buds. Meanwhile, AUX1 and PIN1 were down-regulated in apical buds but up-regulated in axillary buds in GSNOR-suppressed plants. Thus, reduced IAA accumulation was shown in both apical buds and axillary buds of GSNOR-suppressed plants. GSNOR-mediated changes of NO and auxin affected cytokinin biosynthesis, transport, and signaling. And a decreased ratio of auxin: cytokinin was shown in axillary buds of GSNOR-suppressed plants, leading to bud dormancy breaking. We also found that the original NO signaling was generated by nitrate reductase (NR) catalyzing nitrate as substrate. NR-mediated NO reduced the GSNOR activity through S-nitrosylation of Cys-10, then induced a further NO burst, which played the above roles to promote axillary buds outgrowth. Together, GSNOR-mediated NO played important roles in controlling axillary buds outgrowth by altering the homeostasis and signaling of auxin and cytokinin in tomato plants.
Collapse
Affiliation(s)
- Yanyan Yan
- State Key Laboratory of Crop Biology, College of Horticulture Science and Engineering, Shandong Agricultural University, Taian 271018, P.R. China
| | - Qinghua Shi
- State Key Laboratory of Crop Biology, College of Horticulture Science and Engineering, Shandong Agricultural University, Taian 271018, P.R. China
| | - Biao Gong
- State Key Laboratory of Crop Biology, College of Horticulture Science and Engineering, Shandong Agricultural University, Taian 271018, P.R. China
| |
Collapse
|
80
|
Massa CM, Liu Z, Taylor S, Pettit AP, Stakheyeva MN, Korotkova E, Popova V, Atochina-Vasserman EN, Gow AJ. Biological Mechanisms of S-Nitrosothiol Formation and Degradation: How Is Specificity of S-Nitrosylation Achieved? Antioxidants (Basel) 2021; 10:antiox10071111. [PMID: 34356344 PMCID: PMC8301044 DOI: 10.3390/antiox10071111] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 01/21/2023] Open
Abstract
The modification of protein cysteine residues underlies some of the diverse biological functions of nitric oxide (NO) in physiology and disease. The formation of stable nitrosothiols occurs under biologically relevant conditions and time scales. However, the factors that determine the selective nature of this modification remain poorly understood, making it difficult to predict thiol targets and thus construct informatics networks. In this review, the biological chemistry of NO will be considered within the context of nitrosothiol formation and degradation whilst considering how specificity is achieved in this important post-translational modification. Since nitrosothiol formation requires a formal one-electron oxidation, a classification of reaction mechanisms is proposed regarding which species undergoes electron abstraction: NO, thiol or S-NO radical intermediate. Relevant kinetic, thermodynamic and mechanistic considerations will be examined and the impact of sources of NO and the chemical nature of potential reaction targets is also discussed.
Collapse
Affiliation(s)
- Christopher M. Massa
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08848, USA; (C.M.M.); (Z.L.); (S.T.); (A.P.P.)
| | - Ziping Liu
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08848, USA; (C.M.M.); (Z.L.); (S.T.); (A.P.P.)
| | - Sheryse Taylor
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08848, USA; (C.M.M.); (Z.L.); (S.T.); (A.P.P.)
| | - Ashley P. Pettit
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08848, USA; (C.M.M.); (Z.L.); (S.T.); (A.P.P.)
| | - Marena N. Stakheyeva
- RASA Center in Tomsk, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.N.S.); (E.N.A.-V.)
- Institute of Natural Resources, Tomsk Polytechnic University, Lenin Av. 30, 634050 Tomsk, Russia; (E.K.); (V.P.)
| | - Elena Korotkova
- Institute of Natural Resources, Tomsk Polytechnic University, Lenin Av. 30, 634050 Tomsk, Russia; (E.K.); (V.P.)
| | - Valentina Popova
- Institute of Natural Resources, Tomsk Polytechnic University, Lenin Av. 30, 634050 Tomsk, Russia; (E.K.); (V.P.)
| | - Elena N. Atochina-Vasserman
- RASA Center in Tomsk, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.N.S.); (E.N.A.-V.)
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew J. Gow
- Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08848, USA; (C.M.M.); (Z.L.); (S.T.); (A.P.P.)
- RASA Center in Tomsk, Tomsk Polytechnic University, 634050 Tomsk, Russia; (M.N.S.); (E.N.A.-V.)
- Correspondence: ; Tel.: +1-848-445-4612
| |
Collapse
|
81
|
Raffay TM, Bonilla-Fernandez K, Jafri A, Sopi RB, Smith LA, Cui F, O'Reilly M, Zhang R, Hodges CA, MacFarlane PM, Deutsch G, Martin RJ, Gaston B. Bronchopulmonary Dysplasia and Pulmonary Hypertension. The Role of Smooth Muscle adh5. Am J Respir Cell Mol Biol 2021; 65:70-80. [PMID: 33780653 DOI: 10.1165/rcmb.2020-0289oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by alveolar simplification, airway hyperreactivity, and pulmonary hypertension. In our BPD model, we have investigated the metabolism of the bronchodilator and pulmonary vasodilator GSNO (S-nitrosoglutathione). We have shown the GSNO catabolic enzyme encoded by adh5 (alcohol dehydrogenase-5), GSNO reductase, is epigenetically upregulated in hyperoxia. Here, we investigated the distribution of GSNO reductase expression in human BPD and created an animal model that recapitulates the human data. Blinded comparisons of GSNO reductase protein expression were performed in human lung tissues from infants and children with and without BPD. BPD phenotypes were evaluated in global (adh5-/-) and conditional smooth muscle (smooth muscle/adh5-/-) adh5 knockout mice. GSNO reductase was prominently expressed in the airways and vessels of human BPD subjects. Compared with controls, expression was greater in BPD smooth muscle, particularly in vascular smooth muscle (2.4-fold; P = 0.003). The BPD mouse model of neonatal hyperoxia caused significant alveolar simplification, airway hyperreactivity, and right ventricular and vessel hypertrophy. Global adh5-/- mice were protected from all three aspects of BPD, whereas smooth muscle/adh5-/- mice were only protected from pulmonary hypertensive changes. These data suggest adh5 is required for the development of BPD. Expression in the pulmonary vasculature is relevant to the pathophysiology of BPD-associated pulmonary hypertension. GSNO-mimetic agents or GSNO reductase inhibitors, both of which are currently in clinical trials for other conditions, could be considered for further study in BPD.
