1
|
Ma C, Du T, Wang W, Liu Y, An Z, Hou Q, Xing L, Zhang W. Insights into the differences of caspase and apoptosis levels in pork longissimus thoracis muscles with different tenderness: A perspective on S-nitrosylation modification. Food Chem 2025; 471:142810. [PMID: 39793362 DOI: 10.1016/j.foodchem.2025.142810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/25/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
This study investigated the differences of caspase and apoptosis levels in pork with different tenderness from the perspective of S-nitrosylation and further explored their role during pork tenderization. Ten longissimus thoracis muscles selected from 36 individual carcasses based on shear force were divided into high (HT) and low (LT) tenderness groups (n = 5), respectively. Results demonstrated that total nitric oxide synthase activity and protein S-nitrosylation levels of LT group were higher than HT group, while myocyte apoptosis levels were lower in LT group (p < 0.05). Additionally, LT group possessed a lower caspase-3 activity while a higher abundance of intact caspase-3 and greater S-nitrosylation levels of cleaved caspase-3 (p < 0.05). However, none of the above differences were found in caspase-9 (p > 0.05). The lower tenderness in LT group might be associated with reduced caspase-3 activity resulted from increased S-nitrosylation levels of its active subunits, which delayed myocyte apoptosis and lowed the degradation of desmin and troponin T.
Collapse
Affiliation(s)
- Chao Ma
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Tongyao Du
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
| | - Wenxuan Wang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yujia Liu
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhenhong An
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qin Hou
- Key Laboratory of Chinese Cuisine Intangible Cultural Heritage Technology Inheritance, Ministry of Culture and Tourism, College of Tourism and Culinary Science, Yangzhou University, Yangzhou 225127, China
| | - Lujuan Xing
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangang Zhang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
2
|
Zhu XH, Zhou YP, Zhang Q, Zhu MY, Song XW, Li J, Chen J, Shi Y, Sun KJ, Zhang YJ, Zhang J, Xia T, Huang BS, Meng F, Zhou QG. A novel anti-epileptogenesis strategy of temporal lobe epilepsy based on nitric oxide donor. EMBO Mol Med 2025; 17:85-111. [PMID: 39653809 PMCID: PMC11730642 DOI: 10.1038/s44321-024-00168-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 01/15/2025] Open
Abstract
The molecular mechanism underlying the role of hippocampal hilar interneuron degeneration in temporal lobe epilepsy (TLE) remains unclear. Especially, very few studies have focused on the role of neuronal nitric oxide synthase (nNOS, encoded by Nos1) containing hilar interneurons in TLE. In the present study, Nos1 conditional knockout mice were constructed, and we found that selective deletion of Nos1 in hilar interneurons rather than dentate granular cells (DGCs) triggered epileptogenesis. The level of nNOS was downregulated in patients and mice with TLE. Nos1 deletion led to excessive epilepsy-like excitatory input circuit formation and hyperexcitation of DGCs. Replenishment of hilar nNOS protein blocked epileptogenic development and memory impairment in pilocarpine-induced TLE mice. Moreover, chronic treatment with DETA/NONOate, a slowly released exogenous nitric oxide (NO) donor, prevented aberrant neural circuits of DGCs and the consequent epileptogenesis without acute antiseizure effects. Therefore, we concluded that NO donor therapy may be a novel anti-epileptogenesis strategy, different from existing antiseizure medications (ASMs), for curing TLE.
Collapse
Affiliation(s)
- Xian-Hui Zhu
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Ya-Ping Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Changzhou Hygiene Vocational Technology College, Changzhou, 213002, China
| | - Qiao Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Suqian First Hospital, Nanjing Medical University, Suqian, 223800, China
| | - Ming-Yi Zhu
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Centre of Medicinal Preparations, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Xiao-Wei Song
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong First People's Hospital, Nantong, 226001, China
| | - Jun Li
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jiang Chen
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 210032, China
| | - Yun Shi
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 210032, China
| | - Kang-Jian Sun
- Department of Neurosurgery, Nanjing Jinling University, Nanjing, 210002, China
| | - Yong-Jie Zhang
- Department of Human Anatomy, Human Brain Tissue Resource Center of Nanjing Medical University, National Health and Disease Human Brain Tissue Resource Center-sub-center of Nanjing Medical University, Nanjing, 211166, China
| | - Jing Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- The Key Center of Gene Technology Drugs of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Tian Xia
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Bao-Sheng Huang
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China.
| | - Fan Meng
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- The Key Center of Gene Technology Drugs of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China.
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- The Key Center of Gene Technology Drugs of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China.
- Department of Pharmacy of First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
3
|
Hou Q, Ma C, Liu R, Kang Z, Zhang W. Exploring the Effects of S-Nitrosylation on Caspase-3 Modification and Myofibril Degradation of Beef In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21772-21780. [PMID: 39295075 DOI: 10.1021/acs.jafc.4c06663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
This study aimed to explore the effects of S-nitrosylation on caspase-3 modification and its subsequent effects on beef myofibril degradation in vitro. Recombinant caspase-3 was reacted with different concentrations of S-nitrosoglutathione (GSNO, nitric oxide donor) at 37 °C for 30 min and subsequently incubated with purified myofibrillar protein from bovine semimembranosus muscle. Results indicated that the activity of caspase-3 was significantly reduced after GSNO treatments (P < 0.05) and showed a dose-dependent inhibitory effect, which was attributed to the increased S-nitrosylation extent of caspase-3. LC-MS/MS analysis revealed that caspase-3 was S-nitrosylated at cysteine sites 116, 170, 184, 220, and 264. Moreover, the degradation of desmin and troponin-T was notably suppressed by S-nitrosylated caspase-3 (P < 0.05). To conclude, protein S-nitrosylation could modify the cysteine residues of caspase-3, which accounts for the reduced caspase-3 activity and further represses its proteolytic ability on beef myofibrillar protein.
Collapse
Affiliation(s)
- Qin Hou
- School of Tourism and Cuisine, Industrial Engineering Center for Huaiyang Cuisine of Jiangsu Province, Yangzhou University, Yangzhou, Jiangsu 225127, China
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Chao Ma
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Rui Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Zhuangli Kang
- School of Tourism and Cuisine, Industrial Engineering Center for Huaiyang Cuisine of Jiangsu Province, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Wangang Zhang
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education China, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| |
Collapse
|
4
|
Guil-Luna S, Sanchez-Montero MT, Rodríguez-Ariza A. S-Nitrosylation at the intersection of metabolism and autophagy: Implications for cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:189012. [PMID: 37918453 DOI: 10.1016/j.bbcan.2023.189012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/26/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
Metabolic plasticity, which determines tumour growth and metastasis, is now understood to be a flexible and context-specific process in cancer metabolism. One of the major pathways contributing to metabolic adaptations in eucaryotic cells is autophagy, a cellular degradation and recycling process that is activated during periods of starvation or stress to maintain metabolite and biosynthetic intermediate levels. Consequently, there is a close association between the metabolic adaptive capacity of tumour cells and autophagy-related pathways in cancer. Additionally, nitric oxide regulates protein function and signalling through S-nitrosylation, a post-translational modification that can also impact metabolism and autophagy. The primary objective of this review is to provide an up-to-date overview of the role of S-nitrosylation at the intersection of metabolism and autophagy in cancer. First, we will outline the involvement of S-nitrosylation in the metabolic adaptations that occur in tumours. Then, we will discuss the multifaceted role of autophagy in cancer, the interplay between metabolism and autophagy during tumour progression, and the contribution of S-nitrosylation to autophagic dysregulation in cancer. Finally, we will present insights into relevant therapeutic aspects and discuss prospects for the future.
Collapse
Affiliation(s)
- Silvia Guil-Luna
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Department of Comparative Anatomy and Pathology, Faculty of Veterinary Medicine of Córdoba, University of Córdoba, Córdoba, Spain
| | | | - Antonio Rodríguez-Ariza
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| |
Collapse
|
5
|
Seth D, Stomberski CT, McLaughlin PJ, Premont RT, Lundberg K, Stamler JS. Comparison of the Nitric Oxide Synthase Interactomes and S-Nitroso-Proteomes: Furthering the Case for Enzymatic S-Nitrosylation. Antioxid Redox Signal 2023; 39:621-634. [PMID: 37053107 PMCID: PMC10619892 DOI: 10.1089/ars.2022.0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/13/2023] [Accepted: 04/08/2023] [Indexed: 04/14/2023]
Abstract
Aims: S-nitrosylation of proteins is the main mechanism through which nitric oxide (NO) regulates cellular function and likely represents the archetype redox-based signaling system across aerobic and anaerobic organisms. How NO generated by different nitric oxide synthase (NOS) isoforms leads to specificity of S-nitrosylation remains incompletely understood. This study aimed to identify proteins interacting with, and whose S-nitrosylation is mediated by, human NOS isoforms in the same cellular system, thereby illuminating the contribution of individual NOSs to specificity. Results: Of the hundreds of proteins interacting with each NOS, many were also S-nitrosylated. However, a large proportion of S-nitrosylated proteins (SNO-proteins) did not associate with NOS. Moreover, most NOS interactors and SNO-proteins were unique to each isoform. The amount of NO produced by each NOS isoform was unrelated to the numbers of SNO-proteins. Thus, NOSs promoted S-nitrosylation of largely distinct sets of target proteins. Different signaling pathways were enriched downstream of each NOS. Innovation and Conclusion: The interactomes and SNOomes of individual NOS isoforms were largely distinct. Only a small fraction of SNO-proteins interacted with their respective NOS. Amounts of S-nitrosylation were unrelated to the amount of NO generated by NOSs. These data argue against free diffusion of NO or NOS interactions as being necessary or sufficient for S-nitrosylation and favor roles for additional enzymes and/or regulatory elements in imparting SNO-protein specificity. Antioxid. Redox Signal. 39, 621-634.
Collapse
Affiliation(s)
- Divya Seth
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Colin T. Stomberski
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Precious J. McLaughlin
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Richard T. Premont
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Kathleen Lundberg
- Center for Proteomics and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jonathan S. Stamler
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Cross-Regulation of the Cellular Redox System, Oxygen, and Sphingolipid Signalling. Metabolites 2023; 13:metabo13030426. [PMID: 36984866 PMCID: PMC10054022 DOI: 10.3390/metabo13030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Redox-active mediators are now appreciated as powerful molecules to regulate cellular dynamics such as viability, proliferation, migration, cell contraction, and relaxation, as well as gene expression under physiological and pathophysiological conditions. These molecules include the various reactive oxygen species (ROS), and the gasotransmitters nitric oxide (NO∙), carbon monoxide (CO), and hydrogen sulfide (H2S). For each of these molecules, direct targets have been identified which transmit the signal from the cellular redox state to a cellular response. Besides these redox mediators, various sphingolipid species have turned out as highly bioactive with strong signalling potential. Recent data suggest that there is a cross-regulation existing between the redox mediators and sphingolipid molecules that have a fundamental impact on a cell’s fate and organ function. This review will summarize the effects of the different redox-active mediators on sphingolipid signalling and metabolism, and the impact of this cross-talk on pathophysiological processes. The relevance of therapeutic approaches will be highlighted.
