1
|
Liang F, Wang M, Li J, Guo J. The evolution of S-nitrosylation detection methodology and the role of protein S-nitrosylation in various cancers. Cancer Cell Int 2024; 24:408. [PMID: 39702281 DOI: 10.1186/s12935-024-03568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024] Open
Abstract
S-nitrosylation (SNO) modification, a nitric oxide (NO)-mediated post-translational modification (PTM) of proteins, plays an important role in protein microstructure, degradation, activity, and stability. Due to the presence of reducing agents, the SNO modification process mediated by NO derivatives is often reversible and unstable. This reversible transformation between SNO modification and denitrification often influences the structure, activity, and function of proteins. The reversibility of SNO modifications also poses a challenge when verifying changes in the biological functions of proteins. Moreover, SNO modification of key signaling pathway proteins, such as caspase-3, NF-κB, and Bcl-2, can affect tumor proliferation, invasion, and apoptosis. The SNO-modified proteins play important roles in both promoting and inhibiting cancer, which indirectly confirms the duality and complexity of SNO modification functions. This article reviews the biological significance of various SNO-modified proteins in different cancers, providing a theoretical basis for determining whether the related changes of SNO-modified proteins are universal in cancers. Additionally, this review presents a comprehensive and detailed summary of the evolution of detection methods for SNO-modified proteins, providing a possible methodological basis for future research on SNO-modified proteins.
Collapse
Affiliation(s)
- Feng Liang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Min Wang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jie Guo
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Oh CK, Nakamura T, Zhang X, Lipton SA. Redox regulation, protein S-nitrosylation, and synapse loss in Alzheimer's and related dementias. Neuron 2024; 112:3823-3850. [PMID: 39515322 PMCID: PMC11624102 DOI: 10.1016/j.neuron.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/12/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Redox-mediated posttranslational modification, as exemplified by protein S-nitrosylation, modulates protein activity and function in both health and disease. Here, we review recent findings that show how normal aging, infection/inflammation, trauma, environmental toxins, and diseases associated with protein aggregation can each trigger excessive nitrosative stress, resulting in aberrant protein S-nitrosylation and hence dysfunctional protein networks. These redox reactions contribute to the etiology of multiple neurodegenerative disorders as well as systemic diseases. In the CNS, aberrant S-nitrosylation reactions of single proteins or, in many cases, interconnected networks of proteins lead to dysfunctional pathways affecting endoplasmic reticulum (ER) stress, inflammatory signaling, autophagy/mitophagy, the ubiquitin-proteasome system, transcriptional and enzymatic machinery, and mitochondrial metabolism. Aberrant protein S-nitrosylation and transnitrosylation (transfer of nitric oxide [NO]-related species from one protein to another) trigger protein aggregation, neuronal bioenergetic compromise, and microglial phagocytosis, all of which contribute to the synapse loss that underlies cognitive decline in Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Chang-Ki Oh
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tomohiro Nakamura
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xu Zhang
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
3
|
Liu S, Lu Q, Wang M, Guo H, Wang Y, Nong J, Wang S, Xia H, Xia T, Sun H. S-nitrosoglutathione reductase-dependent p65 denitrosation promotes osteoclastogenesis by facilitating recruitment of p65 to NFATc1 promoter. Bone 2024; 181:117036. [PMID: 38311303 DOI: 10.1016/j.bone.2024.117036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/26/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Osteoclasts, the exclusive bone resorptive cells, are indispensable for bone remodeling. Hence, understanding novel signaling modulators regulating osteoclastogenesis is clinically important. Nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) is a master transcription factor in osteoclastogenesis, and binding of NF-κB p65 subunit to NFATc1 promoter is required for its expression. It is well-established that DNA binding activity of p65 can be regulated by various post-translational modifications, including S-nitrosation. Recent studies have demonstrated that S-nitrosoglutathione reductase (GSNOR)-mediated protein denitrosation participated in cell fate commitment by regulating gene transcription. However, the role of GSNOR in osteoclastogenesis remains unexplored and enigmatic. Here, we investigated the effect of GSNOR-mediated denitrosation of p65 on osteoclastogenesis. Our results revealed that GSNOR was up-regulated during osteoclastogenesis in vitro. Moreover, GSNOR inhibition with a chemical inhibitor impaired osteoclast differentiation, podosome belt formation, and bone resorption activity. Furthermore, GSNOR inhibition enhanced the S-nitrosation level of p65, precluded the binding of p65 to NFATc1 promoter, and suppressed NFATc1 expression. In addition, mouse model of lipopolysaccharides (LPS)-induced calvarial osteolysis was employed to evaluate the therapeutic effect of GSNOR inhibitor in vivo. Our results indicated that GSNOR inhibitor treatment alleviated the inflammatory bone loss by impairing osteoclast formation in mice. Taken together, these data have shown that GSNOR activity is required for osteoclastogenesis by facilitating binding of p65 to NFATc1 promoter via promoting p65 denitrosation, suggesting that GSNOR may be a potential therapeutic target in the treatment of osteolytic diseases.
Collapse
Affiliation(s)
- Shumin Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qian Lu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Min Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Huilin Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yiwen Wang
- School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jingwen Nong
- School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Shuo Wang
- School of Stomatology, Wuhan University, Wuhan 430079, China
| | - Haibin Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ting Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Huifang Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Center for Prosthodontics and Implant Dentistry, Optics Valley Branch, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
4
|
Saini S, Sharma P, Singh P, Kumar V, Yadav P, Sharma A. Nitric oxide: An emerging warrior of plant physiology under abiotic stress. Nitric Oxide 2023; 140-141:58-76. [PMID: 37848156 DOI: 10.1016/j.niox.2023.10.001] [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: 06/26/2023] [Revised: 09/05/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023]
Abstract
The natural environment of plants comprises a complex set of various abiotic stresses and their capability to react and survive under this anticipated changing climate is highly flexible and involves a series of balanced interactions between signaling molecules where nitric oxide becomes a crucial component. In this article, we focussed on the role of nitric oxide (NO) in various signal transduction pathways of plants and its positive impact on maintaining cellular homeostasis under various abiotic stresses. Besides this, the recent data on interactions of NO with various phytohormones to control physiological and biochemical processes to attain abiotic stress tolerance have also been considered. These crosstalks modulate the plant's defense mechanism and help in alleviating the negative impact of stress. While focusing on the diverse functions of NO, an effort has been made to explore the functions of NO-mediated post-translational modifications, such as the N-end rule pathway, tyrosine nitration, and S-nitrosylation which revealed the exact mechanism and characterization of proteins that modify various metabolic processes in stressed conditions. Considering all of these factors, the present review emphasizes the role of NO and its interlinking with various phytohormones in maintaining developmental processes in plants, specifically under unfavorable environments.
Collapse
Affiliation(s)
- Sakshi Saini
- Department of Botany, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Priyanka Sharma
- Department of Botany, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Pooja Singh
- Department of Botany, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Vikram Kumar
- Department of Botany, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Priya Yadav
- Department of Botany, Zakir Husain Delhi College, University of Delhi, New Delhi, India.
| | - Asha Sharma
- Department of Botany, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
5
|
Panneerselvan P, Vasanthakumar K, Muthuswamy K, Krishnan V, Subramaniam S. Insights on the functional dualism of nitric oxide in the hallmarks of cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:189001. [PMID: 37858621 DOI: 10.1016/j.bbcan.2023.189001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023]
Abstract
Nitric oxide (NO), a gaseous radical, governs a variety of physiological and pathological processes, including cancer, pro-inflammatory signalling, and vasodilation. The family of nitric oxide synthases (NOS), which comprises the constitutive forms, nNOS and eNOS, and the inducible form, iNOS, produces NO enzymatically. Additionally, NO can be generated non-enzymatically from the nitrate-nitrite-NO pathway. The anti- and pro-oxidant properties of NO and its functional dualism in cancer is due to its highly reactive nature. Numerous malignancies have NOS expression, which interferes with the tumour microenvironment to modulate the tumour's growth in both favourable and unfavourable ways. NO regulates a number of mechanisms in the tumour microenvironment, including metabolism, cell cycle, DNA repair, angiogenesis, and apoptosis/necrosis, depending on its concentration and spatiotemporal profile. This review focuses on the bi-modal impact of nitric oxide on the alteration of a few cancer hallmarks.
Collapse
Affiliation(s)
- Prabha Panneerselvan
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Keerthana Vasanthakumar
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Karthi Muthuswamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Vasanth Krishnan
- Molecular Biology Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Selvakumar Subramaniam
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India.
| |
Collapse
|
6
|
Azizi SA, Qiu T, Brookes NE, Dickinson BC. Regulation of ERK2 activity by dynamic S-acylation. Cell Rep 2023; 42:113135. [PMID: 37715953 PMCID: PMC10591828 DOI: 10.1016/j.celrep.2023.113135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/28/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023] Open
Abstract
Extracellular signal-regulated kinases (ERK1/2) are key effector proteins of the mitogen-activated protein kinase pathway, choreographing essential processes of cellular physiology. Here, we discover that ERK1/2 are subject to S-acylation, a reversible lipid modification of cysteine residues, at C271/C254. The levels of ERK1/2 S-acylation are modulated by epidermal growth factor (EGF) signaling, mirroring its phosphorylation dynamics, and acylation-deficient ERK2 displays altered phosphorylation patterns. We show that ERK1/2 S-acylation is mediated by "writer" protein acyl transferases (PATs) and "eraser" acyl protein thioesterases (APTs) and that chemical inhibition of either lipid addition or removal alters ERK1/2's EGF-triggered transcriptional program. Finally, in a mouse model of metabolic syndrome, we find that ERK1/2 lipidation levels correlate with alterations in ERK1/2 lipidation writer/eraser expression, solidifying a link between ERK1/2 activity, ERK1/2 lipidation, and organismal health. This study describes how lipidation regulates ERK1/2 and offers insight into the role of dynamic S-acylation in cell signaling more broadly.
