1
|
Yamauchi N, Tamai K, Kimura I, Naito A, Tokuda N, Ashida Y, Motohashi N, Aoki Y, Yamada T. High-intensity interval training in the form of isometric contraction improves fatigue resistance in dystrophin-deficient muscle. J Physiol 2023; 601:2917-2933. [PMID: 37184335 DOI: 10.1113/jp284532] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023] Open
Abstract
Duchenne muscular dystrophy is a genetic muscle-wasting disorder characterized by progressive muscle weakness and easy fatigability. Here we examined whether high-intensity interval training (HIIT) in the form of isometric contraction improves fatigue resistance in skeletal muscle from dystrophin-deficient mdx52 mice. Isometric HIIT was performed on plantar flexor muscles in vivo with supramaximal electrical stimulation every other day for 4 weeks (a total of 15 sessions). In the non-trained contralateral gastrocnemius muscle from mdx52 mice, the decreased fatigue resistance was associated with a reduction in the amount of peroxisome proliferator-activated receptor γ coactivator 1-α, citrate synthase activity, mitochondrial respiratory complex II, LC3B-II/I ratio, and mitophagy-related gene expression (i.e. Pink1, parkin, Bnip3 and Bcl2l13) as well as an increase in the phosphorylation levels of Src Tyr416 and Akt Ser473, the amount of p62, and the percentage of Evans Blue dye-positive area. Isometric HIIT restored all these alterations and markedly improved fatigue resistance in mdx52 muscles. Moreover, an acute bout of HIIT increased the phosphorylation levels of AMP-activated protein kinase (AMPK) Thr172, acetyl CoA carboxylase Ser79, unc-51-like autophagy activating kinase 1 (Ulk1) Ser555, and dynamin-related protein 1 (Drp1) Ser616 in mdx52 muscles. Thus, our data show that HIIT with isometric contractions significantly mitigates histological signs of pathology and improves fatigue resistance in dystrophin-deficient muscles. These beneficial effects can be explained by the restoration of mitochondrial function via AMPK-dependent induction of the mitophagy programme and de novo mitochondrial biogenesis. KEY POINTS: Skeletal muscle fatigue is often associated with Duchenne muscular dystrophy (DMD) and leads to an inability to perform daily tasks, profoundly decreasing quality of life. We examined the effect of high-intensity interval training (HIIT) in the form of isometric contraction on fatigue resistance in skeletal muscle from the mdx52 mouse model of DMD. Isometric HIIT counteracted the reduced fatigue resistance as well as dystrophic changes in skeletal muscle of mdx52 mice. This beneficial effect could be explained by the restoration of mitochondrial function via AMP-activated protein kinase-dependent mitochondrial biogenesis and the induction of the mitophagy programme in the dystrophic muscles.
Collapse
Affiliation(s)
- Nao Yamauchi
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Katsuyuki Tamai
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Iori Kimura
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Azuma Naito
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Nao Tokuda
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Yuki Ashida
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
- The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
2
|
Wang SW, Zhang Q, Lu D, Fang YC, Yan XC, Chen J, Xia ZK, Yuan QT, Chen LH, Zhang YM, Nan FJ, Xie X. GPR84 regulates pulmonary inflammation by modulating neutrophil functions. Acta Pharmacol Sin 2023:10.1038/s41401-023-01080-z. [PMID: 37016043 PMCID: PMC10072043 DOI: 10.1038/s41401-023-01080-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/14/2023] [Indexed: 04/06/2023] Open
Abstract
Acute lung injury (ALI) is an acute, progressive hypoxic respiratory failure that could develop into acute respiratory distress syndrome (ARDS) with very high mortality rate. ALI is believed to be caused by uncontrolled inflammation, and multiple types of immune cells, especially neutrophils, are critically involved in the development of ALI. The treatment for ALI/ARDS is very limited, a better understanding of the pathogenesis and new therapies are urgently needed. Here we discover that GPR84, a medium chain fatty acid receptor, plays critical roles in ALI development by regulating neutrophil functions. GPR84 is highly upregulated in the cells isolated from the bronchoalveolar lavage fluid of LPS-induced ALI mice. GPR84 deficiency or blockage significantly ameliorated ALI mice lung inflammation by reducing neutrophils infiltration and oxidative stress. Further studies reveal that activation of GPR84 strongly induced reactive oxygen species production from neutrophils by stimulating Lyn, AKT and ERK1/2 activation and the assembly of the NADPH oxidase. These results reveal an important role of GPR84 in neutrophil functions and lung inflammation and strongly suggest that GPR84 is a potential drug target for ALI.
Collapse
Affiliation(s)
- Si-Wei Wang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Zhang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Dan Lu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - You-Chen Fang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Ci Yan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jing Chen
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhi-Kan Xia
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian-Ting Yuan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lin-Hai Chen
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | | | - Fa-Jun Nan
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
3
|
Role of c-Src and reactive oxygen species in cardiovascular diseases. Mol Genet Genomics 2023; 298:315-328. [PMID: 36700976 DOI: 10.1007/s00438-023-01992-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
Oxidative stress, caused by the over production of oxidants or inactivity of antioxidants, can modulate the redox state of several target proteins such as tyrosine kinases, mitogen-activated protein kinases and tyrosine phosphatases. c-Src is one such non-receptor tyrosine kinase which activates NADPH oxidases (Noxs) in response to various growth factors and shear stress. Interaction between c-Src and Noxs is influenced by cell type and primary messengers such as angiotensin II, which binds to G-protein coupled receptor and activates the intracellular signaling cascade. c-Src stimulated activation of Noxs results in elevated release of intracellular and extracellular reactive oxygen species (ROS). These ROS species disturb vascular homeostasis and cause cardiac hypertrophy, coronary artery disease, atherosclerosis and hypertension. Interaction between c-Src and ROS in the pathobiology of cardiac fibrosis is hypothesized to be influenced by cell type and stimuli. c-Src and ROS have a bidirectional relationship, thus increased ROS levels due to c-Src mediated activation of Noxs can further activate c-Src by promoting the oxidation and sulfenylation of critical cysteine residues. This review highlights the role of c-Src and ROS in mediating downstream signaling pathways underlying cardiovascular diseases. Furthermore, due to the central role of c-Src in activation of various signaling proteins involved in differentiation, migration, proliferation, and cytoskeletal reorganization of vascular cells, it is presented as therapeutic target for treating cardiovascular diseases except cardiac fibrosis.
Collapse
|
4
|
Filina YV, Tikhonova IV, Gabdoulkhakova AG, Rizvanov AA, Safronova VG. Mechanisms of ERK phosphorylation triggered via mouse formyl peptide receptor 2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119356. [PMID: 36087811 DOI: 10.1016/j.bbamcr.2022.119356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/09/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Formyl peptide receptors (FPRs) are expressed in the cells of the innate immune system and provide binding with pathogen and damage-associated molecular patterns with subsequent activation of the phagocytes for defense reactions such as chemotaxis, secretory degranulation and ROS generation. Probably, FPR2 is one of the unique receptors in the organism; it is able to recognize numerous ligands of different chemical structure, and moreover, these ligands can trigger opposite phagocyte responses promoting either pro- or anti-inflammatory reactions. Therefore, FPR2 and its signaling pathways are of intense research interest. We found only slight activation of ERK1/2 in the response to peptide ligand WKYMVM in the accelerating phase of ROS generation and more intense ERK1/2 phosphorylation in the declining phase of it in mouse bone marrow granulocytes. Lipid agonist BML-111 did not induce significant ERK phosphorylation when applied for 10-1800 s. To some extent co-localization of ERK1/2 and NADPH oxidase subunits was observed even in the intact cells and didn't change under FPR2 stimulation by WKYMVM, while direct PKC activation by PMA resulted to more efficient interaction between ERK1/2 and p47phox/p67phox and their translocation to plasma membrane. We have shown that phosphorylation and activation of ERK1/2 in bone marrow granulocytes depended on FPR2-triggered activity of PI3K and PKC, phosphatase DUSP6, and, the most but not the least, on ROS generation. Since blocking of ROS generation led to a slowdown of ERK activation indicating a significant contribution of ROS to the secondary regulation of ERK activity.
Collapse
Affiliation(s)
- Yu V Filina
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation.
| | - I V Tikhonova
- Laboratory of Cellular Neurobiology, Institute of Cell Biophysics of Russian Academy of Sciences, Pushchino, Russian Federation
| | - A G Gabdoulkhakova
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation; Central Research Laboratory, Kazan State Medical Academy, Federal State Budgetary Educational Institution of Further Professional Education "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Kazan, Russian Federation
| | - A A Rizvanov
- Openlab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - V G Safronova
- Laboratory of Cellular Neurobiology, Institute of Cell Biophysics of Russian Academy of Sciences, Pushchino, Russian Federation
| |
Collapse
|
5
|
Liu JF, Chen PC, Ling TY, Hou CH. Hyperthermia increases HSP production in human PDMCs by stimulating ROS formation, p38 MAPK and Akt signaling, and increasing HSF1 activity. Stem Cell Res Ther 2022; 13:236. [PMID: 35659731 PMCID: PMC9166587 DOI: 10.1186/s13287-022-02885-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Background Human placenta-derived multipotent cells (hPDMCs) are isolated from a source uncomplicated by ethical issues and are ideal for therapeutic applications because of their capacity for multilineage differentiation and proven immunosuppressive properties. It is known that heat shock preconditioning induces the upregulation of heat shock proteins (HSPs), which enhance survival and engraftment of embryonic stem cells (ESCs) during transplantation in live animal models, although whether heat shock preconditioning has the same effects in hPDMCs is unclear. Methods The hPDMCs were isolated from placenta of healthy donors. The cells were treated with heat shock (43 °C, 15 min), followed by evaluation of cell viability. Furthermore, the HSPs expression was assessed by Western blot, qPCR. The reactive oxygen species (ROS) production and signal pathway activation were determined by flow cytometry and Western blot, respectively. The regulatory pathways involved in HSPs expression were examined by pretreatment with chemical inhibitors, and siRNAs of MAPK, Akt, and heat shock factor 1 (HSF1), followed by determination of HSPs expression. Results This study demonstrates that heat shock treatment induced ROS generation and HPSs expression in hPDMCs. Heat shock stimulation also increased p38 MAPK and Akt phosphorylation. These effects were reduced by inhibitors of ROS, p38 MAPK and Akt. Moreover, we found that heat shock treatment enhanced nuclear translocation of the HSF1 in hPDMCs, representing activation of HSF1. Pretreatment of hPDMCs with ROS scavengers, SB203580 and Akt inhibitors also reduced the translocation of HSF1 induced by heat shock. Conclusions Our data indicate that heat shock acts via ROS to activate p38 MAPK and Akt signaling, which subsequently activates HSF1, leading to HSP activation and contributing to the protective role of hPDMCs.
