1
|
Singh I, Kim J, Touhidul Islam SM, Fei Q, Singh AK, Won J. The role of S-nitrosoglutathione reductase (GSNOR) in T cell-mediated immunopathology of experimental autoimmune encephalomyelitis (EAE). Neuroscience 2025; 564:1-12. [PMID: 39532197 DOI: 10.1016/j.neuroscience.2024.11.025] [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: 06/27/2024] [Revised: 10/14/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Previously, we reported that both S-nitrosoglutathione (GSNO), a carrier of cellular nitric oxide, and N6022, an injectable form of GSNO reductase (GSNOR) inhibitor that increases endogenous GSNO levels, alleviate experimental autoimmune encephalomyelitis (EAE) in mice by suppressing Th1 and Th17 immune responses. Building on these findings, we explored the role of GSNOR in EAE pathogenesis and evaluated the efficacy of an orally active GSNOR inhibitor (N91115) in treating the EAE disease. EAE mice exhibited heightened expression/activity of GSNOR in the spinal cord, and the knockout of the GSNOR gene resulted in much milder clinical manifestations of EAE, with lower degrees of demyelination and axonal loss, reduced microglial and astrocyte activations, as well as suppressed Th1 and Th17 cell responses, alongside bolstered Treg immune responses. Next, we evaluated the efficacy of N91115 against EAE immunopathology. Consistent with our findings in GSNOR deficient EAE mice, daily N91115 administration reduced clinical EAE severity, with less spinal cord demyelination and axonal loss compared to untreated EAE mice. Furthermore, N91115 treated EAE mice showed diminished Th1 and Th17 immune responses and enhanced Treg responses. This observation underscores the potential of increased GSNOR expression and activity as a risk factor exacerbating EAE immunopathology, while simultaneously highlighting its potential as a target for modifying the disease. Furthermore, the balanced immune regulation provided by orally active N91115 (IL-6/IL-17a vs. IL-10) presents a promising alternative to immunosuppressive drugs, reducing the risk of opportunistic infections.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Spinal Cord/immunology
- Mice, Inbred C57BL
- Female
- Mice, Knockout
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Mice
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Aldehyde Oxidoreductases/metabolism
- Aldehyde Oxidoreductases/antagonists & inhibitors
- Microglia/drug effects
- Microglia/metabolism
- Microglia/pathology
- Alcohol Dehydrogenase
Collapse
Affiliation(s)
- Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| | - Judong Kim
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Qiao Fei
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
2
|
Mazuryk O, Gurgul I, Oszajca M, Polaczek J, Kieca K, Bieszczad-Żak E, Martyka T, Stochel G. Nitric Oxide Signaling and Sensing in Age-Related Diseases. Antioxidants (Basel) 2024; 13:1213. [PMID: 39456466 PMCID: PMC11504650 DOI: 10.3390/antiox13101213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Nitric oxide (NO) is a key signaling molecule involved in numerous physiological and pathological processes within the human body. This review specifically examines the involvement of NO in age-related diseases, focusing on the cardiovascular, nervous, and immune systems. The discussion delves into the mechanisms of NO signaling in these diseases, emphasizing the post-translational modifications of involved proteins, such as S-nitrosation and nitration. The review also covers the dual nature of NO, highlighting both its protective and harmful effects, determined by concentration, location, and timing. Additionally, potential therapies that modulate NO signaling, including the use of NO donors and nitric oxide synthases (NOSs) inhibitors in the treatment of cardiovascular, neurodegenerative, and oncological diseases, are analyzed. Particular attention is paid to the methods for the determination of NO and its derivatives in the context of illness diagnosis and monitoring. The review underscores the complexity and dual role of NO in maintaining cellular balance and suggests areas for future research in developing new therapeutic strategies.
Collapse
Affiliation(s)
- Olga Mazuryk
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Ilona Gurgul
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Maria Oszajca
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Justyna Polaczek
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Konrad Kieca
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ewelina Bieszczad-Żak
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Tobiasz Martyka
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Grażyna Stochel
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| |
Collapse
|
3
|
Pawluk H, Tafelska-Kaczmarek A, Sopońska M, Porzych M, Modrzejewska M, Pawluk M, Kurhaluk N, Tkaczenko H, Kołodziejska R. The Influence of Oxidative Stress Markers in Patients with Ischemic Stroke. Biomolecules 2024; 14:1130. [PMID: 39334896 PMCID: PMC11430825 DOI: 10.3390/biom14091130] [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: 06/25/2024] [Revised: 07/27/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Stroke is the second leading cause of death worldwide, and its incidence is rising rapidly. Acute ischemic stroke is a subtype of stroke that accounts for the majority of stroke cases and has a high mortality rate. An effective treatment for stroke is to minimize damage to the brain's neural tissue by restoring blood flow to decreased perfusion areas of the brain. Many reports have concluded that both oxidative stress and excitotoxicity are the main pathological processes associated with ischemic stroke. Current measures to protect the brain against serious damage caused by stroke are insufficient. For this reason, it is important to investigate oxidative and antioxidant strategies to reduce oxidative damage. This review focuses on studies assessing the concentration of oxidative stress biomarkers and the level of antioxidants (enzymatic and non-enzymatic) and their impact on the clinical prognosis of patients after stroke. Mechanisms related to the production of ROS/RNS and the role of oxidative stress in the pathogenesis of ischemic stroke are presented, as well as new therapeutic strategies aimed at reducing the effects of ischemia and reperfusion.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Agnieszka Tafelska-Kaczmarek
- Department of Organic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100 Torun, Poland
| | - Małgorzata Sopońska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Marta Porzych
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Martyna Modrzejewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Mateusz Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| |
Collapse
|
4
|
Wu J, Jia J, Ji D, Jiao W, Huang Z, Zhang Y. Advances in nitric oxide regulators for the treatment of ischemic stroke. Eur J Med Chem 2023; 262:115912. [PMID: 37931330 DOI: 10.1016/j.ejmech.2023.115912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/14/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023]
Abstract
Ischemic stroke (IS) is a life-threatening disease worldwide. Nitric oxide (NO) derived from l-arginine catalyzed by NO synthase (NOS) is closely associated with IS. Three isomers of NOS (nNOS, eNOS and iNOS) produce different concentrations of NO, resulting in quite unlike effects during IS. Of them, n/iNOSs generate high levels of NO, detrimental to brain by causing nerve cell apoptosis and/or necrosis, whereas eNOS releases small amounts of NO, beneficial to the brain via increasing cerebral blood flow and improving nerve function. As a result, a large variety of NO regulators (NO donors or n/iNOS inhibitors) have been developed for fighting IS. Regrettably, up to now, no review systematically introduces the progresses in this area. This article first outlines dynamic variation rule of NOS/NO in IS, subsequently highlights advances in NO regulators against IS, and finally presents perspectives based on concentration-, site- and timing-effects of NO production to promote this field forward.
Collapse
Affiliation(s)
- Jianbing Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, China
| | - Jian Jia
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, China; Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai, 201203, China
| | - Duorui Ji
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, China
| | - Weijie Jiao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
5
|
Kim J, Qiao F, Singh AK, Won J, Singh I. Efficacies of S-nitrosoglutathione (GSNO) and GSNO reductase inhibitor in SARS-CoV-2 spike protein induced acute lung disease in mice. Front Pharmacol 2023; 14:1304697. [PMID: 38143504 PMCID: PMC10748393 DOI: 10.3389/fphar.2023.1304697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which initially surfaced in late 2019, often triggers severe pulmonary complications, encompassing various disease mechanisms such as intense lung inflammation, vascular dysfunction, and pulmonary embolism. Currently, however, there's no drug addressing all these mechanisms simultaneously. This study explored the multi-targeting potential of S-nitrosoglutathione (GSNO) and N6022, an inhibitor of GSNO reductase (GSNOR) on markers of inflammatory, vascular, and thrombotic diseases related to COVID-19-induced acute lung disease. For this, acute lung disease was induced in C57BL/6 mice through intranasal administration of recombinant SARS-CoV-2 spike protein S1 domain (SP-S1). The mice exhibited fever, body weight loss, and increased blood levels and lung expression of proinflammatory cytokines (e.g., TNF-α and IL-6) as well as increased vascular inflammation mediated by ICAM-1 and VCAM-1 and lung infiltration by immune cells (e.g., neutrophils, monocytes, and activated cytotoxic and helper T cells). Further, the mice exhibited increased lung hyperpermeability (lung Evans blue extravasation) leading to lung edema development as well as elevated blood coagulation factors (e.g., fibrinogen, thrombin, activated platelets, and von Willebrand factor) and lung fibrin deposition. Similar to the patients with COVID-19, male mice showed more severe disease than female mice, along with higher GSNOR expression in the lungs. Optimization of GSNO by treatment with exogenous GSNO or inhibition of GSNOR by N6022 (or GSNO knockout) protects against SP-S1-induced lung diseases in both genders. These findings provide evidence for the potential efficacies of GSNO and GSNOR inhibitors in addressing the multi-mechanistic nature of SARS-CoV-2 SP-associated acute-lung disease.
Collapse
Affiliation(s)
- Judong Kim
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| | - Fei Qiao
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Avtar K. Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
- Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, United States
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
- Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, United States
| |
Collapse
|
6
|
Das S, Ahmad Z, Singh S, Singh S, Wright RE, Giri S, Kumar A. Oral administration of S-nitroso-L-glutathione (GSNO) provides anti-inflammatory and cytoprotective effects during ocular bacterial infections. Cell Mol Life Sci 2023; 80:309. [PMID: 37770649 PMCID: PMC11072052 DOI: 10.1007/s00018-023-04963-w] [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: 05/03/2023] [Revised: 08/20/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023]
Abstract
Bacterial endophthalmitis is a severe complication of eye surgeries that can lead to vision loss. Current treatment involves intravitreal antibiotic injections that control bacterial growth but not inflammation. To identify newer therapeutic targets to promote inflammation resolution in endophthalmitis, we recently employed an untargeted metabolomics approach. This led to the discovery that the levels of S-nitroso-L-glutathione (GSNO) were significantly reduced in an experimental murine Staphylococcus aureus (SA) endophthalmitis model. In this study, we tested the hypothesis whether GSNO supplementation via different routes (oral, intravitreal) provides protection during bacterial endophthalmitis. Our results show that prophylactic administration of GSNO via intravitreal injections ameliorated SA endophthalmitis. Therapeutically, oral administration of GSNO was found to be most effective in reducing intraocular inflammation and bacterial burden. Moreover, oral GSNO treatment synergized with intravitreal antibiotic injections in reducing the severity of endophthalmitis. Furthermore, in vitro experiments using cultured human retinal Muller glia and retinal pigment epithelial (RPE) cells showed that GSNO treatment reduced SA-induced inflammatory mediators and cell death. Notably, both in-vivo and ex-vivo data showed that GSNO strengthened the outer blood-retinal barrier during endophthalmitis. Collectively, our study demonstrates GSNO as a potential therapeutic agent for the treatment of intraocular infections due to its dual anti-inflammatory and cytoprotective properties.
Collapse
Affiliation(s)
- Susmita Das
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 4717 St. Antoine, Detroit, MI, 48201, USA
| | - Zeeshan Ahmad
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 4717 St. Antoine, Detroit, MI, 48201, USA
| | - Sneha Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 4717 St. Antoine, Detroit, MI, 48201, USA
| | - Sukhvinder Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 4717 St. Antoine, Detroit, MI, 48201, USA
| | - Robert Emery Wright
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 4717 St. Antoine, Detroit, MI, 48201, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 4717 St. Antoine, Detroit, MI, 48201, USA.