Collapse
|
7
|
Patel SH, Bachmann M, Kadow S, Wilson GC, Abdel-Salam MML, Xu K, Keitsch S, Soddemann M, Wilker B, Becker KA, Carpinteiro A, Ahmad SA, Szabo I, Gulbins E. Simultaneous targeting of mitochondrial Kv1.3 and lysosomal acid sphingomyelinase amplifies killing of pancreatic ductal adenocarcinoma cells in vitro and in vivo. J Mol Med (Berl) 2023; 101:295-310. [PMID: 36790532 PMCID: PMC10036429 DOI: 10.1007/s00109-023-02290-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 02/16/2023]
Abstract
Pancreas ductal adenocarcinoma (PDAC) remains a malignant tumor with very poor prognosis and low 5-year overall survival. Here, we aimed to simultaneously target mitochondria and lysosomes as a new treatment paradigm of malignant pancreas cancer in vitro and in vivo. We demonstrate that the clinically used sphingosine analog FTY-720 together with PAPTP, an inhibitor of mitochondrial Kv1.3, induce death of pancreas cancer cells in vitro and in vivo. The combination of both drugs results in a marked inhibition of the acid sphingomyelinase and accumulation of cellular sphingomyelin in vitro and in vivo in orthotopic and flank pancreas cancers. Mechanistically, PAPTP and FTY-720 cause a disruption of both mitochondria and lysosomes, an alteration of mitochondrial bioenergetics and accumulation of cytoplasmic Ca2+, events that collectively mediate cell death. Our findings point to an unexpected cross-talk between lysosomes and mitochondria mediated by sphingolipid metabolism. We show that the combination of PAPTP and FTY-720 induces massive death of pancreas cancer cells, thereby leading to a substantially delayed and reduced PDAC growth in vivo. KEY MESSAGES: FTY-720 inhibits acid sphingomyelinase in pancreas cancer cells (PDAC). FTY-720 induces sphingomyelin accumulation and lysosomal dysfunction. The mitochondrial Kv1.3 inhibitor PAPTP disrupts mitochondrial functions. PAPTP and FTY-720 synergistically kill PDAC in vitro. The combination of FTY-720 and PAPTP greatly delays PDAC growth in vivo.
Collapse
Affiliation(s)
- Sameer H Patel
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Magdalena Bachmann
- Department of Biologyand , CNR Institute of Neurosciences, University of Padua, Padua, Italy
| | - Stephanie Kadow
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Gregory C Wilson
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mostafa M L Abdel-Salam
- Department of Biologyand , CNR Institute of Neurosciences, University of Padua, Padua, Italy
| | - Kui Xu
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Simone Keitsch
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Matthias Soddemann
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Barbara Wilker
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Katrin Anne Becker
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Alexander Carpinteiro
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Syed A Ahmad
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ildiko Szabo
- Department of Biologyand , CNR Institute of Neurosciences, University of Padua, Padua, Italy.
| | - Erich Gulbins
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
8
|
Getsy PM, Young AP, Bates JN, Baby SM, Seckler JM, Grossfield A, Hsieh YH, Lewis THJ, Jenkins MW, Gaston B, Lewis SJ. S-nitroso-L-cysteine stereoselectively blunts the adverse effects of morphine on breathing and arterial blood gas chemistry while promoting analgesia. Biomed Pharmacother 2022; 153:113436. [PMID: 36076552 PMCID: PMC9464305 DOI: 10.1016/j.biopha.2022.113436] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 01/05/2023] Open
Affiliation(s)
- Paulina M Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Alex P Young
- Department of Pediatrics, University of Virginia, Charlottesville, VA, USA
| | - James N Bates
- Department of Anesthesia, University of Iowa, Iowa City, IA, USA
| | - Santhosh M Baby
- Galleon Pharmaceuticals, Inc., 213 Witmer Road, Horsham, PA, USA.
| | - James M Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alan Grossfield
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yee-Hsee Hsieh
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Tristan H J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Benjamin Gaston
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA; Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA; Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
9
|
Getsy PM, Baby SM, Gruber RB, Gaston B, Lewis THJ, Grossfield A, Seckler JM, Hsieh YH, Bates JN, Lewis SJ. S-Nitroso-L-Cysteine Stereoselectively Blunts the Deleterious Effects of Fentanyl on Breathing While Augmenting Antinociception in Freely-Moving Rats. Front Pharmacol 2022; 13:892307. [PMID: 35721204 PMCID: PMC9199495 DOI: 10.3389/fphar.2022.892307] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/26/2022] [Indexed: 01/08/2023] Open
Abstract
Endogenous and exogenously administered S-nitrosothiols modulate the activities of central and peripheral systems that control breathing. We have unpublished data showing that the deleterious effects of morphine on arterial blood-gas chemistry (i.e., pH, pCO2, pO2, and sO2) and Alveolar-arterial gradient (i.e., index of gas exchange) were markedly diminished in anesthetized Sprague Dawley rats that received a continuous intravenous infusion of the endogenous S-nitrosothiol, S-nitroso-L-cysteine. The present study extends these findings by showing that unanesthetized adult male Sprague Dawley rats receiving an intravenous infusion of S-nitroso-L-cysteine (100 or 200 nmol/kg/min) markedly diminished the ability of intravenous injections of the potent synthetic opioid, fentanyl (10, 25, and 50 μg/kg), to depress the frequency of breathing, tidal volume, and minute ventilation. Our study also found that the ability of intravenously injected fentanyl (10, 25, and 50 μg/kg) to disturb eupneic breathing, which was measured as a marked increase of the non-eupneic breathing index, was substantially reduced in unanesthetized rats receiving intravenous infusions of S-nitroso-L-cysteine (100 or 200 nmol/kg/min). In contrast, the deleterious effects of fentanyl (10, 25, and 50 μg/kg) on frequency of breathing, tidal volume, minute ventilation and non-eupneic breathing index were fully expressed in rats receiving continuous infusions (200 nmol/kg/min) of the parent amino acid, L-cysteine, or the D-isomer, namely, S-nitroso-D-cysteine. In addition, the antinociceptive actions of the above doses of fentanyl as monitored by the tail-flick latency assay, were enhanced by S-nitroso-L-cysteine, but not L-cysteine or S-nitroso-D-cysteine. Taken together, these findings add to existing knowledge that S-nitroso-L-cysteine stereoselectively modulates the detrimental effects of opioids on breathing, and opens the door for mechanistic studies designed to establish whether the pharmacological actions of S-nitroso-L-cysteine involve signaling processes that include 1) the activation of plasma membrane ion channels and receptors, 2) selective intracellular entry of S-nitroso-L-cysteine, and/or 3) S-nitrosylation events. Whether alterations in the bioavailability and bioactivity of endogenous S-nitroso-L-cysteine is a key factor in determining the potency/efficacy of fentanyl on breathing is an intriguing question.
Collapse
Affiliation(s)
- Paulina M. Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | | | - Ryan B. Gruber
- Galleon Pharmaceuticals, Inc., Horsham, PA, United States
| | - Benjamin Gaston
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tristan H. J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Alan Grossfield
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, United States
| | - James M. Seckler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Yee-Hsee Hsieh
- Division of Pulmonary, Critical Care and Sleep Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - James N. Bates
- Department of Anesthesia, University of Iowa, Iowa City, IA, United States
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
- Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
10
|
Ye H, Wu J, Liang Z, Zhang Y, Huang Z. Protein S-Nitrosation: Biochemistry, Identification, Molecular Mechanisms, and Therapeutic Applications. J Med Chem 2022; 65:5902-5925. [PMID: 35412827 DOI: 10.1021/acs.jmedchem.1c02194] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein S-nitrosation (SNO), a posttranslational modification (PTM) of cysteine (Cys) residues elicited by nitric oxide (NO), regulates a wide range of protein functions. As a crucial form of redox-based signaling by NO, SNO contributes significantly to the modulation of physiological functions, and SNO imbalance is closely linked to pathophysiological processes. Site-specific identification of the SNO protein is critical for understanding the underlying molecular mechanisms of protein function regulation. Although careful verification is needed, SNO modification data containing numerous functional proteins are a potential research direction for druggable target identification and drug discovery. Undoubtedly, SNO-related research is meaningful not only for the development of NO donor drugs but also for classic target-based drug design. Herein, we provide a comprehensive summary of SNO, including its origin and transport, identification, function, and potential contribution to drug discovery. Importantly, we propose new views to develop novel therapies based on potential protein SNO-sourced targets.
Collapse
Affiliation(s)
- Hui Ye
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Jianbing Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Zhuangzhuang Liang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, P.R. China
| |
Collapse
|
11
|
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
|
12
|
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]
|
13
|
Deng Y, Wang Y, Jia F, Liu W, Zhou D, Jin Q, Ji J. Tailoring Supramolecular Prodrug Nanoassemblies for Reactive Nitrogen Species-Potentiated Chemotherapy of Liver Cancer. ACS NANO 2021; 15:8663-8675. [PMID: 33929183 DOI: 10.1021/acsnano.1c00698] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The development of a controllable reactive nitrogen species (RNS) generation system for cancer treatment has remained elusive. Herein, a supramolecular prodrug nanoassemblies (SPNA) strategy that co-delivers a nitric oxide (NO) donor and a superoxide anion (O2•-) inducing chemotherapeutic agent was reported for RNS-potentiated chemotherapy. The mole ratio of platinum(IV) prodrug and NO donor could be precisely tailored in SPNAPt/NO. Platinum(II) and NO would be released intracellularly to produce a highly toxic RNS, peroxynitrite anion (ONOO-). The levels of glutathione reductase (GR) and xeroderma pigmentosum group A (XPA) were down-regulated by ONOO-, thus synergistically decreasing detoxification and blocking DNA damage repair of Pt-based chemotherapy. The RNS-potentiated efficacy of SPNAPt/NO was validated on subcutaneous hepatoma xenograft models and an orthotopic cisplatin-resistant hepatoma model. This co-delivery strategy of NO donor and O2•- inducing chemotherapeutic agents for RNS-mediated therapy provides an insightful direction for cancer treatment.
Collapse
Affiliation(s)
- Yongyan Deng
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Yupeng Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Fan Jia
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Weifeng Liu
- Department of Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310016, People's Republic of China
| | - Dongfang Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, People's Republic of China
| |
Collapse
|
14
|
A systematic review of post-translational modifications in the mitochondrial permeability transition pore complex associated with cardiac diseases. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165992. [PMID: 33091565 DOI: 10.1016/j.bbadis.2020.165992] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022]
Abstract
The mitochondrial permeability transition pore (mPTP) opening is involved in the pathophysiology of multiple cardiac diseases, such as ischemia/reperfusion injury and heart failure. A growing number of evidence provided by proteomic screening techniques has demonstrated the role of post-translational modifications (PTMs) in several key components of the pore in response to changes in the extra/intracellular environment and bioenergetic demand. This could lead to a fine, complex regulatory mechanism that, under pathological conditions, can shift the state of mitochondrial functions and, thus, the cell's fate. Understanding the complex relationship between these PTMs is still under investigation and can provide new, promising therapeutic targets and treatment approaches. This review, using a systematic review of the literature, presents the current knowledge on PTMs of the mPTP and their role in health and cardiac disease.