Collapse
Affiliation(s)
- Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, 110, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan
| | - Po-Chun Chen
- Department of Life Science, National Taiwan Normal University, Taipei, 116, Taiwan.,Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, 111, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan
| | - Thai-Yen Ling
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, No. 1, Jen-Ai Road, Taipei, 100, Taiwan.
| |
Collapse
|
6
|
Harijith A, Basa P, Ha A, Thomas J, Jafri A, Fu P, MacFarlane PM, Raffay TM, Natarajan V, Sudhadevi T. NOX4 Mediates Epithelial Cell Death in Hyperoxic Acute Lung Injury Through Mitochondrial Reactive Oxygen Species. Front Pharmacol 2022; 13:880878. [PMID: 35662702 PMCID: PMC9160661 DOI: 10.3389/fphar.2022.880878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Management of acute respiratory distress involves O2 supplementation, which is lifesaving, but causes severe hyperoxic acute lung injury (HALI). NADPH oxidase (NOX) could be a major source of reactive oxygen species (ROS) in hyperoxia (HO). Epithelial cell death is a crucial step in the development of many lung diseases. Alveolar type II (AT2) cells are the metabolically active epithelial cells of alveoli that serve as a source of AT1 cells following lung injury. The aim of this study was to determine the possible role of AT2 epithelial cell NOX4 in epithelial cell death from HALI. Wild type (WT), Nox4 fl/fl (control), and Nox4 -/- Spc-Cre mice were exposed to room air (NO) or 95% O2 (HO) to investigate the structural and functional changes in the lung. C57BL/6J WT animals subjected to HO showed increased expression of lung NOX4 compared to NO. Significant HALI, increased bronchoalveolar lavage cell counts, increased protein levels, elevated proinflammatory cytokines and increased AT2 cell death seen in hyperoxic Nox4 fl/fl control mice were attenuated in HO-exposed Nox4 -/- Spc-Cre mice. HO-induced expression of NOX4 in MLE cells resulted in increased mitochondrial (mt) superoxide production and cell apoptosis, which was reduced in NOX4 siRNA silenced cells. This study demonstrates a novel role for epithelial cell NOX4 in accelerating lung epithelial cell apoptosis from HALI. Deletion of the Nox4 gene in AT2 cells or silencing NOX4 in lung epithelial cells protected the lungs from severe HALI with reduced apoptosis and decreased mt ROS production in HO. These results suggest NOX4 as a potential target for the treatment of HALI.
Collapse
Affiliation(s)
- Anantha Harijith
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Prathima Basa
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Alison Ha
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jaya Thomas
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Panfeng Fu
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Peter M. MacFarlane
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Thomas M. Raffay
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Internal Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Tara Sudhadevi
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
7
|
NOX1 and NOX2: two enzymes that promote endothelial-to-mesenchymal transition induced by melanoma conditioned media. Pharmacol Res 2022; 177:106097. [DOI: 10.1016/j.phrs.2022.106097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/11/2022]
|
8
|
Gamara J, Davis L, Leong AZ, Pagé N, Rollet-Labelle E, Zhao C, Hongu T, Funakoshi Y, Kanaho Y, Aoudji F, Pelletier M, Bourgoin SG. Arf6 regulates energy metabolism in neutrophils. Free Radic Biol Med 2021; 172:550-561. [PMID: 34245858 DOI: 10.1016/j.freeradbiomed.2021.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/24/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
The small GTPase Arf6 regulates many cellular processes, including cytoskeletal remodeling, receptor endocytosis, and pathogen phagocytosis. Arf6 silencing in neutrophil (PMN)-like cells is well-known to inhibit chemotactic peptide-mediated activation of phospholipase D, the oxidative burst, and β2 integrin-dependent adhesion. In conditional knockout (cKO) mice, the migration to inflammatory sites of Arf6-deficient PMNs was diminished and associated with reduced cell surface expression of β2 integrins. In this study we assessed the impact of Arf6 depletion on the functions and gene expression profile of PMNs isolated from the mouse air pouch. Numerous genes involved in response to oxygen levels, erythrocyte and myeloid differentiation, macrophage chemotaxis, response to chemicals, apoptosis, RNA destabilization, endosome organization, and vesicle transport were differentially expressed in PMNs cKO for Arf6. Lpar6 and Lacc-1 were the most up-regulated and down-regulated genes, respectively. The deletion of Arf6 also decreased Lacc-1 protein level in PMNs, and silencing of Arf6 in THP-1 monocytic cells delayed LPS-mediated Lacc-1 expression. We report that fMLP or zymosan-induced glycolysis and oxygen consumption rate were both decreased in air pouch PMNs but not in bone marrow PMNs of Arf6 cKO mice. Reduced oxygen consumption correlated with a decrease in superoxide and ROS production. Deletion of Arf6 in PMNs also reduced phagocytosis and interfered with apoptosis. The data suggest that Arf6 regulates energy metabolism, which may contribute to impaired phagocytosis, ROS production, and apoptosis in PMN-Arf6 cKO. This study provides new information on the functions and the inflammatory pathways influenced by Arf6 in PMNs.
Collapse
Affiliation(s)
- Jouda Gamara
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2
| | - Lynn Davis
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2
| | - Andrew Z Leong
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2
| | - Nathalie Pagé
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2
| | - Emmanuelle Rollet-Labelle
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2
| | - Chenqi Zhao
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2
| | - Tsunaki Hongu
- German Cancer Research Centre (DFKZ), Group of Metastatic Niches, 69120, Heidelberg, Germany
| | - Yuji Funakoshi
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennohdai, Tsukuba, 305-8575, Japan
| | - Yasunori Kanaho
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennohdai, Tsukuba, 305-8575, Japan
| | - Fawzi Aoudji
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2; Centre ARThrite, Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec, QC, Canada, G1V0A6
| | - Martin Pelletier
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2; Centre ARThrite, Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec, QC, Canada, G1V0A6
| | - Sylvain G Bourgoin
- Division of Infectious Disease and Immunology, CHU de Quebec Research Center, Quebec, QC, Canada, G1V4G2; Centre ARThrite, Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec, QC, Canada, G1V0A6.
| |
Collapse
|
9
|
Pak ES, Uddin MJ, Ha H. Inhibition of Src Family Kinases Ameliorates LPS-Induced Acute Kidney Injury and Mitochondrial Dysfunction in Mice. Int J Mol Sci 2020; 21:ijms21218246. [PMID: 33153232 PMCID: PMC7662942 DOI: 10.3390/ijms21218246] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury (AKI), a critical syndrome characterized by a rapid decrease of kidney function, is a global health problem. Src family kinases (SFK) are proto-oncogenes that regulate diverse biological functions including mitochondrial function. Since mitochondrial dysfunction plays an important role in the development of AKI, and since unbalanced SFK activity causes mitochondrial dysfunction, the present study examined the role of SFK in AKI. Lipopolysaccharides (LPS) inhibited mitochondrial biogenesis and upregulated the expression of NGAL, a marker of tubular epithelial cell injury, in mouse proximal tubular epithelial (mProx) cells. These alterations were prevented by PP2, a pan SFK inhibitor. Importantly, PP2 pretreatment significantly ameliorated LPS-induced loss of kidney function and injury including inflammation and oxidative stress. The attenuation of LPS-induced AKI by PP2 was accompanied by the maintenance of mitochondrial biogenesis. LPS upregulated SFK, especially Fyn and Src, in mouse kidney as well as in mProx cells. These data suggest that Fyn and Src kinases are involved in the pathogenesis of LPS-induced AKI, and that inhibition of Fyn and Src kinases may have a potential therapeutic effect, possibly via improving mitochondrial biogenesis.
Collapse
Affiliation(s)
| | | | - Hunjoo Ha
- Correspondence: ; Tel.: +82-2-3277-4075; Fax: +82-2-3277-2851
| |
Collapse
|
10
|
Abstract
IMPACT STATEMENT Tumor hypoxia promotes cancer cell aggressiveness, and is strongly associated with poor prognosis across multiple tumor types. The hypoxic microenvironments inside tumors also limit the effectiveness of radiotherapy, chemotherapy, and immunotherapy. Several approaches to eliminate hypoxic state in tumors have been proposed to delay cancer progression and improve therapeutic efficacies. This review will summarize current knowledge on hyperoxia, used alone or in combination with other therapeutic modalities, in cancer treatment. Molecular mechanisms and undesired side effects of hyperoxia will also be discussed.
Collapse
Affiliation(s)
- Sei W Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
| | - In K Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang H Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
11
|
Henríquez-Olguín C, Boronat S, Cabello-Verrugio C, Jaimovich E, Hidalgo E, Jensen TE. The Emerging Roles of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 in Skeletal Muscle Redox Signaling and Metabolism. Antioxid Redox Signal 2019; 31:1371-1410. [PMID: 31588777 PMCID: PMC6859696 DOI: 10.1089/ars.2018.7678] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Skeletal muscle is a crucial tissue to whole-body locomotion and metabolic health. Reactive oxygen species (ROS) have emerged as intracellular messengers participating in both physiological and pathological adaptations in skeletal muscle. A complex interplay between ROS-producing enzymes and antioxidant networks exists in different subcellular compartments of mature skeletal muscle. Recent evidence suggests that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a major source of contraction- and insulin-stimulated oxidants production, but they may paradoxically also contribute to muscle insulin resistance and atrophy. Recent Advances: Pharmacological and molecular biological tools, including redox-sensitive probes and transgenic mouse models, have generated novel insights into compartmentalized redox signaling and suggested that NOX2 contributes to redox control of skeletal muscle metabolism. Critical Issues: Major outstanding questions in skeletal muscle include where NOX2 activation occurs under different conditions in health and disease, how NOX2 activation is regulated, how superoxide/hydrogen peroxide generated by NOX2 reaches the cytosol, what the signaling mediators are downstream of NOX2, and the role of NOX2 for different physiological and pathophysiological processes. Future Directions: Future research should utilize and expand the current redox-signaling toolbox to clarify the NOX2-dependent mechanisms in skeletal muscle and determine whether the proposed functions of NOX2 in cells and animal models are conserved into humans.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Susanna Boronat
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
13
|
De Mario A, Peggion C, Massimino ML, Norante RP, Zulian A, Bertoli A, Sorgato MC. The Link of the Prion Protein with Ca 2+ Metabolism and ROS Production, and the Possible Implication in Aβ Toxicity. Int J Mol Sci 2019; 20:ijms20184640. [PMID: 31546771 PMCID: PMC6770541 DOI: 10.3390/ijms20184640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 01/05/2023] Open
Abstract
The cellular prion protein (PrPC) is an ubiquitous cell surface protein mostly expressed in neurons, where it localizes to both pre- and post-synaptic membranes. PrPC aberrant conformers are the major components of mammalian prions, the infectious agents responsible for incurable neurodegenerative disorders. PrPC was also proposed to bind aggregated misfolded proteins/peptides, and to mediate their neurotoxic signal. In spite of long-lasting research, a general consensus on the precise pathophysiologic mechanisms of PrPC has not yet been reached. Here we review our recent data, obtained by comparing primary neurons from PrP-expressing and PrP-knockout mice, indicating a central role of PrPC in synaptic transmission and Ca2+ homeostasis. Indeed, by controlling gene expression and signaling cascades, PrPC is able to optimize glutamate secretion and regulate Ca2+ entry via store-operated channels and ionotropic glutamate receptors, thereby protecting neurons from threatening Ca2+ overloads and excitotoxicity. We will also illustrate and discuss past and unpublished results demonstrating that Aβ oligomers perturb Ca2+ homeostasis and cause abnormal mitochondrial accumulation of reactive oxygen species by possibly affecting the PrP-dependent downregulation of Fyn kinase activity.