Collapse
Affiliation(s)
| | - Koby Bonilla-Fernandez
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio.,San Juan Bautista School of Medicine, Caguas, Puerto Rico
| | - Anjum Jafri
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Laura A Smith
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Feifei Cui
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Rongli Zhang
- Department of Medicine, Institute for Transformative Molecular Medicine, Cardiovascular Research Institute, University Hospitals Cleveland Medical Center, and
| | - Craig A Hodges
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio; and
| | | | - Gail Deutsch
- Department of Pathology, Seattle Children's Hospital & University of Washington, Seattle, Washington
| | | | - Benjamin Gaston
- Division of Pediatric Pulmonology, Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital and Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio.,Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| |
Collapse
|
82
|
Rasool G, Buchholz G, Yasmin T, Shabbir G, Abbasi NA, Malik SI. Overexpression of SlGSNOR impairs in vitro shoot proliferation and developmental architecture in tomato but confers enhanced disease resistance. JOURNAL OF PLANT PHYSIOLOGY 2021; 261:153433. [PMID: 33990008 DOI: 10.1016/j.jplph.2021.153433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/11/2021] [Accepted: 04/23/2021] [Indexed: 06/12/2023]
Abstract
The pervasive presence of nitric oxide (NO) in cells and its role in modifying cystein residues through protein S-nitrosylation is a remarkable redox based signalling mechanism regulating a variety of cellular processes. S-NITROSOGLUTATHIONE REDUCTASE (GSNOR) governs NO bioavailability by the breakdown of S-nitrosoglutathione (GSNO), fine-tunes NO signalling and controls total cellular S-nitrosylated proteins. Most of the published data on GSNOR functional analysis is based on the model plant Arabidopsis with no previous report for its effect on in vitro regeneration of tissue cultured plants. Moreover, the effect of GSNOR overexpression (O.E) on tomato growth, development and disease resistance remains enigmatic. Here we show that SlGSNOR O.E in tomato alters multiple developmental programs from in vitro culture establishment to plant growth and fruit set. Moreover, constitutive SlGSNOR O.E in tomato showed enhanced resistance against early blight (EB) disease caused by Alternaria solani and reduction in hypersensitive response (HR)-mediated cell death after Pseudomonas syringae pv. tomato DC3000 (Pst DC3000) infiltrations. High GSNOR transcript levels led to the inhibition of in vitro shoot proliferation in transformed explants as revealed by the fluorescence microscopy after YFP labelling. Transgenic tomato lines overexpressing SlGSNOR showed defective phenotypes exhibiting stunted plant growth and bushy-type plants due to loss of apical dominance, along with reduced seed germination and delayed flowering. Furthermore, SlGSNOR O.E plants exhibited altered leaf arrangement, fruit shape and modified locules number in tomato fruit. These findings give a novel insight into a multifaceted regulatory role of SlGSNOR in tomato plant development, reproduction and response to pathogens.
Collapse
Affiliation(s)
- Ghulam Rasool
- Department of Plant Breeding and Genetics, Faculty of Crop and Food Sciences, PMAS Arid Agriculture University Rawalpindi, Rawalpindi, 46300, Pakistan
| | - Guenther Buchholz
- RLP AgroScience GmbH, AlPlanta - Institute for Plant Research, Neustadt, Germany
| | - Tayyaba Yasmin
- Department of Biosciences, COMSATS University Islamabad, Park Road, Islamabad, 45550, Pakistan
| | - Ghulam Shabbir
- Department of Plant Breeding and Genetics, Faculty of Crop and Food Sciences, PMAS Arid Agriculture University Rawalpindi, Rawalpindi, 46300, Pakistan
| | - Nadeem Akthar Abbasi
- Department of Horticulture, Faculty of Crop and Food Sciences, PMAS Arid Agriculture University Rawalpindi, Rawalpindi, 46300, Pakistan
| | - Saad Imran Malik
- Department of Plant Breeding and Genetics, Faculty of Crop and Food Sciences, PMAS Arid Agriculture University Rawalpindi, Rawalpindi, 46300, Pakistan.
