1
|
Yang Z, Wang X, Gao C, Wu P, Ahammed GJ, Liu H, Chen S, Cui J. Glutathione is required for nitric oxide-induced chilling tolerance by synergistically regulating antioxidant system, polyamine synthesis, and mitochondrial function in cucumber (Cucumis sativus L.). PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 214:108878. [PMID: 38968841 DOI: 10.1016/j.plaphy.2024.108878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/07/2024]
Abstract
In this paper, we discussed the physiological mechanism of enhanced chilling tolerance with combined treatment of nitric oxide (NO) and reduced glutathione (GSH) in cucumber seedlings. With prolonged low temperature (10 °C/6 °C), oxidative stress improved, which was manifested as an increase the hydrogen peroxide (H2O2) and malondialdehyde (MDA), causing cell membrane damage, particularly after 48 h of chilling stress. Exogenous sodium nitroprusside (SNP, NO donor) enhanced the activity of nitric oxide synthase NOS-like, the contents of GSH and polyamines (PAs), and the cellular redox state, thus regulating the activities of mitochondrial oxidative phosphorylation components (CI, CII, CIV, CV). However, buthionine sulfoximine (BSO, a GSH synthase inhibitor) treatment drastically reversed or attenuated the effects of NO. Importantly, the combination of SNP and GSH treatment had the best effect in alleviating chilling-induced oxidative stress by upregulating the activities of antioxidant enzyme, including superoxidase dismutase (SOD), catalase (CAT), ascorbate peroxidase (APX) and peroxidase (POD) and improved the PAs content, thereby increased activities of CI, CII, CIII, CIV, and CV. This potentially contributes to the maintenance of oxidative phosphorylation originating from mitochondria. In addition, the high activity of S-nitrosoglutathione reductase (GSNOR) in the combined treatment of SNP and GSH possibly mediates the conversion of NO and GSH to S-nitrosoglutathione. Our study revealed that the combined treatment with NO and GSH to synergistically improve the cold tolerance of cucumber seedlings under prolonged low-temperature stress.
Collapse
Affiliation(s)
- Zhifeng Yang
- Department of Horticulture, Agricultural College, Shihezi University, Shihezi, Xinjiang, 832003, China; Key Laboratory of Special Fruits and Vegetables Cultivation Physiology and Germplasm Resources Utilization of Xinjiang Production and Construction Crops, Shihezi, Xinjiang,832003, China
| | - Xiaoyu Wang
- Department of Horticulture, Agricultural College, Shihezi University, Shihezi, Xinjiang, 832003, China; Key Laboratory of Special Fruits and Vegetables Cultivation Physiology and Germplasm Resources Utilization of Xinjiang Production and Construction Crops, Shihezi, Xinjiang,832003, China
| | - Chenpeng Gao
- Department of Horticulture, Agricultural College, Shihezi University, Shihezi, Xinjiang, 832003, China; Key Laboratory of Special Fruits and Vegetables Cultivation Physiology and Germplasm Resources Utilization of Xinjiang Production and Construction Crops, Shihezi, Xinjiang,832003, China
| | - Pei Wu
- Department of Horticulture, Agricultural College, Shihezi University, Shihezi, Xinjiang, 832003, China; Key Laboratory of Special Fruits and Vegetables Cultivation Physiology and Germplasm Resources Utilization of Xinjiang Production and Construction Crops, Shihezi, Xinjiang,832003, China
| | - Golam Jalal Ahammed
- College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang, 471023, China
| | - Huiying Liu
- Department of Horticulture, Agricultural College, Shihezi University, Shihezi, Xinjiang, 832003, China; Key Laboratory of Special Fruits and Vegetables Cultivation Physiology and Germplasm Resources Utilization of Xinjiang Production and Construction Crops, Shihezi, Xinjiang,832003, China
| | - ShuangChen Chen
- College of Horticulture and Plant Protection, Henan University of Science and Technology, Luoyang, 471023, China
| | - Jinxia Cui
- Department of Horticulture, Agricultural College, Shihezi University, Shihezi, Xinjiang, 832003, China; Key Laboratory of Special Fruits and Vegetables Cultivation Physiology and Germplasm Resources Utilization of Xinjiang Production and Construction Crops, Shihezi, Xinjiang,832003, China.
| |
Collapse
|
2
|
Kong W, Lu C. Role of mitochondria in neonatal hypoxic-ischemic encephalopathy. Histol Histopathol 2024; 39:991-1000. [PMID: 38314617 DOI: 10.14670/hh-18-710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Neonatal hypoxic-ischemic encephalopathy, an important cause of death as well as long-term disability in survivors, is caused by oxygen and glucose deprivation, and limited blood flow. Following hypoxic-ischemic injury in the neonatal brain, three main biochemical damages (excitotoxicity, oxidative stress, and exacerbated inflammation) are triggered. Mitochondria are involved in all three cascades. Mitochondria are the nexus of metabolic pathways to offer most of the energy that our body needs. Hypoxic-ischemic injury affects the characteristics of mitochondria, including dynamics, permeability, and ATP production, which also feed back into the process of neonatal hypoxic-ischemic encephalopathy. Mitochondria can be a cellular hub in inflammation, which is another main response of the injured neonatal brain. Some treatments for neonatal hypoxic-ischemic encephalopathy affect the function of mitochondria or target mitochondria, including therapeutic hypothermia and erythropoietin. This review presents the main roles of mitochondria in neonatal hypoxic-ischemic encephalopathy and discusses some potential treatments directed at mitochondria, which may foster the development of new therapeutic strategies for this encephalopathy.
Collapse
Affiliation(s)
- Weijing Kong
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Cheng Lu
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Stykel MG, Ryan SD. Network analysis of S-nitrosylated synaptic proteins demonstrates unique roles in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119720. [PMID: 38582237 DOI: 10.1016/j.bbamcr.2024.119720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Nitric oxide can covalently modify cysteine thiols on target proteins to alter that protein's function in a process called S-nitrosylation (SNO). S-nitrosylation of synaptic proteins plays an integral part in neurotransmission. Here we review the function of the SNO-proteome at the synapse and whether clusters of SNO-modification may predict synaptic dysfunction associated with disease. We used a systematic search strategy to concatenate SNO-proteomic datasets from normal human or murine brain samples. Identified SNO-modified proteins were then filtered against proteins reported in the Synaptome Database, which provides a detailed and experimentally verified annotation of all known synaptic proteins. Subsequently, we performed an unbiased network analysis of all known SNO-synaptic proteins to identify clusters of SNO proteins commonly involved in biological processes or with known disease associations. The resulting SNO networks were significantly enriched in biological processes related to metabolism, whereas significant gene-disease associations were related to Schizophrenia, Alzheimer's, Parkinson's and Huntington's disease. Guided by an unbiased network analysis, the current review presents a thorough discussion of how clustered changes to the SNO-proteome influence health and disease.
Collapse
Affiliation(s)
- Morgan G Stykel
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON, Canada
| | - Scott D Ryan
- Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON, Canada; Hotchkiss Brain Institute, Department of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
4
|
Qiu F, Liu Y, Liu Z. The Role of Protein S-Nitrosylation in Mitochondrial Quality Control in Central Nervous System Diseases. Aging Dis 2024:AD.2024.0099. [PMID: 38739938 DOI: 10.14336/ad.2024.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/25/2024] [Indexed: 05/16/2024] Open
Abstract
S-Nitrosylation is a reversible covalent post-translational modification. Under physiological conditions, S-nitrosylation plays a dynamic role in a wide range of biological processes by regulating the function of substrate proteins. Like other post-translational modifications, S-nitrosylation can affect protein conformation, activity, localization, aggregation, and protein interactions. Aberrant S-nitrosylation can lead to protein misfolding, mitochondrial fragmentation, synaptic damage, and autophagy. Mitochondria are essential organelles in energy production, metabolite biosynthesis, cell death, and immune responses, among other processes. Mitochondrial dysfunction can result in cell death and has been implicated in the development of many human diseases. Recent evidence suggests that S-nitrosylation and mitochondrial dysfunction are important modulators of the progression of several diseases. In this review, we highlight recent findings regarding the aberrant S- nitrosylation of mitochondrial proteins that regulate mitochondrial biosynthesis, fission and fusion, and autophagy. Specifically, we discuss the mechanisms by which S-nitrosylated mitochondrial proteins exercise mitochondrial quality control under pathological conditions, thereby influencing disease. A better understanding of these pathological events may provide novel therapeutic targets to mitigate the development of neurological diseases.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhiheng Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
5
|
Cao F, Li M, Wang W, Yi Y, Chen Y, Liu H. A coumarin-furoxan hybrid as novel nitric oxide donor induced cell apoptosis and ferroptosis in NSCLC by promoting S-nitrosylation of STAT3 and negative regulation of JAK2-STAT3 pathway. Biochem Pharmacol 2024; 222:116068. [PMID: 38387529 DOI: 10.1016/j.bcp.2024.116068] [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: 10/31/2023] [Revised: 12/20/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Non-small cell lung cancer (NSCLC) still lacks effective treatment because of its extensive mutation diversity and frequent drug resistance. Therefore, it is urgent to develop new therapeutic strategies for NSCLC. In this study, we evaluated the inhibitory effect of a new coumarin-furoxan hybrid compound 9, a nitric oxide (NO) donor drug, on NSCLC proliferation and its mechanism. Our results show that compound 9 can inhibit the growth of four NSCLC cell lines and H1975 xenograft model in a dose-dependent manner. Compound 9 effectively releases high concentrations of NO within the mitochondria, leading to cellular oxidative stress, mitochondrial dysfunction, and apoptosis. Moreover, compound 9 inhibits JAK2/STAT3 protein phosphorylation and induces S-nitrosylation modification of STAT3, ultimately resulting in endogenous apoptosis in NSCLC. Additionally, compound 9 significantly induces NSCLC ferroptosis by depleting intracellular GSH, elevating MDA levels, inhibiting SLC7A11/GSH protein expression, and negatively regulating the JAK2/STAT3 pathway. In summary, this study elucidates the inhibitory effects of compound 9 on NSCLC proliferation and provides insights into the underlying mechanisms, offering new possibilities for NSCLC treatment strategies.