Collapse
|
15
|
Walsh BJC, Giedroc DP. H 2S and reactive sulfur signaling at the host-bacterial pathogen interface. J Biol Chem 2020; 295:13150-13168. [PMID: 32699012 PMCID: PMC7504917 DOI: 10.1074/jbc.rev120.011304] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial pathogens that cause invasive disease in the vertebrate host must adapt to host efforts to cripple their viability. Major host insults are reactive oxygen and reactive nitrogen species as well as cellular stress induced by antibiotics. Hydrogen sulfide (H2S) is emerging as an important player in cytoprotection against these stressors, which may well be attributed to downstream more oxidized sulfur species termed reactive sulfur species (RSS). In this review, we summarize recent work that suggests that H2S/RSS impacts bacterial survival in infected cells and animals. We discuss the mechanisms of biogenesis and clearance of RSS in the context of a bacterial H2S/RSS homeostasis model and the bacterial transcriptional regulatory proteins that act as "sensors" of cellular RSS that maintain H2S/RSS homeostasis. In addition, we cover fluorescence imaging- and MS-based approaches used to detect and quantify RSS in bacterial cells. Last, we discuss proteome persulfidation (S-sulfuration) as a potential mediator of H2S/RSS signaling in bacteria in the context of the writer-reader-eraser paradigm, and progress toward ascribing regulatory significance to this widespread post-translational modification.
Collapse
Affiliation(s)
- Brenna J C Walsh
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA.
| |
Collapse
|
16
|
Coazzoli M, Napoli A, Roux-Biejat P, De Palma C, Moscheni C, Catalani E, Zecchini S, Conte V, Giovarelli M, Caccia S, Procacci P, Cervia D, Clementi E, Perrotta C. Acid Sphingomyelinase Downregulation Enhances Mitochondrial Fusion and Promotes Oxidative Metabolism in a Mouse Model of Melanoma. Cells 2020; 9:cells9040848. [PMID: 32244541 PMCID: PMC7226741 DOI: 10.3390/cells9040848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/20/2020] [Accepted: 03/28/2020] [Indexed: 02/07/2023] Open
Abstract
Melanoma is the most severe type of skin cancer. Its unique and heterogeneous metabolism, relying on both glycolysis and oxidative phosphorylation, allows it to adapt to disparate conditions. Mitochondrial function is strictly interconnected with mitochondrial dynamics and both are fundamental in tumour progression and metastasis. The malignant phenotype of melanoma is also regulated by the expression levels of the enzyme acid sphingomyelinase (A-SMase). By modulating at transcriptional level A-SMase in the melanoma cell line B16-F1 cells, we assessed the effect of enzyme downregulation on mitochondrial dynamics and function. Our results demonstrate that A-SMase influences mitochondrial morphology by affecting the expression of mitofusin 1 and OPA1. The enhanced expression of the two mitochondrial fusion proteins, observed when A-SMase is expressed at low levels, correlates with the increase of mitochondrial function via the stimulation of the genes PGC-1alpha and TFAM, two genes that preside over mitochondrial biogenesis. Thus, the reduction of A-SMase expression, observed in malignant melanomas, may determine their metastatic behaviour through the stimulation of mitochondrial fusion, activity and biogenesis, conferring a metabolic advantage to melanoma cells.
Collapse
Affiliation(s)
- Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Alessandra Napoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
- Unit of Clinical Pharmacology, University Hospital “Luigi Sacco”-ASST Fatebenefratelli Sacco, 20157 Milano, Italy
| | - Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milano, Italy;
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Vincenzo Conte
- Department of Biomedical Sciences for Health (SCIBIS), Università degli Studi di Milano, 20133 Milano, Italy; (V.C.); (P.P.)
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
| | - Patrizia Procacci
- Department of Biomedical Sciences for Health (SCIBIS), Università degli Studi di Milano, 20133 Milano, Italy; (V.C.); (P.P.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy
- Correspondence: (E.C.); (C.P.)
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (M.C.); (A.N.); (P.R.-B.); (C.M.); (S.Z.); (M.G.); (S.C.)
- Correspondence: (E.C.); (C.P.)
| |
Collapse
|
17
|
Yu HH, Lin Y, Zeng R, Li X, Zhang T, Tasneem S, Chen C, Qiu YX, Li B, Liao J, Wang YH, Cai X, Wang W. Analgesic and anti-inflammatory effects and molecular mechanisms of Kadsura heteroclita stems, an anti-arthritic Chinese Tujia ethnomedicinal herb. JOURNAL OF ETHNOPHARMACOLOGY 2019; 238:111902. [PMID: 31018145 DOI: 10.1016/j.jep.2019.111902] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/18/2019] [Accepted: 04/19/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by failure of spontaneous resolution of inflammation. The stem of Kadsura heteroclite (KHS) is a well-known anti-arthritic Tujia ethnomedicinal plant, which named Xuetong in folk, has long been used for the prevention and treatment of rheumatic and arthritic diseases. AIM OF THE STUDY The analgesic and anti-inflammatory effects and the potential mechanisms behind such effects of KHS would be investigated by using different animal models. MATERIALS AND METHODS The abdominal writhing episodes of mice induced by intraperitoneal injection of acetic acid and the tail-flick response induced by radiant heat stimulation were used to evaluate the analgesic effect of KHS. The number of abdominal writhing episodes of mice and the latency of tail-flick in rats were measured and recorded. In acute inflammatory models, the ear edema of mice was induced by applying xylene on the ear surface, while the paw edema of male and female rats was induced by subcutaneous injection of carrageenan into the right hind paws of animals. The carrageenan-induced paw swelling in rats were selected as an anti-acute inflammatory mechanism of KHS. Serum levels of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor (TNF-α) were measured by ELISA, and protein expression of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) were detected by Western blot. RESULTS The maximal tolerated single dose of KHS was determined to be 26 g/kg in both sexes of mice. Pharmacological studies showed that KHS at the dose of 200 mg/kg significantly prolonged the reaction time of rats to radiant heat stimulation and suppressed abdominal writhing episodes of mice induced by intraperitoneal injection of acetic acid. KHS at the dose of 200, 400, and 800 mg/kg, showed dose-dependent inhibition of xylene-induced ear swelling in mice. KHS at the dose of 100, 200, 400, and 800 mg/kg demonstrated dose- and time-dependent suppression of paw edema induced by subcutaneous injection of carrageenan in both all rats. Mechanistic studies revealed that the anti-inflammatory effect of KHS was associated with inhibition of the production of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α and effectively decreased the expression of COX and iNOS proteins in the carrageenan-injected rat serum, paw tissues and inflammatory exudates. The positive reference drug, rotundine at a dosage of 100 mg/kg and indomethacin at a dosage of 10 mg/kg were used in both mice and rat models. CONCLUSION These results suggested that KHS has significant effects on analgesia and anti-inflammation with decreasing the pro-inflammation cytokines of IL-1β, IL-6, and TNF-α and inhibiting the proteins expression of COX-2 and iNOS.
Collapse
Affiliation(s)
- Huang-He Yu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China; Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Rong Zeng
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Xin Li
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Ting Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Shumaila Tasneem
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Cong Chen
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Yi-Xing Qiu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Jin Liao
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Yu-Hong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China.
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, PR China.
| |
Collapse
|
18
|
Maiuthed A, Bhummaphan N, Luanpitpong S, Mutirangura A, Aporntewan C, Meeprasert A, Rungrotmongkol T, Rojanasakul Y, Chanvorachote P. Nitric oxide promotes cancer cell dedifferentiation by disrupting an Oct4:caveolin-1 complex: A new regulatory mechanism for cancer stem cell formation. J Biol Chem 2018; 293:13534-13552. [PMID: 29986880 DOI: 10.1074/jbc.ra117.000287] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 06/19/2018] [Indexed: 01/11/2023] Open
Abstract
Cancer stem cells (CSCs) are unique populations of cells that can self-renew and generate different cancer cell lineages. Although CSCs are believed to be a promising target for novel therapies, the specific mechanisms by which these putative therapeutics could intervene are less clear. Nitric oxide (NO) is a biological mediator frequently up-regulated in tumors and has been linked to cancer aggressiveness. Here, we search for targets of NO that could explain its activity. We find that it directly affects the stability and function of octamer-binding transcription factor 4 (Oct4), known to drive the stemness of lung cancer cells. We demonstrated that NO promotes the CSC-regulatory activity of Oct4 through a mechanism that involves complex formation between Oct4 and the scaffolding protein caveolin-1 (Cav-1). In the absence of NO, Oct4 forms a molecular complex with Cav-1, which promotes the ubiquitin-mediated proteasomal degradation of Oct4. NO promotes Akt-dependent phosphorylation of Cav-1 at tyrosine 14, disrupting the Cav-1:Oct4 complex. Site-directed mutagenesis and computational modeling studies revealed that the hydroxyl moiety at tyrosine 14 of Cav-1 is crucial for its interaction with Oct4. Both removal of the hydroxyl via mutation to phenylalanine and phosphorylation lead to an increase in binding free energy (ΔGbind) between Oct4 and Cav-1, destabilizing the complex. Together, these results unveiled a novel mechanism of CSC regulation through NO-mediated stabilization of Oct4, a key stem cell transcription factor, and point to new opportunities to design CSC-related therapeutics.
Collapse
Affiliation(s)
- Arnatchai Maiuthed
- From the Department of Pharmacology and Physiology.,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences
| | - Narumol Bhummaphan
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences.,the Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sudjit Luanpitpong
- the Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700 Thailand, and
| | - Apiwat Mutirangura
- the Center of Excellence in Molecular Genetics of Cancer and Human Diseases, Department of Anatomy, Faculty of Medicine, and
| | | | - Arthitaya Meeprasert
- Structural and Computational Biology Research Group, and Department of Biochemistry, Faculty of Science
| | - Thanyada Rungrotmongkol
- Structural and Computational Biology Research Group, and Department of Biochemistry, Faculty of Science.,Ph.D. Program in Bioinformatics and Computational Biology
| | - Yon Rojanasakul
- WVU Cancer Institute, West Virginia University, Morgantown, West Virginia 26506
| | - Pithi Chanvorachote
- From the Department of Pharmacology and Physiology, .,Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences
| |
Collapse
|
19
|
Pineda E, Perdomo D. Entamoeba histolytica under Oxidative Stress: What Countermeasure Mechanisms Are in Place? Cells 2017; 6:cells6040044. [PMID: 29160807 PMCID: PMC5755502 DOI: 10.3390/cells6040044] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023] Open
Abstract
Entamoeba histolytica is the causative agent of human amoebiasis; it affects 50 million people worldwide and causes approximately 100,000 deaths per year. Entamoeba histolytica is an anaerobic parasite that is primarily found in the colon; however, for unknown reasons, it can become invasive, breaching the gut barrier and migrating toward the liver causing amoebic liver abscesses. During the invasive process, it must maintain intracellular hypoxia within the oxygenated human tissues and cellular homeostasis during the host immune defense attack when it is confronted with nitric oxide and reactive oxygen species. But how? This review will address the described and potential mechanisms available to counter the oxidative stress generated during invasion and the possible role that E. histolytica’s continuous endoplasmic reticulum (Eh-ER) plays during these events.
