1
|
Liu YF, Zhang YX, Zhu YW, Tang AQ, Liang HB, Yang YL, Zhai YK, Ji XY, Wu DD. Hydrogen Sulfide in Musculoskeletal Diseases: Molecular Mechanisms and Therapeutic Opportunities. Antioxid Redox Signal 2024. [PMID: 39276087 DOI: 10.1089/ars.2024.0625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
Significance: Musculoskeletal diseases seriously affect global health, but their importance is greatly underestimated. These diseases often afflict the elderly, leading to disability, paralysis, and other complications. Hydrogen sulfide (H2S) plays an important role in the occurrence and development of musculoskeletal diseases, which may have potential therapeutic significance for these diseases. Recent Advances: Recently, it has been found that many musculoskeletal diseases, such as osteoporosis, periodontitis, muscle atrophy, muscle ischemia-reperfusion injury, muscle contraction under high fever, arthritis, and disc herniation, can be alleviated by treatment with H2S. H2S may be conducive to the development of multiple myeloma. The mechanism of action of H2S in the musculoskeletal system has been partly elucidated. A variety of H2S donors and nano-delivery systems provide promising prospects for H2S-based therapies. Critical Issues: Related research remains at the level of cell or animal experiments, but clinical research is lacking. The roles of H2S in more musculoskeletal disorders remain largely unknown. The serious consequences of musculoskeletal diseases have not been widely concerned. Targeted delivery of H2S remains a challenging task in musculoskeletal diseases. Future Directions: Develop therapeutic drugs for musculoskeletal diseases based on H2S and test their safety, efficacy, and tolerance. Explore the combination of current drugs for musculoskeletal diseases with H2S-releasing components to improve the therapeutic efficacy and avoid side effects. Carry out relevant clinical trials to verify the possibility of its widespread use. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Ao-Qi Tang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Hao-Bo Liang
- College of Orthopedics, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yi-Lun Yang
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yuan-Kun Zhai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, China
| |
Collapse
|
2
|
Li M, Zhao Z, Yi J. Biomaterials Designed to Modulate Reactive Oxygen Species for Enhanced Bone Regeneration in Diabetic Conditions. J Funct Biomater 2024; 15:220. [PMID: 39194658 DOI: 10.3390/jfb15080220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Diabetes mellitus, characterized by enduring hyperglycemia, precipitates oxidative stress, engendering a spectrum of complications, notably increased bone vulnerability. The genesis of reactive oxygen species (ROS), a byproduct of oxygen metabolism, instigates oxidative detriment and impairs bone metabolism in diabetic conditions. This review delves into the mechanisms of ROS generation and its impact on bone homeostasis within the context of diabetes. Furthermore, the review summarizes the cutting-edge progress in the development of ROS-neutralizing biomaterials tailored for the amelioration of diabetic osteopathy. These biomaterials are engineered to modulate ROS dynamics, thereby mitigating inflammatory responses and facilitating bone repair. Additionally, the challenges and therapeutic prospects of ROS-targeted biomaterials in clinical application of diabetic bone disease treatment is addressed.
Collapse
Affiliation(s)
- Mingshan Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Kale VP, Hengst JA, Sharma AK, Golla U, Dovat S, Amin SG, Yun JK, Desai DH. Characterization of Anticancer Effects of the Analogs of DJ4, a Novel Selective Inhibitor of ROCK and MRCK Kinases. Pharmaceuticals (Basel) 2023; 16:1060. [PMID: 37630974 PMCID: PMC10458458 DOI: 10.3390/ph16081060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/17/2023] [Accepted: 07/22/2023] [Indexed: 08/27/2023] Open
Abstract
The Rho associated coiled-coil containing protein kinase (ROCK1 and ROCK2) and myotonic dystrophy-related Cdc-42 binding kinases (MRCKα and MRCKβ) are critical regulators of cell proliferation and cell plasticity, a process intimately involved in cancer cell migration and invasion. Previously, we reported the discovery of a novel small molecule (DJ4) selective multi-kinase inhibitor of ROCK1/2 and MRCKα/β. Herein, we further characterized the anti-proliferative and apoptotic effects of DJ4 in non-small cell lung cancer and triple-negative breast cancer cells. To further optimize the ROCK/MRCK inhibitory potency of DJ4, we generated a library of 27 analogs. Among the various structural modifications, we identified four additional active analogs with enhanced ROCK/MRCK inhibitory potency. The anti-proliferative and cell cycle inhibitory effects of the active analogs were examined in non-small cell lung cancer, breast cancer, and melanoma cell lines. The anti-proliferative effectiveness of DJ4 and the active analogs was further demonstrated against a wide array of cancer cell types using the NCI-60 human cancer cell line panel. Lastly, these new analogs were tested for anti-migratory effects in highly invasive MDA-MB-231 breast cancer cells. Together, our results demonstrate that selective inhibitors of ROCK1/2 (DJE4, DJ-Allyl) inhibited cell proliferation and induced cell cycle arrest at G2/M but were less effective in cell death induction compared with dual ROCK1/2 and MRCKα/β (DJ4 and DJ110).
Collapse
Affiliation(s)
- Vijay Pralhad Kale
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Jeremy A. Hengst
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Arati K. Sharma
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Upendarrao Golla
- Department of Medicine, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Sinisa Dovat
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA;
| | - Shantu G. Amin
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Jong K. Yun
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| | - Dhimant H. Desai
- Department of Pharmacology Penn State College of Medicine, Hershey, PA 17033, USA (J.A.H.); (S.G.A.)
| |
Collapse
|
4
|
Zhang H, Chen Y, Hua W, Gu W, Zhuang H, Li H, Jiang X, Mao Y, Liu Y, Jin D, Bu W. Heterostructures with Built-in Electric Fields for Long-lasting Chemodynamic Therapy. Angew Chem Int Ed Engl 2023; 62:e202300356. [PMID: 36780170 DOI: 10.1002/anie.202300356] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/14/2023]
Abstract
Sustained signal activation by hydroxyl radicals (⋅OH) has great significance, especially for tumor treatment, but remains challenging. Here, a built-in electric field (BIEF)-driven strategy was proposed for sustainable generation of ⋅OH, thereby achieving long-lasting chemodynamic therapy (LCDT). As a proof of concept, a novel Janus-like Fe@Fe3 O4 -Cu2 O heterogeneous catalyst was designed and synthesized, in which the BIEF induced the transfer of electrons in the Fe core to the surface, reducing ≡Cu2+ to ≡Cu+ , thus achieving continuous Fenton-like reactions and ⋅OH release for over 18 h, which is approximately 12 times longer than that of Fe3 O4 -Cu2 O and 72 times longer than that of Cu2 O nanoparticles. In vitro and in vivo antitumor results indicated that sustained ⋅OH levels led to persistent extracellular regulated protein kinases (ERK) signal activation and irreparable oxidative damage to tumor cells, which promoted irreversible tumor apoptosis. Importantly, this strategy provides ideas for developing long-acting nanoplatforms for various applications.
Collapse
Affiliation(s)
- Huilin Zhang
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China.,Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Yang Chen
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Wei Hua
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China
| | - Wenjun Gu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Hongjun Zhuang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China.,Departments of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Huiyan Li
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Xingwu Jiang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Ying Mao
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, New South Wales, 2007, Australia
| | - Wenbo Bu
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China.,Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| |
Collapse
|
5
|
Tocotrienol as a Protecting Agent against Glucocorticoid-Induced Osteoporosis: A Mini Review of Potential Mechanisms. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185862. [PMID: 36144598 PMCID: PMC9506150 DOI: 10.3390/molecules27185862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022]
Abstract
Glucocorticoid-induced osteogenic dysfunction is the main pathologyical mechanism underlying the development of glucocorticoid-induced osteoporosis. Glucocorticoids promote adipogenic differentiation and osteoblast apoptosis through various pathways. Various ongoing studies are exploring the potential of natural products in preventing glucocorticoid-induced osteoporosis. Preclinical studies have consistently shown the bone protective effects of tocotrienol through its antioxidant and anabolic effects. This review aims to summarise the potential mechanisms of tocotrienol in preventing glucocorticoid-induced osteoporosis based on existing in vivo and in vitro evidence. The current literature showed that tocotrienol prevents oxidative damage on osteoblasts exposed to high levels of glucocorticoids. Tocotrienol reduces lipid peroxidation and increases oxidative stress enzyme activities. The reduction in oxidative stress protects the osteoblasts and preserves the bone microstructure and biomechanical strength of glucocorticoid-treated animals. In other animal models, tocotrienol has been shown to activate the Wnt/β-catenin pathway and lower the RANKL/OPG ratio, which are the targets of glucocorticoids. In conclusion, tocotrienol enhances osteogenic differentiation and bone formation in glucocorticoid-treated osteoblasts while improving structural integrity in glucocorticoid-treated rats. This is achieved by preventing oxidative stress and osteoblast apoptosis. However, these preclinical results should be validated in a randomised controlled trial.