Collapse
Affiliation(s)
- Saara-Anne Azizi
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Medical Scientist Training Program, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Tian Qiu
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Noah E Brookes
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Bryan C Dickinson
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
7
|
Low temperature plasma suppresses proliferation, invasion, migration and survival of SK-BR-3 breast cancer cells. Mol Biol Rep 2023; 50:2025-2031. [PMID: 36538172 DOI: 10.1007/s11033-022-08026-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Low temperature plasma (LTP) is a developing field in recent years to play important roles of sterilization, material modification and wound healing. Breast cancer is a common gynecological malignant tumor. Recent studies have shown that LTP is a promising selective anti-cancer treatment. The effect of LTP on breast cancer is still unclear. In this study, We treated breast cancer cell lines with low temperature plasma for different periods of time and analyzed the relevant differences. METHODS AND RESULTS SK-BR-3 cell nutrient solution was firstly treated by ACP for 0, 10, 20, 40, 80 and 120 s, which was next used to cultivateSK-BR-3cells for overnight.we found that LTP was able to suppress cell vitality, proliferation, invasion and migration of SK-BR-3 cells. Also, SK-BR-3 apoptosis was induced by LTP in a time-dependent manner. CONCLUSION These evidences suggest the negative effect of LTP on malignant development of SK-BR-3 cells, and LTP has the potential clinical application for breast cancer treatment.
Collapse
|
8
|
Sadeghi MA, Hemmati S, Nassireslami E, Yousefi Zoshk M, Hosseini Y, Abbasian K, Chamanara M. Targeting neuronal nitric oxide synthase and the nitrergic system in post-traumatic stress disorder. Psychopharmacology (Berl) 2022; 239:3057-3082. [PMID: 36029333 DOI: 10.1007/s00213-022-06212-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/04/2022] [Indexed: 12/22/2022]
Abstract
RATIONALE Current pharmacological approaches to treatment of post-traumatic stress disorder (PTSD) lack adequate effectiveness. As a result, identifying new molecular targets for drug development is necessary. Furthermore, fear learning and memory in PTSD can undergo different phases, such as fear acquisition, consolidation, and extinction. Each phase may involve different cellular pathways and brain regions. As a result, effective management of PTSD requires mindfulness of the timing of drug administration. One of the molecular targets currently under intense investigation is the N-methyl-D-aspartate (NMDA)-type glutamate receptor (NMDAR). However, despite the therapeutic efficacy of drugs targeting NMDAR, their translation into clinical use has been challenging due to their various side effects. One possible solution to this problem is to target signaling proteins downstream to NMDAR to improve targeting specificity. One of these proteins is the neuronal nitric oxide synthase (nNOS), which is activated following calcium influx through the NMDAR. OBJECTIVE In this paper, we review the literature on the pharmacological modulation of nNOS in animal models of PTSD to evaluate its therapeutic potential. Furthermore, we attempt to decipher the inconsistencies observed between the findings of these studies based on the specific phase of fear learning which they had targeted. RESULTS Inhibition of nNOS may inhibit fear acquisition and recall, while not having a significant effect on fear consolidation and extinction. However, it may improve extinction consolidation or reconsolidation blockade. CONCLUSIONS Modulation of nNOS has therapeutic potential against PTSD and warrants further development for use in the clinical setting.
Collapse
Affiliation(s)
- Mohammad Amin Sadeghi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Hemmati
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Yasaman Hosseini
- Cognitive Neuroscience Center, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Kourosh Abbasian
- Management and Health Economics Department, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran. .,Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Nguyen PT, Wacker T, Brown AJP, da Silva Dantas A, Shekhova E. Understanding the Role of Nitronate Monooxygenases in Virulence of the Human Fungal Pathogen Aspergillus fumigatus. J Fungi (Basel) 2022; 8:736. [PMID: 35887491 PMCID: PMC9323177 DOI: 10.3390/jof8070736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 12/23/2022] Open
Abstract
Aspergillus fumigatus is the leading cause of the fungal invasive disease called aspergillosis, which is associated with a high mortality rate that can reach 50% in some groups of immunocompromised individuals. The increasing prevalence of azole-resistant A. fumigatus isolates, both in clinical settings and the environment, highlights the importance of discovering new fungal virulence factors that can potentially become targets for novel antifungals. Nitronate monooxygenases (Nmos) represent potential targets for antifungal compounds as no orthologs of those enzymes are present in humans. Nmos catalyse the denitrification of nitroalkanes, thereby detoxifying these mediators of nitro-oxidative stress, and therefore we tested whether Nmos provide protection for A. fumigatus against host-imposed stresses at sites of infection. The results of inhibition zone assays indicated that Nmo2 and Nmo5 are not essential for the oxidative stress resistance of A. fumigatus in vitro. In addition, the resazurin-based metabolic activity assay revealed that the growth of mutants lacking the nmo2 or nmo5 genes was only slightly reduced in the presence of 0.05 mM peroxynitrite. Nevertheless, both Nmo2 and Nmo5 were shown to contribute to defense against murine bone marrow-derived macrophages, and this was no longer observed when NADPH oxidase, the main generator of reactive oxygen species during infection, was inhibited in macrophages. Furthermore, we revealed that Nnmos promote the virulence of the fungus in the Galleria mellonella model of infection. Both nmo2 and nmo5 knock-out strains were less virulent than the wild-type control as recorded 72 h post-infection. Our results indicate that Nmos play a role in the virulence of A. fumigatus.
Collapse
|
10
|
Morris G, Walder K, Berk M, Carvalho AF, Marx W, Bortolasci CC, Yung AR, Puri BK, Maes M. Intertwined associations between oxidative and nitrosative stress and endocannabinoid system pathways: Relevance for neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2022; 114:110481. [PMID: 34826557 DOI: 10.1016/j.pnpbp.2021.110481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/19/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022]
Abstract
The endocannabinoid system (ECS) appears to regulate metabolic, cardiovascular, immune, gastrointestinal, lung, and reproductive system functions, as well as the central nervous system. There is also evidence that neuropsychiatric disorders are associated with ECS abnormalities as well as oxidative and nitrosative stress pathways. The goal of this mechanistic review is to investigate the mechanisms underlying the ECS's regulation of redox signalling, as well as the mechanisms by which activated oxidative and nitrosative stress pathways may impair ECS-mediated signalling. Cannabinoid receptor (CB)1 activation and upregulation of brain CB2 receptors reduce oxidative stress in the brain, resulting in less tissue damage and less neuroinflammation. Chronically high levels of oxidative stress may impair CB1 and CB2 receptor activity. CB1 activation in peripheral cells increases nitrosative stress and inducible nitric oxide (iNOS) activity, reducing mitochondrial activity. Upregulation of CB2 in the peripheral and central nervous systems may reduce iNOS, nitrosative stress, and neuroinflammation. Nitrosative stress may have an impact on CB1 and CB2-mediated signalling. Peripheral immune activation, which frequently occurs in response to nitro-oxidative stress, may result in increased expression of CB2 receptors on T and B lymphocytes, dendritic cells, and macrophages, reducing the production of inflammatory products and limiting the duration and intensity of the immune and oxidative stress response. In conclusion, high levels of oxidative and nitrosative stress may compromise or even abolish ECS-mediated redox pathway regulation. Future research in neuropsychiatric disorders like mood disorders and deficit schizophrenia should explore abnormalities in these intertwined signalling pathways.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Wolf Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| | - Alison R Yung
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia; School of Health Science, University of Manchester, UK.
| | - Basant K Puri
- University of Winchester, UK, and C.A.R., Cambridge, UK.
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
11
|
Pillars and Gaps of S-Nitrosylation-Dependent Epigenetic Regulation in Physiology and Cancer. Life (Basel) 2021; 11:life11121424. [PMID: 34947954 PMCID: PMC8704633 DOI: 10.3390/life11121424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Nitric oxide (NO) is a diffusible signaling molecule produced by three isoforms of nitric oxide synthase, which release NO during the metabolism of the amino acid arginine. NO participates in pathophysiological responses of many different tissues, inducing concentration-dependent effect. Indeed, while low NO levels generally have protective effects, higher NO concentrations induce cytotoxic/cytostatic actions. In recent years, evidences have been accumulated unveiling S-nitrosylation as a major NO-dependent post-translational mechanism ruling gene expression. S-nitrosylation is a reversible, highly regulated phenomenon in which NO reacts with one or few specific cysteine residues of target proteins generating S-nitrosothiols. By inducing this chemical modification, NO might exert epigenetic regulation through direct effects on both DNA and histones as well as through indirect actions affecting the functions of transcription factors and transcriptional co-regulators. In this light, S-nitrosylation may also impact on cancer cell gene expression programs. Indeed, it affects different cell pathways and functions ranging from the impairment of DNA damage repair to the modulation of the activity of signal transduction molecules, oncogenes, tumor suppressors, and chromatin remodelers. Nitrosylation is therefore a versatile tool by which NO might control gene expression programs in health and disease.
Collapse
|
12
|
Tettey A, Jiang Y, Li X, Li Y. Therapy for Pulmonary Arterial Hypertension: Glance on Nitric Oxide Pathway. Front Pharmacol 2021; 12:767002. [PMID: 34867394 PMCID: PMC8633825 DOI: 10.3389/fphar.2021.767002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with a resultant increase of the mean pulmonary arterial pressure, right ventricular hypertrophy and eventual death. Research in recent years has produced various therapeutic options for its clinical management but the high mortality even under treatment remains a big challenge attributed to the complex pathophysiology. Studies from clinical and non-clinical experiments have revealed that the nitric oxide (NO) pathway is one of the key pathways underlying the pathophysiology of PAH. Many of the essential drugs used in the management of PAH act on this pathway highlighting its significant role in PAH. Meanwhile, several novel compounds targeting on NO pathway exhibits great potential to become future therapy medications. Furthermore, the NO pathway is found to interact with other crucial pathways. Understanding such interactions could be helpful in the discovery of new drug that provide better clinical outcomes.