| |
Collapse
|
83
|
Zhang L, Song H, Li B, Wang M, Di D, Lin X, Kronzucker HJ, Shi W, Li G. Induction of S-nitrosoglutathione reductase protects root growth from ammonium toxicity by regulating potassium homeostasis in Arabidopsis and rice. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:4548-4564. [PMID: 33772588 DOI: 10.1093/jxb/erab140] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/24/2021] [Indexed: 05/12/2023]
Abstract
Ammonium (NH4+) is toxic to root growth in most plants already at moderate levels of supply, but mechanisms of root growth tolerance to NH4+ remain poorly understood. Here, we report that high levels of NH4+ induce nitric oxide (NO) accumulation, while inhibiting potassium (K+) acquisition via SNO1 (sensitive to nitric oxide 1)/SOS4 (salt overly sensitive 4), leading to the arrest of primary root growth. High levels of NH4+ also stimulated the accumulation of GSNOR (S-nitrosoglutathione reductase) in roots. GSNOR overexpression improved root tolerance to NH4+. Loss of GSNOR further induced NO accumulation, increased SNO1/SOS4 activity, and reduced K+ levels in root tissue, enhancing root growth sensitivity to NH4+. Moreover, the GSNOR-like gene, OsGSNOR, is also required for NH4+ tolerance in rice. Immunoblotting showed that the NH4+-induced GSNOR protein accumulation was abolished in the VTC1- (vitamin C1) defective mutant vtc1-1, which is hypersensititive to NH4+ toxicity. GSNOR overexpression enhanced vtc1-1 root tolerance to NH4+. Our findings suggest that induction of GSNOR increases NH4+ tolerance in Arabidopsis roots by counteracting NO-mediated suppression of tissue K+, which depends on VTC1 function.
Collapse
Affiliation(s)
- Lin Zhang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing, China
| | - Haiyan Song
- Academic Affairs Office, Foshan University, Foshan, China
| | - Baohai Li
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, HangzhouChina
| | - Meng Wang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing, China
| | - Dongwei Di
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing, China
| | - Xianyong Lin
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental & Resource Sciences, Zhejiang University, HangzhouChina
| | - Herbert J Kronzucker
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Weiming Shi
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing, China
| | - Guangjie Li
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil Science, Chinese Academy of Sciences, Nanjing, China
| |
Collapse
|
84
|
Sasidharan R, Schippers JHM, Schmidt RR. Redox and low-oxygen stress: signal integration and interplay. PLANT PHYSIOLOGY 2021; 186:66-78. [PMID: 33793937 PMCID: PMC8154046 DOI: 10.1093/plphys/kiaa081] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 05/21/2023]
Abstract
Plants are aerobic organisms relying on oxygen to serve their energy needs. The amount of oxygen available to sustain plant growth can vary significantly due to environmental constraints or developmental programs. In particular, flooding stress, which negatively impacts crop productivity, is characterized by a decline in oxygen availability. Oxygen fluctuations result in an altered redox balance and the formation of reactive oxygen/nitrogen species (ROS/RNS) during the onset of hypoxia and upon re-oxygenation. In this update, we provide an overview of the current understanding of the impact of redox and ROS/RNS on low-oxygen signaling and adaptation. We first focus on the formation of ROS and RNS during low-oxygen conditions. Following this, we examine the impact of hypoxia on cellular and organellar redox systems. Finally, we describe how redox and ROS/RNS participate in signaling events during hypoxia through potential post-translational modifications (PTMs) of hypoxia-relevant proteins. The aim of this update is to define our current understanding of the field and to provide avenues for future research directions.
Collapse
Affiliation(s)
- Rashmi Sasidharan
- Plant Ecophysiology, Institute of Environmental Biology, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Jos H M Schippers
- Department of Molecular Genetics, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, Seeland 06466, Germany
| | - Romy R Schmidt
- Faculty of Biology, Plant Biotechnology Group, Bielefeld University, Bielefeld 33615, Germany
- Author for communication:
| |
Collapse
|
85
|
Fujii J, Homma T, Miyata S, Takahashi M. Pleiotropic Actions of Aldehyde Reductase (AKR1A). Metabolites 2021; 11:343. [PMID: 34073440 PMCID: PMC8227408 DOI: 10.3390/metabo11060343] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
We provide an overview of the physiological roles of aldehyde reductase (AKR1A) and also discuss the functions of aldose reductase (AKR1B) and other family members when necessary. Many types of aldehyde compounds are cytotoxic and some are even carcinogenic. Such toxic aldehydes are detoxified via the action of AKR in an NADPH-dependent manner and the resulting products may exert anti-diabetic and anti-tumorigenic activity. AKR1A is capable of reducing 3-deoxyglucosone and methylglyoxal, which are reactive intermediates that are involved in glycation, a non-enzymatic glycosylation reaction. Accordingly, AKR1A is thought to suppress the formation of advanced glycation end products (AGEs) and prevent diabetic complications. AKR1A and, in part, AKR1B are responsible for the conversion of d-glucuronate to l-gulonate which constitutes a process for ascorbate (vitamin C) synthesis in competent animals. AKR1A is also involved in the reduction of S-nitrosylated glutathione and coenzyme A and thereby suppresses the protein S-nitrosylation that occurs under conditions in which the production of nitric oxide is stimulated. As the physiological functions of AKR1A are currently not completely understood, the genetic modification of Akr1a could reveal the latent functions of AKR1A and differentiate it from other family members.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan;
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan;
| | - Satoshi Miyata
- Miyata Diabetes and Metabolism Clinic, 5-17-21 Fukushima, Fukushima-ku, Osaka 553-0003, Japan;
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| |
Collapse
|
86
|
Thompson CM, Gentry R, Fitch S, Lu K, Clewell HJ. An updated mode of action and human relevance framework evaluation for Formaldehyde-Related nasal tumors. Crit Rev Toxicol 2021; 50:919-952. [PMID: 33599198 DOI: 10.1080/10408444.2020.1854679] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Formaldehyde is a reactive aldehyde naturally present in all plant and animal tissues and a critical component of the one-carbon metabolism pathway. It is also a high production volume chemical used in the manufacture of numerous products. Formaldehyde is also one of the most well-studied chemicals with respect to environmental fate, biology, and toxicology-including carcinogenic potential, and mode of action (MOA). In 2006, a published MOA for formaldehyde-induced nasal tumors in rats concluded that nasal tumors were most likely driven by cytotoxicity and regenerative cell proliferation, with possible contributions from direct genotoxicity. In the past 15 years, new research has better informed the MOA with the publication of in vivo genotoxicity assays, toxicogenomic analyses, and development of ultra-sensitive methods to measure endogenous and exogenous formaldehyde-induced DNA adducts. Herein, we review and update the MOA for nasal tumors, with particular emphasis on the numerous studies published since 2006. These new studies further underscore the involvement of cytotoxicity and regenerative cell proliferation, and further inform the genotoxic potential of inhaled formaldehyde. The data lend additional support for the use of mechanistic data for the derivation of toxicity criteria and/or scientifically supported approaches for low-dose extrapolation for the risk assessment of formaldehyde.