Collapse
|
6
|
Sahu R, Mehan S, Kumar S, Prajapati A, Alshammari A, Alharbi M, Assiri MA, Narula AS. Effect of alpha-mangostin in the prevention of behavioural and neurochemical defects in methylmercury-induced neurotoxicity in experimental rats. Toxicol Rep 2022; 9:977-998. [PMID: 35783250 PMCID: PMC9247835 DOI: 10.1016/j.toxrep.2022.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 12/11/2022] Open
Abstract
Methylmercury (MeHg+) is a known neurotoxin that causes progressive motor neuron degeneration in the central nervous system. Axonal degeneration, oligodendrocyte degeneration, and myelin basic protein (MBP) deficits are among the neuropathological abnormalities caused by MeHg+ in amyotrophic lateral sclerosis (ALS). This results in demyelination and motor neuron death in both humans and animals. Previous experimental studies have confirmed that overexpression of the extracellular signalling regulated kinase (ERK1/2) signalling contributes to glutamate excitotoxicity, inflammatory response of microglial cells, and oligodendrocyte (OL) dysfunction that promotes myelin loss. Alpha-mangostin (AMG), an active ingredient obtained from the tree "Garcinia mangostana Linn," has been used in experimental animals to treat a variety of brain disorders, including Parkinson's and Huntington's disease memory impairment, Alzheimer's disease, and schizophrenia, including Parkinson's disease and Huntington's disease memory impairment, Alzheimer's disease, and schizophrenia. AMG has traditionally been used as an antioxidant, anti-inflammatory, and neuroprotective agent.Accordingly, we investigated the therapeutic potential of AMG (100 and 200 mg/kg) in experimental rats with methylmercury (MeHg+)-induced neurotoxicity. The neuroprotective effect of AMG on behavioural, cellular, molecular, and other gross pathological changes, such as histopathological alterations in MeHg+ -treated rat brains, is presented. The neurological behaviour of experimental rats was evaluated using a Morris water maze (MWM), open field test (OFT), grip strength test (GST), and force swim test (FST). In addition, we investigate AMG's neuroprotective effect by restoring MBP levels in cerebral spinal fluid and whole rat brain homogenate. The apoptotic, pro-inflammatory, and oxidative stress markers were measured in rat blood plasma samples and brain homogenate. According to the findings of this study, AMG decreases ERK-1/2 levels and modulates neurochemical alterations in rat brains, minimising MeHg+ -induced neurotoxicity.
Collapse
Affiliation(s)
- Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sumit Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Aradhana Prajapati
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | | |
Collapse
|
7
|
Zhu C, Shen S, Zhang S, Huang M, Zhang L, Chen X. Autophagy in Bone Remodeling: A Regulator of Oxidative Stress. Front Endocrinol (Lausanne) 2022; 13:898634. [PMID: 35846332 PMCID: PMC9279723 DOI: 10.3389/fendo.2022.898634] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022] Open
Abstract
Bone homeostasis involves bone formation and bone resorption, which are processes that maintain skeletal health. Oxidative stress is an independent risk factor, causing the dysfunction of bone homeostasis including osteoblast-induced osteogenesis and osteoclast-induced osteoclastogenesis, thereby leading to bone-related diseases, especially osteoporosis. Autophagy is the main cellular stress response system for the limination of damaged organelles and proteins, and it plays a critical role in the differentiation, apoptosis, and survival of bone cells, including bone marrow stem cells (BMSCs), osteoblasts, osteoclasts, and osteocytes. High evels of reactive oxygen species (ROS) induced by oxidative stress induce autophagy to protect against cell damage or even apoptosis. Additionally, pathways such as ROS/FOXO3, ROS/AMPK, ROS/Akt/mTOR, and ROS/JNK/c-Jun are involved in the regulation of oxidative stress-induced autophagy in bone cells, including osteoblasts, osteocytes and osteoclasts. This review discusses how autophagy regulates bone formation and bone resorption following oxidative stress and summarizes the potential protective mechanisms exerted by autophagy, thereby providing new insights regarding bone remodeling and potential therapeutic targets for osteoporosis.
Collapse
Affiliation(s)
- Chenyu Zhu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Sports Science, Wenzhou Medical University, Wenzhou, China
| | - Shiwei Shen
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shihua Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- College of Sports and Health, Shandong Sport University, Jinan, China
| | - Mei Huang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lan Zhang
- College of Sports and Health, Shandong Sport University, Jinan, China
- *Correspondence: Xi Chen, ; Lan Zhang,
| | - Xi Chen
- School of Sports Science, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xi Chen, ; Lan Zhang,
| |
Collapse
|
8
|
Auraptene, a Monoterpene Coumarin, Inhibits LTA-Induced Inflammatory Mediators via Modulating NF- κB/MAPKs Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5319584. [PMID: 34824589 PMCID: PMC8610650 DOI: 10.1155/2021/5319584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/01/2021] [Indexed: 12/04/2022]
Abstract
Objective Oxidative stress-mediated inflammatory events involve in the progress of several diseases such as asthma, cancers, and multiple sclerosis. Auraptene (AU), a natural prenyloxycoumarin, possesses numerous pharmacological activities. Here, the anti-inflammatory effects of AU were investigated in lipoteichoic acid- (LTA-) induced macrophage cells (RAW 264.7). Methods The expression of cyclooxygenase (COX-2), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and inducible nitric oxide synthase (iNOS) and the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, p38 MAPK, c-Jun N-terminal kinase (JNK), heme oxygenase (HO-1), p65, and IκBα were all identified by western blotting assay. The level of nitric oxide (NO) was measured by spectrometer analysis. The nuclear translocation of p65 nuclear factor kappa B (NF-κB) was assessed by the confocal microscopic staining method. Native polyacrylamide gel electrophoresis was performed to perceive the activity of antioxidant enzyme catalase (CAT). Results AU expressively reduced NO production and COX-2, TNF-α, IL-1 β, and iNOS expression in LTA-stimulated cells. AU at higher concentration (10 µM) inhibited ERK and JNK, but not p38 phosphorylation induced by LTA. Moreover, AU blocked IκB and p65 phosphorylation, and p65 nuclear translocation. However, AU pretreatment was not effective on antioxidant HO-1 expression, CAT activity, and reduced glutathione (GSH, a nonenzymatic antioxidant), in LTA-induced RAW 264.7 cells. Conclusion The findings of this study advocate that AU shows anti-inflammatory effects via reducing NF-κB/MAPKs signaling pathways.