Collapse
Affiliation(s)
- Fan Cao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mengru Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weijie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Yi
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ying Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
6
|
Malaviya R, Meshanni JA, Sunil VR, Venosa A, Guo C, Abramova EV, Vayas KN, Jiang C, Cervelli JA, Gow AJ, Laskin JD, Laskin DL. Role of macrophage bioenergetics in N-acetylcysteine-mediated mitigation of lung injury and oxidative stress induced by nitrogen mustard. Toxicol Appl Pharmacol 2024; 485:116908. [PMID: 38513841 DOI: 10.1016/j.taap.2024.116908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Nitrogen mustard (NM) is a toxic vesicant that causes acute injury to the respiratory tract. This is accompanied by an accumulation of activated macrophages in the lung and oxidative stress which have been implicated in tissue injury. In these studies, we analyzed the effects of N-acetylcysteine (NAC), an inhibitor of oxidative stress and inflammation on NM-induced lung injury, macrophage activation and bioenergetics. Treatment of rats with NAC (150 mg/kg, i.p., daily) beginning 30 min after administration of NM (0.125 mg/kg, i.t.) reduced histopathologic alterations in the lung including alveolar interstitial thickening, blood vessel hemorrhage, fibrin deposition, alveolar inflammation, and bronchiolization of alveolar walls within 3 d of exposure; damage to the alveolar-epithelial barrier, measured by bronchoalveolar lavage fluid protein and cells, was also reduced by NAC, along with oxidative stress as measured by heme oxygenase (HO)-1 and Ym-1 expression in the lung. Treatment of rats with NAC attenuated the accumulation of macrophages in the lung expressing proinflammatory genes including Ptgs2, Nos2, Il-6 and Il-12; macrophages expressing inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2 and tumor necrosis factor (TNF)α protein were also reduced in histologic sections. Conversely, NAC had no effect on macrophages expressing the anti-inflammatory proteins arginase-1 or mannose receptor, or on NM-induced increases in matrix metalloproteinase (MMP)-9 or proliferating cell nuclear antigen (PCNA), markers of tissue repair. Following NM exposure, lung macrophage basal and maximal glycolytic activity increased, while basal respiration decreased indicating greater reliance on glycolysis to generate ATP. NAC increased both glycolysis and oxidative phosphorylation. Additionally, in macrophages from both control and NM treated animals, NAC treatment resulted in increased S-nitrosylation of ATP synthase, protecting the enzyme from oxidative damage. Taken together, these data suggest that alterations in NM-induced macrophage activation and bioenergetics contribute to the efficacy of NAC in mitigating lung injury.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jaclynn A Meshanni
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Vasanthi R Sunil
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Changjiang Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Elena V Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Kinal N Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Chenghui Jiang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jessica A Cervelli
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
7
|
Chen Y, Wu L, Liu J, Ma L, Zhang W. Adenine nucleotide translocase: Current knowledge in post-translational modifications, regulations and pathological implications for human diseases. FASEB J 2023; 37:e22953. [PMID: 37224026 DOI: 10.1096/fj.202201855rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/01/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Adenine nucleotide translocases (ANTs) are central to mitochondrial integrity and bioenergetic metabolism. This review aims to integrate the progresses and knowledge on ANTs over the last few years, contributing to a potential implication of ANTs for various diseases. Structures, functions, modifications, regulators and pathological implications of ANTs for human diseases are intensively demonstrated here. ANTs have four isoforms (ANT1-4), responsible for exchanging ATP/ADP, possibly composing of pro-apoptotic mPTP as a major component, and mediating FA-dependent uncoupling of proton efflux. ANT can be modified by methylation, nitrosylation and nitroalkylation, acetylation, glutathionylation, phosphorylation, carbonylation and hydroxynonenal-induced modifications. Compounds, including bongkrekic acid, atractyloside calcium, carbon monoxide, minocycline, 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid, cardiolipin, free long-chain fatty acids, agaric acid, long chain acyl-coenzyme A esters, all have an ability to regulate ANT activities. ANT impairment leads to bioenergetic failure and mitochondrial dysfunction, contributing to pathogenesis of diseases, such as diabetes (deficiency), heart disease (deficiency), Parkinson's disease (reduction), Sengers Syndrome (decrease), cancer (isoform shifting), Alzheimer's Disease (coaggregation with Tau), Progressive External Opthalmoplegia (mutation), and Fascioscapulohumeral muscular dystrophy (overexpression). This review improves the understanding of the mechanism of ANT in pathogenesis of human diseases, and opens a window for novel therapeutic strategies targeted on ANT in diseases.
Collapse
Affiliation(s)
- Yingfei Chen
- Grade 2020, Capital Medical University, Beijing, China
| | - Leshuang Wu
- Grade 2019, Dalian Medical University, Dalian, China
| | - Jun Liu
- Department of Epidemiology, Dalian Medical University, Dalian, China
| | - Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian, China
| | - Wenli Zhang
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Wang K, Moore A, Grayson C, Mailloux RJ. S-nitroso-glutathione (GSNO) inhibits hydrogen peroxide production by alpha-ketoglutarate dehydrogenase: An investigation into sex and diet effects. Free Radic Biol Med 2023; 204:287-300. [PMID: 37225107 DOI: 10.1016/j.freeradbiomed.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/06/2023] [Accepted: 05/11/2023] [Indexed: 05/26/2023]
Abstract
Pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase (KGDH) are vital sources of hydrogen peroxide (H2O2) and key sites for redox regulation. Here, we report KGDH is more sensitive to inhibition by S-nitroso-glutathione (GSNO) when compared to PDH and deactivation of both enzymes by nitro modification is affected by sex and diet. Liver mitochondria from male C57BL/6N mice displayed a robust inhibition of H2O2 production after exposure to 500-2000 μM GSNO. H2O2 genesis by PDH was not significantly affected by GSNO. Purified KGDH of porcine heart origin displayed a ∼82% decrease in H2O2 generating activity at 500 μM GSNO, which was mirrored by a decrease in NADH production. By contrast, H2O2- and NADH-producing activity of purified PDH was only minimally affected by an incubation in 500 μM GSNO. Incubations in GSNO had no significant effect on the H2O2-generating activity of KGDH and PDH in female liver mitochondria when compared to samples collected from males, which was attributed to higher GSNO reductase (GSNOR) activity. High fat feeding augmented the GSNO-mediated inhibition of KGDH in liver mitochondria from male mice. Exposure of male mice to a HFD also resulted in a significant decrease in the GSNO-mediated inhibition of H2O2 genesis by PDH, an effect not observed in mice fed a control-matched diet (CD). Female mice displayed higher resistance to the GSNO-induced inhibition of H2O2 production, regardless of being fed a CD or HFD. However, exposure to a HFD did result in a small but significant decrease in H2O2 production by KGDH and PDH when female liver mitochondria were treated with GSNO. Although, the effect was less when compared to their male counterparts. Collectively, we show for the first time GSNO deactivates H2O2 production by α-keto acid dehydrogenases and we demonstrate that sex and diet are determinants for the nitro-inhibition of both KGDH and PDH.
Collapse
Affiliation(s)
- Kevin Wang
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Amanda Moore
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Cathryn Grayson
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada
| | - Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Ste.-Anne-de-Bellevue, Quebec, Canada.
| |
Collapse
|
9
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
10
|
Verde C, Giordano D, Bruno S. NO and Heme Proteins: Cross-Talk between Heme and Cysteine Residues. Antioxidants (Basel) 2023; 12:antiox12020321. [PMID: 36829880 PMCID: PMC9952723 DOI: 10.3390/antiox12020321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Heme proteins are a diverse group that includes several unrelated families. Their biological function is mainly associated with the reactivity of the heme group, which-among several other reactions-can bind to and react with nitric oxide (NO) and other nitrogen compounds for their production, scavenging, and transport. The S-nitrosylation of cysteine residues, which also results from the reaction with NO and other nitrogen compounds, is a post-translational modification regulating protein activity, with direct effects on a variety of signaling pathways. Heme proteins are unique in exhibiting this dual reactivity toward NO, with reported examples of cross-reactivity between the heme and cysteine residues within the same protein. In this work, we review the literature on this interplay, with particular emphasis on heme proteins in which heme-dependent nitrosylation has been reported and those for which both heme nitrosylation and S-nitrosylation have been associated with biological functions.
Collapse
Affiliation(s)
- Cinzia Verde
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Via Pietro Castellino 111, 80131 Napoli, Italy
- Department of Marine Biotechnology, Stazione Zoologica Anton Dohrn (SZN), Villa Comunale, 80121 Napoli, Italy
| | - Daniela Giordano
- Institute of Biosciences and BioResources (IBBR), National Research Council (CNR), Via Pietro Castellino 111, 80131 Napoli, Italy
- Department of Marine Biotechnology, Stazione Zoologica Anton Dohrn (SZN), Villa Comunale, 80121 Napoli, Italy
| | - Stefano Bruno
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
- Biopharmanet-TEC, University of Parma, 43124 Parma, Italy
- Correspondence:
| |
Collapse
|
11
|
Robichaux DJ, Harata M, Murphy E, Karch J. Mitochondrial permeability transition pore-dependent necrosis. J Mol Cell Cardiol 2023; 174:47-55. [PMID: 36410526 PMCID: PMC9868081 DOI: 10.1016/j.yjmcc.2022.11.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
Mitochondrial permeability transition pore (mPTP)-dependent cell death is a form of necrotic cell death that is driven by mitochondrial dysfunction by the opening of the mPTP and is triggered by increases in matrix levels of Ca2+ and reactive oxygen species. This form of cell death has been implicated in ischemic injuries of the heart and brain as well as numerous degenerative diseases in the brain and skeletal muscle. This review focuses on the molecular triggers and regulators of mPTP-dependent necrosis in the context of myocardial ischemia reperfusion injury. Research over the past 50 years has led to the identity of regulators and putative pore-forming components of the mPTP. Finally, downstream consequences of activation of the mPTP as well as ongoing questions and areas of research are discussed. These questions pose a particular interest as targeting the mPTP could potentially represent an efficacious therapeutic strategy to reduce infarct size following an ischemic event.
Collapse
Affiliation(s)
- Dexter J Robichaux
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Mikako Harata
- Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD, USA
| | - Elizabeth Murphy
- Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD, USA
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
13
|
Arasimowicz-Jelonek M, Jagodzik P, Płóciennik A, Sobieszczuk-Nowicka E, Mattoo A, Polcyn W, Floryszak-Wieczorek J. Dynamics of nitration during dark-induced leaf senescence in Arabidopsis reveals proteins modified by tryptophan nitration. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:6853-6875. [PMID: 35981877 DOI: 10.1093/jxb/erac341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Nitric oxide (NO) is a critical molecule that links plant development with stress responses. Herein, new insights into the role of NO metabolism during leaf senescence in Arabidopsis are presented. A gradual decrease in NO emission accompanied dark-induced leaf senescence (DILS), and a transient wave of peroxynitrite (ONOO-) formation was detected by day 3 of DILS. The boosted ONOO- did not promote tryptophan (Trp) nitration, while the pool of 6-nitroTrp-containing proteins was depleted as senescence progressed. Immunoprecipitation combined with mass spectrometry was used to identify 63 and 4 characteristic 6-nitroTrp-containing proteins in control and individually darkened leaves, respectively. The potential in vivo targets of Trp nitration were mainly related to protein biosynthesis and carbohydrate metabolism. In contrast, nitration of tyrosine-containing proteins was intensified 2-fold on day 3 of DILS. Also, nitrative modification of RNA and DNA increased significantly on days 3 and 7 of DILS, respectively. Taken together, ONOO- can be considered a novel pro-senescence regulator that fine-tunes the redox environment for selective bio-target nitration. Thus, DILS-triggered nitrative changes at RNA and protein levels promote developmental shifts during the plant's lifespan and temporal adjustment in plant metabolism under suboptimal environmental conditions.