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
7
|
Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol 2022; 19:1079-1101. [PMID: 36056148 PMCID: PMC9508259 DOI: 10.1038/s41423-022-00902-0] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractThe immune-inflammatory response is associated with increased nitro-oxidative stress. The aim of this mechanistic review is to examine: (a) the role of redox-sensitive transcription factors and enzymes, ROS/RNS production, and the activity of cellular antioxidants in the activation and performance of macrophages, dendritic cells, neutrophils, T-cells, B-cells, and natural killer cells; (b) the involvement of high-density lipoprotein (HDL), apolipoprotein A1 (ApoA1), paraoxonase-1 (PON1), and oxidized phospholipids in regulating the immune response; and (c) the detrimental effects of hypernitrosylation and chronic nitro-oxidative stress on the immune response. The redox changes during immune-inflammatory responses are orchestrated by the actions of nuclear factor-κB, HIF1α, the mechanistic target of rapamycin, the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, mitogen-activated protein kinases, 5' AMP-activated protein kinase, and peroxisome proliferator-activated receptor. The performance and survival of individual immune cells is under redox control and depends on intracellular and extracellular levels of ROS/RNS. They are heavily influenced by cellular antioxidants including the glutathione and thioredoxin systems, nuclear factor erythroid 2-related factor 2, and the HDL/ApoA1/PON1 complex. Chronic nitro-oxidative stress and hypernitrosylation inhibit the activity of those antioxidant systems, the tricarboxylic acid cycle, mitochondrial functions, and the metabolism of immune cells. In conclusion, redox-associated mechanisms modulate metabolic reprogramming of immune cells, macrophage and T helper cell polarization, phagocytosis, production of pro- versus anti-inflammatory cytokines, immune training and tolerance, chemotaxis, pathogen sensing, antiviral and antibacterial effects, Toll-like receptor activity, and endotoxin tolerance.
Collapse
|
8
|
Cheng W, Cui C, Liu G, Ye C, Shao F, Bagchi AK, Mehta JL, Wang X. NF-κB, A Potential Therapeutic Target in Cardiovascular Diseases. Cardiovasc Drugs Ther 2022; 37:571-584. [PMID: 35796905 DOI: 10.1007/s10557-022-07362-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. Atherosclerosis is the basis of major CVDs - myocardial ischemia, heart failure, and stroke. Among numerous functional molecules, the transcription factor nuclear factor κB (NF-κB) has been linked to downstream target genes involved in atherosclerosis. The activation of the NF-κB family and its downstream target genes in response to environmental and cellular stress, hypoxia, and ischemia initiate different pathological events such as innate and adaptive immunity, and cell survival, differentiation, and proliferation. Thus, NF-κB is a potential therapeutic target in the treatment of atherosclerosis and related CVDs. Several biologics and small molecules as well as peptide/proteins have been shown to regulate NF-κB dependent signaling pathways. In this review, we will focus on the function of NF-κB in CVDs and the role of NF-κB inhibitors in the treatment of CVDs.
Collapse
Affiliation(s)
- Weijia Cheng
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Can Cui
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China.,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chenji Ye
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Fang Shao
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450046, China
| | - Ashim K Bagchi
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, 72205, USA.
| | - Xianwei Wang
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Weihui, China. .,Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
9
|
Bladowski M, Szahidewicz-Krupska E, Wiśniewski J, Fortuna P, Chojdak-Łukasiewicz J, Budrewicz S, Fleszar M, Doroszko A. Changes in the Plasma and Platelet Nitric Oxide Biotransformation Metabolites during Ischemic Stroke-A Dynamic Human LC/MS Metabolomic Study. Antioxidants (Basel) 2022; 11:antiox11050955. [PMID: 35624819 PMCID: PMC9137966 DOI: 10.3390/antiox11050955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 12/10/2022] Open
Abstract
Despite improvement in the management of modifiable cardiovascular risk factors, ischemic stroke remains the leading cause of morbidity and mortality in the adult population. The aim of this study was to analyze the time-dependent dynamic differences in expression of the nitric oxide (NO) metabolic pathway in the platelet and plasma compartment between subjects with and without ischemic stroke. Additionally, the interplay between these parameters and platelet aggregation was investigated. A total of 418 patients in acute phase of non-cardioembolic stroke were investigated. Following the inclusion and exclusion criteria, finally 40 subjects with stroke and 39 demographically matched healthy participants were enrolled. Neurological physical examination, followed by assessment of the platelet and plasma levels of the nitric oxide synthase (NOS) inhibitors, including asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA), as well as NOS substrate-L-Arginine were performed dynamically three times within the first 24-h, then on the 3rd and 7th day after the stroke onset, which was compared with the healthy control. The platelet L-Arginine concentration was significantly higher on the 1st and 3rd day of stroke, while the plasma levels were significantly lower on exact days in comparison to the control. The competitive NOS-inhibitors in platelets were stably elevated in stroke subjects, whereas no significant differences in plasma compartment were noted. The arachidonic-acid-induced platelet aggregation was negatively associated with the platelet NOS substrate bioavailability, as assessed by the L−Arginine ADMA-ratio on the 3rd and 7th day. Subjects with non-cardioembolic ischemic stroke are characterized by elevated platelet levels of NOS inhibitors. Management of stroke results in increasing the platelet L-Arginine concentration and subsequent NO bioavailability in the platelet compartment.
Collapse
Affiliation(s)
- Maciej Bladowski
- Department and Clinic of Internal Medicine, Hypertension and Clinical Oncology, Faculty of Medicine, Wroclaw Medical University, Borowska 213 Str., 50-556 Wroclaw, Poland; (M.B.); (E.S.-K.)
| | - Ewa Szahidewicz-Krupska
- Department and Clinic of Internal Medicine, Hypertension and Clinical Oncology, Faculty of Medicine, Wroclaw Medical University, Borowska 213 Str., 50-556 Wroclaw, Poland; (M.B.); (E.S.-K.)
| | - Jerzy Wiśniewski
- Department of Medical Biochemistry, Wroclaw Medical University, Chalubinskiego 10 Str., 50-368 Wroclaw, Poland; (J.W.); (P.F.); (M.F.)
- Department of Biochemistry, Molecular Biology and Biotechnology, Wroclaw University of Science and Technology, Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Paulina Fortuna
- Department of Medical Biochemistry, Wroclaw Medical University, Chalubinskiego 10 Str., 50-368 Wroclaw, Poland; (J.W.); (P.F.); (M.F.)
| | - Justyna Chojdak-Łukasiewicz
- Department and Clinic of Neurology, Wroclaw Medical University, Borowska 213 Str., 50-556 Wroclaw, Poland; (J.C.-Ł.); (S.B.)
| | - Slawomir Budrewicz
- Department and Clinic of Neurology, Wroclaw Medical University, Borowska 213 Str., 50-556 Wroclaw, Poland; (J.C.-Ł.); (S.B.)
| | - Mariusz Fleszar
- Department of Medical Biochemistry, Wroclaw Medical University, Chalubinskiego 10 Str., 50-368 Wroclaw, Poland; (J.W.); (P.F.); (M.F.)
| | - Adrian Doroszko
- Department and Clinic of Internal Medicine, Hypertension and Clinical Oncology, Faculty of Medicine, Wroclaw Medical University, Borowska 213 Str., 50-556 Wroclaw, Poland; (M.B.); (E.S.-K.)
- Correspondence: ; Tel.: +48-71-736-4000
| |
Collapse
|
10
|
Li T, Xu T, Zhao J, Gao H, Xie W. Depletion of iNOS-positive inflammatory cells decelerates neuronal degeneration and alleviates cerebral ischemic damage by suppressing the inflammatory response. Free Radic Biol Med 2022; 181:209-220. [PMID: 35150825 DOI: 10.1016/j.freeradbiomed.2022.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/25/2022] [Accepted: 02/08/2022] [Indexed: 01/05/2023]
Abstract
Ischemic stroke leads to neuronal damage and severe inflammation that activate iNOS expression in different cell types, especially inflammatory cells in the brain. It is shown that NO released from iNOS contributes to the pathological development of cerebral ischemia. However, the role of these iNOS-expressing inflammatory cells in ischemic stroke has not been fully elucidated. Our purpose is to test if ischemia-induced iNOS+ inflammatory cells may exaggerate cerebral inflammation to exacerbate neuronal deficit. We studied the dynamics of iNOS+ cells after stroke and found an early and sustained iNOS expression at lesion site. Since iNOS is highly expressed in inflammatory cells after injury, we depleted the iNOS + inflammatory cells via the selective scavenger GdCl3, and investigated its effect on stroke outcome, neuronal and vascular deficit, and inflammatory response. After GdCl3 treatment, half of iNOS+ inflammatory cells were depleted, including mainly activated microglia/macrophages and some astrocytes. Selective depletion of iNOS+ inflammatory cells resulted in a pronounced reduction in brain damage, resulting in improvement of motor ability. Histologic studies and in vivo two-photon imaging data revealed a slowdown of neuronal degeneration after the depletion of iNOS+ inflammatory cells. In contrast to iNOS inhibition alone, depletion of iNOS+ inflammatory cells profoundly altered the immune microenvironment profile, in addition to reducing NO production. qRT-PCR analysis showed that depletion of iNOS+ inflammatory cells significantly restrained the production of pro-inflammatory cytokines, which moderated the immune microenvironment at the lesion site. Taken together, our data demonstrate that depleting iNOS+ inflammatory cells prevents neuronal damage not only by inhibiting NO, but also importantly by suppressing the inflammatory response, which is beneficial to ischemic injury. These results provide evidence that iNOS+ inflammatory cells, as a vital source of pro-inflammatory cytokines, contribute to the development of ischemic damage and could be a potential therapeutic target for the treatment of ischemia.
Collapse
Affiliation(s)
- Ting Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China.
| | - Ting Xu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| | - Jin Zhao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| | - Hao Gao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| | - Wenguang Xie
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 73000, China
| |
Collapse
|
11
|
Huang Q, Cai G, Liu T, Liu Z. Relationships Among Gut Microbiota, Ischemic Stroke and Its Risk Factors: Based on Research Evidence. Int J Gen Med 2022; 15:2003-2023. [PMID: 35795301 PMCID: PMC9252587 DOI: 10.2147/ijgm.s353276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/08/2022] [Indexed: 11/27/2022] Open
Abstract
Stroke is a highly lethal disease and disabling illness while ischemic stroke accounts for the majority of stroke. It has been found that inflammation plays a key role in the initiation and progression of stroke, and atherosclerotic plaque rupture is considered to be the leading cause of ischemic stroke. Furthermore, chronic inflammatory diseases, such as obesity, type 2 diabetes mellitus (T2DM) and hypertension, are also considered as the high-risk factors for stroke. Recently, the topic on how gut microbiota affects human health has aroused great concern. The initiation and progression of ischemic stroke has been found to have close relation with gut microbiota dysbiosis. Hence, this manuscript briefly summarizes the roles of gut microbiota in ischemic stroke and its related risk factors, and the practicability of preventing and alleviating ischemic stroke by reconstructing gut microbiota.
Collapse
Affiliation(s)
- Qinhong Huang
- First Clinical School, Guangzhou Medical University, Guangzhou, 511436, People’s Republic of China
| | - Guannan Cai
- First Clinical School, Guangzhou Medical University, Guangzhou, 511436, People’s Republic of China
| | - Ting Liu
- Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, Innovation Center for Advanced Interdisciplinary Medicine, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, People’s Republic of China
- Correspondence: Ting Liu; Zhihua Liu, Email ;
| | - Zhihua Liu
- Department of Anorectal Surgery, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, People’s Republic of China
| |
Collapse
|
12
|
Oxidative Stress in the Brain: Basic Concepts and Treatment Strategies in Stroke. Antioxidants (Basel) 2021; 10:antiox10121886. [PMID: 34942989 PMCID: PMC8698986 DOI: 10.3390/antiox10121886] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022] Open
Abstract
The production of free radicals is inevitably associated with metabolism and other enzymatic processes. Under physiological conditions, however, free radicals are effectively eliminated by numerous antioxidant mechanisms. Oxidative stress occurs due to an imbalance between the production and elimination of free radicals under pathological conditions. Oxidative stress is also associated with ageing. The brain is prone to oxidative damage because of its high metabolic activity and high vulnerability to ischemic damage. Oxidative stress, thus, plays a major role in the pathophysiology of both acute and chronic pathologies in the brain, such as stroke, traumatic brain injury or neurodegenerative diseases. The goal of this article is to summarize the basic concepts of oxidative stress and its significance in brain pathologies, as well as to discuss treatment strategies for dealing with oxidative stress in stroke.