Collapse
|
9
|
Zhang X, Jiang Y, Mao J, Ren X, Ji Y, Mao Y, Chen Y, Sun X, Pan Y, Ma J, Huang S. Hydroxytyrosol prevents periodontitis-induced bone loss by regulating mitochondrial function and mitogen-activated protein kinase signaling of bone cells. Free Radic Biol Med 2021; 176:298-311. [PMID: 34610362 DOI: 10.1016/j.freeradbiomed.2021.09.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/24/2021] [Accepted: 09/30/2021] [Indexed: 01/10/2023]
Abstract
Reactive oxygen species (ROS) overproduction promotes the alveolar bone loss during the development of periodontitis. Mitochondria are the principal source of ROS. Hydroxytyrosol (HT), a natural phenolic compound present in olive oil, is well known for its antioxidant and mitochondrial-protective prosperities. Nonetheless, the impact of HT on periodontitis and its related mechanisms underlying bone cell behavior remains unknown. Osteoclasts differentiated from RAW264.7 model and oxidative stress (OS) induced pre-osteoblast MC3T3-E1 cell injury model were treated with and without HT. Cell viability, apoptosis, differentiation, mitochondrial function along with mitogen-activated protein kinase (MAPK) signaling pathway were investigated. Meanwhile, the effect and related mechanisms of HT on bone loss in mice with periodontitis were also detected. HT inhibited osteoclast differentiation and prevented OS induced pre-osteoblast cells injury via regulating mitochondrial function as well as ERK and JNK signaling pathways. Moreover, HT attenuated the alveolar bone loss, increased bone forming activity, inhibited the osteoclasts differentiation and decreased the level of OS in mice with periodontitis. Our findings, for the first time, revealed a novel function of HT in bone remodeling of periodontitis, and highlighted its therapeutical potential for the prevention/treatment of periodontitis.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yun Jiang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jiajie Mao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xuekun Ren
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yinghui Ji
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yixin Mao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yang Chen
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Sun
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yihuai Pan
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Jianfeng Ma
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
10
|
ERK: A Double-Edged Sword in Cancer. ERK-Dependent Apoptosis as a Potential Therapeutic Strategy for Cancer. Cells 2021; 10:cells10102509. [PMID: 34685488 PMCID: PMC8533760 DOI: 10.3390/cells10102509] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
The RAF/MEK/ERK signaling pathway regulates diverse cellular processes as exemplified by cell proliferation, differentiation, motility, and survival. Activation of ERK1/2 generally promotes cell proliferation, and its deregulated activity is a hallmark of many cancers. Therefore, components and regulators of the ERK pathway are considered potential therapeutic targets for cancer, and inhibitors of this pathway, including some MEK and BRAF inhibitors, are already being used in the clinic. Notably, ERK1/2 kinases also have pro-apoptotic functions under certain conditions and enhanced ERK1/2 signaling can cause tumor cell death. Although the repertoire of the compounds which mediate ERK activation and apoptosis is expanding, and various anti-cancer compounds induce ERK activation while exerting their anti-proliferative effects, the mechanisms underlying ERK1/2-mediated cell death are still vague. Recent studies highlight the importance of dual-specificity phosphatases (DUSPs) in determining the pro- versus anti-apoptotic function of ERK in cancer. In this review, we will summarize the recent major findings in understanding the role of ERK in apoptosis, focusing on the major compounds mediating ERK-dependent apoptosis. Studies that further define the molecular targets of these compounds relevant to cell death will be essential to harnessing these compounds for developing effective cancer treatments.
Collapse
|
11
|
Sahu R, Upadhayay S, Mehan S. Inhibition of extracellular regulated kinase (ERK)-1/2 signaling pathway in the prevention of ALS: Target inhibitors and influences on neurological dysfunctions. Eur J Cell Biol 2021; 100:151179. [PMID: 34560374 DOI: 10.1016/j.ejcb.2021.151179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/18/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cell signal transduction pathways are essential modulators of several physiological and pathological processes in the brain. During overactivation, these signaling processes may lead to disease progression. Abnormal protein kinase activation is associated with several biological dysfunctions that facilitate neurodegeneration under different biological conditions. As a result, these signaling pathways are essential in understanding brain disorders' development or progression. Recent research findings indicate the crucial role of extracellular signal-regulated kinase-1/2 (ERK-1/2) signaling during the neuronal development process. ERK-1/2 is a key component of its mitogen-activated protein kinase (MAPK) group, controlling certain neurological activities by regulating metabolic pathways, cell proliferation, differentiation, and apoptosis. ERK-1/2 also influences neuronal elastic properties, nerve growth, and neurological and cognitive processing during brain injuries. The primary goal of this review is to elucidate the activation of ERK1/2 signaling, which is involved in the development of several ALS-related neuropathological dysfunctions. ALS is a rare neurological disorder category that mainly affects the nerve cells responsible for regulating voluntary muscle activity. ALS is progressive, which means that the symptoms are getting worse over time, and there is no cure for ALS and no effective treatment to avoid or reverse. Genetic abnormalities, oligodendrocyte degradation, glial overactivation, and immune deregulation are associated with ALS progression. Furthermore, the current review also identifies ERK-1/2 signaling inhibitors that can promote neuroprotection and neurotrophic effects against the clinical-pathological presentation of ALS. As a result, in the future, the potential ERK-1/2 signaling inhibitors could be used in the treatment of ALS and related neurocomplications.
Collapse
Affiliation(s)
- Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shubham Upadhayay
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
12
|
Bengtson C, Bogaerts A. The Quest to Quantify Selective and Synergistic Effects of Plasma for Cancer Treatment: Insights from Mathematical Modeling. Int J Mol Sci 2021; 22:ijms22095033. [PMID: 34068601 PMCID: PMC8126141 DOI: 10.3390/ijms22095033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/30/2022] Open
Abstract
Cold atmospheric plasma (CAP) and plasma-treated liquids (PTLs) have recently become a promising option for cancer treatment, but the underlying mechanisms of the anti-cancer effect are still to a large extent unknown. Although hydrogen peroxide (H2O2) has been recognized as the major anti-cancer agent of PTL and may enable selectivity in a certain concentration regime, the co-existence of nitrite can create a synergistic effect. We develop a mathematical model to describe the key species and features of the cellular response toward PTL. From the numerical solutions, we define a number of dependent variables, which represent feasible measures to quantify cell susceptibility in terms of the H2O2 membrane diffusion rate constant and the intracellular catalase concentration. For each of these dependent variables, we investigate the regimes of selective versus non-selective, and of synergistic versus non-synergistic effect to evaluate their potential role as a measure of cell susceptibility. Our results suggest that the maximal intracellular H2O2 concentration, which in the selective regime is almost four times greater for the most susceptible cells compared to the most resistant cells, could be used to quantify the cell susceptibility toward exogenous H2O2. We believe our theoretical approach brings novelty to the field of plasma oncology, and more broadly, to the field of redox biology, by proposing new ways to quantify the selective and synergistic anti-cancer effect of PTL in terms of inherent cell features.
Collapse
|
13
|
Zheng D, Cui C, Shao C, Wang Y, Ye C, Lv G. Coenzyme Q10 inhibits RANKL-induced osteoclastogenesis by regulation of mitochondrial apoptosis and oxidative stress in RAW264.7 cells. J Biochem Mol Toxicol 2021; 35:e22778. [PMID: 33754447 DOI: 10.1002/jbt.22778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/06/2021] [Accepted: 03/12/2021] [Indexed: 01/21/2023]
Abstract
Coenzyme Q10 (CoQ10) has been reported to improve bone density and the number of trabeculae in postmenopausal osteoporosis, but the mechanism remains to be elucidated. We aimed to investigate the effects of CoQ10 on receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis and the underlying molecular mechanisms. RAW264.7 cells were treated with different concentrations of RANKL to differentiate into osteoclasts, and then these cells were treated with different concentrations of CoQ10 with or without H2 O2 . Tartrate-resistant acid phosphatase staining was performed to detect osteoclasts. Cell viability was tested by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, cell apoptosis was examined by flow cytometry, and the effects of CoQ10 on protein and messenger RNA expression of mitochondrial apoptosis-associated proteins and osteoclast marker proteins were measured by quantitative reverse transcription polymerase chain reaction and western blot, respectively. Furthermore, enzyme-linked immunosorbent assay was conducted to analyze the activities of malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT). RANKL significantly induced osteoclastogenesis in RAW264.7 cells, with the greatest efficiency at 50 ng/ml. CoQ10 had no significant effects on cell viability but it significantly increased the percentages of cell apoptosis. Mechanically, CoQ10 statistically decreased the levels of Bcl-2 and cytochrome C in mitochondria and upregulated the levels of Bax, cleaved caspase 3, and cytochrome C in the cytoplasm. Moreover, CoQ10 significantly decreased RANKL-induced osteoclastogenesis regardless of exposure to H2 O2 . In addition, CoQ10 statistically reduced MDA activity and elevated the activities of SOD and CAT, as well as the expression of oxidative stress-related proteins. CoQ10 may inhibit RANKL-induced osteoclastogenesis by regulation of mitochondrial apoptosis and oxidative stress in RAW264.7 cells.
Collapse
Affiliation(s)
- Delu Zheng
- Department of Endocrinology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Chenli Cui
- The Operative Surgery Laboratory, Bengbu Medical College, Bengbu, Anhui, China
| | - Chen Shao
- Department of Endocrinology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yanqiu Wang
- Department of Endocrinology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Chengsong Ye
- Department of Endocrinology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Gaoyou Lv
- Department of Endocrinology, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
14
|
Yang G, Xia X, Zhong H, Shen J, Li S. Protective Effect of Tangeretin and 5-Hydroxy-6,7,8,3',4'-Pentamethoxyflavone on Collagen-Induced Arthritis by Inhibiting Autophagy via Activation of the ROS-AKT/mTOR Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:259-266. [PMID: 33372513 DOI: 10.1021/acs.jafc.0c06801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by long duration and repeated relapse. This study explored the preventive effect of tangeretin (TAN) and 5-hydroxy-6,7,8,3',4'-pentamethoxyflavone (5-HPMF) on RA, and the underlying molecular mechanism based on a rat model stimulated by bovine type II collagen (BIIC). After the intervention of TAN or 5-HPMF (TAN/5-HPMF) for 5 weeks, the RA lesions and autophagy levels of the synovial tissue were significantly reduced, and the ROS content and HO-1 expression level were down-regulated simultaneously. The relative expression levels of p-AKT and p-mTOR were down-regulated after TAN/5-HPMF feeding. Meanwhile, the relative expression level of p62 increased by more than two-fold for TAN/5-HPMF treated rats at 200 mg/kg BW comparing with those in BIIC group. Results of immunofluorescence staining and Western blotting further confirmed that TAN/5-HPMF treatment reduced BIIC-induced conversion from LC3I to LC3II. Observations under transmission electron microscope also demonstrated that the autophagy level was reduced upon TAN/5-HPMF intervention. Collectively, these results revealed that TAN and 5-HPMF prevented the pathological process of BIIC-stimulated arthritis through inhibiting the autophagy of synovial cells, achieved via the ROS-AKT/mTOR signal axis. Thus, our findings confirmed the protective potential of TAN and 5-HPMF for RA disease.