Collapse
Affiliation(s)
| | | | | | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, USA
| | | |
Collapse
|
87
|
Pharmacological Inhibition of S-Nitrosoglutathione Reductase Reduces Cardiac Damage Induced by Ischemia-Reperfusion. Antioxidants (Basel) 2021; 10:antiox10040555. [PMID: 33918310 PMCID: PMC8065739 DOI: 10.3390/antiox10040555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 01/09/2023] Open
Abstract
The cardioprotective effects of nitric oxide (NO) have been described through S-nitrosylation of several important proteins in the mitochondria of the cardiomyocyte. S-nitrosoglutathione reductase (GSNOR) is an enzyme involved in the metabolism of S-nitrosothiols by producing denitrosylation, thus limiting the cardioprotective effect of NO. The effect of GSNOR inhibition on the damage by cardiac ischemia–reperfusion is still unclear. We tested the hypothesis that pharmacological inhibition of GSNOR promotes cardioprotection by increasing the levels of protein S-nitrosylation. In a model of ischemia–reperfusion in isolated rat heart, the effect of a GSNOR inhibitor, 5-chloro-3-(2-[4-ethoxyphenyl) (ethyl) amino]-2-oxoethyl)-1H-indole-2-carboxylic acid (C2), was investigated. Ventricular function and hemodynamics were determined, in addition to tissue damage and S-nitrosylation of mitochondrial proteins. Hearts treated with C2 showed a lower release of myocardial damage marker creatine kinase and a reduction in the infarcted area. It also improved post-ischemia ventricular function compared to controls. These results were associated with increasing protein S-nitrosylation, specifically of the mitochondrial complexes III and V. The pharmacological inhibition of GSNOR showed a concentration-dependent cardioprotective effect, being observed in functional parameters and myocardial damage, which was maximal at 1 µmol/L, associated with increased S-nitrosylation of mitochondrial proteins. These data suggest that GSNOR is an interesting pharmacological target for cardiac reperfusion injury.
Collapse
|
88
|
Yoshikawa Y, Nasuno R, Takagi H. An NADPH-independent mechanism enhances oxidative and nitrosative stress tolerance in yeast cells lacking glucose-6-phosphate dehydrogenase activity. Yeast 2021; 38:414-423. [PMID: 33648021 DOI: 10.1002/yea.3558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
The reduced form of nicotinamide adenine dinucleotide phosphate (NADPH), which is required for various redox systems involving antioxidative stress enzymes, is thus important for stress tolerance mechanisms. Here, we analyzed the stress response of the NADPH-depleted cells of Saccharomyces cerevisiae. A cell viability assay showed that the NADPH depletion induced by disruption of the ZWF1 gene encoding glucose-6-phosphate dehydrogenase, which is the major determinant of the intracellular NADPH/NADP+ ratio, enhanced the tolerance of S. cerevisiae to both oxidative and nitrosative stresses. The subsequent analyses demonstrated that the antioxidative transcriptional factor Yap1 was activated and the cytosolic catalase Ctt1, whose expression is regulated by Yap1, was upregulated in zwf1Δ cells irrespective of the presence or absence of stress stimuli. Moreover, deletion of the YAP1 or CTT1 gene inhibited the increased stress tolerance of zwf1Δ cells, indicating that Ctt1 dominantly contributed to the higher stress tolerance of zwf1Δ cells. Our findings suggest that an NADPH-independent mechanism enhances oxidative and nitrosative stress tolerance in ZWF1-lacking yeast cells.
Collapse
Affiliation(s)
- Yuki Yoshikawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Ryo Nasuno
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Hiroshi Takagi
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
89
|
Exploiting S-nitrosylation for cancer therapy: facts and perspectives. Biochem J 2021; 477:3649-3672. [PMID: 33017470 DOI: 10.1042/bcj20200064] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
S-nitrosylation, the post-translational modification of cysteines by nitric oxide, has been implicated in several cellular processes and tissue homeostasis. As a result, alterations in the mechanisms controlling the levels of S-nitrosylated proteins have been found in pathological states. In the last few years, a role in cancer has been proposed, supported by the evidence that various oncoproteins undergo gain- or loss-of-function modifications upon S-nitrosylation. Here, we aim at providing insight into the current knowledge about the role of S-nitrosylation in different aspects of cancer biology and report the main anticancer strategies based on: (i) reducing S-nitrosylation-mediated oncogenic effects, (ii) boosting S-nitrosylation to stimulate cell death, (iii) exploiting S-nitrosylation through synthetic lethality.