Collapse
|
13
|
Nebulization of Low-Dose S-Nitrosoglutathione in Diabetic Stroke Enhances Benefits of Reperfusion and Prevents Post-Thrombolysis Hemorrhage. Biomolecules 2021; 11:biom11111587. [PMID: 34827584 PMCID: PMC8615482 DOI: 10.3390/biom11111587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic has escalated the occurrence of hypoxia including thrombotic stroke worldwide, for which nitric oxide (NO) therapy seems very promising and translatable. Therefore, various modes/routes of NO-delivery are now being tested in different clinical trials for safer, faster, and more effective interventions against ischemic insults. Intravenous (IV) infusion of S-Nitrosoglutathione (GSNO), the major endogenous molecular pool of NO, has been reported to protect against mechanical cerebral ischemia-reperfusion (IR); however, it has been never tested in any kind of “clinically” relevant thromboembolic stroke models with or without comorbidities and in combination with the thrombolytic reperfusion therapy. Moreover, “IV-effects” of higher dose of GSNO following IR-injury have been contradicted to augment stroke injury. Herein, we tested the hypothesis that nebulization of low-dose GSNO will not alter blood pressure (BP) and will mitigate stroke injury in diabetic mice via enhanced cerebral blood flow (CBF) and brain tissue oxygenation (PbtO2). GSNO-nebulization (200 μg/kgbwt) did not alter BP, but augmented the restoration of CBF, improved behavioral outcomes and reduced stroke injury. Moreover, GSNO-nebulization increased early reoxygenation of brain tissue/PbtO2 as measured at 6.5 h post-stroke following thrombolytic reperfusion, and enervated unwanted effects of late thrombolysis in diabetic stroke. We conclude that the GSNO-nebulization is safe and effective for enhancing collateral microvascular perfusion in the early hours following stroke. Hence, nebulized-GSNO therapy has the potential to be developed and translated into an affordable field therapy against ischemic events including strokes, particularly in developing countries with limited healthcare infrastructure.
Collapse
|
14
|
Chiletti R, Bennet M, Kenna K, Angerosa J, Sheeran FL, Brink J, Perrier S, Zannino D, Smolich J, Pepe S, Cheung MM. S-nitroso-glutathione limits apoptosis and reduces pulmonary vascular dysfunction after bypass. Ann Thorac Surg 2021; 114:1468-1474. [PMID: 34416229 DOI: 10.1016/j.athoracsur.2021.07.041] [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: 02/19/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND During hypoxia or acidosis, S-nitrosoglutathione (GSNO) has been shown to protect the cardiomyocyte from IR injury. In a randomised double blinded control study of a porcine model of paediatric CPB, we aimed to evaluate the effects of two different doses (low and high) of GSNO. METHODS Pigs weighing 15-20 kg were exposed to CPB with one hour of aortic cross-clamp. Prior to and during CPB, animals were randomised to receive low dose (up to 20 nmol/kg/min) GSNO (n=8), high dose (up to 60 nmol/kg/min) GSNO (n=6) or normal saline (n=7). Standard cardiac intensive care management was continued for 4 hours post-bypass. RESULTS There was a reduction in myocyte apoptosis after administration of GSNO (p=0.04) with no difference between low and high dose GSNO. The low dose GSNO group had lower pulmonary vascular resistance post-CPB (p=0.007). Mitochondrial Complex I activity normalised to citrate synthase activity was higher after GSNO compared to control (p=0.02), with no difference between low and high dose GSNO. CONCLUSIONS In a porcine model of CPB intravenous administration of GSNO limits myocardial apoptosis through preservation of mitochondrial complex I activity, and improves pulmonary vascular resistance. There appears to be a dose dependent effect to this protection.
Collapse
Affiliation(s)
- Roberto Chiletti
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia;; Paediatric Intensive Care Unit, Royal Children's Hospital, Melbourne, Australia
| | - Martin Bennet
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia;; Cardiac Surgery Unit, Royal Children's Hospital, Melbourne, Australia
| | - Kelly Kenna
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Julie Angerosa
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - Freya L Sheeran
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Johann Brink
- Cardiac Surgery Unit, Starship Hospital, Auckland, New Zealand
| | - Stephanie Perrier
- Cardivascular Surgery, University Hospital of Strasbourg, Strasbourg, France
| | - Diana Zannino
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - Joseph Smolich
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Salvatore Pepe
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia;; Department of Cardiology, Royal Children's Hospital, Melbourne, Australia
| | - Michael Mh Cheung
- Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia;; Department of Paediatrics, University of Melbourne, Melbourne, Australia;; Department of Cardiology, Royal Children's Hospital, Melbourne, Australia.
| |
Collapse
|
15
|
Singh I, Kim J, Saxena N, Choi S, Islam SMT, Singh AK, Khan M, Won J. Vascular and immunopathological role of Asymmetric Dimethylarginine (ADMA) in Experimental Autoimmune Encephalomyelitis. Immunology 2021; 164:602-616. [PMID: 34310708 DOI: 10.1111/imm.13396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 07/01/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous nitric oxide synthase (NOS) inhibitor/uncoupler inducing vascular pathology. Vascular pathology is an important factor for the development and progression of CNS pathology of MS, yet the role of ADMA in MS remains elusive. Patients with multiple sclerosis (MS) are reported to have elevated blood levels of ADMA, and mice with experimental autoimmune encephalomyelitis (EAE, an animal model of MS) generated by auto-immunization of myelin oligodendrocyte glycoprotein (MOG) and blood-brain barrier (BBB) disruption by pertussis toxin also had increased blood ADMA levels in parallel with induction of clinical disease. To explore the role of ADMA in EAE pathogenesis, EAE mice were treated with a daily dose of ADMA. It is of special interest that ADMA treatment enhanced the BBB disruption in EAE mice and exacerbated the clinical and CNS disease of EAE. ADMA treatment also induced the BBB disruption and EAE disease in MOG-immunized mice even without pertussis toxin treatment, suggesting the role of ADMA in BBB dysfunction in EAE. T-cell polarization studies also documented that ADMA treatment promotes TH 1- and TH 17-mediated immune responses but without affecting Treg-mediated immune response in EAE mice as well as in in vitro T-cell culture. Taken together, these data, for the first time, document the vascular and immunopathogenic roles of ADMA in EAE, thus pointing to the potential of ADMA-mediated mechanism as a new target of potential therapy for MS.
Collapse
Affiliation(s)
- Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA.,Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| | - Judong Kim
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Seungho Choi
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.,Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| | - Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
16
|
Wierońska JM, Cieślik P, Kalinowski L. Nitric Oxide-Dependent Pathways as Critical Factors in the Consequences and Recovery after Brain Ischemic Hypoxia. Biomolecules 2021; 11:biom11081097. [PMID: 34439764 PMCID: PMC8392725 DOI: 10.3390/biom11081097] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Brain ischemia is one of the leading causes of disability and mortality worldwide. Nitric oxide (NO•), a molecule that is involved in the regulation of proper blood flow, vasodilation, neuronal and glial activity constitutes the crucial factor that contributes to the development of pathological changes after stroke. One of the early consequences of a sudden interruption in the cerebral blood flow is the massive production of reactive oxygen and nitrogen species (ROS/RNS) in neurons due to NO• synthase uncoupling, which leads to neurotoxicity. Progression of apoptotic or necrotic neuronal damage activates reactive astrocytes and attracts microglia or lymphocytes to migrate to place of inflammation. Those inflammatory cells start to produce large amounts of inflammatory proteins, including pathological, inducible form of NOS (iNOS), which generates nitrosative stress that further contributes to brain tissue damage, forming vicious circle of detrimental processes in the late stage of ischemia. S-nitrosylation, hypoxia-inducible factor 1α (HIF-1α) and HIF-1α-dependent genes activated in reactive astrocytes play essential roles in this process. The review summarizes the roles of NO•-dependent pathways in the early and late aftermath of stroke and treatments based on the stimulation or inhibition of particular NO• synthases and the stabilization of HIF-1α activity.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Paulina Cieślik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Biobank Fahrenheit BBMRI.pl, Medical University of Gdansk, Debinki Street 7, 80-211 Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Debinki Street 7, 80-211 Gdansk, Poland
- BioTechMed Center/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza 11/12, 80-223 Gdansk, Poland
- Correspondence: ; Tel.: +48-58-349-1182
| |
Collapse
|
17
|
Delaitre C, Boisbrun M, Lecat S, Dupuis F. Targeting the Angiotensin II Type 1 Receptor in Cerebrovascular Diseases: Biased Signaling Raises New Hopes. Int J Mol Sci 2021; 22:ijms22136738. [PMID: 34201646 PMCID: PMC8269339 DOI: 10.3390/ijms22136738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/20/2022] Open
Abstract
The physiological and pathophysiological relevance of the angiotensin II type 1 (AT1) G protein-coupled receptor no longer needs to be proven in the cardiovascular system. The renin–angiotensin system and the AT1 receptor are the targets of several classes of therapeutics (such as angiotensin converting enzyme inhibitors or angiotensin receptor blockers, ARBs) used as first-line treatments in cardiovascular diseases. The importance of AT1 in the regulation of the cerebrovascular system is also acknowledged. However, despite numerous beneficial effects in preclinical experiments, ARBs do not induce satisfactory curative results in clinical stroke studies. A better understanding of AT1 signaling and the development of biased AT1 agonists, able to selectively activate the β-arrestin transduction pathway rather than the Gq pathway, have led to new therapeutic strategies to target detrimental effects of AT1 activation. In this paper, we review the involvement of AT1 in cerebrovascular diseases as well as recent advances in the understanding of its molecular dynamics and biased or non-biased signaling. We also describe why these alternative signaling pathways induced by β-arrestin biased AT1 agonists could be considered as new therapeutic avenues for cerebrovascular diseases.
Collapse
Affiliation(s)
- Céline Delaitre
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France;
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS/Université de Strasbourg, 300 Boulevard Sébastien Brant, CS 10413, CEDEX, 67412 Illkirch-Graffenstaden, France;
| | | | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS/Université de Strasbourg, 300 Boulevard Sébastien Brant, CS 10413, CEDEX, 67412 Illkirch-Graffenstaden, France;
| | - François Dupuis
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France;
- Correspondence: ; Tel.: +33-372747272
| |
Collapse
|
18
|
Jayakumar P, Martínez-Moreno CG, Lorenson MY, Walker AM, Morales T. Prolactin Attenuates Neuroinflammation in LPS-Activated SIM-A9 Microglial Cells by Inhibiting NF-κB Pathways Via ERK1/2. Cell Mol Neurobiol 2021; 42:2171-2186. [PMID: 33821330 DOI: 10.1007/s10571-021-01087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/27/2021] [Indexed: 10/21/2022]
Abstract
Prolactin (PRL) is a pleiotropic hormone with multiple functions in several tissues and organs, including the brain. PRL decreases lesion-induced microgliosis and modifies gene expression related to microglial functions in the hippocampus, thereby providing a possible mechanism through which it might participate in neuroimmune modulatory responses and prevent neuronal cell damage. However, the direct contribution of microglial cells to PRL-mediated neuroprotection is still unclear and no studies have yet documented whether PRL can directly activate cellular pathways in microglial cells. The aim of this study is to elucidate in vitro actions of PRL on the immortalized SIM-A9 microglia cell line in basal and LPS-stimulated conditions. PRL alone induced a time-dependent extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Pretreatment with PRL attenuated LPS (200 ng/ml) stimulated pro-inflammatory markers: nitric oxide (NO) levels, inducible nitric oxide synthase (iNOS), interleukins (IL)-6, -1β and tumor necrosis factor (TNF-α) expression at 20 nM dosage. PRL suppressed LPS-induced nuclear factor (NF)-κappaB (NF-κB) p65 subunit phosphorylation and its upstream p-ERK1/2 activity. In conclusion, PRL exhibits anti-inflammatory effects in LPS-stimulated SIM-A9 microglia by downregulating pro-inflammatory mediators corresponding to suppression of LPS-activated ERK1/2 and NF-κB phosphorylation.