Collapse
Affiliation(s)
- Guliang Yang
- National Engineering Laboratory for Rice and Byproducts Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, P. R. China
| | - Xinxin Xia
- National Engineering Laboratory for Rice and Byproducts Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, P. R. China
| | - Haiyan Zhong
- National Engineering Laboratory for Rice and Byproducts Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha, Hunan 410004, P. R. China
| | - Junfeng Shen
- Hubei Key Laboratory of EFGIR, Huanggang Normal University, Huanggang, Hubei 438000, P. R. China
| | - Shiming Li
- Hubei Key Laboratory of EFGIR, Huanggang Normal University, Huanggang, Hubei 438000, P. R. China
| |
Collapse
|
15
|
Han SH, Kim KT. RNF144a induces ERK-dependent cell death under oxidative stress via downregulation of vaccinia-related kinase 3. J Cell Sci 2020; 133:jcs247304. [PMID: 33067254 DOI: 10.1242/jcs.247304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/17/2020] [Indexed: 08/31/2023] Open
Abstract
Vaccinia-related kinase 3 (VRK3) has been reported to be a negative regulator of ERK (ERK1 and ERK2; also known as MAPK3 and MAPK1, respectively) that protects cells from persistent ERK activation and inhibits ERK-dependent apoptosis. Here we report that the E3 ubiquitin-protein ligase RNF144a promotes the degradation of VRK3 via polyubiquitylation and thus affects VRK3-mediated ERK activity. Under oxidative stress, VRK3 migrates from the nucleus to the cytoplasm, which increases its chance of interacting with RNF144a, thereby promoting the degradation of VRK3. Overexpression of RNF144a increases ERK activity via downregulation of VRK3 and promotes ERK-dependent apoptosis. In contrast, depletion of RNF144a increases the protein level of VRK3 and protects cells from excessive ERK activity. These findings suggest that VRK3 protects cells by suppressing oxidative stress-induced ERK, and that RNF144a sensitively regulates this process.
Collapse
Affiliation(s)
- Seung Hyun Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Kyong-Tai Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| |
Collapse
|
16
|
Four-octyl itaconate activates Nrf2 cascade to protect osteoblasts from hydrogen peroxide-induced oxidative injury. Cell Death Dis 2020; 11:772. [PMID: 32943614 PMCID: PMC7499214 DOI: 10.1038/s41419-020-02987-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023]
Abstract
Four-octyl itaconate (4-OI) is the cell-permeable derivative of itaconate that can activate Nrf2 signaling by alkylating Keap1’s cysteine residues. Here, we tested the potential effect of 4-OI on hydrogen peroxide (H2O2)-induced oxidative injury in osteoblasts. In OB-6 cells and primary murine osteoblasts, 4-OI was able to activate Nrf2 signaling cascade and cause Keap1–Nrf2 disassociation, Nrf2 protein stabilization, cytosol accumulation, and nuclear translocation. 4-OI also augmented antioxidant-response element reporter activity and promoted expression of Nrf2-dependent genes (HO1, NQO1, and GCLC). Pretreatment with 4-OI inhibited H2O2-induced reactive oxygen species production, cell death, and apoptosis in osteoblasts. Furthermore, 4-OI inhibited H2O2-induced programmed necrosis by suppressing mitochondrial depolarization, mitochondrial cyclophilin D-ANT1 (adenine nucleotide translocase 1)-p53 association, and cytosol lactate dehydrogenase release in osteoblasts. Ectopic overexpression of immunoresponsive gene 1 (IRG1) increased endogenous itaconate production and activated Nrf2 signaling cascade, thereby inhibiting H2O2-induced oxidative injury and cell death. In OB-6 cells, Nrf2 silencing or CRISPR/Cas9-induced Nrf2 knockout blocked 4-OI-induced osteoblast cytoprotection against H2O2. Conversely, forced Nrf2 activation, by CRISPR/Cas9-induced Keap1 knockout, mimicked 4-OI-induced actions in OB-6 cells. Importantly, 4-OI was ineffective against H2O2 in Keap1-knockout cells. Collectively, 4-OI efficiently activates Nrf2 signaling to inhibit H2O2-induced oxidative injury and death of osteoblasts.
Collapse
|
17
|
Ruiz-Heiland G, Yong JW, von Bremen J, Ruf S. Leptin reduces in vitro cementoblast mineralization and survival as well as induces PGE2 release by ERK1/2 commitment. Clin Oral Investig 2020; 25:1933-1944. [PMID: 32820432 PMCID: PMC7965856 DOI: 10.1007/s00784-020-03501-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022]
Abstract
Objectives Juvenile obesity is a complex clinical condition that is present more and more frequently in the daily orthodontic practice. Over-weighted patients have an impaired bone metabolism, due in part to their increased levels of circulating adipokines. Particularly, leptin has been reported to play a key role in bone physiology. Leptin is ubiquitously present in the body, including blood, saliva, and crevicular fluid. If, and to what extent, it could influence the reaction of cementoblasts during orthodontic-induced forces is yet unknown. Material and methods OCCM-30 cementoblasts were cultivated under compressive forces using different concentrations of leptin. The expression of ObR, Runx-2, Osteocalcin, Rank-L, Sost, Caspase 3, 8, and 9 were analyzed by RT-PCR. Western blots were employed for protein analysis. The ERK1/2 antagonist FR180204 (Calbiochem) was used and cPLA2 activation, PGE2, and cytochrome C release were further evaluated. Results In vitro, when compressive forces are applied, leptin promotes ERK1/2 phosphorylation, as well as upregulates PGE2 and caspase 3 and caspase 9 on OCCM cells. Blockade of ERK1/2 impairs leptin-induced PGE2 secretion and reduced caspase 3 and caspase 9 expression. Conclusions Leptin influences the physiological effect of compressive forces on cementoblasts, exerting in vitro a pro-inflammatory and pro-apoptotic effect. Clinical relevance Our findings indicate that leptin exacerbates the physiological effect of compressive forces on cementoblasts promoting the release of PGE2 and increases the rate of cell apoptosis, and thus, increased levels of leptin may influence the inflammatory response during orthodontically induced tooth movement.
Collapse
Affiliation(s)
- G Ruiz-Heiland
- Department of Orthodontics, University of Giessen, Schlangenzahl 14, 35392, Giessen, Germany.
| | - J W Yong
- Department of Orthodontics, University of Giessen, Schlangenzahl 14, 35392, Giessen, Germany
| | - J von Bremen
- Department of Orthodontics, University of Giessen, Schlangenzahl 14, 35392, Giessen, Germany
| | - S Ruf
- Department of Orthodontics, University of Giessen, Schlangenzahl 14, 35392, Giessen, Germany
| |
Collapse
|
18
|
Hong X, Yu Z, Chen Z, Jiang H, Niu Y, Huang Z. High molecular weight fibroblast growth factor 2 induces apoptosis by interacting with complement component 1 Q subcomponent-binding protein in vitro. J Cell Biochem 2018; 119:8807-8817. [PMID: 30159917 PMCID: PMC6220755 DOI: 10.1002/jcb.27131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/04/2018] [Indexed: 02/05/2023]
Abstract
Fibroblast growth factor 2 (FGF2) is a multifunctional cell growth factor that regulates cell proliferation, differentiation, adhesion, migration, and apoptosis. FGF2 has multiple isoforms, including an 18-kDa low molecular weight isoform (lo-FGF2) and 22-, 23-, 24-, and 34-kDa high molecular weight isoforms (hi-FGF2). Hi-FGF2 overexpression induces chromatin compaction, which requires the mitochondria and leads to apoptosis. Complement component 1 Q subcomponent-binding protein (C1QBP) plays an important role in mitochondria-dependent apoptosis by regulating the opening of the mitochondrial permeability transition pore. However, the interaction between C1QBP and hi-FGF2 and its role in hi-FGF2-mediated apoptosis remain unclear. Here, we found that hi-FGF2 overexpression induced depolarization of the mitochondrial membrane, cytochrome c release into the cytosol, and a considerable increase in C1QBP messenger RNA and protein expression. Furthermore, coimmunoprecipitation results showed that the mitochondrial protein, C1QBP, interacts with hi-FGF2. C1QBP knockdown using small interfering RNA significantly decreased the localization of hi-FGF2 to the mitochondria and increased the rate of apoptosis. Our results highlight a novel mechanism underlying hi-FGF2-induced, mitochondria-driven cell death involving the direct interaction between hi-FGF2 and C1QBP and the upregulation of C1QBP expression.