Collapse
|
90
|
Chen G, An N, Ye W, Huang S, Chen Y, Hu Z, Shen E, Zhu J, Gong W, Tong G, Zhu Y, Fang L, Cai C, Li X, Kim K, Jin L, Xiao J, Cong W. bFGF alleviates diabetes-associated endothelial impairment by downregulating inflammation via S-nitrosylation pathway. Redox Biol 2021; 41:101904. [PMID: 33706169 PMCID: PMC7972985 DOI: 10.1016/j.redox.2021.101904] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/26/2021] [Accepted: 02/15/2021] [Indexed: 01/02/2023] Open
Abstract
Protein S-nitrosylation is a reversible protein modification implicated in both physiological and pathophysiological regulation of protein function. However, the relationship between dysregulated S-nitrosylation homeostasis and diabetic vascular complications remains incompletely understood. Here, we demonstrate that basic fibroblast growth factor (bFGF) is a key regulatory link between S-nitrosylation homeostasis and inflammation, and alleviated endothelial dysfunction and angiogenic defects in diabetes. Subjecting human umbilical vein endothelial cells (HUVECs) to hyperglycemia and hyperlipidemia significantly decreased endogenous S-nitrosylated proteins, including S-nitrosylation of inhibitor kappa B kinase β (IKKβC179) and transcription factor p65 (p65C38), which was alleviated by bFGF co-treatment. Pretreatment with carboxy-PTIO (c-PTIO), a nitric oxide scavenger, abolished bFGF-mediated S-nitrosylation increase and endothelial protection. Meanwhile, nitrosylation-resistant IKKβC179S and p65C38S mutants exacerbated endothelial dysfunction in db/db mice, and in cultured HUVECs subjected to hyperglycemia and hyperlipidemia. Mechanistically, bFGF-mediated increase of S-nitrosylated IKKβ and p65 was attributed to synergistic effects of increased endothelial nitric oxide synthase (eNOS) and thioredoxin (Trx) activity. Taken together, the endothelial protective effect of bFGF under hyperglycemia and hyperlipidemia can be partially attributed to its role in suppressing inflammation via the S-nitrosylation pathway.
Collapse
Affiliation(s)
- Gen Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China; College of Pharmacy, Chonnam National University, Gwangju, 500-757, South Korea
| | - Ning An
- Department of Pharmacy, Ningbo Medical Center Lihuili Hospital, Ningbo, 315041, PR China
| | - Weijian Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PR China
| | - Shuai Huang
- Zhejiang Provincial Key Laboratory of Interventional Pulmonology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Yunjie Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Zhicheng Hu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Enzhao Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Wenjie Gong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Gaozan Tong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Yu Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Lexuan Fang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Chunyuan Cai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Kwonseop Kim
- College of Pharmacy, Chonnam National University, Gwangju, 500-757, South Korea.
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China.
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, PR China.
| |
Collapse
|
91
|
Manrique-Gil I, Sánchez-Vicente I, Torres-Quezada I, Lorenzo O. Nitric oxide function during oxygen deprivation in physiological and stress processes. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:904-916. [PMID: 32976588 PMCID: PMC7876777 DOI: 10.1093/jxb/eraa442] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/16/2020] [Indexed: 05/07/2023]
Abstract
Plants are aerobic organisms that have evolved to maintain specific requirements for oxygen (O2), leading to a correct respiratory energy supply during growth and development. There are certain plant developmental cues and biotic or abiotic stress responses where O2 is scarce. This O2 deprivation known as hypoxia may occur in hypoxic niches of plant-specific tissues and during adverse environmental cues such as pathogen attack and flooding. In general, plants respond to hypoxia through a complex reprogramming of their molecular activities with the aim of reducing the impact of stress on their physiological and cellular homeostasis. This review focuses on the fine-tuned regulation of hypoxia triggered by a network of gaseous compounds that includes O2, ethylene, and nitric oxide. In view of recent scientific advances, we summarize the molecular mechanisms mediated by phytoglobins and by the N-degron proteolytic pathway, focusing on embryogenesis, seed imbibition, and germination, and also specific structures, most notably root apical and shoot apical meristems. In addition, those biotic and abiotic stresses that comprise hypoxia are also highlighted.