Collapse
Affiliation(s)
- Preethi Jayakumar
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Carlos G Martínez-Moreno
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Mary Y Lorenson
- Department of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Ameae M Walker
- Department of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico.
| |
Collapse
|
19
|
Sengupta R, Coppo L, Sircar E, Mishra P, Holmgren A. S‐Denitrosylation by the C‐Terminal Swinging Arm of R1 Subunit: A Novel Mechanism to Restore Ribonucleotide Reductase Activity. ChemistrySelect 2021. [DOI: 10.1002/slct.202100153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Rajib Sengupta
- Division of Biochemistry Department of Medical Biochemistry and Biophysics, Karolinska Institute Stockholm Sweden
- Amity Institute of Biotechnology Amity University, Major Arterial Road, Rajarhat, New Town Kolkata 700135, West Bengal India
| | - Lucia Coppo
- Division of Biochemistry Department of Medical Biochemistry and Biophysics, Karolinska Institute Stockholm Sweden
| | - Esha Sircar
- Amity Institute of Biotechnology Amity University, Major Arterial Road, Rajarhat, New Town Kolkata 700135, West Bengal India
| | - Pradeep Mishra
- Division of Biochemistry Department of Medical Biochemistry and Biophysics, Karolinska Institute Stockholm Sweden
| | - Arne Holmgren
- Division of Biochemistry Department of Medical Biochemistry and Biophysics, Karolinska Institute Stockholm Sweden
| |
Collapse
|
20
|
Hayashida K, Miyara SJ, Shinozaki K, Takegawa R, Yin T, Rolston DM, Choudhary RC, Guevara S, Molmenti EP, Becker LB. Inhaled Gases as Therapies for Post-Cardiac Arrest Syndrome: A Narrative Review of Recent Developments. Front Med (Lausanne) 2021; 7:586229. [PMID: 33585501 PMCID: PMC7873953 DOI: 10.3389/fmed.2020.586229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/04/2020] [Indexed: 01/22/2023] Open
Abstract
Despite recent advances in the management of post-cardiac arrest syndrome (PCAS), the survival rate, without neurologic sequelae after resuscitation, remains very low. Whole-body ischemia, followed by reperfusion after cardiac arrest (CA), contributes to PCAS, for which established pharmaceutical interventions are still lacking. It has been shown that a number of different processes can ultimately lead to neuronal injury and cell death in the pathology of PCAS, including vasoconstriction, protein modification, impaired mitochondrial respiration, cell death signaling, inflammation, and excessive oxidative stress. Recently, the pathophysiological effects of inhaled gases including nitric oxide (NO), molecular hydrogen (H2), and xenon (Xe) have attracted much attention. Herein, we summarize recent literature on the application of NO, H2, and Xe for treating PCAS. Recent basic and clinical research has shown that these gases have cytoprotective effects against PCAS. Nevertheless, there are likely differences in the mechanisms by which these gases modulate reperfusion injury after CA. Further preclinical and clinical studies examining the combinations of standard post-CA care and inhaled gas treatment to prevent ischemia-reperfusion injury are warranted to improve outcomes in patients who are being failed by our current therapies.
Collapse
Affiliation(s)
- Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Santiago J Miyara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Medicine, and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, New York, NY, United States.,Institute of Health Innovations and Outcomes Research, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Koichiro Shinozaki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Ryosuke Takegawa
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Daniel M Rolston
- Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Department of Surgery, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| | - Rishabh C Choudhary
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States
| | - Sara Guevara
- Department of Surgery, Northwell Health, Manhasset, NY, United States
| | - Ernesto P Molmenti
- Department of Surgery, Medicine, and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, New York, NY, United States.,Institute of Health Innovations and Outcomes Research, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, United States.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health System, Manhasset, NY, United States.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, United States
| |
Collapse
|
21
|
Abstract
Sudden cardiac arrest is a leading cause of death worldwide. Although the methods of cardiopulmonary resuscitation have been improved, mortality is still unacceptably high, and many survivors suffer from lasting neurological deficits due to the post-cardiac arrest syndrome (PCAS). Pathophysiologically, generalized vascular endothelial dysfunction accompanied by platelet activation and systemic inflammation has been implicated in the pathogenesis of PCAS. Because endothelial-derived nitric oxide (NO) plays a central role in maintaining vascular homeostasis, the role of NO-dependent signaling has been a focus of the intense investigation. Recent preclinical studies showed that therapeutic interventions that increase vascular NO bioavailability may improve outcomes after cardiac arrest complicated with PCAS. In particular, NO inhalation therapy has been shown to improve neurological outcomes and survival in multiple species. Clinical studies examining the safety and efficacy of inhaled NO in patients sustaining PCAS are warranted.
Collapse
|
22
|
Gholamzadeh R, Aboutaleb N, Nazarinia D. Intravenous injection of apelin-13 improves sensory-motor balance deficits caused by cerebral ischemic reperfusion injury in male wistar rats via restoration of nitric oxide. J Chem Neuroanat 2020; 112:101886. [PMID: 33189869 DOI: 10.1016/j.jchemneu.2020.101886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022]
Abstract
It has been reported that apelin-13 possesses neuroprotective effects against cerebral ischemia/reperfusion injury (IRI). Disabilities in sense, movement and balance are the major stroke complications which, result in a high rate of mortality. Here, effects of intravenous (IV) injection of apelin-13 on the severity of neural death, infarct volume, neurological defects and its association with nitric oxide (NO) were investigated. A rat model of cerebral IRI was created by middle cerebral artery occlusion (MCAO) for 60 min and restoration of blood flow for 23 h. Animals were randomly assigned into six groups: sham, ischemia (MCAO), vehicle (MCAO + PBS) and three treatment groups (MCAO + apelin-13 in 10, 20, 40 μg/kg doses, IV). All injections were carried out via tail vein injection 5 min before reperfusion. Neural loss and infarct volume were evaluated by Nissl and 2,3,5-triphenyltetrazolium chloride (TTC) staining, respectively. Neurological defects were scored by standard modified criteria. Serum NO was measured by colorimetric method. Apelin-13 in doses of 20 and 40 μg/kg significantly reduced neural death, infarct volume and disturbance of sensory-motor balance compared to control and vehicle groups (p < 0.05). Serum NO levels reduced in MCAO groups compared to sham. Apelin-13 restored serum NO levels at 20 μg/kg dose (p < 0.05). Our data showed beneficial effect of IV injection of apelin-13 on sensory-motor balance defects by reducing neural death and restoration of serum NO levels. The present study shows the validity of apelin-13 in treatment of ischemic stroke in different administration methods.
Collapse
Affiliation(s)
- Raheleh Gholamzadeh
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Aboutaleb
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Donya Nazarinia
- Department of Physiology, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
23
|
Protective Mechanism and Treatment of Neurogenesis in Cerebral Ischemia. Neurochem Res 2020; 45:2258-2277. [PMID: 32794152 DOI: 10.1007/s11064-020-03092-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Stroke is the fifth leading cause of death worldwide and is a main cause of disability in adults. Neither currently marketed drugs nor commonly used treatments can promote nerve repair and neurogenesis after stroke, and the repair of neurons damaged by ischemia has become a research focus. This article reviews several possible mechanisms of stroke and neurogenesis and introduces novel neurogenic agents (fibroblast growth factors, brain-derived neurotrophic factor, purine nucleosides, resveratrol, S-nitrosoglutathione, osteopontin, etc.) as well as other treatments that have shown neuroprotective or neurogenesis-promoting effects.
Collapse
|
24
|
Investigation of S-Nitrosoglutathione in stroke: A systematic review and meta-analysis of literature in pre-clinical and clinical research. Exp Neurol 2020; 328:113262. [DOI: 10.1016/j.expneurol.2020.113262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/19/2020] [Accepted: 02/28/2020] [Indexed: 11/21/2022]
|
25
|
Wang X, Ao J, Lu H, Zhao Q, Ma Y, Zhang J, Ren H, Zhang Y. Osteoimmune Modulation and Guided Osteogenesis Promoted by Barrier Membranes Incorporated with S-Nitrosoglutathione (GSNO) and Mesenchymal Stem Cell-Derived Exosomes. Int J Nanomedicine 2020; 15:3483-3496. [PMID: 32523344 PMCID: PMC7237116 DOI: 10.2147/ijn.s248741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022] Open
Abstract
Background The use of polycaprolactone (PCL) for bone defects in a clinical setting is limited due to a lack of bioactivity. Exosomes derived from mesenchymal stem cells (MSCs) have an important immunoregulatory potential and together with S-nitrosoglutathione (GSNO) they possess therapeutic potential for bone regeneration. Materials and Methods In this study, PCL was modified with GSNO and MSC-derived exosomes and the impact on macrophages and osteogenes is evaluated. Results MSC-derived exosomes exhibited a cup-shaped morphology and were internalized by macrophages and human bone marrow-derived mesenchymal stromal cells (hBMSCs). The pattern of internalization of scaffold-immobilized exosomes was similar in RAW264.7 cells and hBMSCs after 4h and 24h of co-culture. Assessment of macrophage morphology under inflammatory conditions by scanning electronic microscopy (SEM) and confocal microscopy demonstrated macrophages were significantly elongated and expression of pro-inflammatory genes markedly decreased when co-cultured with PCL/PDA + GSNO + exosome scaffolds. Furthermore, this scaffold modification significantly enhanced osteogenic differentiation of hBMSCs. Discussion This study demonstrated the possibility of using a GSNO- and exosome-based strategy to adapt barrier membrane scaffolds. PCL/PDA + GSNO + exosome scaffolds may serve as an important barrier membrane for osteogenesis and tissue regeneration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China.,Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JCMR-ZMU & URMC), Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Haiping Lu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Qingyu Zhao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China.,Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JCMR-ZMU & URMC), Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Jun Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| | - Hao Ren
- Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, Guangdong 518119, People's Republic of China
| | - Yi Zhang
- Joint Orthopaedic Research Center of Zunyi Medical University & University of Rochester Medical Center (JCMR-ZMU & URMC), Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China.,Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi 563000, Guizhou, People's Republic of China
| |
Collapse
|
26
|
Khan M, Kumar P, Qiao F, Islam SMT, Singh AK, Won JS, Feng W, Singh I. Targeting GSNOR for functional recovery in a middle-aged mouse model of stroke. Brain Res 2020; 1741:146879. [PMID: 32418890 DOI: 10.1016/j.brainres.2020.146879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/01/2023]
Abstract
The nitric oxide (NO) metabolome and the NO metabolite-based neurovascular protective pathways are dysregulated after stroke. The major NO metabolite S-nitrosoglutahione (GSNO) is essential for S-nitrosylation-based signaling events and the inhibition of S-nitrosoglutahione (GSNO)-metabolizing enzyme GSNO reductase (GSNOR) provides protective effects following cardiac ischemia. However, the role of GSNOR and GSNOR inhibition-mediated increased GSNO/S-nitrosylation is not understood in neurovascular diseases such as stroke. Because age is the major risk factor of stroke and recovery in aged stroke patients is low and slow, we investigated the efficacy of GSNOR inhibition using a GSNOR selective inhibitor N6022 in a clinically relevant middle-aged cerebral ischemia and reperfusion (IR) mouse model of stroke. N6022 (5 mg/kg; iv) treatment of IR mice at 2 h after reperfusion followed by the treatment of the same dose daily for 3 days reduced the infarct volume and decreased the neurological score. Daily treatment of IR animals with N6022 for 2 weeks significantly improved neurological score, brain infarctions/atrophy, survival rate, motor (measured by cylinder test) and cognitive (evaluated by novel object recognition test) functions which paralleled the decreased activity of GSNOR, reduced levels of peroxynitrite and decreased neurological score. These results are the first evidence of a new pathway for the treatment of stroke via the inhibition of GSNOR. Based on the efficacy of N6022 in the stroke animal model and its use in human therapeutic studies without toxicity, we submit that GSNOR is a druggable target, and N6022 is a promising drug candidate for human stroke therapy.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.
| | - Pavan Kumar
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.
| | - Fei Qiao
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| | - S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| | - Je-Seong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| | - Wayne Feng
- Department of Neurology, Duke University School of Medicine, Durham, NC, United States.
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| |
Collapse
|
27
|
Zeman RJ, Wen X, Moorthy CR, Etlinger JD. Therapeutic target for external beam x-irradiation in experimental spinal cord injury. J Neurosurg Spine 2020; 32:649-656. [PMID: 31899880 DOI: 10.3171/2019.11.spine19305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 11/05/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE X-irradiation has been shown to be beneficial to recovery from spinal cord injury (SCI); however, the optimal therapeutic target has not been defined. Experiments were designed to determine the optimal target volume within the injured spinal cord for improving functional recovery and sparing tissue with stereotactic x-irradiation. METHODS SCI was produced in rats at the T10 level. A 20-Gy dose of radiation was delivered with a single, 4-mm-diameter, circular radiation beam centered either on the injury epicenter or 4 or 8 mm caudal or rostral to the injury epicenter. Locomotor function was determined for 6 weeks with the Basso, Beattie, and Bresnahan locomotor scale and tissue sparing by histological analysis of transverse sections along the spinal cords. RESULTS X-irradiation of spinal cord segments at 4 mm, but not 8 mm, caudal or rostral to the contusion epicenter resulted in increases in locomotor recovery. Consistently, significant tissue sparing also occurred with x-irradiation centered at those sites, although irradiation centered 4 mm rostral to the epicenter led to tissue sparing along the greatest length of the spinal cord. Interestingly, regression analysis of these variables demonstrated that the quantitative relationship between the amount of tissue spared and the improvement in locomotion recovery was greatest in a region several millimeters rostral to the injury epicenter. CONCLUSIONS These results indicate that x-irradiation in a region rostral to the injury epicenter is optimal for recovery from SCI. This minimal target should be attractive for therapeutic application since it allows a greatly reduced target volume so that uninjured tissue is not needlessly irradiated.