Collapse
Affiliation(s)
- Agnese De Mario
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Caterina Peggion
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Maria Lina Massimino
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Rosa Pia Norante
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Alessandra Zulian
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy.
| | - Maria Catia Sorgato
- Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
- CNR Neuroscience Institute, Department of Biomedical Science, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
14
|
Gao Y, Raj JU. Src and Epidermal Growth Factor Receptor: Novel Partners in Mediating Chronic Hypoxia-induced Pulmonary Artery Hypertension. Am J Respir Cell Mol Biol 2019; 62:5-7. [PMID: 31298924 PMCID: PMC6938126 DOI: 10.1165/rcmb.2019-0230ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Yuansheng Gao
- Health Science CenterPeking UniversityBeijing, Chinaand
| | - J Usha Raj
- College of MedicineUniversity of Illinois at ChicagoChicago, Illinois
| |
Collapse
|
15
|
Prasad GVRK, Dhar V, Mukhopadhaya A. Vibrio cholerae OmpU Mediates CD36-Dependent Reactive Oxygen Species Generation Triggering an Additional Pathway of MAPK Activation in Macrophages. THE JOURNAL OF IMMUNOLOGY 2019; 202:2431-2450. [PMID: 30867241 DOI: 10.4049/jimmunol.1800389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 02/06/2019] [Indexed: 01/18/2023]
Abstract
OmpU, one of the porins of Gram-negative bacteria Vibrio cholerae, induces TLR1/2-MyD88-NF-κB-dependent proinflammatory cytokine production by monocytes and macrophages of human and mouse origin. In this study, we report that in both the cell types, OmpU-induced proinflammatory responses involve activation of MAPKs (p38 and JNK). Interestingly, we observed that in OmpU-treated macrophages, p38 activation is TLR2 dependent, but JNK activation happens through a separate pathway involving reactive oxygen species (ROS) generation by NADPH oxidase complex and mitochondrial ROS. Further, we observed that OmpU-mediated mitochondrial ROS generation probably depends on OmpU translocation to mitochondria and NADPH oxidase-mediated ROS production is due to activation of scavenger receptor CD36. For the first time, to our knowledge, we are reporting that a Gram-negative bacterial protein can activate CD36 as a pattern recognition receptor. Additionally, we found that in OmpU-treated monocytes, both JNK and p38 activation is linked to the TLR2 activation only. Therefore, the ability of macrophages to employ multiple receptors such as TLR2 and CD36 to recognize a single ligand, as in this case OmpU, probably explains the very basic nature of macrophages being more proinflammatory than monocytes.
Collapse
Affiliation(s)
- G V R Krishna Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, 140306 Punjab, India
| | - Vinica Dhar
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, 140306 Punjab, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, 140306 Punjab, India
| |
Collapse
|
16
|
Parascandolo A, Laukkanen MO. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal 2019; 30:443-486. [PMID: 29478325 PMCID: PMC6393772 DOI: 10.1089/ars.2017.7268] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Reduction/oxidation (redox) balance could be defined as an even distribution of reduction and oxidation complementary processes and their reaction end products. There is a consensus that aberrant levels of reactive oxygen species (ROS), commonly observed in cancer, stimulate primary cell immortalization and progression of carcinogenesis. However, the mechanism how different ROS regulate redox balance is not completely understood. Recent Advances: In the current review, we have summarized the main signaling cascades inducing NADPH oxidase NOX1-5 and superoxide dismutase (SOD) 1-3 expression and their connection to cell proliferation, immortalization, transformation, and CD34+ cell differentiation in thyroid, colon, lung, breast, and hematological cancers. CRITICAL ISSUES Interestingly, many of the signaling pathways activating redox enzymes or mediating the effect of ROS are common, such as pathways initiated from G protein-coupled receptors and tyrosine kinase receptors involving protein kinase A, phospholipase C, calcium, and small GTPase signaling molecules. FUTURE DIRECTIONS The clarification of interaction of signal transduction pathways could explain how cells regulate redox balance and may even provide means to inhibit the accumulation of harmful levels of ROS in human pathologies.
Collapse
|
17
|
Abstract
SIGNIFICANCE G protein-coupled receptors (GPCR) are the largest group of cell surface receptors, which link cells to their environment. Reactive oxygen species (ROS) can act as important cellular signaling molecules. The family of NADPH oxidases generates ROS in response to activated cell surface receptors. Recent Advances: Various signaling pathways linking GPCRs and activation of NADPH oxidases have been characterized. CRITICAL ISSUES Still, a more detailed analysis of G proteins involved in the GPCR-mediated activation of NADPH oxidases is needed. In addition, a more precise discrimination of NADPH oxidase activation due to either upregulation of subunit expression or post-translational subunit modifications is needed. Also, the role of noncanonical modulators of NADPH oxidase activation in the response to GPCRs awaits further analyses. FUTURE DIRECTIONS As GPCRs are one of the most popular classes of investigational drug targets, further detailing of G protein-coupled mechanisms in the activation mechanism of NADPH oxidases as well as better understanding of the link between newly identified NADPH oxidase interaction partners and GPCR signaling will provide new opportunities for improved efficiency and decreased off target effects of therapies targeting GPCRs.
Collapse
Affiliation(s)
- Andreas Petry
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany
| | - Agnes Görlach
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research) , Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
18
|
Lin HC, Li CC, Yang YC, Chiu TH, Liu KL, Lii CK, Chen HW. Andrographis paniculata diterpenoids and ethanolic extract inhibit TNFα-induced ICAM-1 expression in EA.hy926 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 52:157-167. [PMID: 30599895 DOI: 10.1016/j.phymed.2018.09.205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 08/03/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Andrographis paniculata (A. paniculata), a traditional herb in Southeastern Asia, is used to treat inflammation-mediated diseases. PURPOSE The two major bioactive diterpenoids in A. paniculata are andrographolide (AND) and 14-deoxy-11,12-didehydroandrographolide (deAND). Because of the anti-inflammatory evidence for AND, we hypothesized that deAND might possess similar potency for inhibiting monocyte adhesion to the vascular endothelium, which is a critical event for atherosclerotic lesion formation. MATERIAL In the present study, we used 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to determine cell viability. We evaluated the production of intracellular reactive oxygen species (ROS) by using DCFDA assay. We assayed the protein expression by using Western blot analysis, the mRNA expression by using RT-PCR, and the nuclear protein-DNA binding activity by using EMSA. RESULTS We showed that pretreatment of EA.hy926 cells with A. paniculata ethanolic extract (APE), deAND, and AND significantly inhibited TNFα-induced ICAM-1 protein and mRNA expression, ICAM-1 promoter activity, and monocyte adhesion. TNFα-stimulated IKKβ phosphorylation, IκBα phosphorylation and degradation, p65 nuclear translocation, and NFκB nuclear protein-DNA binding activity were attenuated by pretreatment with APE, deAND, and AND. APE, deAND, and AND attenuated TNFα-induced Src phosphorylation and membrane translocation of the NOX subunits p47phox and p67phox. Both APE and AND induced protein expression of heme oxygenase 1 and the glutamate cysteine ligase modifier subunit and enhanced glutathione content. Pretreatment with AND and deAND inhibited TNFα-induced ROS generation. CONCLUSION These results suggest that the mechanism by which APE, deAND, and AND down-regulates TNFα-induced ICAM-1 expression in EA.hy926 cells is via attenuation of activation of the IKK/IκB/NFκB pathway.
Collapse
Affiliation(s)
- Hung-Chih Lin
- Division of Neonatology, College of Medicine and Department of Pediatrics, Children's Hospital of China Medical University and China Medical University Hospital, Taichung 404, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung 402, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ya-Chen Yang
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413, Taiwan
| | - Tzu-Hsuan Chiu
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung 402, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chong-Kuei Lii
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413, Taiwan; Department of Nutrition, China Medical University, Taichung 404, Taiwan.
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
19
|
Zhang KY, Gao P, Sun G, Zhang T, Li X, Liu S, Zhao Q, Lo KKW, Huang W. Dual-Phosphorescent Iridium(III) Complexes Extending Oxygen Sensing from Hypoxia to Hyperoxia. J Am Chem Soc 2018; 140:7827-7834. [PMID: 29874455 DOI: 10.1021/jacs.8b02492] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hypoxia and hyperoxia, referring to states of biological tissues in which oxygen supply is in sufficient and excessive, respectively, are often pathological conditions. Many luminescent oxygen probes have been developed for imaging intracellular and in vivo hypoxia, but their sensitivity toward hyperoxia becomes very low. Here we report a series of iridium(III) complexes in which limited internal conversion between two excited states results in dual phosphorescence from two different excited states upon excitation at a single wavelength. Structural manipulation of the complexes allows rational tuning of the dual-phosphorescence properties and the spectral profile response of the complexes toward oxygen. By manipulating the efficiency of internal conversion between the two emissive states, we obtained a complex exhibiting naked-eye distinguishable green, orange, and red emission in aqueous buffer solution under an atmosphere of N2, air, and O2, respectively. This complex is used for intracellular and in vivo oxygen sensing not only in the hypoxic region but also in normoxic and hyperoxic intervals. To the best of our knowledge, this is the first example of using a molecular probe for simultaneous bioimaging of hypoxia and hyperoxia.
Collapse
Affiliation(s)
- Kenneth Yin Zhang
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Pengli Gao
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Guanglan Sun
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Taiwei Zhang
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Xiangling Li
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Shujuan Liu
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Qiang Zhao
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry , City University of Hong Kong , Tat Chee Avenue , Hong Kong , P. R. China
| | - Wei Huang
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , 9 Wenyuan Road , Nanjing 210023 , P. R. China.,Key Laboratory of Flexible Electronics and Institute of Advanced Materials, Jiangsu National Synergetic Innovation Center for Advanced Materials , Nanjing Tech University , 30 South Puzhu Road , Nanjing 211816 , P. R. China
| |
Collapse
|
20
|
Su CM, Chen CY, Lu T, Sun Y, Li W, Huang YL, Tsai CH, Chang CS, Tang CH. A novel benzofuran derivative, ACDB, induces apoptosis of human chondrosarcoma cells through mitochondrial dysfunction and endoplasmic reticulum stress. Oncotarget 2018; 7:83530-83543. [PMID: 27835579 PMCID: PMC5347786 DOI: 10.18632/oncotarget.13171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022] Open
Abstract
Chondrosarcoma is one of the bone tumor with high mortality in respond to poor radiation and chemotherapy treatment. Here, we analyze the antitumor activity of a novel benzofuran derivative, 2-amino-3-(2-chlorophenyl)-6-(4-dimethylaminophenyl)benzofuran-4-yl acetate (ACDB), in human chondrosarcoma cells. ACDB increased the cell apoptosis of human chondrosarcomas without harm in chondrocytes. ACDB also enhanced endoplasmic reticulum (ER) stress, which was characterized by varieties in the cytosolic calcium levels and induced the expression of glucose-regulated protein (GRP) and calpain. Furthermore, the ACDB-induced chondrosarcoma apoptosis was associated with the upregulation of the B cell lymphoma-2 (Bcl-2) family members including pro- and anti-apoptotic proteins, downregulation of dysfunctional mitochondria that released cytochrome C, and subsequent activation of caspases-3. In addition, the ACDB-mediated cellular apoptosis was suppressed by transfecting cells with glucose-regulated protein (GRP) and calpain siRNA or treating cells with ER stress chelators and caspase inhibitors. Interestingly, animal experiments illustrated a reduction in the tumor volume following ACDB treatment. Together, these results suggest that ACDB may be a novel tumor suppressor of chondrosarcoma, and this study demonstrates that the novel antitumor agent, ACDB, induced apoptosis by mitochondrial dysfunction and ER stress in human chondrosarcoma cells in vitro and in vivo.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China.,Graduate Institute of Basic Medical Science, China Medical University, Taichung Taiwan
| | - Chien-Yu Chen
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung, Taiwan
| | - Tingting Lu
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Yi Sun
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Weimin Li
- Department of Cardiology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- School of Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Shiang Chang
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
21
|
Goncharov NV, Nadeev AD, Jenkins RO, Avdonin PV. Markers and Biomarkers of Endothelium: When Something Is Rotten in the State. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9759735. [PMID: 29333215 PMCID: PMC5733214 DOI: 10.1155/2017/9759735] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022]
Abstract
Endothelium is a community of endothelial cells (ECs), which line the blood and lymphatic vessels, thus forming an interface between the tissues and the blood or lympha. This strategic position of endothelium infers its indispensable functional role in controlling vasoregulation, haemostasis, and inflammation. The state of endothelium is simultaneously the cause and effect of many diseases, and this is coupled with modifications of endothelial phenotype represented by markers and with biochemical profile of blood represented by biomarkers. In this paper, we briefly review data on the functional role of endothelium, give definitions of endothelial markers and biomarkers, touch on the methodological approaches for revealing biomarkers, present an implicit role of endothelium in some toxicological mechanistic studies, and survey the role of reactive oxygen species (ROS) in modulation of endothelial status.