Collapse
Affiliation(s)
- Abraham Tettey
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Yujie Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| |
Collapse
|
13
|
Sharma V, Fernando V, Letson J, Walia Y, Zheng X, Fackelman D, Furuta S. S-Nitrosylation in Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22094600. [PMID: 33925645 PMCID: PMC8124305 DOI: 10.3390/ijms22094600] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
S-nitrosylation is a selective and reversible post-translational modification of protein thiols by nitric oxide (NO), which is a bioactive signaling molecule, to exert a variety of effects. These effects include the modulation of protein conformation, activity, stability, and protein-protein interactions. S-nitrosylation plays a central role in propagating NO signals within a cell, tissue, and tissue microenvironment, as the nitrosyl moiety can rapidly be transferred from one protein to another upon contact. This modification has also been reported to confer either tumor-suppressing or tumor-promoting effects and is portrayed as a process involved in every stage of cancer progression. In particular, S-nitrosylation has recently been found as an essential regulator of the tumor microenvironment (TME), the environment around a tumor governing the disease pathogenesis. This review aims to outline the effects of S-nitrosylation on different resident cells in the TME and the diverse outcomes in a context-dependent manner. Furthermore, we will discuss the therapeutic potentials of modulating S-nitrosylation levels in tumors.
Collapse
|
14
|
Sadeghi MA, Hemmati S, Mohammadi S, Yousefi-Manesh H, Vafaei A, Zare M, Dehpour AR. Chronically altered NMDAR signaling in epilepsy mediates comorbid depression. Acta Neuropathol Commun 2021; 9:53. [PMID: 33762011 PMCID: PMC7992813 DOI: 10.1186/s40478-021-01153-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Depression is the most common psychiatric comorbidity of epilepsy. However, the molecular pathways underlying this association remain unclear. The NMDA receptor (NMDAR) may play a role in this association, as its downstream signaling has been shown to undergo long-term changes following excitotoxic neuronal damage. To study this pathway, we used an animal model of fluoxetine-resistant epilepsy-associated depression (EAD). We determined the molecular changes associated with the development of depressive symptoms and examined their response to various combinations of fluoxetine and a selective neuronal nitric oxide synthase inhibitor, 7-nitroindazole (NI). Depressive symptoms were determined using the forced swim test. Furthermore, expression and phosphorylation levels of markers in the ERK/CREB/ELK1/BDNF/cFOS pathway were measured to determine the molecular changes associated with these symptoms. Finally, oxidative stress markers were measured to more clearly determine the individual contributions of each treatment. While chronic fluoxetine (Flxc) and NI were ineffective alone, their combination had a statistically significant synergistic effect in reducing depressive symptoms. The development of depressive symptoms in epileptic rats was associated with the downregulation of ERK2 expression and ELK1 and CREB phosphorylation. These changes were exactly reversed upon Flxc + NI treatment, which led to increased BDNF and cFOS expression as well. Interestingly, ERK1 did not seem to play a role in these experiments. NI seemed to have augmented Flxc’s antidepressant activity by reducing oxidative stress. Our findings suggest NMDAR signaling alterations are a major contributor to EAD development and a potential target for treating conditions associated with underlying excitotoxic neuronal damage.
Collapse
|
15
|
Exploiting S-nitrosylation for cancer therapy: facts and perspectives. Biochem J 2021; 477:3649-3672. [PMID: 33017470 DOI: 10.1042/bcj20200064] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
S-nitrosylation, the post-translational modification of cysteines by nitric oxide, has been implicated in several cellular processes and tissue homeostasis. As a result, alterations in the mechanisms controlling the levels of S-nitrosylated proteins have been found in pathological states. In the last few years, a role in cancer has been proposed, supported by the evidence that various oncoproteins undergo gain- or loss-of-function modifications upon S-nitrosylation. Here, we aim at providing insight into the current knowledge about the role of S-nitrosylation in different aspects of cancer biology and report the main anticancer strategies based on: (i) reducing S-nitrosylation-mediated oncogenic effects, (ii) boosting S-nitrosylation to stimulate cell death, (iii) exploiting S-nitrosylation through synthetic lethality.
Collapse
|
16
|
Luo C, Ji D, Li Y, Cao Y, Zhang S, Yan W, Xue K, Chai J, Wu Y, Liu H, Wang W. Abnormal nitration and S-sulfhydration modification of Sp1-CSE-H 2S pathway trap the progress of hyperhomocysteinemia into a vicious cycle. Free Radic Biol Med 2021; 164:20-33. [PMID: 33418108 DOI: 10.1016/j.freeradbiomed.2020.12.440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 12/01/2020] [Accepted: 12/25/2020] [Indexed: 12/13/2022]
Abstract
Sp1-CSE-H2S pathway plays an important role in homocysteine-metabolism, whose disorder can result in hyperhomocysteinemia. H2S deficiency in hyperhomocysteinemia has been reported, while the underlying mechanism and whether it in turn affects the progress of hyperhomocysteinemia are unclear. This study focused on the post-translational modification of Sp1/CSE and revealed four major findings: (1) Homocysteine-accumulation augmented CSE's nitration, inhibited its bio-activity, thus caused H2S deficiency. (2) H2S deficiency inhibited the S-sulfhydration of Sp1, down-regulated CSE and decreased H2S further, which in turn weakened CSE's own S-sulfhydration. (3) CSE was S-sulfhydrated at Cys84, Cys109, Cys172, Cys229, Cys252, Cys307 and Cys310, among which the S-sulfhydration of Cys172 and Cys310 didn't affect its enzymatic activity, while the S-sulfhydration of Cys84, Cys109, Cys229, Cys252 and Cys307 was necessary for its bio-activity. (4) H2S deficiency trapped homocysteine-metabolism into a vicious cycle, which could be broken by either blocking nitration or restoring S-sulfhydration. This study detected a new mechanism that caused severe hyperhomocysteinemia, thereby provided new therapeutic strategies for hyperhomocysteinemia.
Collapse
Affiliation(s)
- Chenghua Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Dengyu Ji
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Yan Li
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yan Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Shangyue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Wenjing Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Ke Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Jiayin Chai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China.
| |
Collapse
|
17
|
Vivarelli S, Falzone L, Basile MS, Candido S, Libra M. Nitric Oxide in Hematological Cancers: Partner or Rival? Antioxid Redox Signal 2021; 34:383-401. [PMID: 32027171 DOI: 10.1089/ars.2019.7958] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Significance: Hematological malignancies represent the fourth most diagnosed cancer. Relapse and acquired resistance to anticancer therapy constitute two actual issues that need to be overcome. Nitric oxide (NO) plays a pivotal role in regulating cancer progression. At present, many studies are attempting to uncover the potentials of modulating NO levels to improve the efficacy of currently available treatments against lymphoma, leukemia, and myeloma. Recent Advances: It is becoming progressively clear that NO modulation may help hematological cancer management, either by targeting directly tumor cells or by driving the immune system to eliminate cancer cells. Critical Issues: NO is a dual molecule that can have a tumor-protecting or stimulating effect, depending on its local concentration. Moreover, NO is able to target a wide range of molecules involved in both cancer genesis and evolution. In this review, an overview of the recent findings regarding the pivotal role played by NO and nitric oxide synthase in cancer progression and anticancer therapy is presented, with particular focus on hematological malignancies. Future Directions: It is critical to establish the cancer-specific function of NO and critically drive its modulation to improve cancer management toward a personalized approach. This has a special importance in hematological tumors, where the urgency of finding eradicative therapies is constant. Antioxid. Redox Signal. 34, 383-401.
Collapse
Affiliation(s)
- Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", Napoli, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Research Centre for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Research Centre for Prevention, Diagnosis and Treatment of Cancer, University of Catania, Catania, Italy
| |
Collapse
|
18
|
Placental Adaptive Changes to Protect Function and Decrease Oxidative Damage in Metabolically Healthy Maternal Obesity. Antioxidants (Basel) 2020; 9:antiox9090794. [PMID: 32859037 PMCID: PMC7555720 DOI: 10.3390/antiox9090794] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Pregnancy-related disorders, including preeclampsia and gestational diabetes, are characterized by the presence of an adverse intrauterine milieu that may ultimately result in oxidative and nitrosative stress. This scenario may trigger uncontrolled production of reactive oxygen species (ROS) such as superoxide anion (O●−) and reactive nitrogen species (RNS) such as nitric oxide (NO), along with an inactivation of antioxidant systems, which are associated with the occurrence of relevant changes in placental function through recognized redox post-translational modifications in key proteins. The general objective of this study was to assess the impact of a maternal obesogenic enviroment on the regulation of the placental nitroso-redox balance at the end of pregnancy. We measured oxidative damage markers—thiobarbituric acid-reacting substances (TBARS) and carbonyl groups (C=O) levels; nitrosative stress markers—inducible nitric oxide synthase, nitrosothiol groups, and nitrotyrosine residues levels; and the antioxidant biomarkers—catalase and superoxide dismutase (SOD) activity and expression, and total antioxidant capacity (TAC), in full-term placental villous from both pre-pregnancy normal weight and obese women, and with absence of metabolic complications throughout gestation. The results showed a decrease in C=O and TBARS levels in obese pregnancies. Although total SOD and catalase concentrations were shown to be increased, both activities were significantly downregulated in obese pregnancies, along with total antioxidant capacity. Inducible nitric oxide sintase levels were increased in the obese group compared to the lean group, accompanied by an increase in nitrotyrosine residues levels and lower levels of nitrosothiol groups in proteins such as ERK1/2. These findings reveal a reduction in oxidative damage, accompanied by a decline in antioxidant response, and an increase via NO-mediated nitrative stress in placental tissue from metabolically healthy pregnancies with obesity. All this plausibly points to a placental adaptation of the affected antioxidant response towards a NO-induced alternative pathway, through changes in the ROS/RNS balance, in order to reduce oxidative damage and preserve placental function in pregnancy.