Collapse
Affiliation(s)
- Erika Pineda
- Laboratory of Fundamental Microbiology and Pathogenicity (MFP), University of Bordeaux, CNRS UMR-5234, 33000 Bordeaux, France.
| | - Doranda Perdomo
- Laboratory of Fundamental Microbiology and Pathogenicity (MFP), University of Bordeaux, CNRS UMR-5234, 33000 Bordeaux, France.
| |
Collapse
|
20
|
Lu Y, Dang S, Wang X, Zhang J, Zhang L, Su Q, Zhang H, Lin T, Zhang X, Zhang Y, Sun H, Zhu Z, Li H. NO involvement in the inhibition of ghrelin on voltage-dependent potassium currents in rat hippocampal cells. Brain Res 2017; 1678:40-46. [PMID: 28987626 DOI: 10.1016/j.brainres.2017.09.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/30/2023]
Abstract
Ghrelin is a peptide hormone that plays an important role in promoting appetite, regulating distribution and rate of use of energy, cognition, and mood disorders, but the relevant neural mechanisms of these function are still not clear. In this study, we examined the effect of ghrelin on voltage-dependent potassium (K+) currents in hippocampal cells of 1-3 days SD rats by whole-cell patch-clamp technique, and discussed whether NO was involved in this process. The results showed that ghrelin significantly inhibited the voltage-dependent K+ currents in hippocampal cells, and the inhibitory effect was more significant when l-arginine was co-administered. In contrast, N-nitro- l-arginine methyl ester increased the ghrelin inhibited K+ currents and attenuated the inhibitory effect of ghrelin. While d-arginine (D-AA) showed no significant impact on the ghrelin-induced decrease in K+ current. These results show that ghrelin may play a physiological role by inhibiting hippocampal voltage dependent K+ currents, and the NO pathway may be involved in this process.
Collapse
Affiliation(s)
- Yong Lu
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, China; Center Laboratory, Heze Medical College, Shandong 274000, China
| | - Shaokang Dang
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, China
| | - Xu Wang
- Center Laboratory, Heze Medical College, Shandong 274000, China
| | - Junli Zhang
- Shanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Shanxi 710069, China
| | - Lin Zhang
- Shanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Shanxi 710069, China
| | - Qian Su
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, China
| | - Huiping Zhang
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, China
| | - Tianwei Lin
- Shanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Shanxi 710069, China
| | - Xiaoxiao Zhang
- Shanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Shanxi 710069, China
| | - Yurong Zhang
- Department of Pharmacy, the First Affiliated Hospital of Xi'an Medical College, Shanxi 710077, China
| | - Hongli Sun
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, China
| | - Zhongliang Zhu
- Shanxi Province Biomedicine Key Laboratory, College of Life Sciences, Northwest University, Shanxi 710069, China.
| | - Hui Li
- Department of Neonatology, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, China.
| |
Collapse
|
21
|
Stephan M, Edelmann B, Winoto-Morbach S, Janssen O, Bertsch U, Perrotta C, Schütze S, Fritsch J. Role of caspases in CD95-induced biphasic activation of acid sphingomyelinase. Oncotarget 2017; 8:20067-20085. [PMID: 28223543 PMCID: PMC5386744 DOI: 10.18632/oncotarget.15379] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/24/2017] [Indexed: 12/04/2022] Open
Abstract
Acid sphingomyelinase (A-SMase) plays an important role in the initiation of CD95 signaling by forming ceramide-enriched membrane domains that enable clustering and activation of the death receptors. In TNF-R1 and TRAIL-R1/R2 signaling, A-SMase also contributes to the lysosomal apoptosis pathway triggered by receptor internalization. Here, we investigated the molecular mechanism of CD95-mediated A-SMase activation, demonstrating that A-SMase is located in internalized CD95-receptosomes and is activated by the CD95/CD95L complex in a biphasic manner.Since several caspases have been described to be involved in the activation of A-SMase, we evaluated expression levels of caspase-8, caspase-7 and caspase-3 in CD95-receptosomes. The occurrence of cleaved caspase-8 correlated with the first peak of A-SMase activity and translocation of the A-SMase to the cell surface which could be blocked by the caspase-8 inhibitor IETD.Inhibition of CD95-internalization selectively reduced the second phase of A-SMase activity, suggesting a fusion between internalized CD95-receptosomes and an intracellular vesicular pool of A-SMase. Further analysis demonstrated that caspase-7 activity correlates with the second phase of the A-SMase activity, whereas active caspase-3 is present at early and late internalization time points. Blocking caspases-7/ -3 by DEVD reduced the second phase of A-SMase activation in CD95-receptosomes suggesting the potential role of caspase-7 or -3 for late A-SMase activation.In summary, we describe a biphasic A-SMase activation in CD95-receptosomes indicating (I.) a caspase-8 dependent translocation of A-SMase to plasma membrane and (II.) a caspase-7 and/or -3 dependent fusion of internalized CD95-receptosomes with intracellular A-SMase-containing vesicles.
Collapse
Affiliation(s)
- Mario Stephan
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Bärbel Edelmann
- Department of Hematology and Oncology, University Hospital Magdeburg, Magdeburg, Germany
| | | | - Ottmar Janssen
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Uwe Bertsch
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Milano, Italy
| | - Stefan Schütze
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Jürgen Fritsch
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
22
|
Stram AR, Payne RM. Post-translational modifications in mitochondria: protein signaling in the powerhouse. Cell Mol Life Sci 2016; 73:4063-73. [PMID: 27233499 PMCID: PMC5045789 DOI: 10.1007/s00018-016-2280-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 02/03/2023]
Abstract
There is an intimate interplay between cellular metabolism and the pathophysiology of disease. Mitochondria are essential to maintaining and regulating metabolic function of cells and organs. Mitochondrial dysfunction is implicated in diverse diseases, such as cardiovascular disease, diabetes and metabolic syndrome, neurodegeneration, cancer, and aging. Multiple reversible post-translational protein modifications are located in the mitochondria that are responsive to nutrient availability and redox conditions, and which can act in protein-protein interactions to modify diverse mitochondrial functions. Included in this are physiologic redox signaling via reactive oxygen and nitrogen species, phosphorylation, O-GlcNAcylation, acetylation, and succinylation, among others. With the advent of mass proteomic screening techniques, there has been a vast increase in the array of known mitochondrial post-translational modifications and their protein targets. The functional significance of these processes in disease etiology, and the pathologic response to their disruption, are still under investigation. However, many of these reversible modifications act as regulatory mechanisms in mitochondria and show promise for mitochondrial-targeted therapeutic strategies. This review addresses the current knowledge of post-translational processing and signaling mechanisms in mitochondria, and their implications in health and disease.
Collapse
Affiliation(s)
- Amanda R Stram
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., Room R4-302b, Indianapolis, IN, 46202, USA
| | - R Mark Payne
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., Room R4-302b, Indianapolis, IN, 46202, USA.
| |
Collapse
|
23
|
Hussain A, Mun BG, Imran QM, Lee SU, Adamu TA, Shahid M, Kim KM, Yun BW. Nitric Oxide Mediated Transcriptome Profiling Reveals Activation of Multiple Regulatory Pathways in Arabidopsis thaliana. FRONTIERS IN PLANT SCIENCE 2016; 7:975. [PMID: 27446194 PMCID: PMC4926318 DOI: 10.3389/fpls.2016.00975] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/20/2016] [Indexed: 05/18/2023]
Abstract
Imbalance between the accumulation and removal of nitric oxide and its derivatives is a challenge faced by all plants at the cellular level, and is especially important under stress conditions. Exposure of plants to various biotic and abiotic stresses causes rapid changes in cellular redox tone potentiated by the rise in reactive nitrogen species that serve as signaling molecules in mediating defensive responses. To understand mechanisms mediated by these signaling molecules, we performed a large-scale analysis of the Arabidopsis transcriptome induced by nitrosative stress. We generated an average of 84 and 91 million reads from three replicates each of control and 1 mM S-nitrosocysteine (CysNO)-infiltrated Arabidopsis leaf samples, respectively. After alignment, more than 95% of all reads successfully mapped to the reference and 32,535 genes and 55,682 transcripts were obtained. CysNO infiltration caused differential expression of 6436 genes (3448 up-regulated and 2988 down-regulated) and 6214 transcripts (3335 up-regulated and 2879 down-regulated) 6 h post-infiltration. These differentially expressed genes were found to be involved in key physiological processes, including plant defense against various biotic and abiotic stresses, hormone signaling, and other developmental processes. After quantile normalization of the FPKM values followed by student's T-test (P < 0.05) we identified 1165 DEGs (463 up-regulated and 702 down-regulated) with at least 2-folds change in expression after CysNO treatment. Expression patterns of selected genes involved in various biological pathways were verified using quantitative real-time PCR. This study provides comprehensive information about plant responses to nitrosative stress at transcript level and would prove helpful in understanding and incorporating mechanisms associated with nitrosative stress responses in plants.
Collapse
Affiliation(s)
- Adil Hussain
- Department of Agriculture, Abdul Wali Khan University MardanMardan, Pakistan
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Bong-Gyu Mun
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Qari M. Imran
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Sang-Uk Lee
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Teferi A. Adamu
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Muhammad Shahid
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Kyung-Min Kim
- Laboratory of Plant Molecular Breeding, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| | - Byung-Wook Yun
- Laboratory of Plant Functional Genomics, School of Applied Biosciences, Kyungpook National UniversityDaegu, South Korea
| |
Collapse
|
24
|
Nie X, Lowe DW, Rollins LG, Bentzley J, Fraser JL, Martin R, Singh I, Jenkins D. Sex-specific effects of N-acetylcysteine in neonatal rats treated with hypothermia after severe hypoxia-ischemia. Neurosci Res 2016; 108:24-33. [PMID: 26851769 DOI: 10.1016/j.neures.2016.01.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 12/16/2015] [Accepted: 01/24/2016] [Indexed: 12/29/2022]
Abstract
Approximately half of moderate to severely hypoxic-ischemic (HI) newborns do not respond to hypothermia, the only proven neuroprotective treatment. N-acetylcysteine (NAC), an antioxidant and glutathione precursor, shows promise for neuroprotection in combination with hypothermia, mitigating post-HI neuroinflammation due to oxidative stress. As mechanisms of HI injury and cell death differ in males and females, sex differences must be considered in translational research of neuroprotection. We assessed the potential toxicity and efficacy of NAC in combination with hypothermia, in male and female neonatal rats after severe HI injury. NAC 50mg/kg/d administered 1h after initiation of hypothermia significantly decreased iNOS expression and caspase 3 activation in the injured hemisphere versus hypothermia alone. However, only females treated with hypothermia +NAC 50mg/kg showed improvement in short-term infarct volumes compared with saline treated animals. Hypothermia alone had no effect in this severe model. When NAC was continued for 6 weeks, significant improvement in long-term neuromotor outcomes over hypothermia treatment alone was observed, controlling for sex. Antioxidants may provide insufficient neuroprotection after HI for neonatal males in the short term, while long-term therapy may benefit both sexes.
Collapse
Affiliation(s)
- Xingju Nie
- Center for Biomedical Imaging, Medical University of South Carolina, 165 Ashley Ave, Charleston, SC 29425, United States.
| | - Danielle W Lowe
- Department of Pediatrics, Medical University of South Carolina, 165 Ashley Ave, Charleston, SC 29425, United States.
| | - Laura Grace Rollins
- Department of Psychology, University of Massachusetts, 100 Morrissey Blvd, Boston, MA 02125, United States.
| | - Jessica Bentzley
- Department of Pediatrics, Medical University of South Carolina, 165 Ashley Ave, Charleston, SC 29425, United States.
| | - Jamie L Fraser
- Medical Genetics Training Program, National Human Genome Research Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-2152, United States.
| | - Renee Martin
- Department of Biostatistics and Epidemiology, Medical University of South Carolina, 165 Ashley Ave, Charleston, SC 29425, United States.