Collapse
Affiliation(s)
- Magdalena Arasimowicz-Jelonek
- Department of Plant Ecophysiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Przemysław Jagodzik
- Department of Plant Ecophysiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Artur Płóciennik
- Department of Plant Ecophysiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Ewa Sobieszczuk-Nowicka
- Department of Plant Physiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Autar Mattoo
- Sustainable Agricultural Systems Laboratory, USDA-ARS, Henry A. Wallace Beltsville Agricultural Research Center, Beltsville, MD 20705-2350, USA
| | - Władysław Polcyn
- Department of Plant Physiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | | |
Collapse
|
14
|
Stykel MG, Ryan SD. Nitrosative stress in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:104. [PMID: 35953517 PMCID: PMC9372037 DOI: 10.1038/s41531-022-00370-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022] Open
Abstract
Parkinson’s Disease (PD) is a neurodegenerative disorder characterized, in part, by the loss of dopaminergic neurons within the nigral-striatal pathway. Multiple lines of evidence support a role for reactive nitrogen species (RNS) in degeneration of this pathway, specifically nitric oxide (NO). This review will focus on how RNS leads to loss of dopaminergic neurons in PD and whether RNS accumulation represents a central signal in the degenerative cascade. Herein, we provide an overview of how RNS accumulates in PD by considering the various cellular sources of RNS including nNOS, iNOS, nitrate, and nitrite reduction and describe evidence that these sources are upregulating RNS in PD. We document that over 1/3 of the proteins that deposit in Lewy Bodies, are post-translationally modified (S-nitrosylated) by RNS and provide a broad description of how this elicits deleterious effects in neurons. In doing so, we identify specific proteins that are modified by RNS in neurons which are implicated in PD pathogenesis, with an emphasis on exacerbation of synucleinopathy. How nitration of alpha-synuclein (aSyn) leads to aSyn misfolding and toxicity in PD models is outlined. Furthermore, we delineate how RNS modulates known PD-related phenotypes including axo-dendritic-, mitochondrial-, and dopamine-dysfunctions. Finally, we discuss successful outcomes of therapeutics that target S-nitrosylation of proteins in Parkinson’s Disease related clinical trials. In conclusion, we argue that targeting RNS may be of therapeutic benefit for people in early clinical stages of PD.
Collapse
Affiliation(s)
- Morgan G Stykel
- The Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON N1G 2W1, ON, Canada
| | - Scott D Ryan
- The Department of Molecular and Cellular Biology, The University of Guelph, Guelph, ON N1G 2W1, ON, Canada. .,Neurodegenerative Disease Center, Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
15
|
Borges Araujo AJ, Cerruti GV, Zuccarelli R, Rodriguez Ruiz M, Freschi L, Singh R, Moerschbacher BM, Floh EIS, Wendt dos Santos AL. Proteomic Analysis of S-Nitrosation Sites During Somatic Embryogenesis in Brazilian Pine, Araucaria angustifolia (Bertol.) Kuntze. FRONTIERS IN PLANT SCIENCE 2022; 13:902068. [PMID: 35845673 PMCID: PMC9280032 DOI: 10.3389/fpls.2022.902068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Cysteine S-nitrosation is a redox-based post-translational modification that mediates nitric oxide (NO) regulation of various aspects of plant growth, development and stress responses. Despite its importance, studies exploring protein signaling pathways that are regulated by S-nitrosation during somatic embryogenesis have not been performed. In the present study, endogenous cysteine S-nitrosation site and S-nitrosated proteins were identified by iodo-TMT labeling during somatic embryogenesis in Brazilian pine, an endangered native conifer of South America. In addition, endogenous -S-nitrosothiol (SNO) levels and S-nitrosoglutathione reductase (GSNOR) activity were determined in cell lines with contrasting embryogenic potential. Overall, we identified an array of proteins associated with a large variety of biological processes and molecular functions with some of them already described as important for somatic embryogenesis (Class IV chitinase, pyruvate dehydrogenase E1 and dehydroascorbate reductase). In total, our S-nitrosoproteome analyses identified 18 endogenously S-nitrosated proteins and 50 in vitro S-nitrosated proteins (after GSNO treatment) during cell culture proliferation and embryo development. Furthermore, SNO levels and GSNOR activity were increased during embryo formation. These findings expand our understanding of the Brazilian pine proteome and shed novel insights into the potential use of pharmacological manipulation of NO levels by using NO inhibitors and donors during somatic embryogenesis.
Collapse
Affiliation(s)
| | | | - Rafael Zuccarelli
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Marta Rodriguez Ruiz
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Luciano Freschi
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Ratna Singh
- Department of Plant Biology and Biotechnology, WWU Münster, Münster, Germany
| | | | - Eny Iochevet Segal Floh
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
16
|
Schiffer TA, Löf L, Gallini R, Kamali-Moghaddam M, Carlström M, Palm F. Mitochondrial Respiration-Dependent ANT2-UCP2 Interaction. Front Physiol 2022; 13:866590. [PMID: 35694398 PMCID: PMC9177158 DOI: 10.3389/fphys.2022.866590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Adenine nucleotide translocases (ANTs) and uncoupling proteins (UCPs) are known to facilitate proton leak across the inner mitochondrial membrane. However, it remains to be unravelled whether UCP2/3 contribute to significant amount of proton leak in vivo. Reports are indicative of UCP2 dependent proton-coupled efflux of C4 metabolites from the mitochondrial matrix. Previous studies have suggested that UCP2/3 knockdown (KD) contributes to increased ANT-dependent proton leak. Here we investigated the hypothesis that interaction exists between the UCP2 and ANT2 proteins, and that such interaction is regulated by the cellular metabolic demand. Protein-protein interaction was evaluated using reciprocal co-immunoprecipitation and in situ proximity ligation assay. KD of ANT2 and UCP2 was performed by siRNA in human embryonic kidney cells 293A (HEK293A) cells. Mitochondrial and cellular respiration was measured by high-resolution respirometry. ANT2-UCP2 interaction was demonstrated, and this was dependent on cellular metabolism. Inhibition of ATP synthase promoted ANT2-UCP2 interaction whereas high cellular respiration, induced by adding the mitochondrial uncoupler FCCP, prevented interaction. UCP2 KD contributed to increased carboxyatractyloside (CATR) sensitive proton leak, whereas ANT2 and UCP2 double KD reduced CATR sensitive proton leak, compared to UCP2 KD. Furthermore, proton leak was reduced in double KD compared to UCP2 KD. In conclusion, our results show that there is an interaction between ANT2-UCP2, which appears to be dynamically regulated by mitochondrial respiratory activity. This may have implications in the regulation of mitochondrial efficiency or cellular substrate utilization as increased activity of UCP2 may promote a switch from glucose to fatty acid metabolism.
Collapse
Affiliation(s)
- Tomas A. Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- *Correspondence: Tomas A. Schiffer,
| | - Liza Löf
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Radiosa Gallini
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
S-nitrosoglutathione alleviates hyperglycemia-induced neurobehavioral deficits involving nitro-oxidative stress and aberrant monaminergic system. Nitric Oxide 2022; 122-123:35-44. [DOI: 10.1016/j.niox.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/07/2022] [Accepted: 03/02/2022] [Indexed: 11/22/2022]
|
18
|
Boulghobra D, Dubois M, Alpha-Bazin B, Coste F, Olmos M, Gayrard S, Bornard I, Meyer G, Gaillard JC, Armengaud J, Reboul C. Increased protein S-nitrosylation in mitochondria: a key mechanism of exercise-induced cardioprotection. Basic Res Cardiol 2021; 116:66. [PMID: 34940922 DOI: 10.1007/s00395-021-00906-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) activation in the heart plays a key role in exercise-induced cardioprotection during ischemia-reperfusion, but the underlying mechanisms remain unknown. We hypothesized that the cardioprotective effect of exercise training could be explained by the re-localization of eNOS-dependent nitric oxide (NO)/S-nitrosylation signaling to mitochondria. By comparing exercised (5 days/week for 5 weeks) and sedentary Wistar rats, we found that exercise training increased eNOS level and activation by phosphorylation (at serine 1177) in mitochondria, but not in the cytosolic subfraction of cardiomyocytes. Using confocal microscopy, we confirmed that NO production in mitochondria was increased in response to H2O2 exposure in cardiomyocytes from exercised but not sedentary rats. Moreover, by S-nitrosoproteomic analysis, we identified several key S-nitrosylated proteins involved in mitochondrial function and cardioprotection. In agreement, we also observed that the increase in Ca2+ retention capacity by mitochondria isolated from the heart of exercised rats was abolished by exposure to the NOS inhibitor L-NAME or to the reducing agent ascorbate, known to denitrosylate proteins. Pre-incubation with ascorbate or L-NAME also increased mitochondrial reactive oxygen species production in cardiomyocytes from exercised but not from sedentary animals. We confirmed these results using isolated hearts perfused with L-NAME before ischemia-reperfusion. Altogether, these results strongly support the hypothesis that exercise training increases eNOS/NO/S-nitrosylation signaling in mitochondria, which might represent a key mechanism of exercise-induced cardioprotection.