Collapse
Affiliation(s)
| | | | - Chitti R Moorthy
- 2Radiation Medicine, New York Medical College, Valhalla, New York
| | | |
Collapse
|
28
|
Abstract
Ischemic strokes occur when a major cerebral artery or its branches are occluded, resulting in activation of inflammatory processes that cause secondary tissue injury, breakdown of the blood–brain barrier, edema or hemorrhage. Treatments that inhibit inflammatory processes may thus be highly beneficial. A key regulator of the inflammatory process is the nuclear factor kappa B (NF-κB) pathway. In its active form, NF-κB regulates expression of proinflammatory and proapoptotic genes. The molecules that interact with NF-κB, and the subunits that compose NF-κB itself, represent therapeutic targets that can be modulated to decrease inflammation. This review focuses on our current understanding of the NF-κB pathway and the potential benefits of inhibiting NF-κB in ischemia-reperfusion injury of the brain.
Collapse
|
29
|
Morris G, Puri BK, Walker AJ, Berk M, Walder K, Bortolasci CC, Marx W, Carvalho AF, Maes M. The compensatory antioxidant response system with a focus on neuroprogressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109708. [PMID: 31351160 DOI: 10.1016/j.pnpbp.2019.109708] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
Major antioxidant responses to increased levels of inflammatory, oxidative and nitrosative stress (ONS) are detailed. In response to increasing levels of nitric oxide, S-nitrosylation of cysteine thiol groups leads to post-transcriptional modification of many cellular proteins and thereby regulates their activity and allows cellular adaptation to increased levels of ONS. S-nitrosylation inhibits the function of nuclear factor kappa-light-chain-enhancer of activated B cells, toll-like receptor-mediated signalling and the activity of several mitogen-activated protein kinases, while activating nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2 or NFE2L2); in turn, the redox-regulated activation of Nrf2 leads to increased levels and/or activity of key enzymes and transporter systems involved in the glutathione system. The Nrf2/Kelch-like ECH-associated protein-1 axis is associated with upregulation of NAD(P)H:quinone oxidoreductase 1, which in turn has anti-inflammatory effects. Increased Nrf2 transcriptional activity also leads to activation of haem oxygenase-1, which is associated with upregulation of bilirubin, biliverdin and biliverdin reductase as well as increased carbon monoxide signalling, anti-inflammatory and antioxidant activity. Associated transcriptional responses, which may be mediated by retrograde signalling owing to elevated hydrogen peroxide, include the unfolded protein response (UPR), mitohormesis and the mitochondrial UPR; the UPR also results from increasing levels of mitochondrial and cytosolic reactive oxygen species and reactive nitrogen species leading to nitrosylation, glutathionylation, oxidation and nitration of crucial cysteine and tyrosine causing protein misfolding and the development of endoplasmic reticulum stress. It is shown how these mechanisms co-operate in forming a co-ordinated rapid and prolonged compensatory antioxidant response system.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Adam J Walker
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry, The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ken Walder
- CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Chiara C Bortolasci
- CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Wolfgang Marx
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.
| | - Michael Maes
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
30
|
Hayashida K, Bagchi A, Miyazaki Y, Hirai S, Seth D, Silverman MG, Rezoagli E, Marutani E, Mori N, Magliocca A, Liu X, Berra L, Hindle AG, Donnino MW, Malhotra R, Bradley MO, Stamler JS, Ichinose F. Improvement in Outcomes After Cardiac Arrest and Resuscitation by Inhibition of S-Nitrosoglutathione Reductase. Circulation 2019; 139:815-827. [PMID: 30586713 DOI: 10.1161/circulationaha.117.032488] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The biological effects of nitric oxide are mediated via protein S-nitrosylation. Levels of S-nitrosylated protein are controlled in part by the denitrosylase, S-nitrosoglutathione reductase (GSNOR). The objective of this study was to examine whether GSNOR inhibition improves outcomes after cardiac arrest and cardiopulmonary resuscitation (CA/CPR). METHODS Adult wild-type C57BL/6 and GSNOR-deleted (GSNOR-/-) mice were subjected to potassium chloride-induced CA and subsequently resuscitated. Fifteen minutes after a return of spontaneous circulation, wild-type mice were randomized to receive the GSNOR inhibitor, SPL-334.1, or normal saline as placebo. Mortality, neurological outcome, GSNOR activity, and levels of S-nitrosylated proteins were evaluated. Plasma GSNOR activity was measured in plasma samples obtained from post-CA patients, preoperative cardiac surgery patients, and healthy volunteers. RESULTS GSNOR activity was increased in plasma and multiple organs of mice, including brain in particular. Levels of protein S-nitrosylation were decreased in the brain 6 hours after CA/CPR. Administration of SPL-334.1 attenuated the increase in GSNOR activity in brain, heart, liver, spleen, and plasma, and restored S-nitrosylated protein levels in the brain. Inhibition of GSNOR attenuated ischemic brain injury and improved survival in wild-type mice after CA/CPR (81.8% in SPL-334.1 versus 36.4% in placebo; log rank P=0.031). Similarly, GSNOR deletion prevented the reduction in the number of S-nitrosylated proteins in the brain, mitigated brain injury, and improved neurological recovery and survival after CA/CPR. Both GSNOR inhibition and deletion attenuated CA/CPR-induced disruption of blood brain barrier. Post-CA patients had higher plasma GSNOR activity than did preoperative cardiac surgery patients or healthy volunteers ( P<0.0001). Plasma GSNOR activity was positively correlated with initial lactate levels in postarrest patients (Spearman correlation coefficient=0.48; P=0.045). CONCLUSIONS CA and CPR activated GSNOR and reduced the number of S-nitrosylated proteins in the brain. Pharmacological inhibition or genetic deletion of GSNOR prevented ischemic brain injury and improved survival rates by restoring S-nitrosylated protein levels in the brain after CA/CPR in mice. Our observations suggest that GSNOR is a novel biomarker of postarrest brain injury as well as a molecular target to improve outcomes after CA.
Collapse
Affiliation(s)
- Kei Hayashida
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Aranya Bagchi
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Yusuke Miyazaki
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Shuichi Hirai
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Divya Seth
- Institute for Transformative Molecular Medicine and Department of Medicine, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center (D.S.), Cleveland, OH
| | - Michael G Silverman
- Cardiology Division, Department of Medicine, Massachusetts General Hospital (M.G.S., R.M.), Boston, MA
| | - Emanuele Rezoagli
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Naohiro Mori
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Aurora Magliocca
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Xiaowen Liu
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA (X.L., M.W.D.)
| | - Lorenzo Berra
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Allyson G Hindle
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| | - Michael W Donnino
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA (X.L., M.W.D.)
| | - Rajeev Malhotra
- Cardiology Division, Department of Medicine, Massachusetts General Hospital (M.G.S., R.M.), Boston, MA
| | | | | | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School (K.H., A.B., Y.M., S.H., E.R., E.M., N.M., A.M., L.B., A.G.H., F.I.), Boston, MA
| |
Collapse
|
31
|
Khan M, Dhammu TS, Qiao F, Kumar P, Singh AK, Singh I. S-Nitrosoglutathione Mimics the Beneficial Activity of Endothelial Nitric Oxide Synthase-Derived Nitric Oxide in a Mouse Model of Stroke. J Stroke Cerebrovasc Dis 2019; 28:104470. [PMID: 31680031 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/18/2019] [Accepted: 10/05/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The nitric oxide (NO)-producing activity of endothelial nitric oxide synthase (eNOS) plays a significant role in maintaining endothelial function and protecting against the stroke injury. However, the activity of the eNOS enzyme and the metabolism of major NO metabolite S-nitrosoglutathione (GSNO) are dysregulated after stroke, causing endothelial dysfunction. We investigated whether an administration of exogenous of GSNO or enhancing the level of endogenous GSNO protects against neurovascular injury in wild-type (WT) and eNOS-null (endothelial dysfunction) mouse models of cerebral ischemia-reperfusion (IR). METHODS Transient cerebral ischemic injury was induced by middle cerebral artery occlusion (MCAO) for 60 minutes in male adult WT and eNOS null mice. GSNO (0.1 mg/kg body weight, intravenously) or N6022 (GSNO reductase inhibitor, 5.0 mg/kg body weight, intravenously) was administered 30 minutes before MCAO in preinjury and at the reperfusion in postinjury studies. Brain infarctions, edema, and neurobehavioral functions were evaluated at 24 hours after the reperfusion. RESULTS eNOS-null mice had a higher degree (P< .05) of injury than WT. Pre- or postinjury treatment with either GSNO or N6022 significantly reduced infarct volume, improved neurological and sensorimotor function in both WT and eNOS-null mice. CONCLUSION Reduced brain infarctions and edema, and improved neurobehavioral functions by pre- or postinjury GSNO treatment of eNOS knock out mice indicate that GSNO can attenuate IR injury, likely by mimicking the eNOS-derived NO-dependent anti-ischemic and anti-inflammatory functions. Neurovascular protection by GSNO/N6022 in both pre- and postischemic injury groups support GSNO as a promising drug candidate for the prevention and treatment of stroke injury.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina.
| | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Fei Qiao
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Pavan Kumar
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina; Ralph H Johnson VA Medical Center, Charleston, South Carolina
| | - Inderjit Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina; Ralph H Johnson VA Medical Center, Charleston, South Carolina
| |
Collapse
|
32
|
Novel Insights on the Toxicity of Phycotoxins on the Gut through the Targeting of Enteric Glial Cells. Mar Drugs 2019; 17:md17070429. [PMID: 31340532 PMCID: PMC6669610 DOI: 10.3390/md17070429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 02/08/2023] Open
Abstract
In vitro and in vivo studies have shown that phycotoxins can impact intestinal epithelial cells and can cross the intestinal barrier to some extent. Therefore, phycotoxins can reach cells underlying the epithelium, such as enteric glial cells (EGCs), which are involved in gut homeostasis, motility, and barrier integrity. This study compared the toxicological effects of pectenotoxin-2 (PTX2), yessotoxin (YTX), okadaic acid (OA), azaspiracid-1 (AZA1), 13-desmethyl-spirolide C (SPX), and palytoxin (PlTX) on the rat EGC cell line CRL2690. Cell viability, morphology, oxidative stress, inflammation, cell cycle, and specific glial markers were evaluated using RT-qPCR and high content analysis (HCA) approaches. PTX2, YTX, OA, AZA1, and PlTX induced neurite alterations, oxidative stress, cell cycle disturbance, and increase of specific EGC markers. An inflammatory response for YTX, OA, and AZA1 was suggested by the nuclear translocation of NF-κB. Caspase-3-dependent apoptosis and induction of DNA double strand breaks (γH2AX) were also observed with PTX2, YTX, OA, and AZA1. These findings suggest that PTX2, YTX, OA, AZA1, and PlTX may affect intestinal barrier integrity through alterations of the human enteric glial system. Our results provide novel insight into the toxicological effects of phycotoxins on the gut.