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, 165 Ashley Ave, Charleston, SC 29425, United States.
| | - Dorothea Jenkins
- Department of Pediatrics, Medical University of South Carolina, 165 Ashley Ave, Charleston, SC 29425, United States.
| |
Collapse
|
25
|
Hollow polymer nanoparticles with S-nitrosothiols as scaffolds for nitric oxide release. J Colloid Interface Sci 2015; 459:115-122. [DOI: 10.1016/j.jcis.2015.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 12/31/2022]
|
26
|
Gulbins E, Walter S, Becker KA, Halmer R, Liu Y, Reichel M, Edwards MJ, Müller CP, Fassbender K, Kornhuber J. A central role for the acid sphingomyelinase/ceramide system in neurogenesis and major depression. J Neurochem 2015; 134:183-92. [DOI: 10.1111/jnc.13145] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/15/2015] [Accepted: 04/09/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Erich Gulbins
- Department of Surgery; University of Cincinnati; Cincinnati Ohio USA
- Department of Molecular Biology; University of Duisburg-Essen; Essen Germany
| | - Silke Walter
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Katrin Anne Becker
- Department of Molecular Biology; University of Duisburg-Essen; Essen Germany
| | - Ramona Halmer
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Yang Liu
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy; Friedrich-Alexander-University of Erlangen; Erlangen Germany
| | | | - Christian P. Müller
- Department of Psychiatry and Psychotherapy; Friedrich-Alexander-University of Erlangen; Erlangen Germany
| | - Klaus Fassbender
- Department of Neurology; University Hospital of the Saarland; Homburg/Saar Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy; Friedrich-Alexander-University of Erlangen; Erlangen Germany
| |
Collapse
|
27
|
Managò A, Becker KA, Carpinteiro A, Wilker B, Soddemann M, Seitz AP, Edwards MJ, Grassmé H, Szabò I, Gulbins E. Pseudomonas aeruginosa pyocyanin induces neutrophil death via mitochondrial reactive oxygen species and mitochondrial acid sphingomyelinase. Antioxid Redox Signal 2015; 22:1097-110. [PMID: 25686490 PMCID: PMC4403017 DOI: 10.1089/ars.2014.5979] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Pulmonary infections with Pseudomonas aeruginosa are a serious clinical problem and are often lethal. Because many strains of P. aeruginosa are resistant to antibiotics, therapeutic options are limited. Neutrophils play an important role in the host's early acute defense against pulmonary P. aeruginosa. Therefore, it is important to define the mechanisms by which P. aeruginosa interacts with host cells, particularly neutrophils. RESULTS Here, we report that pyocyanin, a membrane-permeable pigment and toxin released by P. aeruginosa, induces the death of wild-type neutrophils; its interaction with the mitochondrial respiratory chain results in the release of reactive oxygen species (ROS), the activation of mitochondrial acid sphingomyelinase, the formation of mitochondrial ceramide, and the release of cytochrome c from mitochondria. A genetic deficiency in acid sphingomyelinase prevents both the activation of this pathway and pyocyanin-induced neutrophil death. This reduced death, on the other hand, is associated with an increase in the release of interleukin-8 from pyocyanin-activated acid sphingomyelinase-deficient neutrophils but not from wild-type cells. INNOVATION These studies identified the mechanisms by which pyocyanin induces the release of mitochondrial ROS and by which ROS induce neutrophil death via mitochondrial acid sphingomyelinase. CONCLUSION These findings demonstrate a novel mechanism of pyocyanin-induced death of neutrophils and show how this apoptosis balances innate immune reactions.
Collapse
|
28
|
Iyer AKV, Rojanasakul Y, Azad N. Nitrosothiol signaling and protein nitrosation in cell death. Nitric Oxide 2014; 42:9-18. [PMID: 25064181 DOI: 10.1016/j.niox.2014.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 06/01/2014] [Accepted: 07/10/2014] [Indexed: 10/25/2022]
Abstract
Nitric oxide, a reactive free radical, is an important signaling molecule that can lead to a plethora of cellular effects affecting homeostasis. A well-established mechanism by which NO manifests its effect on cellular functions is the post-translational chemical modification of cysteine thiols in substrate proteins by a process known as S-nitrosation. Studies that investigate regulation of cellular functions through NO have increasingly established S-nitrosation as the primary modulatory mechanism in their respective systems. There has been a substantial increase in the number of reports citing various candidate proteins undergoing S-nitrosation, which affects cell-death and -survival pathways in a number of tissues including heart, lung, brain and blood. With an exponentially growing list of proteins being identified as substrates for S-nitrosation, it is important to assimilate this information in different cell/tissue systems in order to gain an overall view of protein regulation of both individual proteins and a class of protein substrates. This will allow for broad mapping of proteins as a function of S-nitrosation, and help delineate their global effects on pathophysiological responses including cell death and survival. This information will not only provide a much better understanding of overall functional relevance of NO in the context of various disease states, it will also facilitate the generation of novel therapeutics to combat specific diseases that are driven by NO-mediated S-nitrosation.
Collapse
Affiliation(s)
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV 26505, USA
| | - Neelam Azad
- Department of Pharmaceutical Sciences, Hampton University, Hampton, VA 23668, USA
| |
Collapse
|
29
|
Preclinical therapeutic potential of a nitrosylating agent in the treatment of ovarian cancer. PLoS One 2014; 9:e97897. [PMID: 24887420 PMCID: PMC4041717 DOI: 10.1371/journal.pone.0097897] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/24/2014] [Indexed: 12/27/2022] Open
Abstract
This study examines the role of s-nitrosylation in the growth of ovarian cancer using cell culture based and in vivo approaches. Using the nitrosylating agent, S-nitrosoglutathione (GSNO), a physiological nitric oxide molecule, we show that GSNO treatment inhibited proliferation of chemoresponsive and chemoresistant ovarian cancer cell lines (A2780, C200, SKVO3, ID8, OVCAR3, OVCAR4, OVCAR5, OVCAR7, OVCAR8, OVCAR10, PE01 and PE04) in a dose dependent manner. GSNO treatment abrogated growth factor (HB-EGF) induced signal transduction including phosphorylation of Akt, p42/44 and STAT3, which are known to play critical roles in ovarian cancer growth and progression. To examine the therapeutic potential of GSNO in vivo, nude mice bearing intra-peritoneal xenografts of human A2780 ovarian carcinoma cell line (2×106) were orally administered GSNO at the dose of 1 mg/kg body weight. Daily oral administration of GSNO significantly attenuated tumor mass (p<0.001) in the peritoneal cavity compared to vehicle (phosphate buffered saline) treated group at 4 weeks. GSNO also potentiated cisplatin mediated tumor toxicity in an A2780 ovarian carcinoma nude mouse model. GSNO’s nitrosylating ability was reflected in the induced nitrosylation of various known proteins including NFκB p65, Akt and EGFR. As a novel finding, we observed that GSNO also induced nitrosylation with inverse relationship at tyrosine 705 phosphorylation of STAT3, an established player in chemoresistance and cell proliferation in ovarian cancer and in cancer in general. Overall, our study underlines the significance of S-nitrosylation of key cancer promoting proteins in modulating ovarian cancer and proposes the therapeutic potential of nitrosylating agents (like GSNO) for the treatment of ovarian cancer alone or in combination with chemotherapeutic drugs.
Collapse
|
30
|
Hertz R, Ben Lulu S, Shahi P, Trebicz-Geffen M, Benhar M, Ankri S. Proteomic identification of S-nitrosylated proteins in the parasite Entamoeba histolytica by resin-assisted capture: insights into the regulation of the Gal/GalNAc lectin by nitric oxide. PLoS One 2014; 9:e91518. [PMID: 24626316 PMCID: PMC3953491 DOI: 10.1371/journal.pone.0091518] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/12/2014] [Indexed: 11/19/2022] Open
Abstract
Entamoeba histolytica is a gastrointestinal protozoan parasite that causes amebiasis, a disease which has a worldwide distribution with substantial morbidity and mortality. Nitrosative stress, which is generated by innate immune cells, is one of the various environmental challenges that E. histolytica encounters during its life cycle. Although the effects of nitric oxide (NO) on the regulation of gene expression in this parasite have been previously investigated, our knowledge on S-nitrosylated proteins in E.histolytica is lacking. In order to fill this knowledge gap, we performed a large-scale detection of S-nitrosylated (SNO) proteins in E.histolytica trophozoites that were treated with the NO donor, S-nitrosocysteine by resin-assisted capture (RAC). We found that proteins involved in glycolysis, gluconeogenesis, translation, protein transport, and adherence to target cells such as the heavy subunit of Gal/GalNac lectin are among the S-nitrosylated proteins that were enriched by SNO-RAC. We also found that the S-nitrosylated cysteine residues in the carbohydrate recognition domain (CRD) of Gal/GalNAc lectin impairs its function and contributes to the inhibition of E.histolytica adherence to host cells. Collectively, these results advance our understanding of the mechanism of reduced E.histolytica adherence to mammalian cells by NO and emphasize the importance of NO as a regulator of key physiological functions in E.histolytica.
Collapse
Affiliation(s)
- Rivka Hertz
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Shani Ben Lulu
- Department of Biochemistry, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Preeti Shahi
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Meirav Trebicz-Geffen
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Moran Benhar
- Department of Biochemistry, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Serge Ankri
- Department of Molecular Microbiology, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
- * E-mail:
| |
Collapse
|
31
|
Tanaka K, Shimizu T, Higashi Y, Nakamura K, Taniuchi K, Dimitriadis F, Shimizu S, Yokotani K, Saito M. Central bombesin possibly induces S-nitrosylation of cyclooxygenase-1 in pre-sympathetic neurons of rat hypothalamic paraventricular nucleus. Life Sci 2014; 100:85-96. [PMID: 24530741 DOI: 10.1016/j.lfs.2014.01.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/16/2014] [Accepted: 01/30/2014] [Indexed: 01/22/2023]
Abstract
AIMS Cyclooxygenase (COX) can be activated by nitric oxide-induced (NO-induced) conversion of cysteine thiol group of COX into S-nitrosothiol. We previously reported the involvement of brain COX/NO synthase (NOS) in centrally administered bombesin-, a stress-related neuropeptide, induced secretion of rat adrenal noradrenaline and adrenaline. To examine a possible involvement of the NO-induced modification of COX in bombesin-induced response, we investigated whether bombesin induces close proximity of COX-1 and neuronal NOS (nNOS) or S-nitroso-cysteine in pre-sympathetic spinally projecting neurons in the rat hypothalamic paraventricular nucleus (PVN), a regulatory center of adrenomedullary outflow. MAIN METHODS In twelve-week-old male Wistar rats, pre-sympathetic spinally projecting neurons in the PVN were labeled with a retrograde tracer Fluoro-Gold (FG). After intracerebroventricular administration of bombesin, we performed double immunohistochemical analysis for Fos and COX-1 or nNOS in FG-labeled PVN neurons. We also performed a fluorescent in situ proximity ligation assay (PLA) for visualizing of close proximity (<40 nm) of COX-1 with nNOS or S-nitroso-cysteine. KEY FINDINGS Bombesin significantly increased the number of Fos-immunoreactive cells in FG-labeled PVN neurons with COX-1 or nNOS immunoreactivity. 7-Nitroindazole, a selective nNOS inhibitor, abolished Fos-immunoreactivity induced by bombesin in COX-1-immunoreactive FG-labeled PVN neurons. Bombesin also induced PLA-positive signals indicating close proximity of COX-1/nNOS and COX-1/S-nitroso-cysteine in FG-labeled PVN neurons. SIGNIFICANCE Centrally administered bombesin possibly induces S-nitrosylation of COX-1 through close proximity of COX-1 and nNOS in pre-sympathetic spinally projecting PVN neurons, thereby activating COX-1 during the bombesin-induced activation of central adrenomedullary outflow in the rat.