Collapse
Affiliation(s)
| | | | - Béatrice Alpha-Bazin
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200, Bagnols-sur-Cèze, France
| | - Florence Coste
- LAPEC EA-4278, Avignon Université, 84000, Avignon, France
| | - Maxime Olmos
- LAPEC EA-4278, Avignon Université, 84000, Avignon, France
| | | | | | - Gregory Meyer
- LAPEC EA-4278, Avignon Université, 84000, Avignon, France
| | - Jean-Charles Gaillard
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200, Bagnols-sur-Cèze, France
| | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200, Bagnols-sur-Cèze, France
| | - Cyril Reboul
- LAPEC EA-4278, Avignon Université, 84000, Avignon, France. .,Cardiovascular Physiology Laboratory, UPR4278, UFR Sciences Technologies Santé, Centre INRAE-Site Agroparc, 228 route de l'Aérodrome, 84914, Avignon Cedex 9, France.
| |
Collapse
|
19
|
Hande S, Sonkar V, Bhoj P, Togre N, Goswami K, Dash D. The Role of Oxidative and Nitrosative Stress of Silver Nanoparticles in Human Parasitic Helminth Brugia malayi: A Mechanistic Insight. Acta Parasitol 2021; 66:1212-1221. [PMID: 33884574 DOI: 10.1007/s11686-021-00394-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 04/12/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Silver nanoparticles (AgNPs) mediated apoptosis is well-known but its rationale is yet to be elucidated. This study explored the mechanistic underpinning of the apoptosis in the Brugia malayi parasitic model. METHOD Silver nanoparticles were synthesized and tested against B. malayi microfilariae (Mf) to explore the role of oxidative and nitrosative stress in its apoptotic effect. RESULTS AgNPs caused significant decrease in reduced glutathione (GSH) level and increase in both protein carbonylation and nitric oxide (NO) level indicating oxidative as well as nitrosative stress. Both GSH and nitric oxide synthase (NOS) inhibitors exhibited marked reversal. Nanoparticles and NO-donor in combination but not the NO-donor alone showed significant antiparasitic effect implying the requisite of combined oxidative and nitrosative stress to induce apoptosis. Synthetically prepared peroxynitrite from NaNO2 to H2O2 showed marked antiparasitic effect in very low dose which could be achieved neither by NaNO2 or H2O2 alone. GSH reversed the effect of peroxynitrite similar to its specific inhibitor, acetaminophen. GSH also reversed the plummet in mitochondrial oxygen consumption by AgNPs. CONCLUSION We conclude that apoptosis by AgNPs is possibly mediated through peroxynitrite dependent depletion of GSH; this provides a significant insight into the pharmacological as well as toxicological impact of AgNPs.
Collapse
Affiliation(s)
- Sneha Hande
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, 442 102, India
| | - Vijay Sonkar
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Uttar Pradesh, Varanasi, 221 005, India
| | - Priyanka Bhoj
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, 442 102, India
| | - Namdev Togre
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, 442 102, India
| | - Kalyan Goswami
- Department of Biochemistry, All India Institute of Medical Sciences, Kalyani, West Bengal, 741 245, India.
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Uttar Pradesh, Varanasi, 221 005, India.
| |
Collapse
|
20
|
Systems Biology Reveals S-Nitrosylation-Dependent Regulation of Mitochondrial Functions in Mice with Shank3 Mutation Associated with Autism Spectrum Disorder. Brain Sci 2021; 11:brainsci11060677. [PMID: 34064215 PMCID: PMC8224296 DOI: 10.3390/brainsci11060677] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/20/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder manifested in repetitive behavior, abnormalities in social interactions, and communication. The pathogenesis of this disorder is not clear, and no effective treatment is currently available. Protein S-nitrosylation (SNO), the nitric oxide (NO)-mediated posttranslational modification, targets key proteins implicated in synaptic and neuronal functions. Previously, we have shown that NO and SNO are involved in the ASD mouse model based on the Shank3 mutation. The energy supply to the brain mostly relies on oxidative phosphorylation in the mitochondria. Recent studies show that mitochondrial dysfunction and oxidative stress are involved in ASD pathology. In this work, we performed SNO proteomics analysis of cortical tissues of the Shank3 mouse model of ASD with the focus on mitochondrial proteins and processes. The study was based on the SNOTRAP technology followed by systems biology analysis. This work revealed that 63 mitochondrial proteins were S-nitrosylated and that several mitochondria-related processes, including those associated with oxidative phosphorylation, oxidative stress, and apoptosis, were enriched. This study implies that aberrant SNO signaling induced by the Shank3 mutation can target a wide range of mitochondria-related proteins and processes that may contribute to the ASD pathology. It is the first study to investigate the role of NO-dependent mitochondrial functions in ASD.
Collapse
|
21
|
Buelna-Chontal M, García-Niño WR, Silva-Palacios A, Enríquez-Cortina C, Zazueta C. Implications of Oxidative and Nitrosative Post-Translational Modifications in Therapeutic Strategies against Reperfusion Damage. Antioxidants (Basel) 2021; 10:749. [PMID: 34066806 PMCID: PMC8151040 DOI: 10.3390/antiox10050749] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022] Open
Abstract
Post-translational modifications based on redox reactions "switch on-off" the biological activity of different downstream targets, modifying a myriad of processes and providing an efficient mechanism for signaling regulation in physiological and pathological conditions. Such modifications depend on the generation of redox components, such as reactive oxygen species and nitric oxide. Therefore, as the oxidative or nitrosative milieu prevailing in the reperfused heart is determinant for protective signaling, in this review we defined the impact of redox-based post-translational modifications resulting from either oxidative/nitrosative signaling or oxidative/nitrosative stress that occurs during reperfusion damage. The role that cardioprotective conditioning strategies have had to establish that such changes occur at different subcellular levels, particularly in mitochondria, is also presented. Another section is devoted to the possible mechanism of signal delivering of modified proteins. Finally, we discuss the possible efficacy of redox-based therapeutic strategies against reperfusion damage.
Collapse
Affiliation(s)
| | | | | | | | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (M.B.-C.); (W.R.G.-N.); (A.S.-P.); (C.E.-C.)
| |
Collapse
|
22
|
Jedelská T, Sedlářová M, Lochman J, Činčalová L, Luhová L, Petřivalský M. Protein S-nitrosation differentially modulates tomato responses to infection by hemi-biotrophic oomycetes of Phytophthora spp. HORTICULTURE RESEARCH 2021; 8:34. [PMID: 33518717 PMCID: PMC7848004 DOI: 10.1038/s41438-021-00469-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 05/04/2023]
Abstract
Regulation of protein function by reversible S-nitrosation, a post-translational modification based on the attachment of nitroso group to cysteine thiols, has emerged among key mechanisms of NO signalling in plant development and stress responses. S-nitrosoglutathione is regarded as the most abundant low-molecular-weight S-nitrosothiol in plants, where its intracellular concentrations are modulated by S-nitrosoglutathione reductase. We analysed modulations of S-nitrosothiols and protein S-nitrosation mediated by S-nitrosoglutathione reductase in cultivated Solanum lycopersicum (susceptible) and wild Solanum habrochaites (resistant genotype) up to 96 h post inoculation (hpi) by two hemibiotrophic oomycetes, Phytophthora infestans and Phytophthora parasitica. S-nitrosoglutathione reductase activity and protein level were decreased by P. infestans and P. parasitica infection in both genotypes, whereas protein S-nitrosothiols were increased by P. infestans infection, particularly at 72 hpi related to pathogen biotrophy-necrotrophy transition. Increased levels of S-nitrosothiols localised in both proximal and distal parts to the infection site, which suggests together with their localisation to vascular bundles a signalling role in systemic responses. S-nitrosation targets in plants infected with P. infestans identified by a proteomic analysis include namely antioxidant and defence proteins, together with important proteins of metabolic, regulatory and structural functions. Ascorbate peroxidase S-nitrosation was observed in both genotypes in parallel to increased enzyme activity and protein level during P. infestans pathogenesis, namely in the susceptible genotype. These results show important regulatory functions of protein S-nitrosation in concerting molecular mechanisms of plant resistance to hemibiotrophic pathogens.
Collapse
Affiliation(s)
- Tereza Jedelská
- Department of Biochemistry, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic
| | - Michaela Sedlářová
- Department of Botany, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic
| | - Jan Lochman
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-625 00, Brno, Czech Republic
| | - Lucie Činčalová
- Department of Biochemistry, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic
| | - Lenka Luhová
- Department of Biochemistry, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic
| | - Marek Petřivalský
- Department of Biochemistry, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic.
| |
Collapse
|
23
|
Gil A, Martín-Montañez E, Valverde N, Lara E, Boraldi F, Claros S, Romero-Zerbo SY, Fernández O, Pavia J, Garcia-Fernandez M. Neuronal Metabolism and Neuroprotection: Neuroprotective Effect of Fingolimod on Menadione-Induced Mitochondrial Damage. Cells 2020; 10:cells10010034. [PMID: 33383658 PMCID: PMC7824129 DOI: 10.3390/cells10010034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Imbalance in the oxidative status in neurons, along with mitochondrial damage, are common characteristics in some neurodegenerative diseases. The maintenance in energy production is crucial to face and recover from oxidative damage, and the preservation of different sources of energy production is essential to preserve neuronal function. Fingolimod phosphate is a drug with neuroprotective and antioxidant actions, used in the treatment of multiple sclerosis. This work was performed in a model of oxidative damage on neuronal cell cultures exposed to menadione in the presence or absence of fingolimod phosphate. We studied the mitochondrial function, antioxidant enzymes, protein nitrosylation, and several pathways related with glucose metabolism and glycolytic and pentose phosphate in neuronal cells cultures. Our results showed that menadione produces a decrease in mitochondrial function, an imbalance in antioxidant enzymes, and an increase in nitrosylated proteins with a decrease in glycolysis and glucose-6-phosphate dehydrogenase. All these effects were counteracted when fingolimod phosphate was present in the incubation media. These effects were mediated, at least in part, by the interaction of this drug with its specific S1P receptors. These actions would make this drug a potential tool in the treatment of neurodegenerative processes, either to slow progression or alleviate symptoms.
Collapse
Affiliation(s)
- Antonio Gil
- Department of Pharmacology and Pediatrics, Faculty of Medicine, Malaga University, 29010 Malaga, Spain; (A.G.); (E.M.-M.); (O.F.)
| | - Elisa Martín-Montañez
- Department of Pharmacology and Pediatrics, Faculty of Medicine, Malaga University, 29010 Malaga, Spain; (A.G.); (E.M.-M.); (O.F.)
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
| | - Nadia Valverde
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
- Department of Human Physiology, Faculty of Medicine, Malaga University, 29010 Malaga, Spain;
| | - Estrella Lara
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
- Department of Human Physiology, Faculty of Medicine, Malaga University, 29010 Malaga, Spain;
| | - Federica Boraldi
- Department of Life Sciences, University of Modena e Reggio Emilia, 41125 Modena, Italy;
| | - Silvia Claros
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
- Department of Human Physiology, Faculty of Medicine, Malaga University, 29010 Malaga, Spain;
| | | | - Oscar Fernández
- Department of Pharmacology and Pediatrics, Faculty of Medicine, Malaga University, 29010 Malaga, Spain; (A.G.); (E.M.-M.); (O.F.)