Collapse
|
19
|
Mizuta Y, Tokuda K, Guo J, Zhang S, Narahara S, Kawano T, Murata M, Yamaura K, Hoka S, Hashizume M, Akahoshi T. Sodium thiosulfate prevents doxorubicin-induced DNA damage and apoptosis in cardiomyocytes in mice. Life Sci 2020; 257:118074. [PMID: 32673667 DOI: 10.1016/j.lfs.2020.118074] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/23/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
AIM Doxorubicin (DOX) induces dose-dependent cardiotoxicity due to reactive oxygen species (ROS)-mediated oxidative stress and subsequent apoptosis of cardiomyocytes. We aimed to assess whether sodium thiosulfate (STS), which has antioxidant and antiapoptotic properties, exerts cardioprotective effects on DOX-induced cardiomyopathy. MAIN METHODS Male C57BL/6N mice were divided into four groups, control, DOX, STS, and DOX + STS, and administered DOX (20 or 30 mg/kg) or normal saline intraperitoneally, followed by an injection of STS (2 g/kg) or normal saline 4 h later. KEY FINDINGS The DOX group showed a poorer 6-day survival and decreased cardiac function than the DOX + STS group. The DOX group showed a marked increase in the plasma creatine kinase isoenzyme myocardial band (CK-MB) and lactate dehydrogenase (LDH) levels 10 h after DOX injection, while the DOX + STS group showed suppression of DOX-induced elevation of CK-MB and LDH levels. The DOX group showed increased 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels in the heart, whereas the DOX + STS group showed increased catalase and superoxide dismutase (SOD) activities and decreased 8-OHdG levels in the heart compared with DOX group, suggesting that STS reduces DOX-induced DNA damage by improving antioxidant enzymes activities in cardiomyocytes. Additionally, the DOX + STS group showed attenuation of cleaved caspase-3 and DNA fragmentation in cardiomyocytes compared with the DOX group, suggesting that STS suppresses DOX-induced apoptosis in cardiomyocytes. SIGNIFICANCE STS exerts cardioprotective effects against DOX-induced cardiac dysfunction partly by improving antioxidant defense and suppressing apoptosis, indicating the therapeutic potential of STS against DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Yukie Mizuta
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Tokuda
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Intensive Care Unit, Kyushu University Hospital, Fukuoka, Japan.
| | - Jie Guo
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shuo Zhang
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sayoko Narahara
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sumio Hoka
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Hashizume
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Tomohiko Akahoshi
- Department of Disaster and Emergency Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
20
|
Ghione S, Mabrouk N, Paul C, Bettaieb A, Plenchette S. Protein kinase inhibitor-based cancer therapies: Considering the potential of nitric oxide (NO) to improve cancer treatment. Biochem Pharmacol 2020; 176:113855. [PMID: 32061562 DOI: 10.1016/j.bcp.2020.113855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/10/2020] [Indexed: 12/14/2022]
Abstract
The deregulation of a wide variety of protein kinases is associated with cancer cell initiation and tumor progression. Owing to their indispensable function in signaling pathways driving malignant cell features, protein kinases constitute major therapeutic targets in cancer. Over the past two decades, intense efforts in drug development have been dedicated to this field. The development of protein kinase inhibitors (PKIs) have been a real breakthrough in targeted cancer therapy. Despite obvious successes across patients with different types of cancer, the development of PKI resistance still prevails. Combination therapies are part of a comprehensive approach to address the problem of drug resistance. The therapeutic use of nitric oxide (NO) donors to bypass PKI resistance in cancer has never been tested in clinic yet but several arguments suggest that the combination of PKIs and NO donors may exert a potential anticancer effect. The present review summarized the current state of knowledge on common targets to both PKIs and NO. Herein, we attempt to provide the rationale underlying a potential combination of PKIs and NO donors for future directions and design of new combination therapies in cancer.
Collapse
Affiliation(s)
- Silvia Ghione
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France; LIIC, EA7269, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Nesrine Mabrouk
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France; LIIC, EA7269, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France; LIIC, EA7269, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France; LIIC, EA7269, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Stéphanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France; LIIC, EA7269, Université de Bourgogne Franche-Comté, 21000 Dijon, France.
| |
Collapse
|
21
|
Behring JB, van der Post S, Mooradian AD, Egan MJ, Zimmerman MI, Clements JL, Bowman GR, Held JM. Spatial and temporal alterations in protein structure by EGF regulate cryptic cysteine oxidation. Sci Signal 2020; 13:eaay7315. [PMID: 31964804 PMCID: PMC7263378 DOI: 10.1126/scisignal.aay7315] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stimulation of plasma membrane receptor tyrosine kinases (RTKs), such as the epidermal growth factor receptor (EGFR), locally increases the abundance of reactive oxygen species (ROS). These ROS then oxidize cysteine residues in proteins to potentiate downstream signaling. Spatial confinement of ROS is an important regulatory mechanism of redox signaling that enables the stimulation of different RTKs to oxidize distinct sets of downstream proteins. To uncover additional mechanisms that specify cysteines that are redox regulated by EGF stimulation, we performed time-resolved quantification of the EGF-dependent oxidation of 4200 cysteine sites in A431 cells. Fifty-one percent of cysteines were statistically significantly oxidized by EGF stimulation. Furthermore, EGF induced three distinct spatiotemporal patterns of cysteine oxidation in functionally organized protein networks, consistent with the spatial confinement model. Unexpectedly, protein crystal structure analysis and molecular dynamics simulations indicated widespread redox regulation of cryptic cysteine residues that are solvent exposed only upon changes in protein conformation. Phosphorylation and increased flux of nucleotide substrates served as two distinct modes by which EGF specified the cryptic cysteine residues that became solvent exposed and redox regulated. Because proteins that are structurally regulated by different RTKs or cellular perturbations are largely unique, these findings suggest that solvent exposure and redox regulation of cryptic cysteine residues contextually delineate redox signaling networks.
Collapse
Affiliation(s)
- Jessica B Behring
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Sjoerd van der Post
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Arshag D Mooradian
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Matthew J Egan
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Maxwell I Zimmerman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jenna L Clements
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jason M Held
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
22
|
Cai H, Han B, Hu Y, Zhao X, He Z, Chen X, Sun H, Yuan J, Li Y, Yang X, Kong W, Kong WJ. Metformin attenuates the D‑galactose‑induced aging process via the UPR through the AMPK/ERK1/2 signaling pathways. Int J Mol Med 2020; 45:715-730. [PMID: 31922237 PMCID: PMC7015132 DOI: 10.3892/ijmm.2020.4453] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022] Open
Abstract
Age‑related hearing loss, also termed central presbycusis, is a progressive neurodegenerative disease; it is a devastating disorder that severely affects the quality of life of elderly individuals. Substantial evidence has indicated that oxidative stress and associated protein folding dysfunction have a marked influence on neurodegenerative diseases. In this study, we aimed to cells to investigate whether metformin protects against age‑related pathologies and to elucidate the underlying mechanisms; specifically, we focused on the role of unfolded protein response (UPR) via the AMPK/ERK1/2 signaling pathways. For this purpose, the biguanide compound, metformin, a medication widely used in the treatment of type 2 diabetes, was administered to rats in a model of mimetic aging. In addition, senescent PC12 were treated with metformin. Although it has been well established that UPR signaling is activated in response to cellular stress and is associated with the pathogenesis of neuronal deterioration, the detailed functions of the UPR in the auditory cortex remain unclear. We found that metformin treatment markedly affected the UPR and the AMPK/ERK1/2 signaling pathway, and maintained the auditory brainstem response (ABR) threshold during the aging process. The results indicated that the regulation of the UPR and AMPK/ERK1/2 signaling pathway by metformin significantly attenuated hearing loss, cell apoptosis and age‑related neurodegeneration. Reversing these harmful effects through the use of metformin suggests its involvement in restoring the antioxidant status and protein homeostasis related to the underlying pathology of presbycusis. The findings of this study may provide a better approach for the treatment of age‑related neurodegeneration diseases.
Collapse
Affiliation(s)
- Hua Cai
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Baoai Han
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yujuan Hu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xueyan Zhao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zuhong He
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xubo Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Haiying Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jie Yuan
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yongqin Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiuping Yang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wei-Jia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
23
|
Emerging role of innate B1 cells in the pathophysiology of autoimmune and neuroimmune diseases: Association with inflammation, oxidative and nitrosative stress and autoimmune responses. Pharmacol Res 2019; 148:104408. [PMID: 31454534 DOI: 10.1016/j.phrs.2019.104408] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022]
|
24
|
Bignon E, Allega MF, Lucchetta M, Tiberti M, Papaleo E. Computational Structural Biology of S-nitrosylation of Cancer Targets. Front Oncol 2018; 8:272. [PMID: 30155439 PMCID: PMC6102371 DOI: 10.3389/fonc.2018.00272] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/02/2018] [Indexed: 12/15/2022] Open
Abstract
Nitric oxide (NO) plays an essential role in redox signaling in normal and pathological cellular conditions. In particular, it is well known to react in vivo with cysteines by the so-called S-nitrosylation reaction. S-nitrosylation is a selective and reversible post-translational modification that exerts a myriad of different effects, such as the modulation of protein conformation, activity, stability, and biological interaction networks. We have appreciated, over the last years, the role of S-nitrosylation in normal and disease conditions. In this context, structural and computational studies can help to dissect the complex and multifaceted role of this redox post-translational modification. In this review article, we summarized the current state-of-the-art on the mechanism of S-nitrosylation, along with the structural and computational studies that have helped to unveil its effects and biological roles. We also discussed the need to move new steps forward especially in the direction of employing computational structural biology to address the molecular and atomistic details of S-nitrosylation. Indeed, this redox modification has been so far an underappreciated redox post-translational modification by the computational biochemistry community. In our review, we primarily focus on S-nitrosylated proteins that are attractive cancer targets due to the emerging relevance of this redox modification in a cancer setting.