Collapse
Affiliation(s)
- Kenjiro Tanaka
- Department of Pharmacology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan.
| | - Youichirou Higashi
- Department of Neurosurgery, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Kumiko Nakamura
- Department of Pharmacology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Keisuke Taniuchi
- Department of Pharmacology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Fotios Dimitriadis
- B' Urologic Department, Papageorgiou General Hospital, Aristotle University School of Medicine, Thessaloniki, Greece
| | - Shogo Shimizu
- Division of Molecular Pharmacology, Tottori University School of Medicine, Yonago, Tottori 683-8503, Japan
| | - Kunihiko Yokotani
- Department of Pharmacology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| | - Motoaki Saito
- Department of Pharmacology, Kochi University School of Medicine, Nankoku, Kochi 783-8505, Japan
| |
Collapse
|
32
|
Rochette S, Gagnon-Arsenault I, Diss G, Landry CR. Modulation of the yeast protein interactome in response to DNA damage. J Proteomics 2013; 100:25-36. [PMID: 24262151 DOI: 10.1016/j.jprot.2013.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 10/10/2013] [Accepted: 11/05/2013] [Indexed: 01/24/2023]
Abstract
UNLABELLED Cells deploy diverse mechanisms to physiologically adapt to potentially detrimental perturbations. These mechanisms include changes in the organization of protein-protein interaction networks (PINs). Most PINs characterized to date are portrayed in a single environmental condition and are thus likely to miss important connections among biological processes. In this report, we show that the yeast DHFR-PCA on high-density arrays allows to detects modulations of protein-protein interactions (PPIs) in different conditions by testing more than 1000 PPIs in standard and in a drug-inducing DNA damage conditions. We identify 156 PPIs that show significant modulation in response to DNA damage. We provide evidence that modulated PPIs involve essential genes (NOP7, EXO84 and LAS17) playing critical roles in response to DNA damage. Additionally, we show that a significant proportion of PPI changes are likely explained by changes in protein localization and, to a lesser extent, protein abundance. The protein interaction modules affected by changing PPIs support the role of mRNA stability and translation, protein degradation and ubiquitylation and the regulation of the actin cytoskeleton in response to DNA damage. Overall, we provide a valuable tool and dataset for the study of the rewiring of PINs in response to environmental perturbations. BIOLOGICAL SIGNIFICANCE We show that the DHFR-PCA is a high-throughput method that allows the detection of changes in PPIs associated with different environmental conditions using DNA damage response as a testbed. We provide a valuable resource for the study of DNA damage in eukaryotic cells. This article is part of a Special Issue: Can Proteomics Fill the Gap Between Genomics and Phenotypes?
Collapse
Affiliation(s)
- Samuel Rochette
- Département de Biologie, PROTEO and Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Québec G1V 0A6, Canada
| | - Isabelle Gagnon-Arsenault
- Département de Biologie, PROTEO and Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Québec G1V 0A6, Canada
| | - Guillaume Diss
- Département de Biologie, PROTEO and Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Québec G1V 0A6, Canada
| | - Christian R Landry
- Département de Biologie, PROTEO and Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec, Québec G1V 0A6, Canada.
| |
Collapse
|
33
|
Paulsen C, Carroll KS. Cysteine-mediated redox signaling: chemistry, biology, and tools for discovery. Chem Rev 2013; 113:4633-79. [PMID: 23514336 PMCID: PMC4303468 DOI: 10.1021/cr300163e] [Citation(s) in RCA: 873] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Indexed: 02/06/2023]
Affiliation(s)
- Candice
E. Paulsen
- Department of Chemistry, The Scripps Research
Institute, Jupiter, Florida, 33458, United States
| | - Kate S. Carroll
- Department of Chemistry, The Scripps Research
Institute, Jupiter, Florida, 33458, United States
| |
Collapse
|
34
|
Diss G, Filteau M, Freschi L, Leducq JB, Rochette S, Torres-Quiroz F, Landry CR. Integrative avenues for exploring the dynamics and evolution of protein interaction networks. Curr Opin Biotechnol 2013; 24:775-83. [PMID: 23571097 DOI: 10.1016/j.copbio.2013.02.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 02/14/2013] [Accepted: 02/24/2013] [Indexed: 01/09/2023]
Abstract
Over the past decade, the study of protein interaction networks (PINs) has shed light on the organizing principles of living cells. However, PINs have been mostly mapped in one single condition. We outline three of the most promising avenues of investigation in this field, namely the study of first, how PINs are rewired by mutations and environmental perturbations; secondly, how inter-species interactions affect PIN achitectures; thirdly, what mechanisms and forces drive PIN evolution. These investigations will unravel the dynamics and condition dependence of PINs and will thus lead to a better functional annotation of network architecture. One major challenge to reach these goals is the integration of PINs with other cellular regulatory networks in the context of complex cellular phenotypes.
Collapse
Affiliation(s)
- Guillaume Diss
- Département de Biologie, Institut de Biologie Intégrative et des Systèmes (IBIS), PROTEO, Université Laval, Québec, Canada G1V 0A6
| | | | | | | | | | | | | |
Collapse
|
35
|
Foster MW, Thompson JW, Forrester MT, Sha Y, McMahon TJ, Bowles DE, Moseley MA, Marshall HE. Proteomic analysis of the NOS2 interactome in human airway epithelial cells. Nitric Oxide 2013; 34:37-46. [PMID: 23438482 DOI: 10.1016/j.niox.2013.02.079] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 01/22/2023]
Abstract
The cytokine-inducible isoform of nitric oxide synthase (NOS2) is constitutively expressed in human respiratory epithelia and is upregulated in inflammatory lung disease. Here, we sought to better define the protein interactions that may be important for NOS2 activity and stability, as well as to identify potential targets of NOS2-derived NO, in the respiratory epithelium. We overexpressed Flag-tagged, catalytically-inactive NOS2 in A549 cells and used mass spectrometry to qualitatively identify NOS2 co-immunoprecipitating proteins. Stable isotope labeling of amino acids in cell culture (SILAC) was used to quantify the coordinate effects of cytokine stimulation on NOS2-protein interactions. Multi-protein networks dominated the NOS2 interactome, and cytokine-inducible interactions with allosteric activators and with the ubiquitin-proteasome system were correlated with cytokine-dependent increases in NO metabolites and in NOS2 ubiquitination. The ubiquitin ligase scaffolding protein, FBXO45, was identified as a novel, direct NOS2 interactor. Similar to the SPRY domain-containing SOCS box (SPSB) proteins, FBXO45 requires Asn27 in the (23)DINNN(27) motif of NOS2 for its interaction. However, FBXO45 is unique from the SPSBs in that it recruits a distinct E3 ligase complex containing MYCBP2 and SKP1. Collectively, these findings demonstrate the general utility of interaction proteomics for defining new aspects of NOS2 physiology.
Collapse
Affiliation(s)
- Matthew W Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Centers, Durham, NC 27710, United States.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Lee SB, Kim CK, Lee KH, Ahn JY. S-nitrosylation of B23/nucleophosmin by GAPDH protects cells from the SIAH1-GAPDH death cascade. ACTA ACUST UNITED AC 2013; 199:65-76. [PMID: 23027902 PMCID: PMC3461512 DOI: 10.1083/jcb.201205015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
S-nitrosylation of B23/nucleophosmin mediates neuroprotective effects by binding SIAH1, displacing GAPDH, and preventing SIAH1 E3 ligase activity. B23/nucleophosmin is a multifunctional protein that participates in cell survival signaling by shuttling between the nucleolus/nucleoplasm and nucleus/cytoplasm. In this paper, we report a novel neuroprotective function of B23 through regulation of the SIAH1–glyceraldehyde-3-phosphate dehydrogenase (GAPDH) death cascade. B23 physiologically bound to both SIAH1 and GAPDH, disrupting the SIAH1–GAPDH complex in the nucleus in response to nitrosative stress. S-nitrosylation of B23 at cysteine 275 by trans-nitrosylation from GAPDH dramatically reduced the interaction between SIAH1 and GAPDH. S-nitrosylation of B23 enhanced B23–SIAH1 binding and mediated the neuroprotective actions of B23 by abrogating the E3 ligase activity of SIAH1. In mice, overexpression of B23 notably inhibited N-methyl-d-aspartate–mediated neurotoxicity, whereas expression of the C275S mutant, which is defective in binding to SIAH1, did not prevent neurotoxicity. Thus, B23 regulates neuronal survival by preventing SIAH1–GAPDH death signaling under stress-induced conditions in the brain.
Collapse
Affiliation(s)
- Sang Bae Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, South Korea
| | | | | | | |
Collapse
|
37
|
Sun SH, Liu SQ, Cai CP, Cai R, Chen L, Zhang QB. Down-regulation of alpha-2u globulin in renal mitochondria of STZ-induced diabetic rats observed by a proteomic method. ANNALES D'ENDOCRINOLOGIE 2012; 73:530-41. [PMID: 23131471 DOI: 10.1016/j.ando.2012.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 06/12/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
AIM To identify the changes of mitochondrial protein expression in diabetic renal parenchyma and to characterize their molecular functions and biological processes in diabetes. METHODS Mitochondrial proteins extracted from renal parenchyma mitochondria of streptozotocin-induced diabetic rats and normal rats were separated by two-dimensional polyacrylamide gel electrophoresis and identified by matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry. RESULTS Eleven proteins from 533 visualized protein spots displayed significant different expressions in mitochondria of diabetic kidneys compared with those in normal ones. Among these altered proteins, two proteins with the most obvious changes in protein expression were identified as alpha-2u globulin (mature protein, named A2) and its proteolytically modified form (named A2-fragment) respectively. These proteins were found in mitochondria of male rat renal parenchyma and were proved to be down-regulated in diabetic rats simultaneously. CONCLUSION Our results suggest that down-regulation of alpha-2u globulin may be associated with an abnormal β-oxidation of long-chain fatty acids during diabetes. The decreased expression of A2-fragment in renal mitochondria of diabetic nephropathy may reduce fatty acid β-oxidation, which leads to a diminished energy supply from mitochondria to kidney tissue and the deposition of a large number of fatty acids in the kidney, ultimately causing and aggravating kidney damage. In conclusion, these findings may be helpful for understanding the molecular mechanism of diabetic nephropathy.