Collapse
Affiliation(s)
- Xiaobing Hong
- The Second Affiliated Hospital, Shantou University Medical CollegeShantouChina
| | - Zelin Yu
- The Second Affiliated Hospital, Shantou University Medical CollegeShantouChina
| | - Zhonglin Chen
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Hongyan Jiang
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Yongdong Niu
- Department of PharmacologyShantou University Medical CollegeShantouChina
| | - Zhanqin Huang
- Department of PharmacologyShantou University Medical CollegeShantouChina
| |
Collapse
|
19
|
Hou X, Xiao H, Zhang Y, Zeng X, Huang M, Chen X, Birnbaumer L, Liao Y. Transient receptor potential channel 6 knockdown prevents apoptosis of renal tubular epithelial cells upon oxidative stress via autophagy activation. Cell Death Dis 2018; 9:1015. [PMID: 30282964 PMCID: PMC6170481 DOI: 10.1038/s41419-018-1052-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/04/2018] [Accepted: 09/10/2018] [Indexed: 01/24/2023]
Abstract
Reactive oxygen species (ROS) are generated under various pathological conditions such as renal ischemia/reperfusion (I/R) injury and provoke damage to multiple cellular organelles and processes. Overproduction of ROS causes oxidative stress and contributes to damages of renal proximal tubular cells (PTC), which are the main cause of the pathogenesis of renal I/R injury. Autophagy is a dynamic process that removes long-lived proteins and damaged organelles via lysosome-mediated degradation, which has an antioxidant effect that relieves oxidative stress. The canonical transient receptor potential channel 6 (TRPC6), a nonselective cation channel that allows passage of Ca2+, plays an important role in renal disease. Yet, the relationship between TRPC6 and autophagy, as well as their functions in renal oxidative stress injury, remains unclear. In this study, we found that oxidative stress triggered TRPC6-dependent Ca2+ influx in PTC to inhibit autophagy, thereby rendering cells more susceptible to death. We also demonstrated that TRPC6 knockout (TRPC6-/-) or inhibition by SAR7334, a TRPC6-selective inhibitor, increased autophagic flux and mitigated oxidative stress-induced apoptosis of PTC. The protective effects of TRPC6 ablation were prevented by autophagy inhibitors Chloroquine and Bafilomycin A1. Moreover, this study also shows that TRPC6 blockage promotes autophagic flux via inhibiting the PI3K/Akt/mTOR and ERK1/2 signaling pathways. This is the first evidence showing that TRPC6-mediated Ca2+ influx plays a novel role in suppressing cytoprotective autophagy triggered by oxidative stress in PTC, and it may become a novel therapeutic target for the treatment of renal oxidative stress injury in the future.
Collapse
Affiliation(s)
- Xin Hou
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.,Department of Anatomy, Medical College, Affiliated Hospital, Hebei University of Engineering, 056002, Handan, China
| | - Haitao Xiao
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases of Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yanhong Zhang
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases of Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Xixi Zeng
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases of Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Mengjun Huang
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases of Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Xiaoyun Chen
- Department of Pathology, First Hospital of Wuhan, 430030, Wuhan, China
| | - Lutz Birnbaumer
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF, Buenos Aires, Argentina. .,Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA.
| | - Yanhong Liao
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China. .,Key Laboratory of Neurological Diseases of Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| |
Collapse
|
20
|
Oxidative stress-induced chromosome breaks within the ABL gene: a model for chromosome rearrangement in nasopharyngeal carcinoma. Hum Genomics 2018; 12:29. [PMID: 29914565 PMCID: PMC6006577 DOI: 10.1186/s40246-018-0160-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background The mechanism underlying chromosome rearrangement in nasopharyngeal carcinoma (NPC) remains elusive. It is known that most of the aetiological factors of NPC trigger oxidative stress. Oxidative stress is a potent apoptotic inducer. During apoptosis, chromatin cleavage and DNA fragmentation occur. However, cells may undergo DNA repair and survive apoptosis. Non-homologous end joining (NHEJ) pathway has been known as the primary DNA repair system in human cells. The NHEJ process may repair DNA ends without any homology, although region of microhomology (a few nucleotides) is usually utilised by this DNA repair system. Cells that evade apoptosis via erroneous DNA repair may carry chromosomal aberration. Apoptotic nuclease was found to be associated with nuclear matrix during apoptosis. Matrix association region/scaffold attachment region (MAR/SAR) is the binding site of the chromosomal DNA loop structure to the nuclear matrix. When apoptotic nuclease is associated with nuclear matrix during apoptosis, it potentially cleaves at MAR/SAR. Cells that survive apoptosis via compromised DNA repair may carry chromosome rearrangement contributing to NPC tumourigenesis. The Abelson murine leukaemia (ABL) gene at 9q34 was targeted in this study as 9q34 is a common region of loss in NPC. This study aimed to identify the chromosome breakages and/or rearrangements in the ABL gene in cells undergoing oxidative stress-induced apoptosis. Results In the present study, in silico prediction of MAR/SAR was performed in the ABL gene. More than 80% of the predicted MAR/SAR sites are closely associated with previously reported patient breakpoint cluster regions (BCR). By using inverse polymerase chain reaction (IPCR), we demonstrated that hydrogen peroxide (H2O2)-induced apoptosis in normal nasopharyngeal epithelial and NPC cells led to chromosomal breakages within the ABL BCR that contains a MAR/SAR. Intriguingly, we detected two translocations in H2O2-treated cells. Region of microhomology was found at the translocation junctions. This observation is consistent with the operation of microhomology-mediated NHEJ. Conclusions Our findings suggested that oxidative stress-induced apoptosis may participate in chromosome rearrangements of NPC. A revised model for oxidative stress-induced apoptosis mediating chromosome rearrangement in NPC is proposed. Electronic supplementary material The online version of this article (10.1186/s40246-018-0160-8) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Ruan JW, Yao C, Bai JY, Zhou XZ. microRNA-29a inhibition induces Gab1 upregulation to protect OB-6 human osteoblasts from hydrogen peroxide. Biochem Biophys Res Commun 2018; 503:607-614. [PMID: 29902453 DOI: 10.1016/j.bbrc.2018.06.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/10/2018] [Indexed: 11/25/2022]
Abstract
The present study determines the role of the Gab1 in hydrogen peroxide (H2O2)-induced death of human osteoblasts. We show that Gab1 is required for H2O2-induced Akt activation to promote osteoblast survival. In OB-6 human osteoblasts, Gab1 silencing (by targeted-shRNA) or complete knockout (by CRISPR-Cas9 KO plasmid) largely attenuated Akt activation by H2O2. Gab1-depleted OB-6 cells were more vulnerable to H2O2. Conversely, forced over-expression of Gab1 by an adenovirus vector increased Akt activation to protect OB-6 cells from H2O2. Significantly, the anti-sense of microRNA-29a ("antagomiR-29a") induced Gab1 expression to facilitate H2O2-induced Akt activation, which protected OB-6 cells from apoptosis. AntagomiR-29a was however ineffective in Gab1-deficient and Akt-inhibited OB-6 cells. Forced over-expression of miR-29a induced Gab1 downregulation to inhibit H2O2-induced Akt activation, causing enhanced OB-6 cell death. miR-29a-induced actions were abolished by an adenovirus constitutively-active Akt1 (Ad-caAkt1) in OB-6 cells. Together, microRNA-29a inhibition induces Gab1 upregulation and Akt activation to protect OB-6 osteoblasts from H2O2.