Collapse
Affiliation(s)
- Nikolay V. Goncharov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Saint Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint Petersburg, Russia
| | - Alexander D. Nadeev
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint Petersburg, Russia
- Institute of Cell Biophysics RAS, Pushchino, Russia
| | - Richard O. Jenkins
- School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | | |
Collapse
|
22
|
Chakraborti S, Sarkar J, Chowdhury A, Chakraborti T. Role of ADP ribosylation factor6- Cytohesin1-PhospholipaseD signaling axis in U46619 induced activation of NADPH oxidase in pulmonary artery smooth muscle cell membrane. Arch Biochem Biophys 2017; 633:1-14. [PMID: 28822840 DOI: 10.1016/j.abb.2017.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/12/2017] [Accepted: 08/15/2017] [Indexed: 01/07/2023]
Abstract
Treatment of human pulmonary artery smooth muscle cells (HPASMCs) with the thromboxane A2 receptor antagonist, SQ29548 inhibited U46619 stimulation of phospholipase D (PLD) and NADPH oxidase activities in the cell membrane. Pretreatment with apocynin inhibited U46619 induced increase in NADPH oxidase activity. The cell membrane contains predominantly PLD2 along with PLD1 isoforms of PLD. Pretreatment with pharmacological and genetic inhibitors of PLD2, but not PLD1, attenuated U46619 stimulation of NADPH oxidase activity. U46619 stimulation of PLD and NADPH oxidase activities were insensitive to BFA and Clostridium botulinum C3 toxin; however, pretreatment with secinH3 inhibited U46619 induced increase in PLD and NADPH oxidase activities suggesting a major role of cytohesin in U46619-induced increase in PLD and NADPH oxidase activities. Arf-1, Arf-6, cytohesin-1 and cytohesin-2 were observed in the cytosolic fraction, but only Arf-6 and cytohesin-1 were translocated to the cell membrane upon treatment with U46619. Coimmunoprecipitation study showed association of Arf-6 with cytohesin-1 in the cell membrane fraction. In vitro binding of GTPγS with Arf-6 required the presence of cytohesin-1 and that occurs in BFA insensitive manner. Overall, BFA insensitive Arf6-cytohesin1 signaling axis plays a pivotal role in U46619-mediated activation of PLD leading to stimulation of NADPH oxidase activity in HPASMCs.
Collapse
Affiliation(s)
- Sajal Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India.
| | - Jaganmay Sarkar
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India.
| | - Animesh Chowdhury
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India.
| | - Tapati Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India.
| |
Collapse
|
23
|
Xu MM, Pu Y, Han D, Shi Y, Cao X, Liang H, Chen X, Li XD, Deng L, Chen ZJ, Weichselbaum RR, Fu YX. Dendritic Cells but Not Macrophages Sense Tumor Mitochondrial DNA for Cross-priming through Signal Regulatory Protein α Signaling. Immunity 2017; 47:363-373.e5. [PMID: 28801234 DOI: 10.1016/j.immuni.2017.07.016] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/09/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023]
Abstract
Inhibition of cytosolic DNA sensing represents a strategy that tumor cells use for immune evasion, but the underlying mechanisms are unclear. Here we have shown that CD47-signal regulatory protein α (SIRPα) axis dictates the fate of ingested DNA in DCs for immune evasion. Although macrophages were more potent in uptaking tumor DNA, increase of DNA sensing by blocking the interaction of SIRPα with CD47 preferentially occurred in dendritic cells (DCs) but not in macrophages. Mechanistically, CD47 blockade enabled the activation of NADPH oxidase NOX2 in DCs, which in turn inhibited phagosomal acidification and reduced the degradation of tumor mitochondrial DNA (mtDNA) in DCs. mtDNA was recognized by cyclic-GMP-AMP synthase (cGAS) in the DC cytosol, contributing to type I interferon (IFN) production and antitumor adaptive immunity. Thus, our findings have demonstrated how tumor cells inhibit innate sensing in DCs and suggested that the CD47-SIRPα axis is critical for DC-driven antitumor immunity.
Collapse
Affiliation(s)
- Meng Michelle Xu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Yang Pu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Dali Han
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Yaoyao Shi
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Xuezhi Cao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Hua Liang
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Xiang Chen
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiao-Dong Li
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Liufu Deng
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Zhijian J Chen
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
24
|
Sharma AK, LaPar DJ, Stone ML, Zhao Y, Mehta CK, Kron IL, Laubach VE. NOX2 Activation of Natural Killer T Cells Is Blocked by the Adenosine A2A Receptor to Inhibit Lung Ischemia-Reperfusion Injury. Am J Respir Crit Care Med 2017; 193:988-99. [PMID: 26757359 DOI: 10.1164/rccm.201506-1253oc] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RATIONALE Ischemia-reperfusion (IR) injury after lung transplantation, which affects both short- and long-term allograft survival, involves activation of NADPH oxidase 2 (NOX2) and activation of invariant natural killer T (iNKT) cells to produce IL-17. Adenosine A2A receptor (A2AR) agonists are known to potently attenuate lung IR injury and IL-17 production. However, mechanisms for iNKT cell activation after IR and A2AR agonist-mediated protection remain unclear. OBJECTIVES We tested the hypothesis that NOX2 mediates IL-17 production by iNKT cells after IR and that A2AR agonism prevents IR injury by blocking NOX2 activation in iNKT cells. METHODS An in vivo murine hilar ligation model of IR injury was used, in which left lungs underwent 1 hour of ischemia and 2 hours of reperfusion. MEASUREMENTS AND MAIN RESULTS Adoptive transfer of iNKT cells from p47(phox-/-) or NOX2(-/-) mice to Jα18(-/-) (iNKT cell-deficient) mice significantly attenuated lung IR injury and IL-17 production. Treatment with an A2AR agonist attenuated IR injury and IL-17 production in wild-type (WT) mice and in Jα18(-/-) mice reconstituted with WT, but not A2AR(-/-), iNKT cells. Furthermore, the A2AR agonist prevented IL-17 production by murine and human iNKT cells after acute hypoxia-reoxygenation by blocking p47(phox) phosphorylation, a critical step for NOX2 activation. CONCLUSIONS NOX2 plays a key role in inducing iNKT cell-mediated IL-17 production and subsequent lung injury after IR. A primary mechanism for A2AR agonist-mediated protection entails inhibition of NOX2 in iNKT cells. Therefore, agonism of A2ARs on iNKT cells may be a novel therapeutic strategy to prevent primary graft dysfunction after lung transplantation.
Collapse
Affiliation(s)
- Ashish K Sharma
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Damien J LaPar
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Matthew L Stone
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Yunge Zhao
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | | | - Irving L Kron
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Victor E Laubach
- Department of Surgery, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
25
|
Lin CC, Lin WN, Cho RL, Wang CY, Hsiao LD, Yang CM. TNF-α-Induced cPLA 2 Expression via NADPH Oxidase/Reactive Oxygen Species-Dependent NF-κB Cascade on Human Pulmonary Alveolar Epithelial Cells. Front Pharmacol 2016; 7:447. [PMID: 27932980 PMCID: PMC5122718 DOI: 10.3389/fphar.2016.00447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/08/2016] [Indexed: 01/13/2023] Open
Abstract
Tumor necrosis factor-α (TNF-α) triggers activation of cytosolic phospholipase A2 (cPLA2) and then enhancing the synthesis of prostaglandin (PG) in inflammatory diseases. However, the detailed mechanisms of TNF-α induced cPLA2 expression were not fully defined in human pulmonary alveolar epithelial cells (HPAEpiCs). We found that TNF-α-stimulated increases in cPLA2 mRNA (5.2 folds) and protein (3.9 folds) expression, promoter activity (4.3 folds), and PGE2 secretion (4.7 folds) in HPAEpiCs, determined by Western blot, real-time PCR, promoter activity assay and PGE2 ELISA kit. These TNF-α-mediated responses were abrogated by the inhibitors of NADPH oxidase [apocynin (APO) and diphenyleneiodonium chloride (DPI)], ROS [N-acetyl cysteine, (NAC)], NF-κB (Bay11-7082) and transfection with siRNA of ASK1, p47 phox , TRAF2, NIK, IKKα, IKKβ, or p65. TNF-α markedly stimulated NADPH oxidase activation and ROS including superoxide and hydrogen peroxide production which were inhibited by pretreatment with a TNFR1 neutralizing antibody, APO, DPI or transfection with siRNA of TRAF2, ASK1, or p47 phox . In addition, TNF-α also stimulated p47 phox phosphorylation and translocation in a time-dependent manner. On the other hand, TNF-α induced TNFR1, TRAF2, ASK1, and p47 phox complex formation in HPAEpiCs, which were attenuated by a TNF-α neutralizing antibody. We found that pretreatment with NAC, DPI, or APO also attenuated the TNF-α-stimulated IKKα/β and NF-κB p65 phosphorylation, NF-κB (p65) translocation, and NF-κB promoter activity in HPAEpiCs. Finally, we observed that TNF-α-stimulated NADPH oxidase activation and ROS generation activates NF-κB through the NIK/IKKα/β pathway. Taken together, our results demonstrated that in HPAEpiCs, up-regulation of cPLA2 by TNF-α is, at least in part, mediated through the cooperation of TNFR1, TRAF2, ASK1, and NADPH oxidase leading to ROS generation and ultimately activates NF-κB pathway.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Basic Medicine, Fu Jen Catholic University New Taipei City, Taiwan
| | - Rou-Ling Cho
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chen-Yu Wang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Anesthetics, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan; Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan; Research Center for Industry of Human Ecology, Research Center for Chinese Herbal Medicine, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and TechnologyTao-Yuan, Taiwan
| |
Collapse
|
26
|
Molecular mechanisms underlying hyperoxia acute lung injury. Respir Med 2016; 119:23-28. [DOI: 10.1016/j.rmed.2016.08.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/12/2022]
|
27
|
Ferreira LF, Laitano O. Regulation of NADPH oxidases in skeletal muscle. Free Radic Biol Med 2016; 98:18-28. [PMID: 27184955 PMCID: PMC4975970 DOI: 10.1016/j.freeradbiomed.2016.05.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/31/2016] [Accepted: 05/12/2016] [Indexed: 12/20/2022]
Abstract
The only known function of NAD(P)H oxidases is to produce reactive oxygen species (ROS). Skeletal muscles express three isoforms of NAD(P)H oxidases (Nox1, Nox2, and Nox4) that have been identified as critical modulators of redox homeostasis. Nox2 acts as the main source of skeletal muscle ROS during contractions, participates in insulin signaling and glucose transport, and mediates the myocyte response to osmotic stress. Nox2 and Nox4 contribute to skeletal muscle abnormalities elicited by angiotensin II, muscular dystrophy, heart failure, and high fat diet. Our review addresses the expression and regulation of NAD(P)H oxidases with emphasis on aspects that are relevant to skeletal muscle. We also summarize: i) the most widely used NAD(P)H oxidases activity assays and inhibitors, and ii) studies that have defined Nox enzymes as protagonists of skeletal muscle redox homeostasis in a variety of health and disease conditions.