Collapse
Affiliation(s)
- Emmanuelle Bignon
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria Francesca Allega
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Marta Lucchetta
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Tiberti
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory Danish Cancer Society Research Center, Copenhagen, Denmark.,Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
ERK1/2 MAPK promotes autophagy to suppress ER stress-mediated apoptosis induced by cadmium in rat proximal tubular cells. Toxicol In Vitro 2018; 52:60-69. [PMID: 29870746 DOI: 10.1016/j.tiv.2018.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/28/2018] [Accepted: 06/01/2018] [Indexed: 12/23/2022]
Abstract
Cadmium (Cd) is a toxic heavy metal and its toxic mechanism is not entirely clear. The goal of the present study was to investigate the toxic mechanism of Cd on rPT cells, and to elucidate the role of ERK1/2 signaling pathway in mediating the relationship between apoptosis and autophagy. We evaluated the cell morphology, cell cycle distribution, apoptosis rates, and the expression of related proteins. We observed that increased Cd concentration disrupted cell morphology, increased apoptosis and induced autophagy. Additionally, activation of JNK1/2 and p38 MAPK promoted apoptosis, while activation of ERK1/2 inhibited apoptosis. Upon inhibition of autophagy, apoptosis rate and the expression of ER proteins related to the apoptosis were increased. Following inhibition of the ERK1/2 signaling pathway, the number of LC3 aggregates, the rate of LC3II/LC3I and the expression of Beclin-1were decreased, but the expression level of ER proteins related to apoptosis were increased. Our results indicated that Cd exposure damages cells also induces apoptosis and autophagy, meanwhile demonstrate that the ERK1/2 signaling pathway plays an important role in this process. Besides, these data suggest that autophagy can inhibit Cd-induced apoptosis and the ERK1/2 signaling pathway can suppress ER stress-mediated apoptosis by activating autophagy.
Collapse
|
26
|
Park JW, Lee CM, Cheng JS, Morgan ET. Posttranslational regulation of CYP2J2 by nitric oxide. Free Radic Biol Med 2018; 121:149-156. [PMID: 29715548 PMCID: PMC5978777 DOI: 10.1016/j.freeradbiomed.2018.04.576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 12/24/2022]
Abstract
Nitric oxide (NO) is an essential signaling molecule in the body, regulating numerous biological processes. Beside its physiological roles, NO affects drug metabolism by modulating the activity and/or expression of cytochrome P450 enzymes. Previously, our lab showed that NO generation caused by inflammatory stimuli results in CYP2B6 degradation via the ubiquitin-proteasome pathway. In the current study, we tested the NO-mediated regulation of CYP2J2 that metabolizes arachidonic acids to bioactive epoxyeicosatrienoic acids, as well as therapeutic drugs such as astemizole and ebastine. To investigate the effects of NO on CYP2J2 expression and activity, Huh7 cells stably transduced with CYP2J2 with a C-terminal V5 tag were treated with dipropylenetriamine-NONOate (DPTA), a NO donor. The level of CYP2J2 proteins were decreased in a time- and concentration-dependent manner, and the activity was also rapidly inhibited. However, mRNA expression was not altered and the protein synthesis inhibitor cycloheximide did not attenuate DPTA-mediated downregulation of CYP2J2. Removal of DPTA from the culture media quickly restored the activity of remaining CYP2J2, and no further CYP2J2 degradation occurred. To determine the mechanism of CYP2J2 down-regulation by NO, cells were treated with DPTA in the presence or absence of protease inhibitors including proteasomal, lysosomal and calpain inhibitors. Remarkably, the down-regulation of CYP2J2 by NO was attenuated by calpeptin, a calpain inhibitor. However, other calpain inhibitors or calcium chelator show no inhibitory effects on the degradation. The proteasome inhibitor bortezomib showed small but significant restoration of CYP2J2 levels although stimulated ubiquitination of CYP2J2 was not detected. In conclusion, these data suggest that NO regulates CYP2J2 posttranslationally and NO-evoked CYP2J2 degradation undergoes ubiquitin-independent proteasomal degradation pathway unlike CYP2B6.
Collapse
Affiliation(s)
- Ji Won Park
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA
| | - Choon-Myung Lee
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA
| | - Joan S Cheng
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA
| | - Edward T Morgan
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA.
| |
Collapse
|
27
|
Wang SB, Venkatraman V, Crowgey EL, Liu T, Fu Z, Holewinski R, Ranek M, Kass DA, O'Rourke B, Van Eyk JE. Protein S-Nitrosylation Controls Glycogen Synthase Kinase 3β Function Independent of Its Phosphorylation State. Circ Res 2018; 122:1517-1531. [PMID: 29563102 DOI: 10.1161/circresaha.118.312789] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 01/11/2023]
Abstract
RATIONALE GSK-3β (glycogen synthase kinase 3β) is a multifunctional and constitutively active kinase known to regulate a myriad of cellular processes. The primary mechanism to regulate its function is through phosphorylation-dependent inhibition at serine-9 residue. Emerging evidence indicates that there may be alternative mechanisms that control GSK-3β for certain functions. OBJECTIVES Here, we sought to understand the role of protein S-nitrosylation (SNO) on the function of GSK-3β. SNO-dependent modulation of the localization of GSK-3β and its ability to phosphorylate downstream targets was investigated in vitro, and the network of proteins differentially impacted by phospho- or SNO-dependent GSK-3β regulation and in vivo SNO modification of key signaling kinases during the development of heart failure was also studied. METHODS AND RESULTS We found that GSK-3β undergoes site-specific SNO both in vitro, in HEK293 cells, H9C2 myoblasts, and primary neonatal rat ventricular myocytes, as well as in vivo, in hearts from an animal model of heart failure and sudden cardiac death. S-nitrosylation of GSK-3β significantly inhibits its kinase activity independent of the canonical phospho-inhibition pathway. S-nitrosylation of GSK-3β promotes its nuclear translocation and access to novel downstream phosphosubstrates which are enriched for a novel amino acid consensus sequence motif. Quantitative phosphoproteomics pathway analysis reveals that nuclear GSK-3β plays a central role in cell cycle control, RNA splicing, and DNA damage response. CONCLUSIONS The results indicate that SNO has a differential effect on the location and activity of GSK-3β in the cytoplasm versus the nucleus. SNO modification of GSK-3β occurs in vivo and could contribute to the pathobiology of heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Sheng-Bing Wang
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Vidya Venkatraman
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.).,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| | - Erin L Crowgey
- Department of Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE (E.L.C.)
| | - Ting Liu
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | | | - Ronald Holewinski
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.).,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| | - Mark Ranek
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - David A Kass
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Brian O'Rourke
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Jennifer E Van Eyk
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.) .,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| |
Collapse
|
28
|
Jin L, Cao Y, Zhang T, Wang P, Ji D, Liu X, Shi H, Hua L, Yu R, Gao S. Effects of ERK1/2 S-nitrosylation on ERK1/2 phosphorylation and cell survival in glioma cells. Int J Mol Med 2017; 41:1339-1348. [PMID: 29286066 PMCID: PMC5819938 DOI: 10.3892/ijmm.2017.3334] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/01/2017] [Indexed: 01/08/2023] Open
Abstract
Aberrant activation of extracellular signal-regulated kinase 1/2 (ERK1/2) by phosphorylation modification can trigger tumor cell development in glioma. S-nitrosylation, which refers to the covalent addition of a nitric oxide (NO) group to a cysteine (Cys) thiol, is an important post-translational modification that occurs on numerous cancer-associated proteins. Protein S-nitrosylation can increase or decrease protein activity and stability, and subsequent signal transduction and cellular processes. However, the association between ERK1/2 S-nitrosylation and ERK1/2 phosphorylation, and the effects of ERK1 S-nitrosylation on glioma cell survival are currently unknown. U251 glioma cells were treated with NO donors sodium nitroprusside (SNP) or S-nitrosoglutathione (GSNO). CCK8 assay was used to assess the cell viability. NO levels in the medium were detected by Griess assay. Western blot analysis and biotin switch assay were employed to detect the ERK1/2 phosphorylation and S-nitrosylation. ERK1 wild-type and mutant plasmids were constructed, and used to transfect the U251 cells. Caspase-3 western blot analysis and flow cytometry were employed to assess cell apoptosis. The present study demonstrated that treatment with the NO donors SNP or GSNO led to an increase in ERK1/2 S-nitrosylation, and a reduction in ERK1/2 phosphorylation, which was accompanied by growth inhibition of U251 glioma cells. Mutational analysis demonstrated that Cys183 was vital for S-nitrosylation of ERK1, and that preventing ERK1 S-nitrosylation by replacing Cys183 with alanine partially reversed GSNO-induced cell apoptosis, and reductions in cell viability and ERK1/2 phosphorylation. In addition, increased ERK1/2 phosphorylation was associated with decreased ERK1/2 S-nitrosylation in human glioma tissues. These findings identified the relationship between ERK1/2 S-nitrosylation and phosphorylation in vitro and in vivo, and revealed a novel mechanism of ERK1/2 underlying tumor cell development and apoptotic resistance of glioma.