Collapse
Affiliation(s)
- Shi-He Sun
- Department of Anatomy, North Sichuan Medical College, Nangchong, 234, Fujiang Road, Nangchong, Sichuan, 637007, China
| | | | | | | | | | | |
Collapse
|
38
|
A Comparative Study on the Sensitive Detection of Hydroxyl Radical Using Thiol-capped CdTe and CdTe/ZnS Quantum Dots. J Fluoresc 2012; 22:1513-9. [DOI: 10.1007/s10895-012-1089-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 06/20/2012] [Indexed: 11/26/2022]
|
39
|
Smith BC, Fernhoff NB, Marletta MA. Mechanism and kinetics of inducible nitric oxide synthase auto-S-nitrosation and inactivation. Biochemistry 2012; 51:1028-40. [PMID: 22242685 DOI: 10.1021/bi201818c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nitric oxide (NO), the product of the nitric oxide synthase (NOS) reaction, was previously shown to result in S-nitrosation of the NOS Zn(2+)-tetrathiolate and inactivation of the enzyme. To probe the potential physiological significance of NOS S-nitrosation, we determined the inactivation time scale of the inducible NOS isoform (iNOS) and found it directly correlates with an increase in the level of iNOS S-nitrosation. A kinetic model of NOS inactivation in which arginine is treated as a suicide substrate was developed. In this model, NO synthesized at the heme cofactor is partitioned between release into solution (NO release pathway) and NOS S-nitrosation followed by NOS inactivation (inactivation pathway). Experimentally determined progress curves of NO formation were fit to the model. The NO release pathway was perturbed through addition of the NO traps oxymyoglobin (MbO(2)) and β2 H-NOX, which yielded partition ratios between NO release and inactivation of ~100 at 4 μM MbO(2) and ~22000 at saturating trap concentrations. The results suggest that a portion of the NO synthesized at the heme cofactor reacts with the Zn(2+)-tetrathiolate without being released into solution. Perturbation of the inactivation pathway through addition of the reducing agent GSH or TCEP resulted in a concentration-dependent decrease in the level of iNOS S-nitrosation that directly correlated with protection from iNOS inactivation. iNOS inactivation was most responsive to physiological concentrations of GSH with an apparent K(m) value of 13 mM. NOS turnover that leads to NOS S-nitrosation might be a mechanism for controlling NOS activity, and NOS S-nitrosation could play a role in the physiological generation of nitrosothiols.
Collapse
Affiliation(s)
- Brian C Smith
- California Institute for Quantitative Biosciences, University of California, Berkeley, California 94720-3220, United States
| | | | | |
Collapse
|
40
|
Osburn S, O'Hair RA, Black SM, Ryzhov V. Post-translational modification in the gas phase: mechanism of cysteine S-nitrosylation via ion-molecule reactions. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2011; 25:3216-22. [PMID: 22006383 PMCID: PMC3908822 DOI: 10.1002/rcm.5219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The gas-phase mechanism of S-nitrosylation of thiols was studied in a quadrupole ion trap mass spectrometer. This was done via ion-molecule reactions of protonated cysteine and many of its derivatives and other thiol ions with neutral tert-butyl nitrite or nitrous acid. Our results showed that the presence of the carboxylic acid functional group, -COOH, in the vicinity of the thiol group is essential for the gas-phase nitrosylation of thiols. When the carboxyl proton is replaced by a methyl group (cysteine methyl ester) no nitrosylation was observed. Other thiols lacking a carboxylic acid functional group displayed no S-nitrosylation, strongly suggesting that the carboxyl hydrogen plays a key role in the nitrosylation process. These results are in excellent agreement with a solution-phase mechanism proposed by Stamler et al. (J. S. Stamler, E. J. Toone, S. A. Lipton, N. J. Sucher. Neuron 1997, 18, 691-696) who suggested a catalytic role for the carboxylic acid group adjacent to cysteine residues and with later additions by Ascenzi et al. (P. Ascenzi, M. Colasanti, T. Persichini, M. Muolo, F. Polticelli, G. Venturini, D. Bordo, M. Bolognesi. Biol. Chem. 2000, 381, 623-627) who postulated that the presence of the carboxyl in the cysteine microenvironment in proteins is crucial for S-nitrosylation. A concerted mechanism for the gas-phase S-nitrosylation was proposed based on our results and was further studied using theoretical calculations. Our calculations showed that this proposed pathway is exothermic by 44.0 kJ mol(-1). This is one of the few recent examples when a gas-phase mechanism matches one in solution.
Collapse
Affiliation(s)
- Sandra Osburn
- Department of Chemistry and Biochemistry, and Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA
| | - Richard A.J. O'Hair
- School of Chemistry, University of Melbourne; Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne; and ARC Centre of Excellence for Free Radical Chemistry and Biotechnology, Melbourne, VIC 3010, Australia
| | - Stephen M. Black
- Pulmonary Vascular Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, GA 30912, USA
| | - Victor Ryzhov
- Department of Chemistry and Biochemistry, and Center for Biochemical and Biophysical Studies, Northern Illinois University, DeKalb, IL 60115, USA
- Correspondence to: V. Ryzhov, Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL 60115, USA.
| |
Collapse
|
41
|
Savidge TC, Urvil P, Oezguen N, Ali K, Choudhury A, Acharya V, Pinchuk I, Torres AG, English RD, Wiktorowicz JE, Loeffelholz M, Kumar R, Shi L, Nie W, Braun W, Herman B, Hausladen A, Feng H, Stamler JS, Pothoulakis C. Host S-nitrosylation inhibits clostridial small molecule-activated glucosylating toxins. Nat Med 2011; 17:1136-41. [PMID: 21857653 DOI: 10.1038/nm.2405] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 05/20/2011] [Indexed: 02/06/2023]
Abstract
The global prevalence of severe Clostridium difficile infection highlights the profound clinical significance of clostridial glucosylating toxins. Virulence is dependent on the autoactivation of a toxin cysteine protease, which is promoted by the allosteric cofactor inositol hexakisphosphate (InsP(6)). Host mechanisms that protect against such exotoxins are poorly understood. It is increasingly appreciated that the pleiotropic functions attributed to nitric oxide (NO), including host immunity, are in large part mediated by S-nitrosylation of proteins. Here we show that C. difficile toxins are S-nitrosylated by the infected host and that S-nitrosylation attenuates virulence by inhibiting toxin self-cleavage and cell entry. Notably, InsP(6)- and inositol pyrophosphate (InsP(7))-induced conformational changes in the toxin enabled host S-nitrosothiols to transnitrosylate the toxin catalytic cysteine, which forms part of a structurally conserved nitrosylation motif. Moreover, treatment with exogenous InsP(6) enhanced the therapeutic actions of oral S-nitrosothiols in mouse models of C. difficile infection. Allostery in bacterial proteins has thus been successfully exploited in the evolutionary development of nitrosothiol-based innate immunity and may provide an avenue to new therapeutic approaches.
Collapse
Affiliation(s)
- Tor C Savidge
- Department of Gastroenterology & Hepatology, University of Texas Medical Branch, Galveston, Texas, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Matsumoto A, Gow AJ. Membrane transfer of S-nitrosothiols. Nitric Oxide 2011; 25:102-7. [PMID: 21377531 PMCID: PMC3130086 DOI: 10.1016/j.niox.2011.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 02/12/2011] [Accepted: 02/24/2011] [Indexed: 02/07/2023]
Abstract
The distinctive function of nitric oxide (NO) in biology is to transmit cellular signals through membranes and regulate cellular functions in adjacent cells. NO conveys signals as a second messenger from a cell where NO is generated to contiguous cells in two ways; one is as gaseous molecule by free diffusion resulting in an activation of soluble guanylate cyclase (NO/cGMP pathway), and another form is by binding with a molecule such as cysteine or protein thiol through S-nitrosylation (SNO pathway). Both pathways transmit much of the biological influence of NO from cell where other messenger molecules but NO are confined, through the plasma membrane to the adjacent cells. Since SNO pathway cannot utilize free-diffusion mechanism to get through the membrane as the molecular size is significantly larger than NO molecule, it utilizes amino acid transporter to convey signals as a form of S-nitrosylated cysteine (CysNO). Although S-nitrosylated glutathione (GSNO) is the molecule which act as a determinant of the total S-nitrosothiol level in cell, transnitrosylation reaction from GSNO to CysNO is an initial requirement to pass through signal through the membrane. Thus, multiplexed combination of these steps and the regulatory factors involved in this system conform and modify the outcome from stimulus-response coupling via the SNO pathway.
Collapse
Affiliation(s)
- Akio Matsumoto
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana Chuo-ku Chiba, 260-8670, Japan,
| | - Andrew J. Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Road, Piscataway, NJ 08854, U.S.A.,
| |
Collapse
|
43
|
Marozkina NV, Gaston B. S-Nitrosylation signaling regulates cellular protein interactions. Biochim Biophys Acta Gen Subj 2011; 1820:722-9. [PMID: 21745537 DOI: 10.1016/j.bbagen.2011.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/13/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND S-Nitrosothiols are made by nitric oxide synthases and other metalloproteins. Unlike nitric oxide, S-nitrosothiols are involved in localized, covalent signaling reactions in specific cellular compartments. These reactions are enzymatically regulated. SCOPE S-Nitrosylation affects interactions involved in virtually every aspect of normal cell biology. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation. MAJOR CONCLUSIONS AND SIGNIFICANCE S-Nitrosylation is a regulated signaling reaction.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- University of Virginia School of Medicine, Division of Pediatric Respiratory Medicine, PO Box 800386, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
44
|
Leon-Bollotte L, Subramaniam S, Cauvard O, Plenchette-Colas S, Paul C, Godard C, Martinez-Ruiz A, Legembre P, Jeannin JF, Bettaieb A. S-nitrosylation of the death receptor fas promotes fas ligand-mediated apoptosis in cancer cells. Gastroenterology 2011; 140:2009-18, 2018.e1-4. [PMID: 21354149 DOI: 10.1053/j.gastro.2011.02.053] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 01/27/2011] [Accepted: 02/14/2011] [Indexed: 12/06/2022]
Abstract
BACKGROUND & AIMS Fas belongs to the family of tumor necrosis factor receptors which induce apoptosis. Many cancer cells express Fas but do not undergo Fas-mediated apoptosis. Nitric oxide reverses this resistance by increasing levels of Fas at the plasma membrane. We studied the mechanisms by which NO affects Fas function. METHODS Colon and mammary cancer cell lines were incubated with the NO donor glyceryl trinitrate or lipid A; S-nitrosylation of Fas was monitored using the biotin switch assay. Fas constructs that contained mutations at cysteine residues that prevent S-nitrosylation were used to investigate the involvement of S-nitrosylation in Fas-mediated cell death. Apoptosis was monitored according to morphologic criteria. RESULTS NO induced S-nitrosylation of cysteine residues 199 and 304 in the cytoplasmic part of Fas. In cancer cells that overexpressed wild-type Fas, S-nitrosylation induced Fas recruitment to lipid rafts and sensitized the cells to Fas ligand. In cells that expressed a mutant form of Fas in which cysteine 304 was replaced by valine residue, NO-mediated translocation of Fas to lipid rafts was affected and the death-inducing signal complex and synergistic effect of glyceryl trinitrate-Fas ligand were inhibited significantly. These effects were not observed in cells that expressed Fas with a mutation at cysteine 199. CONCLUSIONS We identified post-translational modifications (S-nitrosylation of cysteine residues 199 and 304) in the cytoplasmic domain of Fas. S-nitrosylation at cysteine 304 promotes redistribution of Fas to lipid rafts, formation of the death-inducing signal complex, and induction of cell death.