Collapse
|
33
|
Bouressam ML, Lecat S, Raoul A, Gaucher C, Perrin-Sarrado C, Lartaud I, Dupuis F. S-nitrosoglutathione inhibits cerebrovascular angiotensin II-dependent and -independent AT 1 receptor responses: A possible role of S-nitrosation. Br J Pharmacol 2019; 176:2049-2062. [PMID: 30822355 DOI: 10.1111/bph.14644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 01/21/2019] [Accepted: 02/13/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Angiotensin II (AngII) and NO regulate the cerebral circulation. AngII AT1 receptors exert ligand-dependent and ligand-independent (myogenic tone [MT]) vasoconstriction of cerebral vessels. NO induces post-translational modifications of proteins such as S-nitrosation (redox modification of cysteine residues). In cultured cells, S-nitrosation decreases AngII's affinity for the AT1 receptor. The present work evaluated the functional consequences of S-nitrosation on both AngII-dependent and AngII-independent cerebrovascular responses. EXPERIMENTAL APPROACH S-Nitrosation was induced in rat isolated middle cerebral arteries by pretreatment with the NO donors, S-nitrosoglutathione (GSNO) or sodium nitroprusside (SNP). Agonist-dependent activation of AT1 receptors was evaluated by obtaining concentration-response curves to AngII. Ligand-independent activation of AT1 receptors was evaluated by calculating MT (active vs. passive diameter) at pressures ranging from 20 to 200 mmHg in the presence or not of a selective AT1 receptor inverse agonist. KEY RESULTS GSNO or SNP completely abolished the AngII-dependent AT1 receptor-mediated vasoconstriction of cerebral arteries. GSNO had no impact on responses to other vasoconstrictors sharing (phenylephrine, U46619) or not (5-HT) the same signalling pathway. MT was reduced by GSNO, and the addition of losartan did not further decrease MT, suggesting that GSNO blocks AT1 receptor-dependent MT. Ascorbate (which reduces S-nitrosated compounds) restored the response to AngII but not the soluble GC inhibitor ODQ, suggesting that these effects are mediated by S-nitrosation rather than by S-nitrosylation. CONCLUSIONS AND IMPLICATIONS In rat middle cerebral arteries, GSNO pretreatment specifically affects the AT1 receptor and reduces both AngII-dependent and AngII-independent activation, most likely through AT1 receptor S-nitrosation.
Collapse
Affiliation(s)
| | - Sandra Lecat
- BSC UMR7242 "GPCRs, pain and inflammation" team, CNRS, Université de Strasbourg Labex Medalis, Illkirch, France
| | | | | | | | | | | |
Collapse
|
34
|
Shakib N, Khadem Ansari MH, Karimi P, Rasmi Y. Neuroprotective mechanism of low-dose sodium nitrite in oxygen-glucose deprivation model of cerebral ischemic stroke in PC12 cells. EXCLI JOURNAL 2019; 18:229-242. [PMID: 31217786 PMCID: PMC6558507 DOI: 10.17179/excli2018-1947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/26/2019] [Indexed: 01/29/2023]
Abstract
The purpose of this study was to clarify the mechanisms of the protective effects of low-dose sodium nitrite (SN) on oxygen and glucose deprivation (OGD)-induced endoplasmic reticulum (ER) stress in PC12 cells. The PC12 cells were exposed to 4 h of OGD and treated with 100 μmol SN. The expression and activity of ER stress markers, including PKR-like endoplasmic reticulum kinase (PERK), transcription factor 6 (ATF6), CCAAT/enhancer binding protein homologous protein (CHOP), as well as caspase-12 and -3, were detected by immunoblotting assay. Fluorescence staining was used to detect the levels of reactive oxygen species (ROS) and Ca2+ release from the ER. Cell viability was also evaluated by MTT assay. It was found that SN significantly inhibited ROS production and Ca2+ release from the ER in OGD-injured PC12 cells. Moreover, ER stress marker expression and cleaved fragments of caspase-3 and -12 in OGD-injured PC12 cells were decreased after SN treatment. These findings were accompanied by a significant increase in cell viability. It seems that SN exerts a neuroprotective effect at least partially through reduction of ROS-mediated ER stress caused by OGD insult.
Collapse
Affiliation(s)
- Nader Shakib
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
35
|
Muthukaman N, Deshmukh S, Tondlekar S, Tambe M, Pisal D, Sarode N, Mhatre S, Chakraborti S, Shah D, Bhosale VM, Kulkarni A, Mahat MYA, Jadhav SB, Gudi GS, Khairatkar-Joshi N, Gharat LA. Discovery of 5-(2-chloro-4'-(1H-imidazol-1-yl)-[1,1'-biphenyl]-4-yl)-1H-tetrazole as potent and orally efficacious S-nitrosoglutathione reductase (GSNOR) inhibitors for the potential treatment of COPD. Bioorg Med Chem Lett 2018; 28:3766-3773. [PMID: 30340896 DOI: 10.1016/j.bmcl.2018.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/28/2018] [Accepted: 10/10/2018] [Indexed: 02/05/2023]
Abstract
Endogenous nitrosothiols (SNOs) including S-nitrosoglutathione (GSNO) serve as reservoir for bioavailable nitric oxide (NO) and mediate NO-based signaling, inflammatory status and smooth muscle function in the lung. GSNOR inhibition increases pulmonary GSNO and induces bronchodilation while reducing inflammation in lung diseases. In this letter, design, synthesis and structure-activity relationships (SAR) of novel imidazole-biaryl-tetrazole based GSNOR inhibitors are described. Many potent inhibitors (30, 39, 41, 42, 44, 45 and 58) were identified with low nanomolar activity (IC50s: <15 nM) along with adequate metabolic stability. Lead compounds 30 and 58 exhibited good exposure and oral bioavailability in mouse pharmacokinetic (PK) study. Compound 30 was selected for further profiling and revealed comparable mouse and rat GSNOR potency, high selectivity against alcohol dehydrogenase (ADH) and carbonyl reductase (CBR1) family of enzymes, low efflux ratio and permeability in PAMPA, a high permeability in CALU-3 assay, significantly low hERG activity and minimal off-target activity. Further, an in vivo efficacy of compound 30 is disclosed in cigarette smoke (CS) induced mouse model for COPD.
Collapse
Affiliation(s)
- Nagarajan Muthukaman
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Sanjay Deshmukh
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Shital Tondlekar
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Macchindra Tambe
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Dnyandeo Pisal
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Neelam Sarode
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Siddharth Mhatre
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Samitabh Chakraborti
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Daisy Shah
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Vikram M Bhosale
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Abhay Kulkarni
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Mahamad Yunnus A Mahat
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Satyawan B Jadhav
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Girish S Gudi
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Neelima Khairatkar-Joshi
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Laxmikant A Gharat
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India.
| |
Collapse
|
36
|
Khan M, Dhammu TS, Singh I, Singh AK. Amelioration of spinal cord injury in rats by blocking peroxynitrite/calpain activity. BMC Neurosci 2018; 19:50. [PMID: 30103682 PMCID: PMC6090709 DOI: 10.1186/s12868-018-0450-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/07/2018] [Indexed: 01/09/2023] Open
Abstract
Background Spinal cord injury (SCI) is one of the leading causes of disability and chronic pain. In SCI-induced pathology, homeostasis of the nitric oxide (NO) metabolome is lost. Major NO metabolites such as S-nitrosoglutathione (GSNO) and peroxynitrite are reported to play pivotal roles in regulating the activities of key cysteine proteases, calpains. While peroxynitrite (a metabolite of NO and superoxide) up regulates the activities of calpains leading to neurodegeneration, GSNO (a metabolite of NO and glutathione) down regulates the activities of calpains leading to neuroprotection. In this study, effect of GSNO on locomotor function and pain threshold and their relationship with the levels of peroxynitrite and the activity of calpain in the injured spinal cord were investigated using a 2-week rat model of contusion SCI.
Results SCI animals were initially treated with GSNO at 2 h after the injury followed by a once daily dose of GSNO for 14 days. Locomotor function was evaluated by “Basso Beattie and Bresnahan (BBB) locomotor rating scale” and pain by mechanical allodynia. Peroxynitrite level, as expression of 3-nitrotyrosine (3-NT), calpain activity, as the degradation products of calpain substrate alpha II spectrin, and nNOS activity, as the expression phospho nNOS, were measured by western blot analysis. Treatment with GSNO improved locomotor function and mitigated pain. The treatment also reduced the levels of peroxynitrite (3-NT) and decreased activity of calpains. Reduced levels of peroxynitrite resulted from the GSNO-mediated inhibition of aberrant activity of neuronal nitric oxide synthase (nNOS). Conclusions The data indicates that higher levels of 3-NT and aberrant activities of nNOS and calpains correlated with SCI pathology and functional deficits. Treatment with GSNO improved locomotor function and mitigated mechanical allodynia acutely post-injury. Because GSNO shows potential to ameliorate experimental SCI, we discuss implications for GSNO therapy in clinical SCI research.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, 508 Children's Research Institute, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, 29425, USA.
| | - Tajinder S Dhammu
- Department of Pediatrics, 508 Children's Research Institute, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, 29425, USA
| | - Inderjit Singh
- Department of Pediatrics, 508 Children's Research Institute, Medical University of South Carolina, 173 Ashley Ave, Charleston, SC, 29425, USA.,Ralph H Johnson VA Medical Center, Charleston, SC, USA
| | - Avtar K Singh
- Ralph H Johnson VA Medical Center, Charleston, SC, USA.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
37
|
Ahmad S, Alam O, Naim MJ, Shaquiquzzaman M, Alam MM, Iqbal M. Pyrrole: An insight into recent pharmacological advances with structure activity relationship. Eur J Med Chem 2018; 157:527-561. [PMID: 30119011 DOI: 10.1016/j.ejmech.2018.08.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/15/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022]
Abstract
Pyrrole is a heterocyclic ring template with multiple pharmacophores that provides a way for the generation of library of enormous lead molecules. Owing to its vast pharmacological profile, pyrrole and its analogues have drawn much attention of the researchers/chemists round the globe to be explored exhaustively for the benefit of mankind. This review focusses on recent advancements; pertaining to pyrrole scaffold, discussing various aspects of structure activity relationship and its bioactivities.
Collapse
Affiliation(s)
- Shujauddin Ahmad
- Medicinal Chemistry and Molecular Modelling Lab, Dept. of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 62, India
| | - Ozair Alam
- Medicinal Chemistry and Molecular Modelling Lab, Dept. of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 62, India.
| | - Mohd Javed Naim
- Medicinal Chemistry and Molecular Modelling Lab, Dept. of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 62, India
| | - Mohammad Shaquiquzzaman
- Medicinal Chemistry and Molecular Modelling Lab, Dept. of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 62, India
| | - M Mumtaz Alam
- Medicinal Chemistry and Molecular Modelling Lab, Dept. of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, 62, India
| | - Muzaffar Iqbal
- Dept. of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
38
|
Khan M, Shunmugavel A, Dhammu TS, Khan H, Singh I, Singh AK. Combined treatment with GSNO and CAPE accelerates functional recovery via additive antioxidant activities in a mouse model of TBI. J Neurosci Res 2018; 96:1900-1913. [PMID: 30027580 DOI: 10.1002/jnr.24279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/01/2018] [Accepted: 06/15/2018] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is the major cause of physical disability and emotional vulnerability. Treatment of TBI is lacking due to its multimechanistic etiology, including derailed mitochondrial and cellular energy metabolism. Previous studies from our laboratory show that an endogenous nitric oxide (NO) metabolite S-nitrosoglutathione (GSNO) provides neuroprotection and improves neurobehavioral function via anti-inflammatory and anti-neurodegenerative mechanisms. To accelerate the rate and enhance the degree of recovery, we investigated combining GSNO with caffeic acid phenethyl ester (CAPE), a potent antioxidant compound, using a male mouse model of TBI, controlled cortical impact in mice. The combination therapy accelerated improvement of cognitive and depressive-like behavior compared with GSNO or CAPE monotherapy. Separately, both GSNO and CAPE improved mitochondrial integrity/function and decreased oxidative damage; however, the combination therapy had greater effects on Drp1 and MnSOD. Additionally, while CAPE alone activated AMPK, this activation was heightened in combination with GSNO. CAPE treatment of normal animals also significantly increased the expression levels of pAMPK, pACC (activation of AMPK substrate ACC), and pLKB1 (activation of upstream to AMPK kinase LKB1), indicating that CAPE activates AMPK via LKB1. These results show that while GSNO and CAPE provide neuroprotection and improve functional recovery separately, the combination treatment invokes greater recovery by significantly improving mitochondrial functions and activating the AMPK enzyme. Both GSNO and CAPE are in human consumption without any known adverse effects; therefore, a combination therapy-based multimechanistic approach is worthy of investigation in human TBI.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | | | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Hamza Khan
- College of Medicine, University of South Carolina, Columbia, South Carolina
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson VA Medical Center, Charleston, South Carolina
| | - Avtar K Singh
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
39
|
Singh I, Nath N, Saxena N, Singh AK, Won JS. Regulation of IL-10 and IL-17 mediated experimental autoimmune encephalomyelitis by S-nitrosoglutathione. Immunobiology 2018; 223:549-554. [PMID: 29960806 DOI: 10.1016/j.imbio.2018.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 06/24/2018] [Indexed: 01/07/2023]
Abstract
In this study, we investigated IL-10 and IL-17 specific immunomodulatory potential of S-nitrosoglutathione (GSNO), a physiological nitric oxide carrier molecule, in experimental autoimmune encephalomyelitis (EAE). In active EAE model, GSNO treatment attenuated EAE severity and splenic CD4+ T cells isolated from these mice exhibited decreased IL-17 expression without affecting the IFN-γ expression compared to the cells from untreated EAE mice. Similarly, adoptive transfer of these cells to nave mice resulted in reduction in IL-17 expression in the spinal cords of recipient mice with milder EAE severity. CD4+ T cells isolated from GSNO treated EAE mice, as compared to untreated EAE mice, still expressed lower levels of IL-17 under TH17 skewing conditions, but expressed similar levels of IFN-γ under TH1 skewing condition. Interestingly, under both TH17 and TH1 skewing condition, CD4+ T cells isolated from GSNO treated EAE mice, as compared to untreated EAE mice, expressed higher levels of IL-10 and adoptive transfer of these TH17 and TH1 skewed cells seemingly exhibited milder EAE disease. In addition, adoptive transfer of CD4+ T cells from GSNO treated EAE mice to active EAE mice also ameliorated EAE disease with induction of spinal cord expression of IL-10 and reduction in of IL-17, thus suggesting the participation of IL-10 mechanism in GSNO mediated immunomodulation. GSNO treatment of mice passively immunized with CD4+ T cells either from GSNO treated EAE mice or untreated mice further ameliorated EAE disease, supporting efficacy of GSNO for prophylaxis and therapy in EAE. Overall, these data document a modulatory role of GSNO in IL-17/IL-10 axis of EAE and other autoimmune diseases.