Collapse
Affiliation(s)
- Isabel Manrique-Gil
- Departamento de Botánica y Fisiología Vegetal, Instituto Hispano-Luso de Investigaciones Agrarias (CIALE), Facultad de Biología, Universidad de Salamanca. C/ Río Duero 12, Salamanca, Spain
| | - Inmaculada Sánchez-Vicente
- Departamento de Botánica y Fisiología Vegetal, Instituto Hispano-Luso de Investigaciones Agrarias (CIALE), Facultad de Biología, Universidad de Salamanca. C/ Río Duero 12, Salamanca, Spain
| | - Isabel Torres-Quezada
- Departamento de Botánica y Fisiología Vegetal, Instituto Hispano-Luso de Investigaciones Agrarias (CIALE), Facultad de Biología, Universidad de Salamanca. C/ Río Duero 12, Salamanca, Spain
| | - Oscar Lorenzo
- Departamento de Botánica y Fisiología Vegetal, Instituto Hispano-Luso de Investigaciones Agrarias (CIALE), Facultad de Biología, Universidad de Salamanca. C/ Río Duero 12, Salamanca, Spain
- Correspondence:
| |
Collapse
|
92
|
Li ZC, Ren QW, Guo Y, Ran J, Ren XT, Wu NN, Xu HY, Liu X, Liu JZ. Dual Roles of GSNOR1 in Cell Death and Immunity in Tetraploid Nicotiana tabacum. FRONTIERS IN PLANT SCIENCE 2021; 12:596234. [PMID: 33643341 PMCID: PMC7902495 DOI: 10.3389/fpls.2021.596234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/04/2021] [Indexed: 06/12/2023]
Abstract
S-nitrosoglutathione reductase 1 (GSNOR1) is the key enzyme that regulates cellular homeostasis of S-nitrosylation. Although extensively studied in Arabidopsis, the roles of GSNOR1 in tetraploid Nicotiana species have not been investigated previously. To study the function of NtGSNOR1, we knocked out two NtGSNOR1 genes simultaneously in Nicotiana tabacum using clustered regularly interspaced short palindromic repeats (CRISPR)/caspase 9 (Cas9) technology. To our surprise, spontaneous cell death occurred on the leaves of the CRISPR/Cas9 lines but not on those of the wild-type (WT) plants, suggesting that NtGSNOR1 negatively regulates cell death. The natural cell death on the CRISPR/Cas9 lines could be a result from interactions between overaccumulated nitric oxide (NO) and hydrogen peroxide (H2O2). This spontaneous cell death phenotype was not affected by knocking out two Enhanced disease susceptibility 1 genes (NtEDS11a/1b) and thus was independent of the salicylic acid (SA) pathway. Unexpectedly, we found that the NtGSNOR1a/1b knockout plants displayed a significantly (p < 0.001) enhanced resistance to paraquat-induced cell death compared to WT plants, suggesting that NtGSNOR1 functions as a positive regulator of the paraquat-induced cell death. The increased resistance to the paraquat-induced cell death of the NtGSNOR1a/1b knockout plants was correlated with the reduced level of H2O2 accumulation. Interestingly, whereas the N gene-mediated resistance to Tobacco mosaic virus (TMV) was significantly enhanced (p < 0.001), the resistance to Pseudomonas syringae pv. tomato DC3000 was significantly reduced (p < 0.01) in the NtGSNOR1a/1b knockout lines. In summary, our results indicate that NtGSNOR1 functions as both positive and negative regulator of cell death under different conditions and displays distinct effects on resistance against viral and bacterial pathogens.
Collapse
Affiliation(s)
- Zhen-Chao Li
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Qian-Wei Ren
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Yan Guo
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Ran
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xiao-Tian Ren
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Ni-Ni Wu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Hui-Yang Xu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xia Liu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
- Zhejiang Provincial Key Laboratory of Biotechnology on Specialty Economic Plants, Zhejiang Normal University, Jinhua, China
| | - Jian-Zhong Liu
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
- Zhejiang Provincial Key Laboratory of Biotechnology on Specialty Economic Plants, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
93
|
Chatterji A, Sengupta R. Cellular S-denitrosylases: Potential role and interplay of Thioredoxin, TRP14, and Glutaredoxin systems in thiol-dependent protein denitrosylation. Int J Biochem Cell Biol 2021; 131:105904. [DOI: 10.1016/j.biocel.2020.105904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
|
94
|
Li B, Sun C, Lin X, Busch W. The Emerging Role of GSNOR in Oxidative Stress Regulation. TRENDS IN PLANT SCIENCE 2021; 26:156-168. [PMID: 33004257 DOI: 10.1016/j.tplants.2020.09.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 05/19/2023]
Abstract
Oxidative stress is a common event in aerobic organisms and a fundamental and unavoidable cost of the aerobic lifestyle. Reactive oxygen and nitrogen species (ROS/RNS) and iron (Fe) are the most common agents that trigger oxidative stress. A conserved enzyme in the S-nitrosoglutathione (GSNO) metabolism, GSNO reductase (GSNOR), modulates a multitude of abiotic and biotic stress responses. In this review, we focus on the emerging role of GSNOR as a master regulator in oxidative stress through its regulation of the interaction of ROS, RNS, and Fe, and highlight recent discoveries in post-translational modifications of GSNOR and functional variations of natural GSNOR variants during oxidative stress. Recent advances in understanding GSNOR regulation show promise for the modulation of oxidative stress in plants.
Collapse
Affiliation(s)
- Baohai Li
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental and Resource Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| | - Chengliang Sun
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental and Resource Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xianyong Lin
- MOE Key Laboratory of Environment Remediation and Ecological Health, College of Environmental and Resource Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| | - Wolfgang Busch
- Plant Biology Laboratory and Integrative Biology Laboratory, Salk Institute for Biological Studies, 10010 N Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
95
|
Bhatia V, Elnagary L, Dakshinamurti S. Tracing the path of inhaled nitric oxide: Biological consequences of protein nitrosylation. Pediatr Pulmonol 2021; 56:525-538. [PMID: 33289321 DOI: 10.1002/ppul.25201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/28/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a comprehensive regulator of vascular and airway tone. Endogenous NO produced by nitric oxide synthases regulates multiple signaling cascades, including activation of soluble guanylate cyclase to generate cGMP, relaxing smooth muscle cells. Inhaled NO is an established therapy for pulmonary hypertension in neonates, and has been recently proposed for the treatment of hypoxic respiratory failure and acute respiratory distress syndrome due to COVID-19. In this review, we summarize the effects of endogenous and exogenous NO on protein S-nitrosylation, which is the selective and reversible covalent attachment of a nitrogen monoxide group to the thiol side chain of cysteine. This posttranslational modification targets specific cysteines based on the acid/base sequence of surrounding residues, with significant impacts on protein interactions and function. S-nitrosothiol (SNO) formation is tightly compartmentalized and enzymatically controlled, but also propagated by nonenzymatic transnitrosylation of downstream protein targets. Redox-based nitrosylation and denitrosylation pathways dynamically regulate the equilibrium of SNO-proteins. We review the physiological roles of SNO proteins, including nitrosohemoglobin and autoregulation of blood flow through hypoxic vasodilation, and pathological effects of nitrosylation including inhibition of critical vasodilator enzymes; and discuss the intersection of NO source and dose with redox environment, in determining the effects of protein nitrosylation.