Collapse
Affiliation(s)
- Lissbeth Leon-Bollotte
- Ecole Pratique des Hautes Etudes, Tumor Immunology and Immunotherapy Laboratory, Dijon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nitration of tryptophan in ribosomal proteins is a novel post-translational modification of differentiated and naïve PC12 cells. Nitric Oxide 2011; 25:176-82. [PMID: 21642007 DOI: 10.1016/j.niox.2011.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Revised: 05/19/2011] [Accepted: 05/21/2011] [Indexed: 12/30/2022]
Abstract
Neuron growth factor (NGF) signaling in PC12 cell, which is derived from pheochromocytoma of rat adrenal medulla, induces expression of neuronal nitric oxide synthase (nNOS) and nitric oxide (NO) production. Subsequently, NO causes differentiation of PC12 cell to neuronal cell with morphological changes, such as neurite extension. In this study, we showed that 6-nitrotryptophan-containing proteins were produced in PC12 cell (naïve PC12 cell) and/or NGF-induced PC12 cell (differentiated PC12 cell). Western blot analysis of the protein extract of naïve PC12 cell and differentiated PC12 cell using anti 6-nitrotryptophan antibody showed several immunoreactive bands, which were subsequently subjected to trypsin digestion and LC-ESI-MS-MS analysis. The peptides from five ribosomal proteins, namely, 60S ribosomal protein L7 (Trp154), 60S acidic ribosomal protein P1 (Trp43), 40S ribosomal protein S2 (Trp60), 40S ribosomal protein S6 (Trp45), and 40S ribosomal protein S19 (Trp52), were identified as nitrotryptophan residue-containing proteins with significant ion score levels (p<0.05). Among these, tryptophan nitration was observed only in differentiated PC12 cell for S19 protein, and only in naïve PC12 cell for L7 protein. Tryptophan nitration of the other ribosomal proteins P1, S2, and S6 was observed in both naive and differentiated PC12 cells. The positive signal of nitrotryptophan-containing proteins in the Western blotting around 16 kDa (Band 1), which includes 40S ribosomal protein S19, was suppressed by treatment with NOS inhibitor, L-NAME. The tryptophan nitration of 40S ribosomal protein was not observed by LC-ESI-MS-MS analysis of this sample. This is the first study to identify several specific sites of nitrated tryptophan on proteins not only in viable culture cells but also in a physiological process: cell differentiation.
Collapse
|
46
|
Purwar N, McGarry JM, Kostera J, Pacheco AA, Schmidt M. Interaction of nitric oxide with catalase: structural and kinetic analysis. Biochemistry 2011; 50:4491-503. [PMID: 21524057 PMCID: PMC3140772 DOI: 10.1021/bi200130r] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
We present the structures of bovine catalase in its native form and complexed with ammonia and nitric oxide, obtained by X-ray crystallography. Using the NO generator 1-(N,N-diethylamino)diazen-1-ium-1,2-diolate, we were able to generate sufficiently high NO concentrations within the catalase crystals that substantial occupation was observed despite a high dissociation rate. Nitric oxide seems to be slightly bent from the heme normal that may indicate some iron(II) character in the formally ferric catalase. Microspectrophotometric investigations inline with the synchrotron X-ray beam reveal photoreduction of the central heme iron. In the cases of the native and ammonia-complexed catalase, reduction is accompanied by a relaxation phase. This is likely not the case for the catalase NO complex. The kinetics of binding of NO to catalase were investigated using NO photolyzed from N,N′-bis(carboxymethyl)-N,N′-dinitroso-p-phenylenediamine using an assay that combines catalase with myoglobin binding kinetics. The off rate is 1.5 s–1. Implications for catalase function are discussed.
Collapse
Affiliation(s)
- Namrta Purwar
- Department of Physics, University of Wisconsin-Milwaukee, 1900 East Kenwood Boulevard, Milwaukee, WI 53211, USA
| | | | | | | | | |
Collapse
|
47
|
Beiser DG, Orbelyan GA, Inouye BT, Costakis JG, Hamann KJ, McNally EM, Vanden Hoek TL. Genetic deletion of NOS3 increases lethal cardiac dysfunction following mouse cardiac arrest. Resuscitation 2011; 82:115-21. [PMID: 20951489 PMCID: PMC3018673 DOI: 10.1016/j.resuscitation.2010.08.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 08/23/2010] [Accepted: 08/30/2010] [Indexed: 12/23/2022]
Abstract
STUDY AIMS Cardiac arrest mortality is significantly affected by failure to obtain return of spontaneous circulation (ROSC) despite cardiopulmonary resuscitation (CPR). Severe myocardial dysfunction and cardiovascular collapse further affects mortality within hours of initial ROSC. Recent work suggests that enhancement of nitric oxide (NO) signaling within minutes of CPR can improve myocardial function and survival. We studied the role of NO signaling on cardiovascular outcomes following cardiac arrest and resuscitation using endothelial NO synthase knockout (NOS3(-/-)) mice. METHODS Adult female wild-type (WT) and NOS3(-/-) mice were anesthetized, intubated, and instrumented with left-ventricular pressure-volume catheters. Cardiac arrest was induced with intravenous potassium chloride. CPR was performed after 8min of untreated arrest. ROSC rate, cardiac function, whole-blood nitrosylhemoglobin (HbNO) concentrations, heart NOS3 content and phosphorylation (p-NOS3), cyclic guanosine monophosphate (cGMP), and phospho-troponin I (p-TnI) were measured. RESULTS Despite equal quality CPR, NOS3(-/-) mice displayed lower rates of ROSC compared to WT (47.6% [10/21] vs. 82.4% [14/17], p<0.005). Among ROSC animals, NOS3(-/-) vs. WT mice exhibited increased left-ventricular dysfunction and 120min mortality. Prior to ROSC, myocardial effectors of NO signaling including cGMP and p-TnI were decreased in NOS3(-/-) vs. WT mice (p<0.05). Following ROSC in WT mice, significant NOS3-dependent increases in circulating HbNO were seen by 120min. Significant increases in cardiac p-NOS3 occurred between end-arrest and 15min post-ROSC, while total NOS3 content was increased by 120min post-ROSC (p<0.05). CONCLUSIONS Genetic deletion of NOS3 decreases ROSC rate and worsens post-ROSC left-ventricular function. Poor cardiovascular outcomes are associated with differences in NOS3-dependent myocardial cGMP signaling and circulating NO metabolites.
Collapse
Affiliation(s)
- David G Beiser
- Emergency Resuscitation Center, Section of Emergency Medicine, University of Chicago, 5841 S. Maryland Ave., MC 5068, Chicago, IL 60637, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
White PJ, Charbonneau A, Cooney GJ, Marette A. Nitrosative modifications of protein and lipid signaling molecules by reactive nitrogen species. Am J Physiol Endocrinol Metab 2010; 299:E868-78. [PMID: 20876760 DOI: 10.1152/ajpendo.00510.2010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This review is the last of four review articles addressing covalent modifications of proteins and lipids. Two of the reviews in this series were previously published (15, 28) and dealt with modifications of signaling proteins by GlcNAcylation and serine phosphorylation. In the current issue of the Journal, we complete this series with two reviews, one by Riahi et al. (102a) on the signaling and cellular functions of 4-hydroxyalkenals, key products of lipid peroxidation processes, and our present review on the effects of nitrosative modifications of protein and lipid signaling molecules by reactive nitrogen species. The aim of this Perspectives review is to highlight the significant role that reactive nitrogen species may play in the regulation of cellular metabolism through this important class of posttranslational modification. The potential role of nitrosative modifications in the regulation of insulin signal transduction, mitochondrial energy metabolism, mRNA transcription, stress signaling, and endoplasmic reticulum function will each be discussed. Since nitrosative modifications are not restricted to proteins, the current understanding of a recently described genus of "nitro-fatty acids" will also be addressed.
Collapse
Affiliation(s)
- Phillip J White
- The Quebec Heart and Lung Institute, Hôpital Laval, Quebec, Canada G1V 4G5
| | | | | | | |
Collapse
|
49
|
Suliman HB, Babiker A, Withers CM, Sweeney TE, Carraway MS, Tatro LG, Bartz RR, Welty-Wolf KE, Piantadosi CA. Nitric oxide synthase-2 regulates mitochondrial Hsp60 chaperone function during bacterial peritonitis in mice. Free Radic Biol Med 2010; 48:736-46. [PMID: 20043987 PMCID: PMC2842938 DOI: 10.1016/j.freeradbiomed.2009.12.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 12/17/2009] [Accepted: 12/22/2009] [Indexed: 01/06/2023]
Abstract
Nitric oxide synthase-2 (NOS2) plays a critical role in reactive nitrogen species generation and cysteine modifications that influence mitochondrial function and signaling during inflammation. Here, we investigated the role of NOS2 in hepatic mitochondrial biogenesis during Escherichia coli peritonitis in mice. NOS2(-/-) mice displayed smaller mitochondrial biogenesis responses than Wt mice during E. coli infection according to differences in mRNA levels for the PGC-1 alpha coactivator, nuclear respiratory factor-1, mitochondrial transcription factor-A (Tfam), and mtDNA polymerase (Pol gamma). NOS2(-/-) mice did not significantly increase mitochondrial Tfam and Pol gamma protein levels during infection in conjunction with impaired mitochondrial DNA (mtDNA) transcription, loss of mtDNA copy number, and lower State 3 respiration rates. NOS2 blockade in mitochondrial-GFP reporter mice disrupted Hsp60 localization to mitochondria after E. coli exposure. Mechanistically, biotin-switch and immunoprecipitation studies demonstrated NOS2 binding to and S-nitros(yl)ation of Hsp60 and Hsp70. Specifically, NOS2 promoted Tfam accumulation in mitochondria by regulation of Hsp60-Tfam binding via S-nitros(yl)ation. In hepatocytes, site-directed mutagenesis identified (237)Cys as a critical residue for Hsp60 S-nitros(yl)ation. Thus, the role of NOS2 in inflammation-induced mitochondrial biogenesis involves both optimal gene expression for nuclear-encoded mtDNA-binding proteins and functional regulation of the Hsp60 chaperone that enables their importation for mtDNA transcription and replication.
Collapse
Affiliation(s)
- Hagir B Suliman
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lee J, Jung E, Lee J, Huh S, Kim YS, Kim YW, Kim YS, Park D. Anti-adipogenesis by 6-thioinosine is mediated by downregulation of PPAR gamma through JNK-dependent upregulation of iNOS. Cell Mol Life Sci 2010; 67:467-81. [PMID: 19941061 PMCID: PMC11115604 DOI: 10.1007/s00018-009-0196-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 10/27/2009] [Accepted: 10/27/2009] [Indexed: 12/14/2022]
Abstract
Adipocyte dysfunction is associated with the development of obesity. This study shows that 6-thioinosine inhibits adipocyte differentiation. The mRNA levels of PPAR gamma and C/EBPalpha, but not C/EBPbeta and delta, were reduced by 6-thioinosine. Moreover, the mRNA levels of PPAR gamma target genes (LPL, CD36, aP2, and LXRalpha) were down-regulated by 6-thioinosine. We also demonstrated that 6-thioinosine inhibits the transactivation activity and the mRNA level of PPAR gamma. Additionally, attempts to elucidate a possible mechanism underlying the 6-thioinosine-mediated effects revealed that 6-thioinosine induced iNOS gene expression without impacting eNOS expression, and that this was mediated through activation of AP-1, especially, JNK. In addition, 6-thioinosine was found to operate upstream of MEKK-1 in JNK activation signaling. Taken together, these findings suggest that the inhibition of adipocyte differentiation by 6-thioinosine occurs primarily through the reduced expression of PPAR gamma, which is mediated by upregulation of iNOS via the activation of JNK.
Collapse
Affiliation(s)
- Jongsung Lee
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eunsun Jung
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
| | - Jienny Lee
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
| | - Sungran Huh
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
| | - Young-Soo Kim
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
| | - Yong-Woo Kim
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Deokhoon Park
- Biospectrum Life Science Institute, Gunpo City, Gyunggi Do 435-833 Republic of Korea
| |
Collapse
|