Collapse
Affiliation(s)
- Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Universidade Federal do Vale do São Francisco, Petrolina, PE, Brazil
| |
Collapse
|
28
|
The lectin Siglec-G inhibits dendritic cell cross-presentation by impairing MHC class I-peptide complex formation. Nat Immunol 2016; 17:1167-75. [PMID: 27548433 DOI: 10.1038/ni.3535] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
Abstract
CD8α(+) dendritic cells (DCs) are specialized at cross-presenting extracellular antigens on major histocompatibility complex (MHC) class I molecules to initiate cytotoxic T lymphocyte (CTL) responses; however, details of the mechanisms that regulate cross-presentation remain unknown. We found lower expression of the lectin family member Siglec-G in CD8α(+) DCs, and Siglec-G deficient (Siglecg(-/-)) mice generated more antigen-specific CTLs to inhibit intracellular bacterial infection and tumor growth. MHC class I-peptide complexes were more abundant on Siglecg(-/-) CD8α(+) DCs than on Siglecg(+/+) CD8α(+) DCs. Mechanistically, phagosome-expressed Siglec-G recruited the phosphatase SHP-1, which dephosphorylated the NADPH oxidase component p47(phox) and inhibited the activation of NOX2 on phagosomes. This resulted in excessive hydrolysis of exogenous antigens, which led to diminished formation of MHC class I-peptide complexes for cross-presentation. Therefore, Siglec-G inhibited DC cross-presentation by impairing such complex formation, and our results add insight into the regulation of cross-presentation in adaptive immunity.
Collapse
|
29
|
Osanai T, Mikami K, Kitajima M, Urushizaka M, Kawasaki K, Tomisawa T, Itaki C, Noto Y, Magota K, Tomita H. Nutritional regulation of coupling factor 6, a novel vasoactive and proatherogenic peptide. Nutrition 2016; 37:74-78. [PMID: 28359367 DOI: 10.1016/j.nut.2016.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/24/2022]
Abstract
High sodium, high glucose, and obesity are important risk factors for age-related diseases such as cardiovascular disease (CVDs), stroke, and cancer. Coupling factor 6 (CF6) is released from vascular endothelial cells and functions as a circulating peptide that inhibits prostacyclin and nitric oxide generation by intracellular acidosis. High glucose elevates CF6 by activation of protein kinase C and p38 mitogen-activated protein kinase, whereas CF6 causes type 2 diabetes mellitus, resulting in a high glucose vicious cycle. Low glucose increases inhibitory factor peptide 1, an endogenous inhibitor of CF6. High salt intake increases CF6 through nuclear factor κB signaling, whereas CF6 induces salt-sensitive hypertension and salt-induced congestive heart failure. Oral administration of vitamin C cancels salt-induced increase in CF6, and estrogen replacement leads to the delayed onset of CF6-induced salt-sensitive hypertension and the rescue from cardiac systolic dysfunction. Because CF6 contributes to the onset of CVDs, nutritional regulation of CF6 will shed light on the understanding of preventive strategy and mechanisms for CVDs and a target for therapy.
Collapse
Affiliation(s)
- Tomohiro Osanai
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan.
| | - Kasumi Mikami
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Maiko Kitajima
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Mayumi Urushizaka
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Kumiko Kawasaki
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Toshiko Tomisawa
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Chieko Itaki
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Yuka Noto
- Department of Nursing Science, Hirosaki University Graduate School of Health Science, Hirosaki, Japan
| | - Koji Magota
- Daiichi Sankyo Co., Ltd., Biologics Technology Research Laboratories Group 1, Pharmaceutical Technology Division, Gunma, Japan
| | - Hirofumi Tomita
- Department of Cardiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
30
|
Li LF, Chang YL, Chen NH, Wang CY, Chang GJ, Lin MC, Chang CH, Huang CC, Chuang JH, Yang YP, Chiou SH, Liu YY. Inhibition of Src and forkhead box O1 signaling by induced pluripotent stem-cell therapy attenuates hyperoxia-augmented ventilator-induced diaphragm dysfunction. Transl Res 2016; 173:131-147.e1. [PMID: 27055225 DOI: 10.1016/j.trsl.2016.03.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/13/2016] [Accepted: 03/15/2016] [Indexed: 11/19/2022]
Abstract
Mechanical ventilation (MV) with hyperoxia is required for providing life support to patients with acute lung injury (ALI). However, MV may cause diaphragm weakness through muscle injury and atrophy, an effect termed ventilator-induced diaphragm dysfunction (VIDD). Src protein tyrosine kinase and class O of forkhead box 1 (FoxO1) mediate acute inflammatory responses and muscle protein degradation induced by oxidative stress. Induced pluripotent stem cells (iPSCs) have been reported to improve hyperoxia-augmented ALI; however, the mechanisms regulating the interactions among VIDD, hyperoxia, and iPSCs are unclear. In this study, we hypothesized that iPSC therapy can ameliorate hyperoxia-augmented VIDD by suppressing the Src-FoxO1 pathway. Male C57BL/6 mice, either wild-type or Src-deficient, aged between 6 and 8 weeks were exposed to MV (6 or 10 mL/kg) with or without hyperoxia for 2-8 h after the administration of 5 × 10(7) cells/kg Oct4/Sox2/Parp1 mouse iPSCs or iPSC-derived conditioned medium (iPSC-CM). Nonventilated mice were used as controls. MV during hyperoxia aggravated VIDD, as demonstrated by the increases in Src activation, FoxO1 dephosphorylation, malondialdehyde, caspase-3, atrogin-1 and muscle ring finger-1 production, microtubule-associated protein light chain 3-II, disorganized myofibrils, disrupted mitochondria, autophagy, and myonuclear apoptosis; however, MV with hyperoxia reduced mitochondrial cytochrome C, diaphragm muscle fiber size, and contractility (P < 0.05). Hyperoxia-exacerbated VIDD was attenuated in Src-deficient mice and by iPSCs and iPSC-CM (P < 0.05). Our data indicate that iPSC therapy attenuates MV-induced diaphragmatic injury that occurs during hyperoxia-augmented VIDD by inhibiting the Src-FoxO1 signaling pathway.
Collapse
Affiliation(s)
- Li-Fu Li
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yuh-Lih Chang
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ning-Hung Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chien-Ying Wang
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chih-Hao Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chung-Chi Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jen-Hua Chuang
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Pin Yang
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yung-Yang Liu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Chest Department, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
31
|
Harijith A, Pendyala S, Ebenezer DL, Ha AW, Fu P, Wang YT, Ma K, Toth PT, Berdyshev EV, Kanteti P, Natarajan V. Hyperoxia-induced p47phox activation and ROS generation is mediated through S1P transporter Spns2, and S1P/S1P1&2 signaling axis in lung endothelium. Am J Physiol Lung Cell Mol Physiol 2016; 311:L337-51. [PMID: 27343196 DOI: 10.1152/ajplung.00447.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Hyperoxia-induced lung injury adversely affects ICU patients and neonates on ventilator assisted breathing. The underlying culprit appears to be reactive oxygen species (ROS)-induced lung damage. The major contributor of hyperoxia-induced ROS is activation of the multiprotein enzyme complex NADPH oxidase. Sphingosine-1-phosphate (S1P) signaling is known to be involved in hyperoxia-mediated ROS generation; however, the mechanism(s) of S1P-induced NADPH oxidase activation is unclear. Here, we investigated various steps in the S1P signaling pathway mediating ROS production in response to hyperoxia in lung endothelium. Of the two closely related sphingosine kinases (SphKs)1 and 2, which synthesize S1P from sphingosine, only Sphk1(-/-) mice conferred protection against hyperoxia-induced lung injury. S1P is metabolized predominantly by S1P lyase and partial deletion of Sgpl1 (Sgpl1(+/-)) in mice accentuated lung injury. Hyperoxia stimulated S1P accumulation in human lung microvascular endothelial cells (HLMVECs), and downregulation of S1P transporter spinster homolog 2 (Spns2) or S1P receptors S1P1&2, but not S1P3, using specific siRNA attenuated hyperoxia-induced p47(phox) translocation to cell periphery and ROS generation in HLMVECs. These results suggest a role for Spns2 and S1P1&2 in hyperoxia-mediated ROS generation. In addition, p47(phox) (phox:phagocyte oxidase) activation and ROS generation was also reduced by PF543, a specific SphK1 inhibitor in HLMVECs. Our data indicate a novel role for Spns2 and S1P1&2 in the activation of p47(phox) and production of ROS involved in hyperoxia-mediated lung injury in neonatal and adult mice.
Collapse
Affiliation(s)
- Anantha Harijith
- Department of Pediatrics, National Jewish Health, Denver, Colorado; Department of Pharmacology, National Jewish Health, Denver, Colorado;
| | - Srikanth Pendyala
- Department of Pharmacology, National Jewish Health, Denver, Colorado
| | - David L Ebenezer
- Department of Biochemistry & Molecular Genetics, National Jewish Health, Denver, Colorado
| | - Alison W Ha
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Panfeng Fu
- Department of Pharmacology, National Jewish Health, Denver, Colorado
| | - Yue-Ting Wang
- Department of Medicinal Chemistry, National Jewish Health, Denver, Colorado
| | - Ke Ma
- Department of Pathology, National Jewish Health, Denver, Colorado
| | - Peter T Toth
- Department of Pathology, National Jewish Health, Denver, Colorado
| | | | - Prasad Kanteti
- Department of Pharmacology, National Jewish Health, Denver, Colorado
| | - Viswanathan Natarajan
- Department of Pharmacology, National Jewish Health, Denver, Colorado; Department of Biochemistry & Molecular Genetics, National Jewish Health, Denver, Colorado; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
32
|
Abstract
Since its discovery in 1999, a number of studies have evaluated the role of Nox1 NADPH oxidase in the cardiovascular system. Nox1 is activated in vascular cells in response to several different agonists, with its activity regulated at the transcriptional level as well as by NADPH oxidase complex formation, protein stabilization and post-translational modification. Nox1 has been shown to decrease the bioavailability of nitric oxide, transactivate the epidermal growth factor receptor, induce pro-inflammatory signalling, and promote cell migration and proliferation. Enhanced expression and activity of Nox1 under pathologic conditions results in excessive production of reactive oxygen species and dysregulated cellular function. Indeed, studies using genetic models of Nox1 deficiency or overexpression have revealed roles for Nox1 in the pathogenesis of cardiovascular diseases ranging from atherosclerosis to hypertension, restenosis and ischaemia/reperfusion injury. These data suggest that Nox1 is a potential therapeutic target for vascular disease, and drug development efforts are ongoing to identify a specific bioavailable inhibitor of Nox1.