Collapse
Affiliation(s)
- Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| | - Narender Nath
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Je-Seong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
40
|
Bonetti J, Zhou Y, Parent M, Clarot I, Yu H, Fries-Raeth I, Leroy P, Lartaud I, Gaucher C. Intestinal absorption of S-nitrosothiols: Permeability and transport mechanisms. Biochem Pharmacol 2018; 155:21-31. [PMID: 29935960 DOI: 10.1016/j.bcp.2018.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/19/2018] [Indexed: 12/29/2022]
Abstract
S-Nitrosothiols, a class of NO donors, demonstrate potential benefits for cardiovascular diseases. Drugs for such chronic diseases require long term administration preferentially through the oral route. However, the absorption of S-nitrosothiols by the intestine, which is the first limiting barrier for their vascular bioavailability, is rarely evaluated. Using an in vitro model of intestinal barrier, based on human cells, the present work aimed at elucidating the mechanisms of intestinal transport (passive or active, paracellular or transcellular pathway) and at predicting the absorption site of three S-nitrosothiols: S-nitrosoglutathione (GSNO), S-nitroso-N-acetyl-l-cysteine (NACNO) and S-nitroso-N-acetyl-d-penicillamine (SNAP). These S-nitrosothiols include different skeletons carrying the nitroso group, which confer different physico-chemical characteristics and biological activities (antioxidant and anti-inflammatory). According to the values of apparent permeability coefficient, the three S-nitrosothiols belong to the medium class of permeability. The evaluation of the bidirectional apparent permeability demonstrated a passive diffusion of the three S-nitrosothiols. GSNO and NACNO preferentially cross the intestinal barrier though the transcellular pathway, while SNAP followed both the trans- and paracellular pathways. Finally, the permeability of NACNO was favoured at pH 6.4, which is close to the pH of the jejunal part of the intestine. Through this study, we determined the absorption mechanisms of S-nitrosothiols and postulated that they can be administrated through the oral route.
Collapse
Affiliation(s)
| | - Yi Zhou
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | | | - Igor Clarot
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | - Haiyan Yu
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | | | - Pierre Leroy
- Université de Lorraine, CITHEFOR, F-54000 Nancy, France
| | | | | |
Collapse
|
41
|
Saxena N, Won J, Choi S, Singh AK, Singh I. S-nitrosoglutathione reductase (GSNOR) inhibitor as an immune modulator in experimental autoimmune encephalomyelitis. Free Radic Biol Med 2018; 121:57-68. [PMID: 29694854 PMCID: PMC6083447 DOI: 10.1016/j.freeradbiomed.2018.04.558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Abstract
We previously reported that S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier, attenuated TH17-mediated immune responses in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Cellular GSNO homeostasis is regulated via its synthesis by reaction between nitric oxide and glutathione and its enzymatic catabolism by GSNO reductase (GSNOR). In this study, we evaluated potential of reversible inhibitor of GSNOR (N6022) in comparison with exogenous GSNO in immunopathogenesis of EAE. Daily treatment of EAE mice with N6022 or exogenous GSNO significantly attenuated the clinical disease of EAE, but N6022 treatment showed greater efficacy than GSNO. Both N6022 and exogenous GSNO treatments increased the spleen levels of GSNO, as documented by increased protein-associated S-nitrosothiols, and inhibited polarization and CNS effector function of proinflammatory TH17 cells while inducing the polarization and CNS effector function of anti-inflammatory CD4+ CD25+ FOXP3- regulatory T (Treg) cells. Moreover, N6022 further attenuated TH1 while inducing TH2 and CD4+ CD25+ FOXP3+ Treg in their polarization and CNS effector functions. Similar to GSNO, the N6022 treatment protected against the EAE disease induced demyelination. However, neither exogenous GSNO nor N6022 treatment did not cause significant systemic lymphopenic effect as compared to FTY720. Taken together, these data document that optimization of cellular GSNO homeostasis by GSNOR inhibitor (N6022) in NO metabolizing cells attenuates EAE disease via selective inhibition of pro-inflammatory subsets of CD4+ cells (TH1/TH17) while upregulating anti-inflammatory subsets of CD4+ cells (TH2/Treg) without causing lymphopenic effects and thus offers a potential treatment option for MS/EAE.
Collapse
MESH Headings
- Alcohol Dehydrogenase/antagonists & inhibitors
- Animals
- Benzamides/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Mice
- Mice, Inbred C57BL
- Protein S/metabolism
- Pyrroles/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/drug effects
- Th1 Cells/enzymology
- Th1 Cells/immunology
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Seungho Choi
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| |
Collapse
|
42
|
Aggarwal A, Singh I, Sandhir R. Protective effect of S-nitrosoglutathione administration against hyperglycemia induced disruption of blood brain barrier is mediated by modulation of tight junction proteins and cell adhesion molecules. Neurochem Int 2018; 118:205-216. [PMID: 29792953 DOI: 10.1016/j.neuint.2018.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 04/26/2018] [Accepted: 05/18/2018] [Indexed: 11/30/2022]
Abstract
Diabetes is associated with increased blood brain barrier (BBB) permeability resulting in neurological deficits. The present study investigated the role of S-nitrosoglutathione (GSNO) on tight junction proteins and cell adhesion molecules in streptozotocin-induced diabetic mice. Diabetes was induced by intraperitoneal injection of streptozotocin (40 mg/kg body weight) for 5 days in mice. GSNO was administered daily (100 μg/kg body weight, orally) for 8 weeks after the induction of diabetes. A significant decline was observed in the cognitive ability of diabetic animals assessed using radial arm maze test. A significant increase was observed in nitrotyrosine levels in cortex and hippocampus of diabetic mice. Relative mRNA and protein expression of tight junction proteins viz; zona occludens-1 (ZO-1) and occludin were significantly lower in the microvessels isolated from cortex and hippocampus of diabetic animals, whereas expression of claudin-5 was unaltered. Immunofluorescence of tight junction proteins confirmed loss of ZO-1 and occludin in the diabetic brain. Furthermore, significant increase in interstitial cell adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 mRNA and protein levels was observed in diabetic animals. Ultrastructure of microvessels from diabetic brain was also altered thereby confirming BBB disruption. GSNO administration to diabetic animals, on the other hand, was able to ameliorate loss of ZO-1 and occludin as well as normalize ICAM-1 and VCAM-1 expression, restore BBB integrity, and improve cognitive deficits. The findings clearly suggest that GSNO is a therapeutic molecule with potential to protect BBB and prevent diabetes induced neurological deficits.
Collapse
Affiliation(s)
- Aanchal Aggarwal
- Department of Biochemistry, Basic Medical Science Building, Panjab University, Chandigarh, India
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Science Building, Panjab University, Chandigarh, India.
| |
Collapse
|
43
|
Turan I, Sayan Ozacmak H, Ozacmak VH, Barut F, Ozacmak ID. The effects of S-nitrosoglutathione on intestinal ischemia reperfusion injury and acute lung injury in rats: Roles of oxidative stress and NF-κB. Tissue Cell 2018; 52:35-41. [PMID: 29857826 DOI: 10.1016/j.tice.2018.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/16/2018] [Accepted: 03/24/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal ischemia and reperfusion (I/R) induces oxidative stress, inflammatory response, and acute lung injury. S-nitrosoglutathione (GSNO), a nitric oxide donor, has been documented to have protective effects on experimental ischemia models. AIM The aim of this study was to examine the effect of GSNO on I/R-induced intestine and lung damage and detect the potential mechanisms emphasizing the protective role of GSNO. METHODS Intestinal I/R was induced by occluding the superior mesenteric artery for 30 min followed by reperfusion for 180 min. GSNO was administered intravenously before reperfusion period (0.25 mg/kg). The levels of lipid peroxidation, reduced glutathione, and myeloperoxidase (MPO), histopathological evaluation and immunohistochemical expressions of both nuclear factor KappaB (NF-κB) and inducible nitric oxide (iNOS) in intestine and lung tissues were assessed. RESULTS Histolopathologic evaluation demonstrated that intestinal I/R induced severe damages in the intestine and the lung tissues. Histopathological scores decreased with GSNO treatment. GSNO treatment reduced lipid peroxidation and MPO levels and inhibited expression of NF-κB and iNOS in the intestine. CONCLUSION Our results suggest that GSNO treatment may ameliorate the intestinal and lung injury in rats, at least in part, by inhibiting inflammatory response and oxidative stress.
Collapse
Affiliation(s)
- Inci Turan
- Department of Physiology, Bulent Ecevit University Faculty of Medicine, Turkey.
| | - Hale Sayan Ozacmak
- Department of Physiology, Bulent Ecevit University Faculty of Medicine, Turkey
| | - V Haktan Ozacmak
- Department of Physiology, Bulent Ecevit University Faculty of Medicine, Turkey
| | - Figen Barut
- Department of Pathology, Bulent Ecevit University Faculty of Medicine, Turkey
| | - I Diler Ozacmak
- Or-Ahayim Private Balat Hospital, Department of General surgery, Bulent Ecevit University Faculty of Medicine, Turkey
| |
Collapse
|
44
|
Abstract
Stroke is considered to be an acute cerebrovascular disease, including ischemic stroke and hemorrhagic stroke. The high incidence and poor prognosis of stroke suggest that it is a highly disabling and highly lethal disease which can pose a serious threat to human health. Nitric oxide (NO), a common gas in nature, which is often thought as a toxic gas, because of its intimate relationship with the pathological processes of many diseases, especially in the regulation of blood flow and cell inflammation. However, recent years have witnessed an increased interest that NO plays a significant and positive role in stroke as an essential gas signal molecule. In view of the fact that the neuroprotective effect of NO is closely related to its concentration, cell type and time, only in the appropriate circumstances can NO play a protective effect. The purpose of this review is to summarize the roles of NO in ischemic stroke and hemorrhagic stroke.