Collapse
Affiliation(s)
- Jian-Wei Ruan
- Department of Orthopedics, The Second Affiliated Hospital of Suzhou University, Suzhou, China; Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, China
| | - Chen Yao
- Orthopedic Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin-Yu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Suzhou University, Suzhou, China.
| |
Collapse
|
22
|
Zhu F, Liu Z, Ren Y. Mechanism of melatonin combined with calcium carbonate on improving osteoporosis in aged rats. Exp Ther Med 2018; 16:192-196. [PMID: 29977362 PMCID: PMC6030893 DOI: 10.3892/etm.2018.6141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/05/2017] [Indexed: 02/05/2023] Open
Abstract
The effects of melatonin and calcium carbonate on aged rats with osteoporosis (OP) were assessed. Forty female Sprague-Dawley (SD) rats aged 15 months were randomly divided into a model group (group OP), melatonin group (group M), calcium carbonate group (group Ca) and melatonin combined with calcium carbonate group (group M+Ca), while 10 rats aged 3 months were set as the control group (group NC). The changes of bone density and bone mineral level of lumbar vertebra and bilateral femur in rats of each group were observed. The levels of serum calcium, phosphorus, superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) in rats of each group were determined. Compared with those in group NC, bone density of lumbar vertebra and bilateral femur and bone mineral level were distinctly reduced, serum calcium and activities of SOD and GSH-Px were obviously decreased, and MDA content was remarkably increased in rats of groups OP, M and Ca; the differences were statistically significant (P<0.05 or P<0.01); compared with that in group OP, bone density of lumbar vertebra and bilateral femur and bone mineral level were remarkably increased, serum calcium and activities of SOD and GSH-Px were obviously increased, and MDA content was remarkably decreased in rats of groups M, Ca and M+Ca; the differences were statistically significant (P<0.05 or P<0.01); compared with those in groups M and Ca, bone density of lumbar vertebra and bilateral femur and bone mineral level were obviously elevated, serum calcium and activities of SOD and GSH-Px were evidently elevated, and MDA content was remarkably decreased in rats of group M+Ca; the differences were statistically significant (P<0.05). Melatonin and calcium carbonate can significantly improve antioxidative ability in rats with osteoporosis, increase bone density, elevate serum calcium level and reduce bone mineral loss, thus preventing and treating osteoporosis, and the combination displays more remarkable effects.
Collapse
Affiliation(s)
- Fuqiang Zhu
- Department of Spine Surgery, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Zhendong Liu
- Department of Orthopedics, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Yuxin Ren
- Department of Spine Surgery, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| |
Collapse
|
23
|
Olive compounds attenuate oxidative damage induced in HEK-293 cells via MAPK signaling pathway. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
24
|
Yan X, Wu H, Wu Z, Hua F, Liang D, Sun H, Yang Y, Huang D, Bian JS. The New Synthetic H 2S-Releasing SDSS Protects MC3T3-E1 Osteoblasts against H 2O 2-Induced Apoptosis by Suppressing Oxidative Stress, Inhibiting MAPKs, and Activating the PI3K/Akt Pathway. Front Pharmacol 2017; 8:07. [PMID: 28163684 PMCID: PMC5247634 DOI: 10.3389/fphar.2017.00007] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) are important in osteoporosis development. Oxidative stress induces apoptosis of osteoblasts and arrest of their differentiation. Both Danshensu (DSS) and hydrogen sulfide (H2S) produce significant antioxidant effect in various systems. In this study, we synthesized SDSS, a novel H2S-releasing compound derived from DSS, and studied its antioxidant effect in an H2O2-induced MC3T3-E1 osteoblastic cell injury model. We first characterized the H2S releasing property of SDSS in both in vivo and in vitro models. HPLC chromatogram showed that intravenous injection of SDSS in adult rats released ADT-OH, a well proved H2S sustained-release moiety, within several minutes in the rat plasma. Using an H2S selective fluorescent probe, we further confirmed that SDSS released H2S in MC3T3-E1 osteoblastic cells. Biological studies revealed that SDSS had no significant toxic effect but produced protective effects against H2O2-induced MC3T3-E1 cell apoptosis. SDSS also reversed the arrest of cell differentiation caused by H2O2 treatment. This was caused by the stimulatory effect of SDSS on bone sialoprotein, runt-related transcription factor 2, collagen expression, alkaline phosphatase activity, and bone nodule formation. Further studies revealed that SDSS reversed the reduced superoxide dismutase activity and glutathione content, and the increased ROS production in H2O2 treated cells. In addition, SDSS significantly attenuated H2O2-induced activation of p38-, ERK1/2-, and JNK-MAPKs. SDSS also stimulated phosphatidylinositol 3-kinase/Akt signaling pathway. Blockade of this pathway attenuated the cytoprotective effect of SDSS. In conclusion, SDSS protects MC3T3-E1 cells against H2O2-induced apoptosis by suppressing oxidative stress, inhibiting MAPKs, and activating the phosphatidylinositol 3-kinase/Akt pathway.
Collapse
Affiliation(s)
- Xiaofei Yan
- Department of Biochemistry and Molecular Biology, Medical College of Xi'an Jiaotong UniversityXi'an, China; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Haixia Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore; Department of Food Science, Faculty of Science, National University of SingaporeSingapore, Singapore
| | - Zhiyuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| | - Fei Hua
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| | - Dong Liang
- Department of Food Science, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University Xuzhou, China
| | - Yong Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University Nanjing, China
| | - Dejian Huang
- Department of Food Science, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| |
Collapse
|
25
|
Zhu FB, Wang JY, Zhang YL, Hu YG, Yue ZS, Zeng LR, Zheng WJ, Hou Q, Yan SG, Quan RF. Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteoblasts. Mol Med Rep 2016; 14:5377-5384. [PMID: 27840925 DOI: 10.3892/mmr.2016.5908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/17/2016] [Indexed: 11/05/2022] Open
Abstract
Monotropein, the primary iridoid glycoside isolated from Morindacitrifolia, has been previously reported to possess potent antioxidant and antiosteoporotic properties. However, there is no direct evidence correlating the antiosteoporotic effect of monotropein with its observed antioxidant capacity, and the molecular mechanisms involved in mediating these processes remain unclear. Therefore, the aim of the present study was to investigate the protective effects of monotropein against oxidative stress in osteoblasts and the mechanisms involved in mediating this process. Osteoblast viability was evaluated using the MTT assay. The mitochondrial membrane potential and reactive oxygen species were detected by flow cytometry analyses. Western blotting and enzyme‑linked immunosorbent assays were performed to detect protein expression levels. A significant reduction in osteoblast viability was observed at 24 h following exposure to various concentrations (100‑1,000 µM) of H2O2 compared with untreated osteoblasts. The cytotoxic effect of H2O2 was notably reversed when osteoblasts were pretreated with 1‑10 µg/ml monotropein. Pretreatment with 1-10 µg/ml monotropein increased the mitochondrial membrane potential and reduced the generation of reactive oxygen species in osteoblasts following exposure to H2O2. In addition, the H2O2‑induced increase in apoptotic markers (caspase-3 and caspase-9) and H2O2-induced reduction in sirtuin 1 levels were significantly reversed following pretreatment of cells with monotropein. Furthermore, monotropein significantly reduced H2O2‑induced stimulation of NF‑κB expression, in addition to the expression of a number of proinflammatory mediators. These results indicate that monotropein suppresses apoptosis and the inflammatory response in H2O2‑induced osteoblasts through the activation of the mitochondrial apoptotic signaling pathway and inhibition of the NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Fang-Bing Zhu
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Jian-Yue Wang
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Ying-Liang Zhang
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Yun-Gen Hu
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Zhen-Shuang Yue
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Lin-Ru Zeng
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Wen-Jie Zheng
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Qiao Hou
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Shi-Gui Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Ren-Fu Quan
- Department of Orthopedic Surgery, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang 311200, P.R. China
| |
Collapse
|
26
|
Drynda A, Ren Q, Buchhorn GH, Lohmann CH. The induction of CXCR4 expression in human osteoblast-like cells (MG63) by CoCr particles is regulated by the PLC-DAG-PKC pathway. J Biomed Mater Res B Appl Biomater 2016; 105:2326-2332. [PMID: 27504737 DOI: 10.1002/jbm.b.33770] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 06/13/2016] [Accepted: 07/26/2016] [Indexed: 01/13/2023]
Abstract
BACKGROUND Osteolysis which leads to aseptic loosening of implants is a fundamental problem in joint replacement surgery (arthroplasty) and the leading cause for implant failure and revision surgery. Metal (CoCr) particles separated from implants by wear cause osteolysis and the failure of orthopedic implants, but the molecular mechanism is not clear. The chemokine receptor CXCR4 has been shown to play a pivotal role in periprosthetic osteolysis. The aim of this study was to determine which signal transduction pathway (PLC-DAG-PKC or MAPK/ERK) induces CXCR4 expression in osteoblast-like cells (MG63) cells. METHODS MG63 and Jurkat cells were stimulated with different amounts of particles (107 , 106 , and 105 ) for different time periods (30 min to 24 h), in the presence and absence of specific inhibitors (chelerythrine for the PLC-DAG-PKC pathway and PD98059 for the MAPK/ERK pathway). The expression of CXCR4-specific mRNA was determined by real-time polymerase chain reaction (PCR), and the PKC activity was measured by Western Blot using an antibody specific for PKC-related phosphorylation. RESULTS Real-time PCR data showed that CXCR4 mRNA expression in MG63 cells induced by CoCr particles was significantly diminished by the PKC-specific inhibitor chelerythrine. This effect was not observed with the MAPK/ERK inhibitor PD98059. The involvement of PKC was also confirmed by an intensified phosphorylation pattern after stimulation with CoCr particles. In Jurkat cells, none of the inhibitors exhibited any effect. CONCLUSION The induction of CXCR4-specific mRNA expression in MG63 cells after stimulation with CoCr particles is regulated by the PLC-DAG-PKC pathway and not by the MAPK/ERK pathway. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2326-2332, 2017.