Collapse
Affiliation(s)
- Vikram Bhatia
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Lara Elnagary
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Shyamala Dakshinamurti
- Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada.,Section of Neonatology, Departments of Pediatrics and Physiology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
96
|
Mintz J, Vedenko A, Rosete O, Shah K, Goldstein G, Hare JM, Ramasamy R, Arora H. Current Advances of Nitric Oxide in Cancer and Anticancer Therapeutics. Vaccines (Basel) 2021; 9:94. [PMID: 33513777 PMCID: PMC7912608 DOI: 10.3390/vaccines9020094] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a short-lived, ubiquitous signaling molecule that affects numerous critical functions in the body. There are markedly conflicting findings in the literature regarding the bimodal effects of NO in carcinogenesis and tumor progression, which has important consequences for treatment. Several preclinical and clinical studies have suggested that both pro- and antitumorigenic effects of NO depend on multiple aspects, including, but not limited to, tissue of generation, the level of production, the oxidative/reductive (redox) environment in which this radical is generated, the presence or absence of NO transduction elements, and the tumor microenvironment. Generally, there are four major categories of NO-based anticancer therapies: NO donors, phosphodiesterase inhibitors (PDE-i), soluble guanylyl cyclase (sGC) activators, and immunomodulators. Of these, NO donors are well studied, well characterized, and also the most promising. In this study, we review the current knowledge in this area, with an emphasis placed on the role of NO as an anticancer therapy and dysregulated molecular interactions during the evolution of cancer, highlighting the strategies that may aid in the targeting of cancer.
Collapse
Affiliation(s)
- Joel Mintz
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL 33328, USA;
| | - Anastasia Vedenko
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.V.); (J.M.H.)
| | - Omar Rosete
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Khushi Shah
- College of Arts and Sciences, University of Miami, Miami, FL 33146, USA;
| | - Gabriella Goldstein
- College of Health Professions and Sciences, University of Central Florida, Orlando, FL 32816, USA;
| | - Joshua M. Hare
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.V.); (J.M.H.)
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Medicine, Cardiology Division, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Himanshu Arora
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (A.V.); (J.M.H.)
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
97
|
Hayashida K, Miyara SJ, Shinozaki K, Takegawa R, Yin T, Rolston DM, Choudhary RC, Guevara S, Molmenti EP, Becker LB. Inhaled Gases as Therapies for Post-Cardiac Arrest Syndrome: A Narrative Review of Recent Developments. Front Med (Lausanne) 2021; 7:586229. [PMID: 33585501 PMCID: PMC7873953 DOI: 10.3389/fmed.2020.586229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/04/2020] [Indexed: 01/22/2023] Open
Abstract
Despite recent advances in the management of post-cardiac arrest syndrome (PCAS), the survival rate, without neurologic sequelae after resuscitation, remains very low. Whole-body ischemia, followed by reperfusion after cardiac arrest (CA), contributes to PCAS, for which established pharmaceutical interventions are still lacking. It has been shown that a number of different processes can ultimately lead to neuronal injury and cell death in the pathology of PCAS, including vasoconstriction, protein modification, impaired mitochondrial respiration, cell death signaling, inflammation, and excessive oxidative stress. Recently, the pathophysiological effects of inhaled gases including nitric oxide (NO), molecular hydrogen (H2), and xenon (Xe) have attracted much attention. Herein, we summarize recent literature on the application of NO, H2, and Xe for treating PCAS. Recent basic and clinical research has shown that these gases have cytoprotective effects against PCAS. Nevertheless, there are likely differences in the mechanisms by which these gases modulate reperfusion injury after CA. Further preclinical and clinical studies examining the combinations of standard post-CA care and inhaled gas treatment to prevent ischemia-reperfusion injury are warranted to improve outcomes in patients who are being failed by our current therapies.
Collapse
Affiliation(s)
- Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Santiago J Miyara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Medicine, and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, New York, NY, United States.,Institute of Health Innovations and Outcomes Research, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Koichiro Shinozaki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Ryosuke Takegawa
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Daniel M Rolston
- Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Department of Surgery, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| | - Rishabh C Choudhary
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Sara Guevara
- Department of Surgery, Northwell Health, Manhasset, NY, United States
| | - Ernesto P Molmenti
- Department of Surgery, Medicine, and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, New York, NY, United States.,Institute of Health Innovations and Outcomes Research, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| |
Collapse
|
98
|
Abstract
Sudden cardiac arrest is a leading cause of death worldwide. Although the methods of cardiopulmonary resuscitation have been improved, mortality is still unacceptably high, and many survivors suffer from lasting neurological deficits due to the post-cardiac arrest syndrome (PCAS). Pathophysiologically, generalized vascular endothelial dysfunction accompanied by platelet activation and systemic inflammation has been implicated in the pathogenesis of PCAS. Because endothelial-derived nitric oxide (NO) plays a central role in maintaining vascular homeostasis, the role of NO-dependent signaling has been a focus of the intense investigation. Recent preclinical studies showed that therapeutic interventions that increase vascular NO bioavailability may improve outcomes after cardiac arrest complicated with PCAS. In particular, NO inhalation therapy has been shown to improve neurological outcomes and survival in multiple species. Clinical studies examining the safety and efficacy of inhaled NO in patients sustaining PCAS are warranted.