Collapse
Affiliation(s)
- Lei Jin
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yujia Cao
- Department of Neurosurgery, People's Hospital of Gaoxin District, Suzhou, Jiangsu 215011, P.R. China
| | - Tong Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Peng Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Daofei Ji
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xuejiao Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Hengliang Shi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Lei Hua
- Department of Neurosurgery, Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Shangfeng Gao
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
29
|
Keyes JD, Parsonage D, Yammani RD, Rogers LC, Kesty C, Furdui CM, Nelson KJ, Poole LB. Endogenous, regulatory cysteine sulfenylation of ERK kinases in response to proliferative signals. Free Radic Biol Med 2017; 112:534-543. [PMID: 28843779 PMCID: PMC5623068 DOI: 10.1016/j.freeradbiomed.2017.08.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/11/2017] [Accepted: 08/22/2017] [Indexed: 01/04/2023]
Abstract
ERK-dependent signaling is key to many pathways through which extracellular signals are transduced into cell-fate decisions. One conundrum is the way in which disparate signals induce specific responses through a common, ERK-dependent kinase cascade. While studies have revealed intricate ways of controlling ERK signaling through spatiotemporal localization and phosphorylation dynamics, additional modes of ERK regulation undoubtedly remain to be discovered. We hypothesized that fine-tuning of ERK signaling could occur by cysteine oxidation. We report that ERK is actively and directly oxidized by signal-generated H2O2 during proliferative signaling, and that ERK oxidation occurs downstream of a variety of receptor classes tested in four cell lines. Furthermore, within the tested cell lines and proliferative signals, we observed that both activation loop-phosphorylated and non-phosphorylated ERK undergo sulfenylation in cells and that dynamics of ERK sulfenylation is dependent on the cell growth conditions prior to stimulation. We also tested the effect of endogenous ERK oxidation on kinase activity and report that phosphotransfer reactions are reversibly inhibited by oxidation by as much as 80-90%, underscoring the importance of considering this additional modification when assessing ERK activation in response to extracellular signals.
Collapse
Affiliation(s)
- Jeremiah D Keyes
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - Derek Parsonage
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - Rama D Yammani
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - LeAnn C Rogers
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA
| | - Chelsea Kesty
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA
| | - Cristina M Furdui
- Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Kimberly J Nelson
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - Leslie B Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA.
| |
Collapse
|
30
|
Zhou L, Shi M, Zhao L, Lin Z, Tang Z, Sun H, Chen T, Lv Z, Xu J, Huang Y, Yu X. Clonorchis sinensis lysophospholipase A upregulates IL-25 expression in macrophages as a potential pathway to liver fibrosis. Parasit Vectors 2017. [PMID: 28623940 PMCID: PMC5474055 DOI: 10.1186/s13071-017-2228-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Liver fibrosis is an excessive wound-healing reaction that requires the participation of inflammatory cells and hepatic stellate cells (HSCs). The pathogenesis of liver fibrosis caused by viruses and alcohol has been well characterized, but the molecular mechanisms underlying liver fibrosis induced by the liver fluke Clonorchis sinensis are poorly understood. Lysophospholipase A (LysoPLA), which deacylates lysophospholipids, plays a critical role in mediating the virulence and pathogenesis of parasites and fungi; however, the roles of C. sinensis lysophospholipase A (CsLysoPLA) in C. sinensis-induced liver fibrosis remain unknown. Methods A mouse macrophage cell line (RAW264.7) was cultured and treated with CsLysoPLA. IL-25 and members of its associated signaling pathway were detected by performing quantitative real-time PCR, Western blotting and immunofluorescent staining. A human hepatic stellate cell line (LX-2) was cultured and exposed to IL-25. LX-2 cell activation markers were examined via quantitative real-time PCR, Western blotting and immunofluorescent staining. Migration was analyzed in transwell plates. Results Treating RAW264.7 cells with CsLysoPLA significantly induced IL-25 expression. Elevated PKA, B-Raf, and ERK1/2 mRNA levels and phosphorylated B-Raf and ERK1/2 were detected in CsLysoPLA-stimulated RAW264.7 cells. The PKA inhibitor H-89 weakened B-Raf and ERK1/2 phosphorylation whereas the AKT activator SC79 attenuated ERK1/2 phosphorylation in RAW264.7 cells. Both H-89 and SC79 inhibited CsLysoPLA-induced IL-25 upregulation. In addition, stimulation of LX-2 cells with IL-25 upregulated the expression of mesenchymal cell markers, including α-smooth muscle actin (α-SMA) and collagen type I (Collagen-I), and promoted cell migration. Conclusions CsLysoPLA activates HSCs by upregulating IL-25 in macrophages through the PKA-dependent B-Raf/ERK1/2 pathway and potentially promotes hepatic fibrosis during C. sinensis infection.
Collapse
Affiliation(s)
- Lina Zhou
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Mengchen Shi
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Lu Zhao
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zhipeng Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zeli Tang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Hengchang Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Tingjin Chen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zhiyue Lv
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Jin Xu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Yan Huang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China.
| | - Xinbing Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory for Tropical Diseases Control, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China.
| |
Collapse
|
31
|
Hwang JH, Park SJ, Ko WG, Kang SM, Lee DB, Bang J, Park BJ, Wee CB, Kim DJ, Jang IS, Ko JH. Cordycepin induces human lung cancer cell apoptosis by inhibiting nitric oxide mediated ERK/Slug signaling pathway. Am J Cancer Res 2017; 7:417-432. [PMID: 28401001 PMCID: PMC5385633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 06/07/2023] Open
Abstract
Nitric oxide (NO) is an important signaling molecule and a component of the inflammatory cascade. Besides, it is also involved in tumorigenesis. Aberrant upregulation and activation of the ERK cascade by NO often leads to tumor cell development. However, the role of ERK inactivation induced by the negative regulation of NO during apoptosis is not completely understood. In this study, treatment of A549 and PC9 human lung adenocarcinoma cell lines with cordycepin led to a reduction in their viability. Analysis of the effect of cordycepin treatment on ERK/Slug signaling activity in the A549 cell line revealed that LPS-induced inflammatory microenvironments could stimulate the expression of TNF-α, CCL5, IL-1β, IL-6, IL-8 and upregulate NO, phospho-ERK (p-ERK), and Slug expression. In addition, constitutive expression of NO was observed. Cordycepin inhibited LPS-induced stimulation of iNOS, NO, p-ERK, and Slug expression. L-NAME, an inhibitor of NOS, inhibited p-ERK and Slug expression. It was also found that cordycepin-mediated inhibition of ERK downregulated Slug, whereas overexpression of ERK led to an upregulation of Slug levels in the cordycepin-treated A549 cells. Inhibition of Slug by siRNA induced Bax and caspase-3, leading to cordycepin-induced apoptosis. Cordycepin-mediated inhibition of ERK led to a reduction in phospho-GSK3β (p-GSK3β) and Slug levels, whereas LiCl, an inhibitor of GSK3β, upregulated p-GSK3β and Slug. Overall, the results obtained indicate that cordycepin inhibits the ERK/Slug signaling pathway through the activation of GSK3β which, in turn, upregulates Bax, leading to apoptosis of the lung cancer cells.
Collapse
Affiliation(s)
- Jung Hoo Hwang
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Soo Jung Park
- Department of Sasang Constitutional Medicine, Woosuk UniversityWanju 55338, Jeonbuk, Republic of Korea
| | - Won Gyu Ko
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Seong-Mun Kang
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Da Bin Lee
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Junho Bang
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Byung-Joo Park
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Chung-Beum Wee
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| | - Dae Joon Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande ValleyEdinburg, TX 78539, USA
| | - Ik-Soon Jang
- Division of Bioconvergence Analysis, Korea Basic Science InstituteDaejeon 305-333, Republic of Korea
| | - Jae-Hong Ko
- Department of Physiology, Chung-Ang University College of MedicineSeoul 06974, Republic of Korea
| |
Collapse
|
32
|
Ritter LR, Chrestensen CA, Salerno JC. A mathematical model of endothelial nitric oxide synthase activation with time delay exhibiting Hopf bifurcation and oscillations. Math Biosci 2016; 281:62-73. [PMID: 27614021 DOI: 10.1016/j.mbs.2016.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 11/25/2022]
Abstract
Nitric oxide (NO) is a gaseous compound that serves as a signaling molecule in cellular interactions. In the vasculature, NO is synthesized from endogenous agents by endothelial nitric oxide synthase (eNOS) where it plays key roles in several functions related to homeostasis, adaptation, and development. Recent experimental studies have revealed cycles of increasing and decreasing NO production when eNOS is stimulated by factors such as glucose or insulin. We offer a mathematical model of a generic amino acid receptor site on eNOS wherein this species is subject to activation/deactivation by a pair of interactive kinase and phosphatase species. The enzyme kinetic model is presented as a system of ordinary differential equations including time delay to allow for various intermediate, unspecified complexes. We show that under conditions on the model parameters, varying the delay time may give rise to a Hopf bifurcation. Properties of the bifurcating solutions are explored via a center manifold reduction, and a numerical illustration is provided.
Collapse
Affiliation(s)
- L R Ritter
- Department of Mathematics, Kennesaw State University, 1100 S. Marietta Pkwy, MD #9085, Marietta, GA 30060, USA.
| | - C A Chrestensen
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA 30144, USA.
| | - J C Salerno
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA
| |
Collapse
|
33
|
Gasotransmitters in Gametogenesis and Early Development: Holy Trinity for Assisted Reproductive Technology-A Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1730750. [PMID: 27579148 PMCID: PMC4992752 DOI: 10.1155/2016/1730750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/03/2016] [Indexed: 11/23/2022]
Abstract
Creation of both gametes, sperm and oocyte, and their fusion during fertilization are essential step for beginning of life. Although molecular mechanisms regulating gametogenesis, fertilization, and early embryonic development are still subjected to intensive study, a lot of phenomena remain unclear. Based on our best knowledge and own results, we consider gasotransmitters to be essential for various signalisation in oocytes and embryos. In accordance with nitric oxide (NO) and hydrogen sulfide (H2S) physiological necessity, their involvement during oocyte maturation and regulative role in fertilization followed by embryonic development have been described. During these processes, NO- and H2S-derived posttranslational modifications represent the main mode of their regulative effect. While NO represent the most understood gasotransmitter and H2S is still intensively studied gasotransmitter, appreciation of carbon monoxide (CO) role in reproduction is still missing. Overall understanding of gasotransmitters including their interaction is promising for reproductive medicine and assisted reproductive technologies (ART), because these approaches contend with failure of in vitro assisted reproduction.