Collapse
Affiliation(s)
- Andreas Drynda
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Qiang Ren
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Christoph H Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
27
|
Hwang IH, Kwon YK, Cho CK, Lee YW, Sung JS, Joo JC, Lee KB, Yoo HS, Jang IS. Modified Panax ginseng Extract Inhibits uPAR-Mediated α5β1-Integrin Signaling by Modulating Caveolin-1 to Induce Early Apoptosis in Lung Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:1081-97. [PMID: 27430913 DOI: 10.1142/s0192415x16500609] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Urokinase receptor (uPAR) is enhanced in many human cancer cells and is frequently an indicator of poor prognosis. Activation of [Formula: see text]1-integrin requires caveolin-1 and is regulated by uPAR. However, the underlying molecular mechanism responsible for the interaction between uPAR and [Formula: see text]1-integrin remains obscure. We found that modified regular Panax ginseng extract (MRGX) had a negative modulating effect on the uPAR/[Formula: see text]1-integrin interaction, disrupted the uPAR/integrin interaction by modulating caveoline-1, and caused early apoptosis in cancer cells. Additionally, we found that siRNA-mediated caveoline-1 downregulation inhibited uPAR-mediated [Formula: see text]1-integrin signaling, whereas caveoline-1 up-regulation stimulated the signaling, which suppressed p53 expression, thereby indicating negative crosstalk exists between the integrin [Formula: see text]1 and the p53 pathways. Thus, these findings identify a novel mechanism whereby the inhibition of [Formula: see text]1 integrin and the activation of p53 modulate the expression of the anti-apoptotic proteins that are crucially involved in inducing apoptosis in A549 lung cancer cells. Furthermore, MRGX causes changes in the expressions of members of the Bcl-2 family (Bax and Bcl-2) in a pro-apoptotic manner. In addition, MGRX-mediated inhibition of [Formula: see text]1 integrin attenuates ERK phosphorylation (p-ERK), which up-regulates caspase-8 and Bax. Therefore, ERK may affect mitochondria through a negative regulation of caspase-8 and Bax. Taken together, these findings reveal that MRGX is involved in uPAR-[Formula: see text]1-integrin signaling by modulating caveolin-1 signaling to induce early apoptosis in A549 lung-cancer cells and strongly indicate that MRGX might be useful as a herbal medicine and may lead to the development of new herbal medicine that would suppress the growth of lung-cancer cells.
Collapse
Affiliation(s)
- In-Hu Hwang
- Department of Physiology, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Yong-Kyun Kwon
- East-West Cancer Center, Daejeon University, Daejeon 302-120, Republic of Korea
| | - Chong-Kwan Cho
- East-West Cancer Center, Daejeon University, Daejeon 302-120, Republic of Korea
| | - Yeon-Weol Lee
- East-West Cancer Center, Daejeon University, Daejeon 302-120, Republic of Korea
| | - Jung-Suk Sung
- Department of Life Science, Dongguk University, Goyang, Gyeonggi-do 10326, Republic of Korea
| | - Jong-Cheon Joo
- Department of Sasang Constitutional Medicine, Wonkwang University Oriental Medical Hospital, Jeonju 54887, Republic of Korea
| | - Kyung-Bok Lee
- Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Korea
| | - Hwa-Seung Yoo
- East-West Cancer Center, Daejeon University, Daejeon 302-120, Republic of Korea
| | - Ik-Soon Jang
- Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Korea
| |
Collapse
|
28
|
Ardura JA, Portal-Núñez S, Castelbón-Calvo I, Martínez de Toda I, De la Fuente M, Esbrit P. Parathyroid Hormone-Related Protein Protects Osteoblastic Cells From Oxidative Stress by Activation of MKP1 Phosphatase. J Cell Physiol 2016; 232:785-796. [PMID: 27357344 DOI: 10.1002/jcp.25473] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 06/29/2016] [Indexed: 11/06/2022]
Abstract
Oxidative damage is an important contributor to the morphological and functional changes in osteoporotic bone. Aging increases the levels of reactive oxygen species (ROS) that cause oxidative stress and induce osteoblast apoptosis. ROS modify several signaling responses, including mitogen-activated protein kinase (MAPK) activation, related to cell survival. Both parathyroid hormone (PTH) and its bone counterpart, PTH-related protein (PTHrP), can regulate MAPK activation by modulating MAPK phosphatase-1 (MKP1). Thus, we hypothesized that PTHrP might protect osteoblasts from ROS-induced apoptosis by targeting MKP1. In osteoblastic MC3T3-E1 and MG-63 cells, H2 O2 triggered p38, JNK, ERK and p66Shc phosphorylation, and cell apoptosis. Meanwhile, PTHrP (1-37) rapidly but transiently increased ERK and Akt phosphorylation without affecting p38, JNK, or p66Shc activation. H2 O2 -induced p38 and ERK phosphorylation and apoptosis were both decreased by pre-treatment with specific kinase inhibitors or PTHrP (1-37) in both osteoblastic cell types. These dephosphorylating and prosurvival actions of PTHrP (1-37) were prevented by a phosphatase inhibitor cocktail, the phosphatase MKP1 inhibitor sanguinarine or a MKP1 siRNA. PTHrP (1-37) promptly enhanced MKP1 protein and gene expression and MKP1-dependent catalase activity in osteoblastic cells. Furthermore, exposure to PTHrP (1-37) adsorbed in an implanted hydroxyapatite-based ceramic into a tibial defect in aging rats increased MKP1 and catalase gene expression in the healing bone area. Our findings demonstrate that PTHrP counteracts the pro-apoptotic actions of ROS by a mechanism dependent on MKP1-induced dephosphorylation of MAPKs in osteoblasts. J. Cell. Physiol. 232: 785-796, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Juan A Ardura
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, UAM and Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Medicina Molecular Aplicada (IMMA)-Universidad San Pablo CEU, Madrid, Spain
| | - Sergio Portal-Núñez
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, UAM and Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| | - Irantzu Castelbón-Calvo
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, UAM and Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| | | | - Mónica De la Fuente
- Faculty of Biology, Animal Physiology II, Complutense University, Madrid, Spain
| | - Pedro Esbrit
- Laboratorio de Metabolismo Mineral y Óseo, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, UAM and Red Temática de Investigación Cooperativa de Envejecimiento y Fragilidad (RETICEF)-Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Tan SN, Sim SP, Khoo ASB. Potential role of oxidative stress-induced apoptosis in mediating chromosomal rearrangements in nasopharyngeal carcinoma. Cell Biosci 2016; 6:35. [PMID: 27231526 PMCID: PMC4880972 DOI: 10.1186/s13578-016-0103-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 05/10/2016] [Indexed: 12/27/2022] Open
Abstract
Background Genetic aberrations have been identified in nasopharyngeal carcinoma (NPC), however, the underlying mechanism remains elusive. There are increasing evidences that the apoptotic nuclease caspase-activated deoxyribonuclease (CAD) is one of the players leading to translocation in leukemia. Oxidative stress, which has been strongly implicated in carcinogenesis, is a potent apoptotic inducer. Most of the NPC etiological factors are known to induce oxidative stress. Although apoptosis is a cell death process, cells possess the potential to survive apoptosis upon DNA repair. Eventually, the surviving cells may carry rearranged chromosomes. We hypothesized that oxidative stress-induced apoptosis may cause chromosomal breaks mediated by CAD. Upon erroneous DNA repair, cells that survive apoptosis may harbor chromosomal rearrangements contributing to NPC pathogenesis. This study focused on the AF9 gene at 9p22, a common deletion region in NPC. We aimed to propose a possible model for molecular mechanism underlying the chromosomal rearrangements in NPC. Results In the present study, we showed that hydrogen peroxide (H2O2) induced apoptosis in NPC (HK1) and normal nasopharyngeal epithelial (NP69) cells, as evaluated by flow cytometric analyses. Activity of caspases 3/7 was detected in H2O2-treated cells. This activity was inhibited by caspase inhibitor (CI). By nested inverse polymerase chain reaction (IPCR), we demonstrated that oxidative stress-induced apoptosis in HK1 and NP69 cells resulted in cleavages within the breakpoint cluster region (BCR) of the AF9 gene. The gene cleavage frequency detected in the H2O2-treated cells was found to be significantly higher than untreated control. We further found that treatment with CI, which indirectly inhibits CAD, significantly reduced the chromosomal breaks in H2O2-cotreated cells. Intriguingly, a few breakpoints were mapped within the AF9 region that was previously reported to translocate with the mixed lineage leukemia (MLL) gene in acute lymphoblastic leukemia (ALL) patient. Conclusions In conclusion, our findings suggested that oxidative stress-induced apoptosis could be one of the mechanisms underlying the chromosomal rearrangements in NPC. CAD may play an important role in chromosomal cleavages mediated by oxidative stress-induced apoptosis. A potential model for oxidative stress-induced apoptosis mediating chromosomal rearrangements in NPC is proposed.