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
| | - Jose Pavia
- Department of Pharmacology and Pediatrics, Faculty of Medicine, Malaga University, 29010 Malaga, Spain; (A.G.); (E.M.-M.); (O.F.)
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
- Correspondence: (J.P.); (M.G.-F.)
| | - Maria Garcia-Fernandez
- Neuroscience Unit, Biomedical Research Institute of Malaga (IBIMA), Malaga University Hospital, 29010 Malaga, Spain; (N.V.); (E.L.); (S.C.)
- Department of Human Physiology, Faculty of Medicine, Malaga University, 29010 Malaga, Spain;
- Correspondence: (J.P.); (M.G.-F.)
| |
Collapse
|
24
|
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
|
25
|
Gibhardt CS, Ezeriņa D, Sung HM, Messens J, Bogeski I. Redox regulation of the mitochondrial calcium transport machinery. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
26
|
Wolf C, López del Amo V, Arndt S, Bueno D, Tenzer S, Hanschmann EM, Berndt C, Methner A. Redox Modifications of Proteins of the Mitochondrial Fusion and Fission Machinery. Cells 2020; 9:cells9040815. [PMID: 32230997 PMCID: PMC7226787 DOI: 10.3390/cells9040815] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial fusion and fission tailors the mitochondrial shape to changes in cellular homeostasis. Players of this process are the mitofusins, which regulate fusion of the outer mitochondrial membrane, and the fission protein DRP1. Upon specific stimuli, DRP1 translocates to the mitochondria, where it interacts with its receptors FIS1, MFF, and MID49/51. Another fission factor of clinical relevance is GDAP1. Here, we identify and discuss cysteine residues of these proteins that are conserved in phylogenetically distant organisms and which represent potential sites of posttranslational redox modifications. We reveal that worms and flies possess only a single mitofusin, which in vertebrates diverged into MFN1 and MFN2. All mitofusins contain four conserved cysteines in addition to cysteine 684 in MFN2, a site involved in mitochondrial hyperfusion. DRP1 and FIS1 are also evolutionarily conserved but only DRP1 contains four conserved cysteine residues besides cysteine 644, a specific site of nitrosylation. MFF and MID49/51 are only present in the vertebrate lineage. GDAP1 is missing in the nematode genome and contains no conserved cysteine residues. Our analysis suggests that the function of the evolutionarily oldest proteins of the mitochondrial fusion and fission machinery, the mitofusins and DRP1 but not FIS1, might be altered by redox modifications.
Collapse
Affiliation(s)
- Christina Wolf
- Institute of Molecular Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany; (C.W.); (D.B.)
| | - Víctor López del Amo
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, USA;
| | - Sabine Arndt
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany; (S.A.); (S.T.)
| | - Diones Bueno
- Institute of Molecular Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany; (C.W.); (D.B.)
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany; (S.A.); (S.T.)
| | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (E.-M.H.); (C.B.)
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany; (E.-M.H.); (C.B.)
| | - Axel Methner
- Institute of Molecular Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany; (C.W.); (D.B.)
- Correspondence:
| |
Collapse
|
27
|
Calbet JAL, Martín-Rodríguez S, Martin-Rincon M, Morales-Alamo D. An integrative approach to the regulation of mitochondrial respiration during exercise: Focus on high-intensity exercise. Redox Biol 2020; 35:101478. [PMID: 32156501 PMCID: PMC7284910 DOI: 10.1016/j.redox.2020.101478] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 12/14/2022] Open
Abstract
During exercise, muscle ATP demand increases with intensity, and at the highest power output, ATP consumption may increase more than 100-fold above the resting level. The rate of mitochondrial ATP production during exercise depends on the availability of O2, carbon substrates, reducing equivalents, ADP, Pi, free creatine, and Ca2+. It may also be modulated by acidosis, nitric oxide and reactive oxygen and nitrogen species (RONS). During fatiguing and repeated sprint exercise, RONS production may cause oxidative stress and damage to cellular structures and may reduce mitochondrial efficiency. Human studies indicate that the relatively low mitochondrial respiratory rates observed during sprint exercise are not due to lack of O2, or insufficient provision of Ca2+, reduced equivalents or carbon substrates, being a suboptimal stimulation by ADP the most plausible explanation. Recent in vitro studies with isolated skeletal muscle mitochondria, studied in conditions mimicking different exercise intensities, indicate that ROS production during aerobic exercise amounts to 1-2 orders of magnitude lower than previously thought. In this review, we will focus on the mechanisms regulating mitochondrial respiration, particularly during high-intensity exercise. We will analyze the factors that limit mitochondrial respiration and those that determine mitochondrial efficiency during exercise. Lastly, the differences in mitochondrial respiration between men and women will be addressed.
Collapse
Affiliation(s)
- Jose A L Calbet
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas de Gran Canaria, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" (s/n), 35017, Las Palmas de Gran Canaria, Canary Islands, Spain; Department of Physical Performance, The Norwegian School of Sport Sciences, Postboks, 4014 Ulleval Stadion, 0806 Oslo, Norway.
| | - Saúl Martín-Rodríguez
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas de Gran Canaria, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" (s/n), 35017, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - Marcos Martin-Rincon
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas de Gran Canaria, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" (s/n), 35017, Las Palmas de Gran Canaria, Canary Islands, Spain
| | - David Morales-Alamo
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas de Gran Canaria, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" (s/n), 35017, Las Palmas de Gran Canaria, Canary Islands, Spain
| |
Collapse
|
28
|
Dynnik VV, Grishina EV, Fedotcheva NI. The mitochondrial NO-synthase/guanylate cyclase/protein kinase G signaling system underpins the dual effects of nitric oxide on mitochondrial respiration and opening of the permeability transition pore. FEBS J 2019; 287:1525-1536. [PMID: 31602795 DOI: 10.1111/febs.15090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/07/2019] [Accepted: 10/09/2019] [Indexed: 12/30/2022]
Abstract
The available data on the involvement of nitric oxide (NO) and mitochondrial calcium-dependent NO synthase (mtNOS) in the control of mitochondrial respiration and the permeability transition pore (mPTP) are contradictory. We have proposed that the mitochondrial mtNOS/guanylate cyclase/protein kinase G signaling system (mtNOS-SS) is also implicated in the control of respiration and mPTP, providing the interplay between NO and mtNOS-SS, which, in turn, may result in inconsistent effects of NO. Therefore, using rat liver mitochondria, we applied specific inhibitors of the enzymes of this signaling system to evaluate its role in the control of respiration and mPTP opening. Steady-state respiration was supported by pyruvate, glutamate, or succinate in the presence of hexokinase, glucose, and ADP. When applied at low concentrations, l-arginine (to 500 µm) and NO donors (to 50 µm) activated the respiration and increased the threshold concentrations of calcium and d,l-palmitoylcarnitine required for the dissipation of the mitochondrial membrane potential and pore opening. Both effects were eliminated by the inhibitors of NO synthase, guanylate cyclase, and kinase G, which denotes the involvement of mtNOS-SS in the activation of respiration and deceleration of mPTP opening. At high concentrations, l-arginine and NO donors inhibited the respiration and promoted pore opening, indicating that adverse effects induced by an NO excess dominate over the protection provided by mtNOS-SS. Thus, these results demonstrate the opposite impact of NO and mtNOS-SS on the respiration and mPTP control, which can explain the dual effects of NO.
Collapse
Affiliation(s)
- Vladimir V Dynnik
- Department of Bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Russia
| | - Elena V Grishina
- Department of Bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Russia
| | - Nadezhda I Fedotcheva
- Department of Bioenergetics, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Russia
| |
Collapse
|
29
|
S-Nitrosylation: An Emerging Paradigm of Redox Signaling. Antioxidants (Basel) 2019; 8:antiox8090404. [PMID: 31533268 PMCID: PMC6769533 DOI: 10.3390/antiox8090404] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a highly reactive molecule, generated through metabolism of L-arginine by NO synthase (NOS). Abnormal NO levels in mammalian cells are associated with multiple human diseases, including cancer. Recent studies have uncovered that the NO signaling is compartmentalized, owing to the localization of NOS and the nature of biochemical reactions of NO, including S-nitrosylation. S-nitrosylation is a selective covalent post-translational modification adding a nitrosyl group to the reactive thiol group of a cysteine to form S-nitrosothiol (SNO), which is a key mechanism in transferring NO-mediated signals. While S-nitrosylation occurs only at select cysteine thiols, such a spatial constraint is partially resolved by transnitrosylation, where the nitrosyl moiety is transferred between two interacting proteins to successively transfer the NO signal to a distant location. As NOS is present in various subcellular locales, a stress could trigger concerted S-nitrosylation and transnitrosylation of a large number of proteins involved in divergent signaling cascades. S-nitrosylation is an emerging paradigm of redox signaling by which cells confer protection against oxidative stress.
Collapse
|
30
|
Román-Anguiano NG, Correa F, Cano-Martínez A, de la Peña-Díaz A, Zazueta C. Cardioprotective effects of Prolame and SNAP are related with nitric oxide production and with diminution of caspases and calpain-1 activities in reperfused rat hearts. PeerJ 2019; 7:e7348. [PMID: 31392096 PMCID: PMC6673759 DOI: 10.7717/peerj.7348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/25/2019] [Indexed: 01/26/2023] Open
Abstract
Cardiac tissue undergoes changes during ischemia-reperfusion (I-R) that compromise its normal function. Cell death is one of the consequences of such damage, as well as diminution in nitric oxide (NO) content. This signaling molecule regulates the function of the cardiovascular system through dependent and independent effects of cyclic guanosine monophosphate (cGMP). The independent cGMP pathway involves post-translational modification of proteins by S-nitrosylation. Studies in vitro have shown that NO inhibits the activity of caspases and calpains through S-nitrosylation of a cysteine located in their catalytic site, so we propose to elucidate if the regulatory mechanisms of NO are related with changes in S-nitrosylation of cell death proteins in the ischemic-reperfused myocardium. We used two compounds that increase the levels of NO by different mechanisms: Prolame, an amino-estrogenic compound with antiplatelet and anticoagulant effects that induces the increase of NO levels in vivo by activating the endothelial nitric oxide synthase (eNOS) and that has not been tested as a potential inhibitor of apoptosis. On the other hand, S-Nitroso-N-acetylpenicillamine (SNAP), a synthetic NO donor that has been shown to decrease cell death after inducing hypoxia-reoxygenation in cell cultures. Main experimental groups were Control, I-R, I-R+Prolame and I-R+SNAP. Additional groups were used to evaluate the NO action pathways. Contractile function represented as heart rate and ventricular pressure was evaluated in a Langendorff system. Infarct size was measured with 2,3,5-triphenyltetrazolium chloride stain. NO content was determined indirectly by measuring nitrite levels with the Griess reaction and cGMP content was measured by Enzyme-Linked ImmunoSorbent Assay. DNA integrity was evaluated by DNA laddering visualized on an agarose gel and by Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling assay. Activities of caspase-3, caspase-8, caspase-9 and calpain-1 were evaluated spectrophotometrically and the content of caspase-3 and calpain-1 by western blot. S-nitrosylation of caspase-3 and calpain-1 was evaluated by labeling S-nitrosylated cysteines. Our results show that both Prolame and SNAP increased NO content and improved functional recovery in post-ischemic hearts. cGMP-dependent and S-nitrosylation pathways were activated in both groups, but the cGMP-independent pathway was preferentially activated by SNAP, which induced higher levels of NO than Prolame. Although SNAP effectively diminished the activity of all the proteases, a correlative link between the activity of these proteases and S-nitrosylation was not fully established.