Collapse
|
34
|
Plenchette S, Romagny S, Laurens V, Bettaieb A. [NO and cancer: itinerary of a double agent]. Med Sci (Paris) 2016; 32:625-33. [PMID: 27406774 DOI: 10.1051/medsci/20163206027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Protein S-nitrosylation is now recognized as a ubiquitous regulatory mechanism. Like any post-translational modifications, S-nitrosylation is critical for the control of numerous cellular processes. It is now clear that S-nitrosylation is playing a double game, enhancing or inhibiting the tumor growth or the induction of cell death. Thanks to research aimed at demonstrating NO cytotoxic effects, new therapeutic strategies based on NO donor drugs have emerged. Although therapeutic NO donors can target a large number of proteins, the cellular mechanism is still not fully understood. This review reflects the current state of knowledge on S-nitrosylated proteins that take part of the oncogenic and apoptotic signaling, putting forward proteins with potential interest in cancer therapy.
Collapse
Affiliation(s)
- Stéphanie Plenchette
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Sabrina Romagny
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Véronique Laurens
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| | - Ali Bettaieb
- Université de Bourgogne Franche-Comté, LIIC EA7269, 7, boulevard Jeanne d'Arc, F-21000 Dijon, France - EPHE, PSL Research University, F-75014 Paris, France
| |
Collapse
|
35
|
Rizvi F, Mathur A, Krishna S, Siddiqi MI, Kakkar P. Suppression in PHLPP2 induction by morin promotes Nrf2-regulated cellular defenses against oxidative injury to primary rat hepatocytes. Redox Biol 2015; 6:587-598. [PMID: 26513344 PMCID: PMC4633887 DOI: 10.1016/j.redox.2015.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 11/15/2022] Open
Abstract
Recent advances indicate a possible role of phytochemicals as modulatory factors in signaling pathways. We have previously demonstrated PHLPP2-mediated suppression of Nrf2 responses during oxidant attack. The present study was designed to explore Nrf2-potentiating mechanism of morin, a flavonol, via its possible role in intervening PHLPP2-regulated Akt/GSK3β/Fyn kinase axis. Efficacy of morin was evaluated against oxidative stress-mediated damage to primary hepatocytes by tert-butyl hydroperoxide (tBHP) and acetaminophen. The anti-cytotoxic effects of morin were found to be a consequence of fortification of Nrf2-regulated antioxidant defenses since morin failed to sustain activities of redox enzyme in Nrf2 silenced hepatocytes. Morin promoted Nrf2 stability and its nuclear retention by possibly modulating PHLPP2 activity which subdues cellular Nrf2 responses by activating Fyn kinase. Pull-down assay using morin-conjugated beads indicated the binding affinity of morin towards PHLPP2. Molecular docking also revealed the propensity of morin to occupy the active site of PHLPP2 enzyme. Thus, dietary phytochemical morin was observed to counteract oxidant-induced hepatocellular damage by promoting Nrf2-regulated transcriptional induction. The findings support the novel role of morin in potentiating Nrf2 responses by limiting PHLPP2 and hence Fyn kinase activation. Therefore, morin may be exploited in developing novel therapeutic strategy aimed at enhancing Nrf2 responses. Cytoprotection against tBHP-evoked oxidative stress by morin is Nrf2-regulated. Morin prevents Nrf2-destabilization via PHLPP2-Akt-GSK3β-Fyn kinase pathway. In silico docking studies confirmed PHLPP2 activity inhibition by morin. Morin mitigates APAP induced cytotoxicity by suppressing PHLPP2 pathway.
Collapse
Affiliation(s)
- Fatima Rizvi
- Herbal Research Laboratory, Food Drug and Chemical Toxicology Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Post Box No. 80, M.G. Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow, India
| | - Alpana Mathur
- Herbal Research Laboratory, Food Drug and Chemical Toxicology Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Post Box No. 80, M.G. Marg, Lucknow 226001, Uttar Pradesh, India
| | - Shagun Krishna
- Molecular and Structural Biology Division, Council of Scientific and Industrial Research-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, Uttar Pradesh, India
| | - Mohammad Imran Siddiqi
- Molecular and Structural Biology Division, Council of Scientific and Industrial Research-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, Uttar Pradesh, India
| | - Poonam Kakkar
- Herbal Research Laboratory, Food Drug and Chemical Toxicology Group, Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Post Box No. 80, M.G. Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research, CSIR-IITR campus, Lucknow, India.
| |
Collapse
|
36
|
Penna C, Angotti C, Pagliaro P. Protein S-nitrosylation in preconditioning and postconditioning. Exp Biol Med (Maywood) 2015; 239:647-62. [PMID: 24668550 DOI: 10.1177/1535370214522935] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The coronary artery disease is a leading cause of death and morbidity worldwide. This disease has a complex pathophysiology that includes multiple mechanisms. Among these is the oxidative/nitrosative stress. Paradoxically, oxidative/nitrosative signaling plays a major role in cardioprotection against ischemia/reperfusion injury. In this context, the gas transmitter nitric oxide may act through several mechanisms, such as guanylyl cyclase activation and via S-nitrosylation of proteins. The latter is a covalent modification of a protein cysteine thiol by a nitric oxide-group that generates an S-nitrosothiol. Here, we report data showing that nitric oxide and S-nitrosylation of proteins play a pivotal role not only in preconditioning but also in postconditioning cardioprotection.
Collapse
|
37
|
Single-walled carbon nanotubes (SWCNTs) induce vasodilation in isolated rat aortic rings. Toxicol In Vitro 2015; 29:657-62. [DOI: 10.1016/j.tiv.2015.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 01/23/2015] [Accepted: 02/03/2015] [Indexed: 11/23/2022]
|
38
|
Kawabata M, Umemoto N, Shimada Y, Nishimura Y, Zhang B, Kuroyanagi J, Miyabe M, Tanaka T. Downregulation of stanniocalcin 1 is responsible for sorafenib-induced cardiotoxicity. Toxicol Sci 2014; 143:374-84. [PMID: 25370841 DOI: 10.1093/toxsci/kfu235] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sorafenib is associated with adverse cardiac effects, including left ventricular dysfunction. However, the precise mechanism remains unclear. Here, we aimed to establish the genes responsible for this cardiotoxicity using zebrafish and human cardiomyocytes. Fluorescent cardiac imaging using pigmentless zebrafish with green fluorescent protein hearts revealed that the ventricular dimensions of the longitudinal axis with sorafenib were significantly shorter than those of the control group. Transcriptome analysis of their hearts revealed that stanniocalcin 1 (stc1) was downregulated by sorafenib. stc1 knockdown in zebrafish revealed that reduction of stc1 decreased the longitudinal dimensions of zebrafish ventricles, similar to that which occurs during sorafenib treatment. STC1 downregulation and cytotoxicity were also seen in human cardiomyocytes exposed to sorafenib. To clarify the molecular function of stc1 in sorafenib-induced cardiotoxicity, we focused on oxidative stress in cardiomyocytes treated with sorafenib. Reactive oxygen species (ROS) production significantly increased in both species of human cardiomyocytes and zebrafish exposed to sorafenib and STC1 knockdown compared with the controls. Finally, we found that forced expression of stc1 normalized impairment, decreasing the longitudinal dimensions in zebrafish treated with sorafenib. Our study demonstrated that STC1 plays a protective role against ventricular dysfunction and ROS overproduction, which are induced by sorafenib treatment. We discovered for the first time that STC1 downregulation is responsible for sorafenib-induced cardiotoxicity through activated ROS generation.
Collapse
Affiliation(s)
- Miko Kawabata
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Noriko Umemoto
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Yasuhito Shimada
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-
| | - Yuhei Nishimura
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-
| | - Beibei Zhang
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Junya Kuroyanagi
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Masayuki Miyabe
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Toshio Tanaka
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-
| |
Collapse
|
39
|
Nogueira-Pedro A, Dias CC, Regina H, Segreto C, Addios PC, Lungato L, D'Almeida V, Barros CC, Higa EMS, Buri MV, Ferreira AT, Paredes-Gamero EJ. Nitric oxide-induced murine hematopoietic stem cell fate involves multiple signaling proteins, gene expression, and redox modulation. Stem Cells 2014; 32:2949-60. [PMID: 24964894 DOI: 10.1002/stem.1773] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/20/2014] [Accepted: 05/30/2014] [Indexed: 12/22/2022]
Abstract
There are a growing number of reports showing the influence of redox modulation in cellular signaling. Although the regulation of hematopoiesis by reactive oxygen species (ROS) and reactive nitrogen species (RNS) has been described, their direct participation in the differentiation of hematopoietic stem cells (HSCs) remains unclear. In this work, the direct role of nitric oxide (NO(•)), a RNS, in the modulation of hematopoiesis was investigated using two sources of NO(•) , one produced by endothelial cells stimulated with carbachol in vitro and another using the NO(•)-donor S-nitroso-N-acetyl-D,L-penicillamine (SNAP) in vivo. Two main NO(•) effects were observed: proliferation of HSCs-especially of the short-term HSCs-and its commitment and terminal differentiation to the myeloid lineage. NO(•)-induced proliferation was characterized by the increase in the number of cycling HSCs and hematopoietic progenitor cells positive to BrdU and Ki-67, upregulation of Notch-1, Cx43, PECAM-1, CaR, ERK1/2, Akt, p38, PKC, and c-Myc. NO(•)-induced HSCs differentiation was characterized by the increase in granulocytic-macrophage progenitors, granulocyte-macrophage colony forming units, mature myeloid cells, upregulation of PU.1, and C/EBPα genes concomitantly to the downregulation of GATA-3 and Ikz-3 genes, activation of Stat5 and downregulation of the other analyzed proteins mentioned above. Also, redox status modulation differed between proliferation and differentiation responses, which is likely associated with the transition of the proliferative to differentiation status. Our findings provide evidence of the role of NO(•) in inducing HSCs proliferation and myeloid differentiation involving multiple signaling.