Collapse
Affiliation(s)
- Zhou-Qing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ru-Tao Mou
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Dong-Xia Feng
- Department of Scott & White Clinic-Temple, Temple, TX, USA
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
45
|
Benign Effect of Extremely Low-Frequency Electromagnetic Field on Brain Plasticity Assessed by Nitric Oxide Metabolism during Poststroke Rehabilitation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2181942. [PMID: 29138675 PMCID: PMC5613626 DOI: 10.1155/2017/2181942] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/02/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO) is one of the most important signal molecules, involved in both physiological and pathological processes. As a neurotransmitter in the central nervous system, NO regulates cerebral blood flow, neurogenesis, and synaptic plasticity. The aim of our study was to investigate the effect of the extremely low-frequency electromagnetic field (ELF-EMF) on generation and metabolism of NO, as a neurotransmitter, in the rehabilitation of poststroke patients. Forty-eight patients were divided into two groups: ELF-EMF and non-ELF-EMF. Both groups underwent the same 4-week rehabilitation program. Additionally, the ELF-EMF group was exposed to an extremely low-frequency electromagnetic field of 40 Hz, 7 mT, for 15 min/day. Levels of 3-nitrotyrosine, nitrate/nitrite, and TNFα in plasma samples were measured, and NOS2 expression was determined in whole blood samples. Functional status was evaluated before and after a series of treatments, using the Activity Daily Living, Geriatric Depression Scale, and Mini-Mental State Examination. We observed that application of ELF-EMF significantly increased 3-nitrotyrosine and nitrate/nitrite levels, while expression of NOS2 was insignificantly decreased in both groups. The results also show that ELF-EMF treatments improved functional and mental status. We conclude that ELF-EMF therapy is capable of promoting recovery in poststroke patients.
Collapse
|
46
|
Bodas M, Silverberg D, Walworth K, Brucia K, Vij N. Augmentation of S-Nitrosoglutathione Controls Cigarette Smoke-Induced Inflammatory-Oxidative Stress and Chronic Obstructive Pulmonary Disease-Emphysema Pathogenesis by Restoring Cystic Fibrosis Transmembrane Conductance Regulator Function. Antioxid Redox Signal 2017; 27:433-451. [PMID: 28006950 PMCID: PMC5564030 DOI: 10.1089/ars.2016.6895] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Cigarette smoke (CS)-mediated acquired cystic fibrosis transmembrane conductance regulator (CFTR)-dysfunction, autophagy-impairment, and resulting inflammatory-oxidative/nitrosative stress leads to chronic obstructive pulmonary disease (COPD)-emphysema pathogenesis. Moreover, nitric oxide (NO) signaling regulates lung function decline, and low serum NO levels that correlates with COPD severity. Hence, we aim to evaluate here the effects and mechanism(s) of S-nitrosoglutathione (GSNO) augmentation in regulating inflammatory-oxidative stress and COPD-emphysema pathogenesis. RESULTS Our data shows that cystic fibrosis transmembrane conductance regulator (CFTR) colocalizes with aggresome bodies in the lungs of COPD subjects with increasing emphysema severity (Global Initiative for Chronic Obstructive Lung Disease [GOLD] I - IV) compared to nonemphysema controls (GOLD 0). We further demonstrate that treatment with GSNO or S-nitrosoglutathione reductase (GSNOR)-inhibitor (N6022) significantly inhibits cigarette smoke extract (CSE; 5%)-induced decrease in membrane CFTR expression by rescuing it from ubiquitin (Ub)-positive aggresome bodies (p < 0.05). Moreover, GSNO restoration significantly (p < 0.05) decreases CSE-induced reactive oxygen species (ROS) activation and autophagy impairment (decreased accumulation of ubiquitinated proteins in the insoluble protein fractions and restoration of autophagy flux). In addition, GSNO augmentation inhibits protein misfolding as CSE-induced colocalization of ubiquitinated proteins and LC3B (in autophagy bodies) is significantly reduced by GSNO/N6022 treatment. We verified using the preclinical COPD-emphysema murine model that chronic CS (Ch-CS)-induced inflammation (interleukin [IL]-6/IL-1β levels), aggresome formation (perinuclear coexpression/colocalization of ubiquitinated proteins [Ub] and p62 [impaired autophagy marker], and CFTR), oxidative/nitrosative stress (p-Nrf2, inducible nitric oxide synthase [iNOS], and 3-nitrotyrosine expression), apoptosis (caspase-3/7 activity), and alveolar airspace enlargement (Lm) are significantly (p < 0.05) alleviated by augmenting airway GSNO levels. As a proof of concept, we demonstrate that GSNO augmentation suppresses Ch-CS-induced perinuclear CFTR protein accumulation (p < 0.05), which restores both acquired CFTR dysfunction and autophagy impairment, seen in COPD-emphysema subjects. INNOVATION GSNO augmentation alleviates CS-induced acquired CFTR dysfunction and resulting autophagy impairment. CONCLUSION Overall, we found that augmenting GSNO levels controls COPD-emphysema pathogenesis by reducing CS-induced acquired CFTR dysfunction and resulting autophagy impairment and chronic inflammatory-oxidative stress. Antioxid. Redox Signal. 27, 433-451.
Collapse
Affiliation(s)
- Manish Bodas
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - David Silverberg
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Kyla Walworth
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Kathryn Brucia
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan
| | - Neeraj Vij
- 1 College of Medicine, Central Michigan University , Mt. Pleasant, Michigan.,2 Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
47
|
Exogenous S-nitrosoglutathione attenuates inflammatory response and intestinal epithelial barrier injury in endotoxemic rats. J Trauma Acute Care Surg 2017; 80:977-84. [PMID: 26891162 DOI: 10.1097/ta.0000000000001008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Gut barrier injury in sepsis is a major contributor to distant organ dysfunction and bad clinical outcomes. Enteric glia-derived S-nitrosoglutathione (GSNO) has been recognized as a novel modulator of gut barrier integrity. In this study, we tested the potential therapeutic effect and mechanism of exogenous GSNO on endotoxin-induced inflammatory response and intestinal barrier injury in a rat model of endotoxemia. METHODS Male Sprague-Dawley rats were randomly assigned to four groups as follows: control (saline only), GSNO, lipopolysaccharide (LPS), and LPS + GSNO. Femoral venous injection of LPS (10 mg/kg) or saline was followed by GSNO (1 mg/kg) or saline injection 15 minutes later. Distal ileum tissues and blood were harvested after 3 hours of LPS/saline injection. The intestinal barrier function was measured histologically and by intestinal permeability to fluorescein isothiocyanate dextran. The ultrastructural change of the epithelial tight junction was observed using transmission electron microscope, and the expression level of tight junction protein ZO-1 was analyzed using immunofluorescence and Western blot. Systemic and intestinal inflammation was measured by analyzing the tumor necrosis factor and interleukin 1β levels in plasma and distal ileum tissue, respectively. The levels of nuclear factor κB (NF-κB) and myosin light-chain kinase in the distal ileum were measured by Western blot. RESULTS Compared with the endotoxemic rats, the addition of GSNO reduced the intestinal injury observed in histologic sections, decreased permeability to fluorescein isothiocyanate dextran, attenuated damage of the junction between epithelia, and protected against the LPS-induced expression decrease of ZO-1. Furthermore, addition of GSNO reduced plasma and intestinal tumor necrosis factor and interleukin 1β levels as well as inhibited the LPS-induced up-regulation of myosin light-chain kinase expression and NF-κB p65 level in the intestine. CONCLUSION The data indicate that GSNO protects against the LPS-induced systemic inflammatory response and attenuated intestinal inflammation and epithelial barrier injury in rats, possibly through the inhibition of the NF-κB pathway.
Collapse
|
48
|
Arbeláez-Quintero I, Palacios M. To Use or Not to Use Metformin in Cerebral Ischemia: A Review of the Application of Metformin in Stroke Rodents. Stroke Res Treat 2017; 2017:9756429. [PMID: 28634570 PMCID: PMC5467394 DOI: 10.1155/2017/9756429] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/22/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
Ischemic strokes are major causes of death and disability. Searching for potential therapeutic strategies to prevent and treat stroke is necessary, given the increase in overall life expectancy. Epidemiological reports indicate that metformin is an oral antidiabetic medication that can reduce the incidence of ischemic events in patients with diabetes mellitus. Its mechanism of action has not been elucidated, but metformin pleiotropic effects involve actions in addition to glycemic control. AMPK activation has been described as one of the pharmacological mechanisms that explain the action of metformin and that lead to neuroprotective effects. Most experiments done in the cerebral ischemia model, via middle cerebral artery occlusion in rodents (MCAO), had positive results favoring metformin's neuroprotective role and involve several cellular pathways like oxidative stress, endothelial nitric oxide synthase activation, activation of angiogenesis and neurogenesis, autophagia, and apoptosis. We will review the pharmacological properties of metformin and its possible mechanisms that lead to neuroprotection in cerebral ischemia.
Collapse
Affiliation(s)
| | - Mauricio Palacios
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia
| |
Collapse
|
49
|
Li D, Luo L, Xu M, Wu J, Chen L, Li J, Liu Z, Lu G, Wang Y, Qiao L. AMPK activates FOXO3a and promotes neuronal apoptosis in the developing rat brain during the early phase after hypoxia-ischemia. Brain Res Bull 2017; 132:1-9. [PMID: 28499802 DOI: 10.1016/j.brainresbull.2017.05.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/29/2017] [Accepted: 05/05/2017] [Indexed: 12/21/2022]
Abstract
AMP-activated protein kinase (AMPK) is a key metabolic and stress sensor/effector. Few investigations have been performed to study the role of AMPK in developing rat brain with hypoxia-ischemia (HI). Forkhead transcriptional factor (FOXO3a) has been revealed to be a critical effector of AMPK-mediated celluar apoptosis. However, it is not clear whether AMPK/FOXO3a pathway is involved in neuronal apoptosis in the developing rat brain after HI. In this study, we generated hypoxia-ischemia brain damage (HIBD) model using postnatal day 7 rats. We found that activation of AMPK was accompanied by the decrease of p-mTOR, p-Akt and p-FOXO3a, which induced FOXO3a translocation into the nucleus and up-regulated the expression of Bim and cleaved caspase 3 (CC3). Furthermore, we discovered that AMPK inhibition by Compound C, a selective inhibitor for AMPK activity, significantly increased the phosphorylation levels of mTOR, Akt and FOXO3a, attenuated the nuclear translocation of FOXO3a, and inhibited Bim and CC3 expression after HI. Moreover, AMPK inhibition reduced cellular apoptosis, attenuated brain infarct volume and promoted neurological recovery in the developing rat brain after HI. Our findings suggest that AMPK participates in the regulation of FOXO3a-mediated neuronal apoptosis in the developing rat brain after HI. Agents targeting AMPK may offer promise for rescuing neurons from HI-induced damage.
Collapse
Affiliation(s)
- Deyuan Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Lili Luo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Min Xu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Jinlin Wu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Lina Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Zhongqiang Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Guoyan Lu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Yang Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China
| | - Lina Qiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University), Ministry of Education, Chengdu,Sichuan 610041, China.
| |
Collapse
|
50
|
Khan M, Khan H, Singh I, Singh AK. Hypoxia inducible factor-1 alpha stabilization for regenerative therapy in traumatic brain injury. Neural Regen Res 2017; 12:696-701. [PMID: 28616019 PMCID: PMC5461600 DOI: 10.4103/1673-5374.206632] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Mild traumatic brain injury (TBI), also called concussion, initiates sequelae leading to motor deficits, cognitive impairments and subtly compromised neurobehaviors. While the acute phase of TBI is associated with neuroinflammation and nitroxidative burst, the chronic phase shows a lack of stimulation of the neurorepair process and regeneration. The deficiency of nitric oxide (NO), the consequent disturbed NO metabolome, and imbalanced mechanisms of S-nitrosylation are implicated in blocking the mechanisms of neurorepair processes and functional recovery in the both phases. Hypoxia inducible factor-1 alpha (HIF-1α), a master regulator of hypoxia/ischemia, stimulates the process of neurorepair and thus aids in functional recovery after brain trauma. The activity of HIF-1α is regulated by NO via the mechanism of S-nitrosylation of HIF-1α. S-nitrosylation is dynamically regulated by NO metabolites such as S-nitrosoglutathione (GSNO) and peroxynitrite. GSNO stabilizes, and peroxynitrite destabilizes HIF-1α. Exogenously administered GSNO was found not only to stabilize HIF-1α and to induce HIF-1α-dependent genes but also to stimulate the regeneration process and to aid in functional recovery in TBI animals.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Hamza Khan
- College of Medicine, University of South Carolina, Columbia, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.,Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|