Collapse
Affiliation(s)
| | - Francisco Correa
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| | - Aurora de la Peña-Díaz
- Departamento de Biología Molecular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México.,Departamento de Farmacología, Universidad Nacional Autónoma de México, México, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiologia Ignacio Chávez, México, México
| |
Collapse
|
31
|
Thomas NO, Shay KP, Hagen TM. Age-related loss of mitochondrial glutathione exacerbates menadione-induced inhibition of Complex I. Redox Biol 2019; 22:101155. [PMID: 30851669 PMCID: PMC6406584 DOI: 10.1016/j.redox.2019.101155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
The role of mitochondrial GSH (mGSH) in the enhanced age-related susceptibility to xenobiotic toxicity is not well defined. We determined mGSH status and indices of mitochondrial bioenergetics in hepatocytes from young and old F344 rats treated with 300 μM menadione, a concentration that causes 50% cell death in old. At this concentration, mGSH was significantly lost only in hepatocytes from old rats, and with near total depletion due to lower basal mGSH in aged cells. In old hepatocytes, menadione caused mitochondrial membrane potential to collapse, as well as significant deficits in maximal O2 consumption and respiratory reserve capacity, indicators of cellular bioenergetic resiliency. Further examination revealed that the menadione-mediated loss of respiratory reserve capacity in aged hepatocytes was from significant inhibition of Complex I activity and increased proton leak, for which an increase in Complex II activity was not able to compensate. These data demonstrate an age-related increase in mitochondrial susceptibility to a redox-cycling challenge, particularly in regards to Complex I activity, and provide a plausible mechanism to link this vulnerability to mGSH perturbations.
Collapse
Affiliation(s)
- Nicholas O Thomas
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331-6512, USA; Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, 97331-7305, USA
| | - Kate P Shay
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331-6512, USA
| | - Tory M Hagen
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331-6512, USA; Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, 97331-7305, USA.
| |
Collapse
|
32
|
Saletti R, Reina S, Pittalà MG, Magrì A, Cunsolo V, Foti S, De Pinto V. Post-translational modifications of VDAC1 and VDAC2 cysteines from rat liver mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:806-816. [DOI: 10.1016/j.bbabio.2018.06.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 12/14/2022]
|
33
|
Role of Nitric Oxide and Hydrogen Sulfide in Ischemic Stroke and the Emergent Epigenetic Underpinnings. Mol Neurobiol 2018; 56:1749-1769. [PMID: 29926377 DOI: 10.1007/s12035-018-1141-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 05/22/2018] [Indexed: 02/06/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are the key gasotransmitters with an imperious role in the maintenance of cerebrovascular homeostasis. A decline in their levels contributes to endothelial dysfunction that portends ischemic stroke (IS) or cerebral ischemia/reperfusion (CI/R). Nevertheless, their exorbitant production during CI/R is associated with exacerbation of cerebrovascular injury in the post-stroke epoch. NO-producing nitric oxide synthases are implicated in IS pathology and their activity is regulated, inter alia, by various post-translational modifications and chromatin-based mechanisms. These account for heterogeneous alterations in NO production in a disease setting like IS. Interestingly, NO per se has been posited as an endogenous epigenetic modulator. Further, there is compelling evidence for an ingenious crosstalk between NO and H2S in effecting the canonical (direct) and non-canonical (off-target collateral) functions. In this regard, NO-mediated S-nitrosylation and H2S-mediated S-sulfhydration of specific reactive thiols in an expanding array of target proteins are the principal modalities mediating the all-pervasive influence of NO and H2S on cell fate in an ischemic brain. An integrated stress response subsuming unfolded protein response and autophagy to cellular stressors like endoplasmic reticulum stress, in part, is entrenched in such signaling modalities that substantiate the role of NO and H2S in priming the cells for stress response. The precis presented here provides a comprehension on the multifarious actions of NO and H2S and their epigenetic underpinnings, their crosstalk in maintenance of cerebrovascular homeostasis, and their "Janus bifrons" effect in IS milieu together with plausible therapeutic implications.
Collapse
|
34
|
Nitric Oxide and Mitochondrial Function in Neurological Diseases. Neuroscience 2018; 376:48-71. [DOI: 10.1016/j.neuroscience.2018.02.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/20/2018] [Accepted: 02/09/2018] [Indexed: 12/17/2022]
|
35
|
Wilson RJ, Drake JC, Cui D, Lewellen BM, Fisher CC, Zhang M, Kashatus DF, Palmer LA, Murphy MP, Yan Z. Mitochondrial protein S-nitrosation protects against ischemia reperfusion-induced denervation at neuromuscular junction in skeletal muscle. Free Radic Biol Med 2018; 117:180-190. [PMID: 29432799 PMCID: PMC5896769 DOI: 10.1016/j.freeradbiomed.2018.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 01/08/2023]
Abstract
Deterioration of neuromuscular junction (NMJ) integrity and function is causal to muscle atrophy and frailty, ultimately hindering quality of life and increasing the risk of death. In particular, NMJ is vulnerable to ischemia reperfusion (IR) injury when blood flow is restricted followed by restoration. However, little is known about the underlying mechanism(s) and hence the lack of effective interventions. New evidence suggests that mitochondrial oxidative stress plays a causal role in IR injury, which can be precluded by enhancing mitochondrial protein S-nitrosation (SNO). To elucidate the role of IR and mitochondrial protein SNO in skeletal muscle, we utilized a clinically relevant model and showed that IR resulted in significant muscle and motor nerve injuries with evidence of elevated muscle creatine kinase in the serum, denervation at NMJ, myofiber degeneration and regeneration, as well as muscle atrophy. Interestingly, we observed that neuromuscular transmission improved prior to muscle recovery, suggesting the importance of the motor nerve in muscle functional recovery. Injection of a mitochondria-targeted S-nitrosation enhancing agent, MitoSNO, into ischemic muscle prior to reperfusion reduced mitochondrial oxidative stress in the motor nerve and NMJ, attenuated denervation at NMJ, and resulted in accelerated functional recovery of the muscle. These findings demonstrate that enhancing mitochondrial protein SNO protects against IR-induced denervation at NMJ in skeletal muscle and accelerates functional regeneration. This could be an efficacious intervention for protecting neuromuscular injury under the condition of IR and other related pathological conditions.
Collapse
Affiliation(s)
- Rebecca J Wilson
- Departments of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Joshua C Drake
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Di Cui
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Bevan M Lewellen
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Carleigh C Fisher
- Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Mei Zhang
- Departments of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - David F Kashatus
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Lisa A Palmer
- Departments of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Zhen Yan
- Departments of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Departments of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Departments of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Center for Skeletal Muscle Research at Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
36
|
Mitochondria-Bound Hexokinase (mt-HK) Activity Differ in Cortical and Hypothalamic Synaptosomes: Differential Role of mt-HK in H 2O 2 Depuration. Mol Neurobiol 2017; 55:5889-5900. [PMID: 29119535 DOI: 10.1007/s12035-017-0807-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/18/2017] [Indexed: 01/06/2023]
Abstract
Glucose and oxygen are vital for the brain, as these molecules provide energy and metabolic intermediates that are necessary for cell function. The glycolysis pathway and mitochondria play a pivotal role in cell energy metabolism, which is closely related to reactive oxygen species (ROS) production. Hexokinase (HK) is a key enzyme involved in glucose metabolism that modulates the level of brain mitochondrial ROS by recycling ADP for oxidative phosphorylation (OxPhos). Here, we hypothesize that the control of mitochondrial metabolism by hexokinase differs in distinct areas of the brain, such as the cortex and hypothalamus, in which ROS might function as signaling molecules. Thus, we investigated mitochondrial metabolism of synaptosomes derived from both brain regions. Cortical synaptosomes (CSy) show a predominance of glutamatergic synapses, while in the hypothalamic synaptosomes (HSy), the GABAergic synapses predominate. Significant differences of oxygen consumption and ROS production were related to higher mitochondrial complex II activity (succinate dehydrogenase-SDH) in CSy rather than to mitochondrial number. Mitochondrial HK (mt-HK) activity was higher in CSy than in HSy regardless the substrate added. Mitochondrial O2 consumption related to mt-HK activation by 2-deoxyglucose was also higher in CSy. In the presence of substrate for complex II, the activation of synaptosomal mt-HK promoted depuration of ROS in both HSy and CSy, while ROS depuration did not occur in HSy when substrate for complex I was used. The impact of the mt-HK inhibition by glucose-6-phosphate (G6P) was the same in synaptosomes from both areas. Together, the differences found between CSy and HSy indicate specific roles of mt-HK and SDH on the metabolism of each brain region, what probably depends on the main metabolic route that is used by the neurons.
Collapse
|
37
|
Increased Root Canal Endotoxin Levels are Associated with Chronic Apical Periodontitis, Increased Oxidative and Nitrosative Stress, Major Depression, Severity of Depression, and a Lowered Quality of Life. Mol Neurobiol 2017; 55:2814-2827. [PMID: 28455694 DOI: 10.1007/s12035-017-0545-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/07/2017] [Indexed: 10/19/2022]
Abstract
Evidence indicates that major depression is accompanied by increased translocation of gut commensal Gram-negative bacteria (leaky gut) and consequent activation of oxidative and nitrosative (O&NS) pathways. This present study examined the associations among chronic apical periodontitis (CAP), root canal endotoxin levels (lipopolysaccharides, LPS), O&NS pathways, depressive symptoms, and quality of life. Measurements included advanced oxidation protein products (AOPP), nitric oxide metabolites (NOx), lipid peroxides (LOOH), -sulfhydryl (SH) groups, total radical trapping antioxidant parameter (TRAP), and paraoxonase (PON)1 activity in participants with CAP, with and without depression, as well as healthy controls (no depression, no CAP). Root canal LPS levels were positively associated with CAP, clinical depression, severity of depression (as measured with the Hamilton Depression Rating Scale (HDRS) and the Beck Depression Inventory) and O&NS biomarkers, especially NOx and TRAP. CAP-related depression was accompanied by increased levels of NOx, LOOH, AOPP, and TRAP. In CAP participants, there was a strong correlation (r = 0.734, p < 0.001) between root canal LPS and the HDRS score. There were significant and positive associations between CAP or root canal endotoxin with the vegetative and physio-somatic symptoms of the HDRS as well as a significant inverse association between root canal endotoxin and quality of life with strong effects on psychological, environmental, and social domains. It is concluded that increased root canal LPS accompanying CAP may cause depression and a lowered quality of life, which may be partly explained by activated O&NS pathways, especially NOx thereby enhancing hypernitrosylation and thus neuroprogressive processes. Dental health and "leaky teeth" may be intimately linked to the etiology and course of depression, while significantly impacting quality of life.