Collapse
|
40
|
Chen J, Cao J, Luo Y, Xie L, Song J, Xue W, Jia R, Song J. Expression of ERK and p-ERK proteins of ERK signaling pathway in the kidneys of fluoride-exposed carp (Cyprinus carpio). Acta Histochem 2014; 116:1337-41. [PMID: 25190107 DOI: 10.1016/j.acthis.2014.08.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/14/2014] [Accepted: 08/17/2014] [Indexed: 11/30/2022]
Abstract
Chronic exposure to fluoride can result in a variety of adverse effects in fish. Previously we indicated that high fluoride caused damage and apoptosis in the kidneys of the common carp, Cyprinus carpio. In this study, the effects of fluoride on the expression and localization of ERK and p-ERK proteins in the ERK signaling pathway were determined using Western blotting and immunohistochemical methods in the kidneys of carp exposed to 0, 40, 80, 120mg/L fluoride, respectively. Western blotting analysis found that compared with the controls, the levels of ERK1 and ERK2 proteins were relatively unchanged in fluoride-exposed fish, while p-ERK1 and p-ERK2 protein levels decreased significantly with the increased fluoride concentrations. The immunohistochemical analysis found the proteins of ERK and p-ERK were predominantly localized in the cytoplasm of epithelial cells in the renal tubules of C. carpio. Compared with the control group, the levels of ERK protein were relatively constant, yet the levels of p-ERK protein and p-ERK/ERK ratio were reduced with fluoride exposure dose. These findings indicate that the renal damage in carp exposed to fluoride is mediated via the ERK pathway. Fluoride exposure could inactivate ERK, inhibit the expression of p-ERK protein, and induce renal damage in C. carpio.
Collapse
Affiliation(s)
- Jianjie Chen
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Jinling Cao
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Yongju Luo
- Guangxi Academy of Fishery Sciences, Nanning 530021, Guangxi, PR China
| | - Lingtian Xie
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, Liaoning, PR China.
| | - Jing Song
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Wenjuan Xue
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Ruhui Jia
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Jie Song
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| |
Collapse
|
41
|
Cao J, Chen J, Wang J, Klerks P, Xie L. Effects of sodium fluoride on MAPKs signaling pathway in the gills of a freshwater teleost, Cyprinus carpio. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 152:164-172. [PMID: 24780358 DOI: 10.1016/j.aquatox.2014.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/01/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
Exposure to elevated levels of fluoride can cause a variety of adverse effects in fish. Previously we showed that fluoride causes injuries and apoptosis in the gills of Cyprinus carpio. In this study, the effects of fluoride on caspase-3 activity and on accumulation of proteins in the MAPKs pathways were evaluated using Western blotting and immunohistochemistry methods in vivo and in vitro. In vivo experiments showed that the caspase-3 activity increased with fluoride exposure level in a dose-dependent pattern Western blotting and immunohistochemistry results indicated that ERK relative activation tended to decrease as a function of fluoride exposure concentration. In contrast, relative activation of JNK increased with fluoride exposure level. Fluoride exposure did not appear to affect p38 activation. Furthermore, pretreatment of branchial cells with MAPK-specific inhibitors effectively prevented JNK induction and ERK inhibition, respectively, as well as reversed caspase-3 activity in fluoride-treated branchial cells. Our results indicate that activation of JNK and inactivation of ERK were caused by increased ROS and decreased antioxidant capacity in the gills of chronically exposed C. carpio described previously, which eventually caused the observed apoptosis in the fluoride-exposed gills and cells in C. carpio. JNK activation and ERK inactivation mechanism play a crucial role in gill impairment induced by chronic fluorosis. These findings contribute to a better understanding of the initial molecular and cellular events in the gill of fish chronically exposed to fluoride.
Collapse
Affiliation(s)
- Jinling Cao
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| | - Jianjie Chen
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jundong Wang
- State Key Laboratory of Ecological Animal Husbandry and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China.
| | - Paul Klerks
- Department of Biology, University of Louisiana at Lafayette, Box 42521, LA 70504, USA
| | - Lingtian Xie
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China.
| |
Collapse
|
42
|
Zhao K, Ju Y, Li S, Altaany Z, Wang R, Yang G. S-sulfhydration of MEK1 leads to PARP-1 activation and DNA damage repair. EMBO Rep 2014; 15:792-800. [PMID: 24778456 DOI: 10.1002/embr.201338213] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The repair of DNA damage is fundamental to normal cell development and replication. Hydrogen sulfide (H2S) is a novel gasotransmitter that has been reported to protect cellular aging. Here, we show that H2S attenuates DNA damage in human endothelial cells and fibroblasts by S-sulfhydrating MEK1 at cysteine 341, which leads to PARP-1 activation. H2S-induced MEK1 S-sulfhydration facilitates the translocation of phosphorylated ERK1/2 into nucleus, where it activates PARP-1 through direct interaction. Mutation of MEK1 cysteine 341 inhibits ERK phosphorylation and PARP-1 activation. In the presence of H2S, activated PARP-1 recruits XRCC1 and DNA ligase III to DNA breaks to mediate DNA damage repair, and cells are protected from senescence.
Collapse
Affiliation(s)
- Kexin Zhao
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology Lakehead University, Thunder Bay, ON, Canada
| | - YoungJun Ju
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology Lakehead University, Thunder Bay, ON, Canada
| | - Shuangshuang Li
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology Lakehead University, Thunder Bay, ON, Canada
| | - Zaid Altaany
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada Department of Biology, Lakehead University, Thunder Bay, ON, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada Department of Biology, Lakehead University, Thunder Bay, ON, Canada
| | - Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Lakehead University, Thunder Bay, ON, Canada The School of Kinesiology Lakehead University, Thunder Bay, ON, Canada
| |
Collapse
|
43
|
Silver nanoparticles induce anti-proliferative effects on airway smooth muscle cells. Role of nitric oxide and muscarinic receptor signaling pathway. Toxicol Lett 2013; 224:246-56. [PMID: 24188929 DOI: 10.1016/j.toxlet.2013.10.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 12/28/2022]
Abstract
Silver nanoparticles (AgNPs) are used to manufacture materials with new properties and functions. However, little is known about their toxic or beneficial effects on human health, especially in the respiratory system, where its smooth muscle (ASM) regulates the airway contractility by different mediators, such as acetylcholine (ACh) and nitric oxide (NO). The aim of this study was to evaluate the effects of AgNPs on ASM cells. Exposure to AgNPs induced ACh-independent expression of the inducible nitric oxide synthase (iNOS) at 100 μg/mL, associated with excessive production of NO. AgNPs induced the muscarinic receptor activation, since its blockage with atropine and blockage of its downstream signaling pathway inhibited the NO production. AgNPs at 10 and 100 μg/mL induced ACh-independent prolonged cytotoxicity and decreased cellular proliferation mediated by the muscarinic receptor-iNOS pathway. However, the concentration of 100 μg/mL of AgNPs induced muscarinic receptor-independent apoptosis, suggesting the activation of multiple pathways. These data indicate that AgNPs induce prolonged cytotoxic and anti-proliferative effects on ASM cells, suggesting an activation of the muscarinic receptor-iNOS pathway. Further investigation is required to understand the full mechanisms of action of AgNPs on ASM under specific biological conditions.
Collapse
|
44
|
Thiyagarajan M, Anderson H, Gonzales XF. Induction of apoptosis in human myeloid leukemia cells by remote exposure of resistive barrier cold plasma. Biotechnol Bioeng 2013; 111:565-74. [PMID: 24022746 DOI: 10.1002/bit.25114] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/23/2013] [Accepted: 09/03/2013] [Indexed: 12/31/2022]
Abstract
Cold atmospheric plasma (CAP), an ambient temperature ionized gas, is gaining extensive interest as a promising addition to anti-tumor therapy primarily due to the ability to generate and control delivery of electrons, ions, excited molecules, UV photons, and reactive species such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) to a specific site. The heterogeneous composition of CAP offers the opportunity to mediate several signaling pathways that regulate tumor cells. Consequently, the array of CAP generated products has limited the identification of the mechanisms of action on tumor cells. The aim of this work is to assess the cell death response of human myeloid leukemia cells by remote exposure to CAP generated RNS by utilizing a novel resistive barrier discharge system that primarily produces RNS. The effect of variable treatments of CAP generated RNS was tested in THP-1 cell (human monocytic leukemia cell line), a model for hematological malignancy. The number of viable cells was evaluated with erythrosine-B staining, while apoptosis and necrosis was assessed by endonuclease cleavage observed by agarose gel electrophoresis and detection of cells with the exclusionary dye propidium iodide and fluorescently labeled annexin-V by flow cytometry and fluorescent microscopy. Our observations indicate that treatment dosage levels of 45 s of exposure to CAP emitted RNS-induced apoptotic cell death and for higher dosage conditions of ≥50 s of exposure to CAP induced necrosis. Overall the results suggest that CAP emitted RNS play a significant role in the anti-tumor potential of CAP.
Collapse
Affiliation(s)
- Magesh Thiyagarajan
- Plasma Engineering Research Lab (PERL), Texas A&M University-Corpus Christi, EN 222D Engineering Building, 6300 Ocean Drive, Unit 5797, Corpus Christi, Texas, 78412.
| | | | | |
Collapse
|