Collapse
|
33
|
Petersen KE, Rakipovski G, Raun K, Lykkesfeldt J. Does Glucagon-like Peptide-1 Ameliorate Oxidative Stress in Diabetes? Evidence Based on Experimental and Clinical Studies. Curr Diabetes Rev 2016; 12:331-358. [PMID: 26381142 PMCID: PMC5101636 DOI: 10.2174/1573399812666150918150608] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) has shown to influence the oxidative stress status in a number of in vitro, in vivo and clinical studies. Well-known effects of GLP-1 including better glycemic control, decreased food intake, increased insulin release and increased insulin sensitivity may indirectly contribute to this phenomenon, but glucose-independent effects on ROS level, production and antioxidant capacity have been suggested to also play a role. The potential 'antioxidant' activity of GLP-1 along with other proposed glucose-independent modes of action related to ameliorating redox imbalance remains a controversial topic but could hold a therapeutic potential against micro- and macrovascular diabetic complications. This review discusses the presently available knowledge from experimental and clinical studies on the effects of GLP-1 on oxidative stress in diabetes and diabetes-related complications.
Collapse
Affiliation(s)
| | | | | | - Jens Lykkesfeldt
- Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark.
| |
Collapse
|
34
|
Enhancement of Src Family Kinase Activity is Essential for p38 MAP Kinase-Mediated Dedifferentiation Signal of Parotid Acinar Cells . ACTA ACUST UNITED AC 2016. [DOI: 10.5466/ijoms.14.33] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
35
|
Li LF, Lee CS, Liu YY, Chang CH, Lin CW, Chiu LC, Kao KC, Chen NH, Yang CT. Activation of Src-dependent Smad3 signaling mediates the neutrophilic inflammation and oxidative stress in hyperoxia-augmented ventilator-induced lung injury. Respir Res 2015; 16:112. [PMID: 26377087 PMCID: PMC4574227 DOI: 10.1186/s12931-015-0275-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/10/2015] [Indexed: 11/26/2022] Open
Abstract
Background Mechanical ventilation and concomitant administration of hyperoxia in patients with acute respiratory distress syndrome can damage the alveolar epithelial and capillary endothelial barrier by producing inflammatory cytokines and reactive oxygen species. The Src tyrosine kinase and Smad3 are crucial inflammatory regulators used for ventilator-induced lung injury (VILI). The mechanisms regulating interactions between high-tidal-volume mechanical ventilation, hyperoxia, and acute lung injury (ALI) are unclear. We hypothesized that high-tidal-volume mechanical stretches and hyperoxia augment lung inflammation through upregulation of the Src and Smad3 pathways. Methods Wild-type or Src-deficient C57BL/6 mice, aged between 6 and 8 weeks, were exposed to high-tidal-volume (30 mL/kg) ventilation with room air or hyperoxia for 1–4 h after 2-mg/kg Smad3 inhibitor (SIS3) administration. Nonventilated mice were used as control subjects. Results We observed that the addition of hyperoxia to high-tidal-volume mechanical ventilation further induced microvascular permeability, neutrophil infiltration, macrophage inflammatory protein-2 and matrix metalloproteinase-9 (MMP-9) production, malondialdehyde, nicotinamide adenine dinucleotide phosphate oxidase activity, MMP-9 mRNA expression, hypoxemia, and Src and Smad3 activation (P < 0.05). Hyperoxia-induced augmentation of VILI was attenuated in Src-deficient mice and mice with pharmacological inhibition of Smad3 activity by SIS3 (P < 0.05). Mechanical ventilation of Src-deficient mice with hyperoxia further reduced the activation of Smad3. Conclusions Our data suggest that hyperoxia-increased high-tidal-volume ventilation-induced ALI partially depends on the Src and Smad3 pathways.
Collapse
Affiliation(s)
- Li-Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan.,Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chung-Shu Lee
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Hao Chang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan
| | - Chang-Wei Lin
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan
| | - Li-Chung Chiu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan
| | - Kuo-Chin Kao
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan.,Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ning-Hung Chen
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan.,Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng-Ta Yang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, 5 Fu-Hsing Street, Kweishan, Taoyuan, 333, Taiwan. .,Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
36
|
Bielefeld EC. Protection from noise-induced hearing loss with Src inhibitors. Drug Discov Today 2015; 20:760-5. [PMID: 25637168 DOI: 10.1016/j.drudis.2015.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/09/2014] [Accepted: 01/20/2015] [Indexed: 01/23/2023]
Abstract
Noise-induced hearing loss is a major cause of acquired hearing loss around the world and pharmacological approaches to protecting the ear from noise are under investigation. Noise results in a combination of mechanical and metabolic damage pathways in the cochlea. The Src family of protein tyrosine kinases could be active in both pathways and Src inhibitors have successfully prevented noise-induced cochlear damage and hearing loss in animal models. The long-term goal is to optimize delivery methods into the cochlea to reduce invasiveness and limit side-effects before human clinical testing can be considered. At their current early stage of research investigation, Src inhibitors represent an exciting class of compounds for inclusion in a multifaceted pharmacological approach to protecting the ear from noise.
Collapse
Affiliation(s)
- Eric C Bielefeld
- Department of Speech and Hearing Science, The Ohio State University, 110 Pressey Hall, 1070 Carmack Road, Columbus, OH 43210, USA.
| |
Collapse
|
37
|
MacKay CE, Knock GA. Control of vascular smooth muscle function by Src-family kinases and reactive oxygen species in health and disease. J Physiol 2014; 593:3815-28. [PMID: 25384773 DOI: 10.1113/jphysiol.2014.285304] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/22/2014] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are now recognised as second messenger molecules that regulate cellular function by reversibly oxidising specific amino acid residues of key target proteins. Amongst these are the Src-family kinases (SrcFKs), a multi-functional group of non-receptor tyrosine kinases highly expressed in vascular smooth muscle (VSM). In this review we examine the evidence supporting a role for ROS-induced SrcFK activity in normal VSM contractile function and in vascular remodelling in cardiovascular disease. VSM contractile responses to G-protein-coupled receptor stimulation, as well as hypoxia in pulmonary artery, are shown to be dependent on both ROS and SrcFK activity. Specific phosphorylation targets are identified amongst those that alter intracellular Ca(2+) concentration, including transient receptor potential channels, voltage-gated Ca(2+) channels and various types of K(+) channels, as well as amongst those that regulate actin cytoskeleton dynamics and myosin phosphatase activity, including focal adhesion kinase, protein tyrosine kinase-2, Janus kinase, other focal adhesion-associated proteins, and Rho guanine nucleotide exchange factors. We also examine a growing weight of evidence in favour of a key role for SrcFKs in multiple pro-proliferative and anti-apoptotic signalling pathways relating to oxidative stress and vascular remodelling, with a particular focus on pulmonary hypertension, including growth-factor receptor transactivation and downstream signalling, hypoxia-inducible factors, positive feedback between SrcFK and STAT3 signalling and positive feedback between SrcFK and NADPH oxidase dependent ROS production. We also discuss evidence for and against the potential therapeutic targeting of SrcFKs in the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Charles E MacKay
- Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Greg A Knock
- Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
38
|
Kim MJ, Ryu JC, Kwon Y, Lee S, Bae YS, Yoon JH, Ryu JH. Dual oxidase 2 in lung epithelia is essential for hyperoxia-induced acute lung injury in mice. Antioxid Redox Signal 2014; 21:1803-18. [PMID: 24766345 PMCID: PMC4203470 DOI: 10.1089/ars.2013.5677] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIMS Acute lung injury (ALI) induced by excessive hyperoxia has been employed as a model of oxidative stress imitating acute respiratory distress syndrome. Under hyperoxic conditions, overloading quantities of reactive oxygen species (ROS) are generated in both lung epithelial and endothelial cells, leading to ALI. Some NADPH oxidase (NOX) family enzymes are responsible for hyperoxia-induced ROS generation in lung epithelial and endothelial cells. However, the molecular mechanisms of ROS production in type II alveolar epithelial cells (AECs) and ALI induced by hyperoxia are poorly understood. RESULTS In this study, we show that dual oxidase 2 (DUOX2) is a key NOX enzyme that affects hyperoxia-induced ROS production, particularly in type II AECs, leading to lung injury. In DUOX2 mutant mice (DUOX2(thyd/thyd)) or mice in which DUOX2 expression is knocked down in the lungs, hyperoxia-induced ALI was significantly lower than in wild-type (WT) mice. DUOX2 was mainly expressed in type II AECs, but not endothelial cells, and hyperoxia-induced ROS production was markedly reduced in primary type II AECs isolated from DUOX2(thyd/thyd) mice. Furthermore, DUOX2-generated ROS are responsible for caspase-mediated cell death, inducing ERK and JNK phophorylation in type II AECs. INNOVATION To date, no role for DUOX2 has been defined in hyperoxia-mediated ALI despite it being a NOX homologue and major ROS source in lung epithelium. CONCLUSION Here, we present the novel finding that DUOX2-generated ROS induce AEC death, leading to hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Min-Ji Kim
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Chan Ryu
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Younghee Kwon
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Suhee Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Yun Soo Bae
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Joo-Heon Yoon
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
- The Airway Mucus Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Hwan Ryu
- Research Center for Natural Human Defense System, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
39
|
Liu YY, Li LF, Fu JY, Kao KC, Huang CC, Chien Y, Liao YW, Chiou SH, Chang YL. Induced pluripotent stem cell therapy ameliorates hyperoxia-augmented ventilator-induced lung injury through suppressing the Src pathway. PLoS One 2014; 9:e109953. [PMID: 25310015 PMCID: PMC4195701 DOI: 10.1371/journal.pone.0109953] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/12/2014] [Indexed: 01/20/2023] Open
Abstract
Background High tidal volume (VT) mechanical ventilation (MV) can induce the recruitment of neutrophils, release of inflammatory cytokines and free radicals, and disruption of alveolar epithelial and endothelial barriers. It is proposed to be the triggering factor that initiates ventilator-induced lung injury (VILI) and concomitant hyperoxia further aggravates the progression of VILI. The Src protein tyrosine kinase (PTK) family is one of the most critical families to intracellular signal transduction related to acute inflammatory responses. The anti-inflammatory abilities of induced pluripotent stem cells (iPSCs) have been shown to improve acute lung injuries (ALIs); however, the mechanisms regulating the interactions between MV, hyperoxia, and iPSCs have not been fully elucidated. In this study, we hypothesize that Src PTK plays a critical role in the regulation of oxidants and inflammation-induced VILI during hyperoxia. iPSC therapy can ameliorate acute hyperoxic VILI by suppressing the Src pathway. Methods Male C57BL/6 mice, either wild-type or Src-deficient, aged between 2 and 3 months were exposed to high VT (30 mL/kg) ventilation with or without hyperoxia for 1 to 4 h after the administration of Oct4/Sox2/Parp1 iPSCs at a dose of 5×107 cells/kg of mouse. Nonventilated mice were used for the control groups. Results High VT ventilation during hyperoxia further aggravated VILI, as demonstrated by the increases in microvascular permeability, neutrophil infiltration, macrophage inflammatory protein-2 (MIP-2) and plasminogen activator inhibitor-1 (PAI-1) production, Src activation, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and malaldehyde (MDA) level. Administering iPSCs attenuated ALI induced by MV during hyperoxia, which benefited from the suppression of Src activation, oxidative stress, acute inflammation, and apoptosis, as indicated by the Src-deficient mice. Conclusion The data suggest that iPSC-based therapy is capable of partially suppressing acute inflammatory and oxidant responses that occur during hyperoxia-augmented VILI through the inhibition of Src-dependent signaling pathway.