Collapse
|
38
|
Galkin A, Moncada S. Modulation of the conformational state of mitochondrial complex I as a target for therapeutic intervention. Interface Focus 2017; 7:20160104. [PMID: 28382200 DOI: 10.1098/rsfs.2016.0104] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In recent years, there have been significant advances in our understanding of the functions of mitochondrial complex I other than the generation of energy. These include its role in generation of reactive oxygen species, involvement in the hypoxic tissue response and its possible regulation by nitric oxide (NO) metabolites. In this review, we will focus on the hypoxic conformational change of this mitochondrial enzyme, the so-called active/deactive transition. This conformational change is physiological and relevant to the understanding of certain pathological conditions including, in the cardiovascular system, ischaemia/reperfusion (I/R) damage. We will discuss how complex I can be affected by NO metabolites and will outline some potential mitochondria-targeted therapies in I/R damage.
Collapse
Affiliation(s)
- Alexander Galkin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 401 East 61st Street, 5th floor, New York, NY 10065, USA; Queens University Belfast, School of Biological Sciences, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Salvador Moncada
- Manchester Cancer Research Centre , University of Manchester , Wilmslow Road, Manchester M20 4QL , UK
| |
Collapse
|
39
|
Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev 2017; 74:1-20. [PMID: 28093238 DOI: 10.1016/j.neubiorev.2017.01.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
40
|
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: 113] [Impact Index Per Article: 12.6] [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
|
41
|
Morris G, Berk M, Klein H, Walder K, Galecki P, Maes M. Nitrosative Stress, Hypernitrosylation, and Autoimmune Responses to Nitrosylated Proteins: New Pathways in Neuroprogressive Disorders Including Depression and Chronic Fatigue Syndrome. Mol Neurobiol 2016; 54:4271-4291. [PMID: 27339878 DOI: 10.1007/s12035-016-9975-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/13/2016] [Indexed: 12/30/2022]
Abstract
Nitric oxide plays an indispensable role in modulating cellular signaling and redox pathways. This role is mainly effected by the readily reversible nitrosylation of selective protein cysteine thiols. The reversibility and sophistication of this signaling system is enabled and regulated by a number of enzymes which form part of the thioredoxin, glutathione, and pyridoxine antioxidant systems. Increases in nitric oxide levels initially lead to a defensive increase in the number of nitrosylated proteins in an effort to preserve their function. However, in an environment of chronic oxidative and nitrosative stress (O&NS), nitrosylation of crucial cysteine groups within key enzymes of the thioredoxin, glutathione, and pyridoxine systems leads to their inactivation thereby disabling denitrosylation and transnitrosylation and subsequently a state described as "hypernitrosylation." This state leads to the development of pathology in multiple domains such as the inhibition of enzymes of the electron transport chain, decreased mitochondrial function, and altered conformation of proteins and amino acids leading to loss of immune tolerance and development of autoimmunity. Hypernitrosylation also leads to altered function or inactivation of proteins involved in the regulation of apoptosis, autophagy, proteomic degradation, transcription factor activity, immune-inflammatory pathways, energy production, and neural function and survival. Hypernitrosylation, as a consequence of chronically elevated O&NS and activated immune-inflammatory pathways, can explain many characteristic abnormalities observed in neuroprogressive disease including major depression and chronic fatigue syndrome/myalgic encephalomyelitis. In those disorders, increased bacterial translocation may drive hypernitrosylation and autoimmune responses against nitrosylated proteins.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, SA152LW, Wales, UK
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
- Orygen Youth Health Research Centre and the Centre of Youth Mental Health, Poplar Road 35, Parkville, 3052, Australia
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Kenneth Myer Building, Royal Parade 30, Parkville, 3052, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Level 1 North, Main Block, Parkville, 3052, Australia
| | - Hans Klein
- Department of Psychiatry, University of Groningen, UMCG, Groningen, The Netherlands
| | - Ken Walder
- Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds, Australia
| | - Piotr Galecki
- Department of Adult Psychiatry, Medical University of Lodz, Łódź, Poland
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Department of Psychiatry, Faculty of Medicine, State University of Londrina, Londrina, Brazil.
- Department of Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria.
- Revitalis, Waalre, The Netherlands.
- IMPACT Strategic Research Center, Barwon Health, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
42
|
Dröse S, Stepanova A, Galkin A. Ischemic A/D transition of mitochondrial complex I and its role in ROS generation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:946-57. [PMID: 26777588 PMCID: PMC4893024 DOI: 10.1016/j.bbabio.2015.12.013] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/11/2015] [Accepted: 12/25/2015] [Indexed: 12/12/2022]
Abstract
Mitochondrial complex I (NADH:ubiquinone oxidoreductase) is a key enzyme in cellular energy metabolism and provides approximately 40% of the proton-motive force that is utilized during mitochondrial ATP production. The dysregulation of complex I function – either genetically, pharmacologically, or metabolically induced – has severe pathophysiological consequences that often involve an imbalance in the production of reactive oxygen species (ROS). Slow transition of the active (A) enzyme to the deactive, dormant (D) form takes place during ischemia in metabolically active organs such as the heart and brain. The reactivation of complex I occurs upon reoxygenation of ischemic tissue, a process that is usually accompanied by an increase in cellular ROS production. Complex I in the D-form serves as a protective mechanism preventing the oxidative burst upon reperfusion. Conversely, however, the D-form is more vulnerable to oxidative/nitrosative damage. Understanding the so-called active/deactive (A/D) transition may contribute to the development of new therapeutic interventions for conditions like stroke, cardiac infarction, and other ischemia-associated pathologies. In this review, we summarize current knowledge on the mechanism of A/D transition of mitochondrial complex I considering recently available structural data and site-specific labeling experiments. In addition, this review discusses in detail the impact of the A/D transition on ROS production by complex I and the S-nitrosation of a critical cysteine residue of subunit ND3 as a strategy to prevent oxidative damage and tissue damage during ischemia–reperfusion injury. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt. The current knowledge on active/deactive (A/D) transition of complex I is reviewed. The mechanism and driving force of the A/D conformational change are discussed. The A/D transition can affect ROS production and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Stefan Dröse
- Clinic of Anesthesiology, Intensive-Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main 60590, Germany.
| | - Anna Stepanova
- Medical Biology Centre, School of Biological Sciences, Queens University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Alexander Galkin
- Medical Biology Centre, School of Biological Sciences, Queens University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 407 East 61st Street, New York, NY 10065, USA.
| |
Collapse
|
43
|
Wang Y, Zhou Z, Leylek T, Tan H, Sun Y, Parkinson F, Wang JF. Protein cysteine S-nitrosylation inhibits vesicular uptake of neurotransmitters. Neuroscience 2015; 311:374-81. [DOI: 10.1016/j.neuroscience.2015.10.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/25/2015] [Accepted: 10/28/2015] [Indexed: 12/31/2022]
|
44
|
Banerjee S, Melnyk SB, Krager KJ, Aykin-Burns N, Letzig LG, James LP, Hinson JA. The neuronal nitric oxide synthase inhibitor NANT blocks acetaminophen toxicity and protein nitration in freshly isolated hepatocytes. Free Radic Biol Med 2015; 89:750-7. [PMID: 26454079 PMCID: PMC5012542 DOI: 10.1016/j.freeradbiomed.2015.09.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 11/23/2022]
Abstract
3-Nitrotyrosine (3NT) in liver proteins of mice treated with hepatotoxic doses of acetaminophen (APAP) has been postulated to be causative in toxicity. Nitration is by a reactive nitrogen species formed from nitric oxide (NO). The source of the NO is unclear. iNOS knockout mice were previously found to be equally susceptible to APAP toxicity as wildtype mice and iNOS inhibitors did not decrease toxicity in mice or in hepatocytes. In this work we examined the potential role of nNOS in APAP toxicity in hepatocytes using the specific nNOS inhibitor NANT (10 µM)(N-[(4S)-4-amino-5-[(2-aminoethyl)amino]pentyl]-N'-nitroguanidinetris (trifluoroacetate)). Primary hepatocytes (1 million/ml) from male B6C3F1 mice were incubated with APAP (1mM). Cells were removed and assayed spectrofluorometrically for reactive nitrogen and oxygen species using diaminofluorescein (DAF) and Mitosox red, respectively. Cytotoxicity was determined by LDH release into media. Glutathione (GSH, GSSG), 3NT, GSNO, acetaminophen-cysteine adducts, NAD, and NADH were measured by HPLC. APAP significantly increased cytotoxicity at 1.5-3.0 h. The increase was blocked by NANT. NANT did not alter APAP mediated GSH depletion or acetaminophen-cysteine adducts in proteins which indicated that NANT did not inhibit metabolism. APAP significantly increased spectroflurometric evidence of reactive nitrogen and oxygen formation at 0.5 and 1.0 h, respectively, and increased 3NT and GSNO at 1.5-3.0 h. These increases were blocked by NANT. APAP dramatically increased NADH from 0.5-3.0 h and this increase was blocked by NANT. Also, APAP decreased the Oxygen Consumption Rate (OCR), decreased ATP production, and caused a loss of mitochondrial membrane potential, which were all blocked by NANT.