Collapse
|
99
|
Cirotti C, Rizza S, Giglio P, Poerio N, Allega MF, Claps G, Pecorari C, Lee J, Benassi B, Barilà D, Robert C, Stamler JS, Cecconi F, Fraziano M, Paull TT, Filomeni G. Redox activation of ATM enhances GSNOR translation to sustain mitophagy and tolerance to oxidative stress. EMBO Rep 2021; 22:e50500. [PMID: 33245190 PMCID: PMC7788447 DOI: 10.15252/embr.202050500] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
The denitrosylase S-nitrosoglutathione reductase (GSNOR) has been suggested to sustain mitochondrial removal by autophagy (mitophagy), functionally linking S-nitrosylation to cell senescence and aging. In this study, we provide evidence that GSNOR is induced at the translational level in response to hydrogen peroxide and mitochondrial ROS. The use of selective pharmacological inhibitors and siRNA demonstrates that GSNOR induction is an event downstream of the redox-mediated activation of ATM, which in turn phosphorylates and activates CHK2 and p53 as intermediate players of this signaling cascade. The modulation of ATM/GSNOR axis, or the expression of a redox-insensitive ATM mutant influences cell sensitivity to nitrosative and oxidative stress, impairs mitophagy and affects cell survival. Remarkably, this interplay modulates T-cell activation, supporting the conclusion that GSNOR is a key molecular effector of the antioxidant function of ATM and providing new clues to comprehend the pleiotropic effects of ATM in the context of immune function.
Collapse
Affiliation(s)
- Claudia Cirotti
- Department of BiologyTor Vergata UniversityRomeItaly
- Laboratory of Cell SignalingIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa LuciaRomeItaly
| | - Salvatore Rizza
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
| | - Paola Giglio
- Department of BiologyTor Vergata UniversityRomeItaly
| | - Noemi Poerio
- Department of BiologyTor Vergata UniversityRomeItaly
| | - Maria Francesca Allega
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
- Present address:
Cancer Research UK Beatson InstituteGarscube EstateGlasgowUK
| | | | - Chiara Pecorari
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
| | - Ji‐Hoon Lee
- Department of Molecular BiosciencesThe University of Texas at AustinAustinTXUSA
| | - Barbara Benassi
- Division of Health Protection TechnologiesENEA‐CasacciaRomeItaly
| | - Daniela Barilà
- Department of BiologyTor Vergata UniversityRomeItaly
- Laboratory of Cell SignalingIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa LuciaRomeItaly
| | - Caroline Robert
- INSERM, U981VillejuifFrance
- Université Paris SudUniversité Paris‐SaclayKremlin‐BicêtreFrance
- Oncology DepartmentGustave RoussyUniversité Paris‐SaclayVillejuifFrance
| | - Jonathan S Stamler
- Institute for Transformative Molecular MedicineCase Western Reserve University and Harrington Discovery InstituteUniversity Hospitals Case Medical CenterClevelandOHUSA
| | - Francesco Cecconi
- Department of BiologyTor Vergata UniversityRomeItaly
- Cell Stress and Survival UnitDanish Cancer Society Research CenterCopenhagenDenmark
- Department of Pediatric Hematology and OncologyIRCCS Bambino Gesù Children's HospitalRomeItaly
| | | | - Tanya T Paull
- Department of Molecular BiosciencesThe University of Texas at AustinAustinTXUSA
| | - Giuseppe Filomeni
- Department of BiologyTor Vergata UniversityRomeItaly
- Redox Signaling and Oxidative Stress GroupDanish Cancer Society Research CenterCopenhagenDenmark
- Center for Healthy AgingCopenhagen UniversityCopenhagenDenmark
| |
Collapse
|
100
|
Qi H, Xia FN, Xiao S. Autophagy in plants: Physiological roles and post-translational regulation. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2021; 63:161-179. [PMID: 32324339 DOI: 10.1111/jipb.12941] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/22/2020] [Indexed: 05/20/2023]
Abstract
In eukaryotes, autophagy helps maintain cellular homeostasis by degrading and recycling cytoplasmic materials via a tightly regulated pathway. Over the past few decades, significant progress has been made towards understanding the physiological functions and molecular regulation of autophagy in plant cells. Increasing evidence indicates that autophagy is essential for plant responses to several developmental and environmental cues, functioning in diverse processes such as senescence, male fertility, root meristem maintenance, responses to nutrient starvation, and biotic and abiotic stress. Recent studies have demonstrated that, similar to nonplant systems, the modulation of core proteins in the plant autophagy machinery by posttranslational modifications such as phosphorylation, ubiquitination, lipidation, S-sulfhydration, S-nitrosylation, and acetylation is widely involved in the initiation and progression of autophagy. Here, we provide an overview of the physiological roles and posttranslational regulation of autophagy in plants.
Collapse
Affiliation(s)
- Hua Qi
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Fan-Nv Xia
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shi Xiao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| |
Collapse
|