Collapse
Affiliation(s)
- Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- * E-mail: (LFL); (YLC)
| | - Jui-Ying Fu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Chin Kao
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chung-Chi Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yueh Chien
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Wen Liao
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Lih Chang
- Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- * E-mail: (LFL); (YLC)
| |
Collapse
|
40
|
ROS and RNS signaling in skeletal muscle: critical signals and therapeutic targets. ANNUAL REVIEW OF NURSING RESEARCH 2014; 31:367-87. [PMID: 24894146 DOI: 10.1891/0739-6686.31.367] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The health of skeletal muscle is promoted by optimal nutrition and activity/exercise through the activation of molecular signaling pathways. Reactive oxygen species (ROS) or reactive nitrogen species (RNS) have been shown to modulate numerous biochemical processes including glucose uptake, gene expression, calcium signaling, and contractility. In pathological conditions, ROS/RNS signaling excess or dysfunction contributes to contractile dysfunction and myopathy in skeletal muscle. Here we provide a brief review of ROS/RNS chemistry and discuss concepts of ROS/RNS signaling and its role in physiological and pathophysiological processes within striated muscle.
Collapse
|
41
|
Bayley R, Kite KA, McGettrick HM, Smith JP, Kitas GD, Buckley CD, Young SP. The autoimmune-associated genetic variant PTPN22 R620W enhances neutrophil activation and function in patients with rheumatoid arthritis and healthy individuals. Ann Rheum Dis 2014; 74:1588-95. [PMID: 24665115 DOI: 10.1136/annrheumdis-2013-204796] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/01/2014] [Indexed: 12/30/2022]
Abstract
OBJECTIVES A genetic variant of the leukocyte phosphatase PTPN22 (R620W) is strongly associated with autoimmune diseases including rheumatoid arthritis (RA). Functional studies on the variant have focussed on lymphocytes, but it is most highly expressed in neutrophils. We have investigated the effects of the variant on neutrophil function in health and in patients with RA. METHODS Healthy individuals and patients with RA were genotyped for PTPN22 (R620W) and neutrophils isolated from peripheral blood. Neutrophil adhesion and migration across inflamed endothelium were measured. Calcium (Ca(2+)) release and reactive oxygen species (ROS) production in response to fMLP stimulation were also assessed. RESULTS Expression of R620W enhanced neutrophil migration through cytokine activated endothelium (non-R620W=24%, R620W=45% migrating cells, p<0.001). Following fMLP stimulation, neutrophils that were heterozygous and homozygous for R620W released significantly more Ca(2+) when compared to non-R620W neutrophils, in healthy individuals and patients with RA. fMLP stimulation, after TNF-α priming, provoked more ROS from neutrophils heterozygous for R620W in patients with RA (non-R620W vs R620W=∼1.75-fold increase) and healthy individuals (non-R620W vs R620W=fourfold increase) and this increase was statistically significant in healthy individuals (p<0.001) but not in patients with RA (p<0.25). CONCLUSIONS Expression of PTPN22 (R620W) enhanced neutrophil effector functions in health and RA, with migration, Ca(2+) release and production of ROS increased. Neutrophils are found in large numbers in the RA joint, and this hyperactivity of R620W cells may directly contribute to the joint damage, as well as to the initiation and perpetuation of the chronic immune-mediated inflammatory processes driving the disease.
Collapse
Affiliation(s)
- Rachel Bayley
- Rheumatology Research Group, Centre for Translational Inflammation Research, College of Medical and Dental Sciences, School of Immunity and Infection, University of Birmingham, Birmingham, UK
| | - Kerry A Kite
- Rheumatology Research Group, Centre for Translational Inflammation Research, College of Medical and Dental Sciences, School of Immunity and Infection, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Rheumatology Research Group, Centre for Translational Inflammation Research, College of Medical and Dental Sciences, School of Immunity and Infection, University of Birmingham, Birmingham, UK
| | - Jacqueline P Smith
- Department of Rheumatology, Dudley Group of Hospitals NHS Trust, Russell's Hall Hospital, Dudley, West Midlands, UK
| | - George D Kitas
- Department of Rheumatology, Dudley Group of Hospitals NHS Trust, Russell's Hall Hospital, Dudley, West Midlands, UK
| | - Christopher D Buckley
- Rheumatology Research Group, Centre for Translational Inflammation Research, College of Medical and Dental Sciences, School of Immunity and Infection, University of Birmingham, Birmingham, UK
| | - Stephen P Young
- Rheumatology Research Group, Centre for Translational Inflammation Research, College of Medical and Dental Sciences, School of Immunity and Infection, University of Birmingham, Birmingham, UK
| |
Collapse
|
42
|
Usatyuk PV, Fu P, Mohan V, Epshtein Y, Jacobson JR, Gomez-Cambronero J, Wary KK, Bindokas V, Dudek SM, Salgia R, Garcia JGN, Natarajan V. Role of c-Met/phosphatidylinositol 3-kinase (PI3k)/Akt signaling in hepatocyte growth factor (HGF)-mediated lamellipodia formation, reactive oxygen species (ROS) generation, and motility of lung endothelial cells. J Biol Chem 2014; 289:13476-91. [PMID: 24634221 DOI: 10.1074/jbc.m113.527556] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hepatocyte growth factor (HGF) mediated signaling promotes cell proliferation and migration in a variety of cell types and plays a key role in tumorigenesis. As cell migration is important to angiogenesis, we characterized HGF-mediated effects on the formation of lamellipodia, a pre-requisite for migration using human lung microvascular endothelial cells (HLMVECs). HGF, in a dose-dependent manner, induced c-Met phosphorylation (Tyr-1234/1235, Tyr-1349, Ser-985, Tyr-1003, and Tyr-1313), activation of PI3k (phospho-Yp85) and Akt (phospho-Thr-308 and phospho-Ser-473) and potentiated lamellipodia formation and HLMVEC migration. Inhibition of c-Met kinase by SU11274 significantly attenuated c-Met, PI3k, and Akt phosphorylation, suppressed lamellipodia formation and endothelial cell migration. LY294002, an inhibitor of PI3k, abolished HGF-induced PI3k (Tyr-458), and Akt (Thr-308 and Ser-473) phosphorylation and suppressed lamellipodia formation. Furthermore, HGF stimulated p47(phox)/Cortactin/Rac1 translocation to lamellipodia and ROS generation. Moreover, inhibition of c-Met/PI3k/Akt signaling axis and NADPH oxidase attenuated HGF- induced lamellipodia formation, ROS generation and cell migration. Ex vivo experiments with mouse aortic rings revealed a role for c-Met signaling in HGF-induced sprouting and lamellipodia formation. Taken together, these data provide evidence in support of a significant role for HGF-induced c-Met/PI3k/Akt signaling and NADPH oxidase activation in lamellipodia formation and motility of lung endothelial cells.
Collapse
|
43
|
Genetic Interactions of STAT3 and Anticancer Drug Development. Cancers (Basel) 2014; 6:494-525. [PMID: 24662938 PMCID: PMC3980611 DOI: 10.3390/cancers6010494] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays critical roles in tumorigenesis and malignant evolution and has been intensively studied as a therapeutic target for cancer. A number of STAT3 inhibitors have been evaluated for their antitumor activity in vitro and in vivo in experimental tumor models and several approved therapeutic agents have been reported to function as STAT3 inhibitors. Nevertheless, most STAT3 inhibitors have yet to be translated to clinical evaluation for cancer treatment, presumably because of pharmacokinetic, efficacy, and safety issues. In fact, a major cause of failure of anticancer drug development is lack of efficacy. Genetic interactions among various cancer-related pathways often provide redundant input from parallel and/or cooperative pathways that drives and maintains survival environments for cancer cells, leading to low efficacy of single-target agents. Exploiting genetic interactions of STAT3 with other cancer-related pathways may provide molecular insight into mechanisms of cancer resistance to pathway-targeted therapies and strategies for development of more effective anticancer agents and treatment regimens. This review focuses on functional regulation of STAT3 activity; possible interactions of the STAT3, RAS, epidermal growth factor receptor, and reduction-oxidation pathways; and molecular mechanisms that modulate therapeutic efficacies of STAT3 inhibitors.
Collapse
|
44
|
Sato Y, Fujimoto S, Mukai E, Sato H, Tahara Y, Ogura K, Yamano G, Ogura M, Nagashima K, Inagaki N. Palmitate induces reactive oxygen species production and β-cell dysfunction by activating nicotinamide adenine dinucleotide phosphate oxidase through Src signaling. J Diabetes Investig 2014; 5:19-26. [PMID: 24843732 PMCID: PMC4025235 DOI: 10.1111/jdi.12124] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/20/2013] [Accepted: 05/28/2013] [Indexed: 12/25/2022] Open
Abstract
AIMS/INTRODUCTION Chronic hyperlipidemia impairs pancreatic β-cell function, referred to as lipotoxicity. We have reported an important role of endogenous reactive oxygen species (ROS) overproduction by activation of Src, a non-receptor tyrosine kinase, in impaired glucose-induced insulin secretion (GIIS) from diabetic rat islets. In the present study, we investigated the role of ROS production by Src signaling in palmitate-induced dysfunction of β-cells. MATERIALS AND METHODS After rat insulinoma INS-1D cells were exposed to 0.6 mmol/L palmitate for 24 h (palmitate exposure); GIIS, ROS production and nicotinamide adenine dinucleotide phosphate oxidase (NOX) activity were examined with or without exposure to10 μmol/L 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2), a Src inhibitior, for 30 or 60 min. RESULTS Exposure to PP2 recovered impaired GIIS and decreased ROS overproduction as a result of palmitate exposure. Palmitate exposure increased activity of NOX and protein levels of NOX2, a pathological ROS source in β-cells. Palmitate exposure increased the protein level of p47 (phox) , a regulatory protein of NOX2, in membrane fraction compared with control, which was reduced by PP2. Transfection of small interfering ribonucleic acid of p47 (phox) suppressed the augmented p47 (phox) protein level in membrane fraction, decreased augmented ROS production and increased impaired GΙIS by palmitate exposure. In addition, exposure to PP2 ameliorated impaired GIIS and decreased ROS production in isolated islets of KK-A(y) mice, an obese diabetic model with hyperlipidemia. CONCLUSIONS Activation of NOX through Src signaling plays an important role in ROS overproduction and impaired GΙIS caused by chronic exposure to palmitate, suggesting a lipotoxic mechanism of β-cell dysfunction of obese mice.