Collapse
Affiliation(s)
- Sudip Banerjee
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences Little Rock, AR 72205
| | - Stepan B Melnyk
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, AR 72205
| | - Kimberly J Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205
| | - Lynda G Letzig
- Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, AR 72205
| | - Laura P James
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences Little Rock, AR 72205; Department of Pediatrics, Arkansas Children's Hospital Research Institute, Little Rock, AR 72205
| | - Jack A Hinson
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences Little Rock, AR 72205.
| |
Collapse
|
45
|
Okazaki M, Kurabayashi K, Asanuma M, Saito Y, Dodo K, Sodeoka M. VDAC3 gating is activated by suppression of disulfide-bond formation between the N-terminal region and the bottom of the pore. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:3188-96. [PMID: 26407725 DOI: 10.1016/j.bbamem.2015.09.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 09/09/2015] [Accepted: 09/18/2015] [Indexed: 12/30/2022]
Abstract
The voltage-dependent anion channels (VDACs), VDAC1, VDAC2, and VDAC3, are pore-forming proteins that control metabolite flux between mitochondria and cytoplasm. VDAC1 and VDAC2 have voltage-dependent gating activity, whereas VDAC3 is thought to have weak activity. The aim of this study was to analyze the channel properties of all three human VDAC isoforms and to clarify the channel function of VDAC3. Bacterially expressed recombinant human VDAC proteins were reconstituted into artificial planar lipid bilayers and their gating activities were evaluated. VDAC1 and VDAC2 had typical voltage-dependent gating activity, whereas the gating of VDAC3 was weak, as reported. However, gating of VDAC3 was evoked by dithiothreitol (DTT) and S-nitrosoglutathione (GSNO), which are thought to suppress disulfide-bond formation. Several cysteine mutants of VDAC3 also exhibited typical voltage-gating. Our results indicate that channel gating was induced by reduction of a disulfide-bond linking the N-terminal region to the bottom of the pore. Thus, channel gating of VDAC3 might be controlled by redox sensing under physiological conditions.
Collapse
Affiliation(s)
- Masateru Okazaki
- Sodeoka Live Cell Chemistry Project, ERATO, JST, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Synthetic Organic Chemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Katsue Kurabayashi
- Sodeoka Live Cell Chemistry Project, ERATO, JST, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Synthetic Organic Chemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Miwako Asanuma
- Sodeoka Live Cell Chemistry Project, ERATO, JST, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Synthetic Organic Chemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yohei Saito
- Sodeoka Live Cell Chemistry Project, ERATO, JST, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Synthetic Organic Chemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kosuke Dodo
- Sodeoka Live Cell Chemistry Project, ERATO, JST, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Synthetic Organic Chemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mikiko Sodeoka
- Sodeoka Live Cell Chemistry Project, ERATO, JST, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Synthetic Organic Chemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
46
|
de Lima Portella R, Lynn Bickta J, Shiva S. Nitrite Confers Preconditioning and Cytoprotection After Ischemia/Reperfusion Injury Through the Modulation of Mitochondrial Function. Antioxid Redox Signal 2015; 23:307-27. [PMID: 26094636 DOI: 10.1089/ars.2015.6260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Nitrite is now recognized as an intrinsic signaling molecule that mediates a number of biological processes. One of the most reproducible effects of nitrite is its ability to mediate cytoprotection after ischemia/reperfusion (I/R). This robust phenomenon has been reproduced by a number of investigators in varying animal models focusing on different target organs. Furthermore, nitrite's cytoprotective versatility is highlighted by its ability to mediate delayed preconditioning and remote conditioning in addition to acute protection. RECENT ADVANCES In the last 10 years, significant progress has been made in elucidating the mechanisms underlying nitrite-mediated ischemic tolerance. CRITICAL ISSUES The mitochondrion, which is essential to both the progression of I/R injury and the protection afforded by preconditioning, has emerged as a major subcellular target for nitrite. This review will outline the role of the mitochondrion in I/R injury and preconditioning, review the accumulated preclinical studies demonstrating nitrite-mediated cytoprotection, and finally focus on the known interactions of nitrite with mitochondria and their role in the mechanism of nitrite-mediated ischemic tolerance. FUTURE DIRECTIONS These studies set the stage for current clinical trials testing the efficacy of nitrite to prevent warm and cold I/R injury.
Collapse
Affiliation(s)
- Rafael de Lima Portella
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Janelle Lynn Bickta
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Abstract
The cysteine (Cys) proteome is a major component of the adaptive interface between the genome and the exposome. The thiol moiety of Cys undergoes a range of biologic modifications enabling biological switching of structure and reactivity. These biological modifications include sulfenylation and disulfide formation, formation of higher oxidation states, S-nitrosylation, persulfidation, metalation, and other modifications. Extensive knowledge about these systems and their compartmentalization now provides a foundation to develop advanced integrative models of Cys proteome regulation. In particular, detailed understanding of redox signaling pathways and sensing networks is becoming available to allow the discrimination of network structures. This research focuses attention on the need for atlases of Cys modifications to develop systems biology models. Such atlases will be especially useful for integrative studies linking the Cys proteome to imaging and other omics platforms, providing a basis for improved redox-based therapeutics. Thus, a framework is emerging to place the Cys proteome as a complement to the quantitative proteome in the omics continuum connecting the genome to the exposome.
Collapse
Affiliation(s)
- Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Joshua D Chandler
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
48
|
Sun J, Nguyen T, Aponte AM, Menazza S, Kohr MJ, Roth DM, Patel HH, Murphy E, Steenbergen C. Ischaemic preconditioning preferentially increases protein S-nitrosylation in subsarcolemmal mitochondria. Cardiovasc Res 2015; 106:227-36. [PMID: 25694588 DOI: 10.1093/cvr/cvv044] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/08/2015] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) and protein S-nitrosylation (SNO) have been shown to play important roles in ischaemic preconditioning (IPC)-induced acute cardioprotection. The majority of proteins that show increased SNO following IPC are localized to the mitochondria, and our recent studies suggest that caveolae transduce acute NO/SNO cardioprotective signalling in IPC hearts. Due to the close association between subsarcolemmal mitochondria (SSM) and the sarcolemma/caveolae, we tested the hypothesis that SSM, rather than the interfibrillar mitochondria (IFM), are major targets for NO/SNO signalling derived from caveolae-associated eNOS. Following either control perfusion or IPC, SSM and IFM were isolated from Langendorff perfused mouse hearts, and SNO was analysed using a modified biotin switch method with fluorescent maleimide fluors. In perfusion control hearts, the SNO content was higher in SSM compared with IFM (1.33 ± 0.19, ratio of SNO content Perf-SSM vs. Perf-IFM), and following IPC SNO content significantly increased preferentially in SSM, but not in IFM (1.72 ± 0.17 and 1.07 ± 0.04, ratio of SNO content IPC-SSM vs. Perf-IFM, and IPC-IFM vs. Perf-IFM, respectively). Consistent with these findings, eNOS, caveolin-3, and connexin-43 were detected in SSM, but not in IFM, and IPC resulted in a further significant increase in eNOS/caveolin-3 levels in SSM. Interestingly, we did not observe an IPC-induced increase in SNO or eNOS/caveolin-3 in SSM isolated from caveolin-3(-/-) mouse hearts, which could not be protected with IPC. In conclusion, these results suggest that SSM may be the preferential target of sarcolemmal signalling-derived post-translational protein modification (caveolae-derived eNOS/NO/SNO), thus providing an important role in IPC-induced cardioprotection.
Collapse
Affiliation(s)
- Junhui Sun
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Tiffany Nguyen
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Angel M Aponte
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA Proteomics Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sara Menazza
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Mark J Kohr
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - David M Roth
- Department of Anesthesiology, VA San Diego Healthcare System and University of California at San Diego, La Jolla, CA 92093, USA
| | - Hemal H Patel
- Department of Anesthesiology, VA San Diego Healthcare System and University of California at San Diego, La Jolla, CA 92093, USA
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Bldg10/Rm8N206, Bethesda, MD 20892, USA
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| |
Collapse
|
49
|
Skeff MA, Brito GAC, de Oliveira MG, Braga CM, Cavalcante MM, Baldim V, Holanda-Afonso RC, Silva-Boghossian CM, Colombo AP, Ribeiro RA, Moura-Neto V, Leitão RFC. S-nitrosoglutathione accelerates recovery from 5-fluorouracil-induced oral mucositis. PLoS One 2014; 9:e113378. [PMID: 25478918 PMCID: PMC4257535 DOI: 10.1371/journal.pone.0113378] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/16/2014] [Indexed: 11/19/2022] Open
Abstract
Introduction Mucositis induced by anti-neoplastic drugs is an important, dose-limiting and costly side-effect of cancer therapy. Aim To evaluate the effect of the topical application of S-nitrosoglutathione (GSNO), a nitric oxide donor, on 5-fluorouracil (5-FU)-induced oral mucositis in hamsters. Materials and Methods Oral mucositis was induced in male hamsters by two intraperitoneal administrations of 5-FU on the first and second days of the experiment (60 and 40 mg/kg, respectively) followed by mechanical trauma on the fourth day. Animals received saline, HPMC or HPMC/GSNO (0.1, 0.5 or 2.0 mM) 1 h prior to the 5-FU injection and twice a day for 10 or 14 days. Samples of cheek pouches were harvested for: histopathological analysis, TNF-α and IL-1β levels, immunohistochemical staining for iNOS, TNF-α, IL-1β, Ki67 and TGF-β RII and a TUNEL assay. The presence and levels of 39 bacterial taxa were analyzed using the Checkerboard DNA-DNA hybridization method. The profiles of NO released from the HPMC/GSNO formulations were characterized using chemiluminescence. Results The HPMC/GSNO formulations were found to provide sustained release of NO for more than 4 h at concentration-dependent rates of 14 to 80 nmol/mL/h. Treatment with HPMC/GSNO (0.5 mM) significantly reduced mucosal damage, inflammatory alterations and cell death associated with 5-FU-induced oral mucositis on day 14 but not on day 10. HPMC/GSNO administration also reversed the inhibitory effect of 5-FU on cell proliferation on day 14. In addition, we observed that the chemotherapy significantly increased the levels and/or prevalence of several bacterial species. Conclusion Topical HPMC/GSNO accelerates mucosal recovery, reduces inflammatory parameters, speeds up re-epithelization and decreases levels of periodontopathic species in mucosal ulcers.
Collapse
Affiliation(s)
- Maria Adriana Skeff
- Laboratory of Cell Morphogenesis, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Gerly A. C. Brito
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Cintia M. Braga
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Matheus M. Cavalcante
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Victor Baldim
- Institute of Chemistry, University of Campinas, UNICAMP, Campinas, SP, Brazil
| | - Rosenilde C. Holanda-Afonso
- Laboratory of Cell Morphogenesis, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carina M. Silva-Boghossian
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Faculty of Dentistry, University of Grande Rio, Duque de Caxias, RJ, Brazil
| | - Ana Paula Colombo
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ronaldo A. Ribeiro
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Vivaldo Moura-Neto
- Laboratory of Cell Morphogenesis, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, RJ, Brazil
| | - Renata F. C. Leitão
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
- * E-mail:
| |
Collapse
|