Collapse
Affiliation(s)
- Yuichi Sato
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Shimpei Fujimoto
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
- Department of Endocrinology, Metabolism and NephrologyKochi Medical SchoolKochi UniversityNankokuJapan
| | - Eri Mukai
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroki Sato
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Yumiko Tahara
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Kasane Ogura
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Gen Yamano
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Masahito Ogura
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazuaki Nagashima
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Nobuya Inagaki
- Department of Diabetes and Clinical NutritionGraduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
45
|
Tai W, Ye X, Bao X, Zhao B, Wang X, Zhang D. Inhibition of Src tyrosine kinase activity by squamosamide derivative FLZ attenuates neuroinflammation in both in vivo and in vitro Parkinson's disease models. Neuropharmacology 2013; 75:201-12. [DOI: 10.1016/j.neuropharm.2013.07.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/26/2013] [Accepted: 07/16/2013] [Indexed: 11/26/2022]
|
46
|
Wagner B, Gorin Y. Src tyrosine kinase mediates platelet-derived growth factor BB-induced and redox-dependent migration in metanephric mesenchymal cells. Am J Physiol Renal Physiol 2013; 306:F85-97. [PMID: 24197068 DOI: 10.1152/ajprenal.00371.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The adult kidney is derived from the interaction between the metanephric blastema and the ureteric bud. Platelet-derived growth factor (PDGF) receptor β is essential for the development of the mature glomerular tuft, as mice deficient for this receptor lack mesangial cells. This study investigated the role of Src tyrosine kinase in PDGF-mediated reactive oxygen species (ROS) generation and migration of metanephric mesenchymal cells (MMCs). Cultured embryonic MMCs from wild-type and PDGF receptor-deficient embryos were established. Migration was determined via wound-healing assay. Unlike PDGF AA, PDGF BB-induced greater migration in MMCs with respect to control. This was abrogated by neutralizing an antibody to PDGF BB. Phosphatidylinositol 3-kinase (PI3K) inhibitors suppressed PDGF BB-induced migration. Conversely, mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors had no effect. Src inhibitors inhibited PDGF-induced cell migration, PI3K activity, and Akt phosphorylation. Adenoviral dominant negative Src (AD DN Src) abrogated PDGF BB-induced Akt phosphorylation. Hydrogen peroxide stimulated cell migration. PDGF BB-induced wound closure was inhibited by the antioxidants N-acetyl-l-cysteine, tiron, and the flavoprotein inhibitor diphenyleneiodonium. These cells express the NADPH oxidase homolog Nox4. Inhibiting Nox4 with antisense oligonucleotides or small interfering RNA (siRNA) suppressed PDGF-induced wound closure. Inhibition of Src with siRNA reduced PDGF BB-induced ROS generation as assessed by 2',7'-dichlorodihydrofluorescein diacetate fluorescence. Furthermore, PDGF BB-stimulated ROS generation and migration were similarly suppressed by Ad DN Src. In MMCs, PDGF BB-induced migration is mediated by PI3K and Src in a redox-dependent manner involving Nox4. Src may be upstream to PI3K and Nox4.
Collapse
Affiliation(s)
- Brent Wagner
- South Texas Veterans Health Care System, Div. of Nephrology MC 7882, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900.
| | | |
Collapse
|
47
|
Harijith A, Pendyala S, Reddy NM, Bai T, Usatyuk PV, Berdyshev E, Gorshkova I, Huang LS, Mohan V, Garzon S, Kanteti P, Reddy SP, Raj JU, Natarajan V. Sphingosine kinase 1 deficiency confers protection against hyperoxia-induced bronchopulmonary dysplasia in a murine model: role of S1P signaling and Nox proteins. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1169-1182. [PMID: 23933064 DOI: 10.1016/j.ajpath.2013.06.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/05/2013] [Accepted: 06/24/2013] [Indexed: 12/23/2022]
Abstract
Bronchopulmonary dysplasia of the premature newborn is characterized by lung injury, resulting in alveolar simplification and reduced pulmonary function. Exposure of neonatal mice to hyperoxia enhanced sphingosine-1-phosphate (S1P) levels in lung tissues; however, the role of increased S1P in the pathobiological characteristics of bronchopulmonary dysplasia has not been investigated. We hypothesized that an altered S1P signaling axis, in part, is responsible for neonatal lung injury leading to bronchopulmonary dysplasia. To validate this hypothesis, newborn wild-type, sphingosine kinase1(-/-) (Sphk1(-/-)), sphingosine kinase 2(-/-) (Sphk2(-/-)), and S1P lyase(+/-) (Sgpl1(+/-)) mice were exposed to hyperoxia (75%) from postnatal day 1 to 7. Sphk1(-/-), but not Sphk2(-/-) or Sgpl1(+/-), mice offered protection against hyperoxia-induced lung injury, with improved alveolarization and alveolar integrity compared with wild type. Furthermore, SphK1 deficiency attenuated hyperoxia-induced accumulation of IL-6 in bronchoalveolar lavage fluids and NADPH oxidase (NOX) 2 and NOX4 protein expression in lung tissue. In vitro experiments using human lung microvascular endothelial cells showed that exogenous S1P stimulated intracellular reactive oxygen species (ROS) generation, whereas SphK1 siRNA, or inhibitor against SphK1, attenuated hyperoxia-induced S1P generation. Knockdown of NOX2 and NOX4, using specific siRNA, reduced both basal and S1P-induced ROS formation. These results suggest an important role for SphK1-mediated S1P signaling-regulated ROS in the development of hyperoxia-induced lung injury in a murine neonatal model of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Anantha Harijith
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; Department of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| | - Srikanth Pendyala
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Narsa M Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Tao Bai
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Peter V Usatyuk
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Evgeny Berdyshev
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Irina Gorshkova
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Long Shuang Huang
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Vijay Mohan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Steve Garzon
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois
| | - Prasad Kanteti
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Sekhar P Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - J Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
48
|
Lizotte F, Paré M, Denhez B, Leitges M, Guay A, Geraldes P. PKCδ impaired vessel formation and angiogenic factor expression in diabetic ischemic limbs. Diabetes 2013; 62:2948-57. [PMID: 23557702 PMCID: PMC3717846 DOI: 10.2337/db12-1432] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 03/29/2013] [Indexed: 12/17/2022]
Abstract
Decreased collateral vessel formation in diabetic peripheral limbs is characterized by abnormalities of the angiogenic response to ischemia. Hyperglycemia is known to activate protein kinase C (PKC), affecting the expression and activity of growth factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF). The current study investigates the role of PKCδ in diabetes-induced poor collateral vessel formation and inhibition of angiogenic factors expression and actions. Ischemic adductor muscles of diabetic Prkcd(+/+) mice exhibited reduced blood reperfusion, vascular density, and number of small vessels compared with nondiabetic Prkcd(+/+) mice. By contrast, diabetic Prkcd(-/-) mice showed significant increased blood flow, capillary density, and number of capillaries. Although expression of various PKC isoforms was unchanged, activation of PKCδ was increased in diabetic Prkcd(+/+) mice. VEGF and PDGF mRNA and protein expression were decreased in the muscles of diabetic Prkcd(+/+) mice and were normalized in diabetic Prkcd(-/-) mice. Furthermore, phosphorylation of VEGF receptor 2 (VEGFR2) and PDGF receptor-β (PDGFR-β) were blunted in diabetic Prkcd(+/+) mice but elevated in diabetic Prkcd(-/-) mice. The inhibition of VEGFR2 and PDGFR-β activity was associated with increased SHP-1 expression. In conclusion, our data have uncovered the mechanisms by which PKCδ activation induced poor collateral vessel formation, offering potential novel targets to regulate angiogenesis therapeutically in diabetic patients.
Collapse
Affiliation(s)
- Farah Lizotte
- Clinical Research Center Étienne Le-Bel and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Paré
- Clinical Research Center Étienne Le-Bel and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Benoit Denhez
- Clinical Research Center Étienne Le-Bel and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Michael Leitges
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| | - Andréanne Guay
- Clinical Research Center Étienne Le-Bel and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Pedro Geraldes
- Clinical Research Center Étienne Le-Bel and Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
49
|
Su CM, Wang SW, Lee TH, Tzeng WP, Hsiao CJ, Liu SC, Tang CH. Trichodermin induces cell apoptosis through mitochondrial dysfunction and endoplasmic reticulum stress in human chondrosarcoma cells. Toxicol Appl Pharmacol 2013; 272:335-44. [PMID: 23806212 DOI: 10.1016/j.taap.2013.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 12/23/2022]
Abstract
Chondrosarcoma is the second most common primary bone tumor, and it responds poorly to both chemotherapy and radiation treatment. Nalanthamala psidii was described originally as Myxosporium in 1926. This is the first study to investigate the anti-tumor activity of trichodermin (trichothec-9-en-4-ol, 12,13-epoxy-, acetate), an endophytic fungal metabolite from N. psidii against human chondrosarcoma cells. We demonstrated that trichodermin induced cell apoptosis in human chondrosarcoma cell lines (JJ012 and SW1353 cells) instead of primary chondrocytes. In addition, trichodermin triggered endoplasmic reticulum (ER) stress protein levels of IRE1, p-PERK, GRP78, and GRP94, which were characterized by changes in cytosolic calcium levels. Furthermore, trichodermin induced the upregulation of Bax and Bid, the downregulation of Bcl-2, and the dysfunction of mitochondria, which released cytochrome c and activated caspase-3 in human chondrosarcoma. In addition, animal experiments illustrated reduced tumor volume, which led to an increased number of terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-positive cells and an increased level of cleaved PARP protein following trichodermin treatment. Together, this study demonstrates that trichodermin is a novel anti-tumor agent against human chondrosarcoma cells both in vitro and in vivo via mitochondrial dysfunction and ER stress.
Collapse
Affiliation(s)
- Chen-Ming Su
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
50
|
Reactive oxygen species, Nox and angiotensin II in angiogenesis: implications for retinopathy. Clin Sci (Lond) 2013; 124:597-615. [PMID: 23379642 DOI: 10.1042/cs20120212] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pathological angiogenesis is a key feature of many diseases including retinopathies such as ROP (retinopathy of prematurity) and DR (diabetic retinopathy). There is considerable evidence that increased production of ROS (reactive oxygen species) in the retina participates in retinal angiogenesis, although the mechanisms by which this occurs are not fully understood. ROS is produced by a number of pathways, including the mitochondrial electron transport chain, cytochrome P450, xanthine oxidase and uncoupled nitric oxide synthase. The family of NADPH oxidase (Nox) enzymes are likely to be important given that their primary function is to produce ROS. Seven isoforms of Nox have been identified named Nox1-5, Duox (dual oxidase) 1 and Duox2. Nox1, Nox2 and Nox4 have been most extensively studied and are implicated in the development of conditions such as hypertension, cardiovascular disease and diabetic nephropathy. In recent years, evidence has accumulated to suggest that Nox1, Nox2 and Nox4 participate in pathological angiogenesis; however, there is no clear consensus about which Nox isoform is primarily responsible. In terms of retinopathy, there is growing evidence that Nox contribute to vascular injury. The RAAS (renin-angiotensin-aldosterone system), and particularly AngII (angiotensin II), is a key stimulator of Nox. It is known that a local RAAS exists in the retina and that blockade of AngII and aldosterone attenuate pathological angiogenesis in the retina. Whether the RAAS influences the production of ROS derived from Nox in retinopathy is yet to be fully determined. These topics will be reviewed with a particular emphasis on ROP and DR.
Collapse
|