Collapse
Affiliation(s)
- Sang-Nee Tan
- Faculty of Medicine and Health Sciences, Department of Paraclinical Sciences, Universiti Malaysia Sarawak, Sarawak, Malaysia
| | - Sai-Peng Sim
- Faculty of Medicine and Health Sciences, Department of Paraclinical Sciences, Universiti Malaysia Sarawak, Sarawak, Malaysia
| | - Alan S B Khoo
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Yao A, Shen Y, Wang A, Chen S, Zhang H, Chen F, Chen Z, Wei H, Zou Z, Shan Y, Zhang X. Sulforaphane induces apoptosis in adipocytes via Akt/p70s6k1/Bad inhibition and ERK activation. Biochem Biophys Res Commun 2015; 465:696-701. [PMID: 26296464 DOI: 10.1016/j.bbrc.2015.08.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/12/2015] [Indexed: 10/23/2022]
Abstract
Sulforaphane (SFN), an isothiocyanate isolated from cruciferous vegetables, possesses anti-oxidant and anti-cancer bioactivities. Moreover, SFN exerts its pro-apoptotic effects in some cancer lines. However, the effects and mechanisms of SFN on the regulation of apoptosis of adipocytes are still unknown. In this study, we found that SFN induced significant apoptosis in 3T3-L1 adipocytes and markedly decreased the cellular lipid content. Western blot demonstrated that SFN-induced apoptosis was mediated via the mitochondrial apoptosis pathway based on increased cleavage of poly-ADP-ribose-polymerase (PARP), release of cytochrome c into the cytoplasm, and activation of caspase-3, as well as decreased Bcl-2/Bax ratio. In addition, SFN markedly decreased phosphorylation of Akt and downstream proteins, p70s6k1 and Bad, and increased phosphorylation of ERK. Therefore, our findings clarified that SFN could induce 3T3-L1 adipocyte apoptosis via down-regulation of the Akt/p70s6k1/Bad pathway and up-regulation of the ERK pathway, suggesting SFN may be a promising agent for the treatment or prevention of obesity.
Collapse
Affiliation(s)
- Anjun Yao
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Yingzhuo Shen
- Medical School of Ningbo University, Ningbo, Zhejiang, China; YinZhou District Center of Disease Control, Ningbo, Zhejiang, China
| | - Anshi Wang
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Shiyong Chen
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Huiqin Zhang
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Fen Chen
- The Affiliated Hospital of School of Medicine of Ningbo University, Ningbo, Zhejiang, China
| | | | - Hua Wei
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Zuquan Zou
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Yujuan Shan
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Xiaohong Zhang
- Medical School of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
31
|
Rahim AH, Setiawan B, Dewi FRP, Noor Z. Regulation by Phloroglucinol of Nrf2/Maf-Mediated Expression of Antioxidant Enzymes and Inhibition of Osteoclastogenesis via the RANKL/RANK Signaling Pathway: In Silico study. Acta Inform Med 2015; 23:228-32. [PMID: 26483597 PMCID: PMC4584081 DOI: 10.5455/aim.2015.23.228-232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/15/2015] [Indexed: 12/22/2022] Open
Abstract
Introduction: Phloroglucinol is an antioxidant compound with many positive effects on health. The purpose of this study was to determine the role of phloroglucinol in osteoclastogenesis via the RANKL/RANK signaling pathway and the activity of the transcription factor Nrf2. Material and methods: Analysis was performed in silico using the primary method of docking by the use of Hex 8.0 software and Haddock web server. Analysis of interactions was then performed to determine interactions between the ligand and its receptors by using the software LigPlus and LigandScout 3.1. Results: Results indicated that phloroglucinol compound was thought to inhibit osteoclastogenesis via three mechanisms: inhibiting RANKL−RANK interaction, sustaining the RANKL−OPG bond, and increasing the activity of the transcription factor Nrf2.
Collapse
Affiliation(s)
- Agus Hadian Rahim
- Department of Orthopaedics and Traumatology, Hasan Sadikin General Hospital, Medical Faculty Padjadjaran University, Bandung, West Java, Indonesia
| | - Bambang Setiawan
- Research Center for Osteoporosis, Department of Medical Chemistry and Biochemistry, Medical Faculty Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| | | | - Zairin Noor
- Research Center for Osteoporosis, Department of Orthopaedics and Traumatology, Ulin General Hospital, Medical Faculty Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| |
Collapse
|
32
|
Zhang S, Li D, Yang JY, Yan TB. Plumbagin protects against glucocorticoid-induced osteoporosis through Nrf-2 pathway. Cell Stress Chaperones 2015; 20:621-9. [PMID: 25939783 PMCID: PMC4463920 DOI: 10.1007/s12192-015-0585-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/14/2015] [Accepted: 03/18/2015] [Indexed: 11/25/2022] Open
Abstract
Long-term and high-dose glucocorticoids (GCs) supplementation has been linked to osteoporosis. In this study, we studied the protective role of plumbagin against GC-induced cell damage in MC3T3-E1 cells. The effect of dexamethasone (DEX) and plumbagin on cell viability was determined. DEX showed as IC-50 value of 95 μM. Further, 10 μM plumbagin treatment effectively ameliorated DEX-induced cell death by increasing the cell viability to 92 %. A further effect of plumbagin on DEX-induced oxidative stress was determined through reactive oxygen species (ROS) level, lipid peroxide content, and antioxidant status. Nrf-2 nuclear localization was analyzed through immunofluorescence. Protein expression of redox regulator Nrf-2 and their target genes HO-1 and NQO1 and osteogenic markers (OCN, OPN Runx-2) were determined by Western blot. Apoptotic effect was analyzed by mitochondrial membrane potential and caspase activities (3, 8, and 9). The results showed that DEX treatment showed a significant increase in oxidative stress through increased ROS levels and downregulation of cytoprotective antioxidant proteins and antioxidant enzyme activities. Further DEX treatment downregulated the osteogenic markers and upregulated apoptosis through decreased mitochondrial membrane potential and upregulation of caspase activities. Plumbagin treatment significantly reversed the levels of oxidative stress and apoptotic markers and protected against DEX-induced cell damage. Further, plumbagin treatment significantly improved the expression of osteogenic markers compared to DEX treatment. In conclusion, the present study shows that plumbagin offers significant protective role against DEX-induced cellular damage via regulating oxidative stress, apoptosis, and osteogenic markers.
Collapse
Affiliation(s)
- Shuai Zhang
- />Department of Orthopedics, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Road, Jinan, Shandong 250012 China
| | - Dong Li
- />Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021 China
| | - Jing-Yan Yang
- />Department of Pathology, The Second Hospital of Shandong University, Jinan, Shandong 250033 China
| | - Ting-Bin Yan
- />Department of Orthopedics, Qilu Hospital of Shandong University, No. 107 Wenhuaxi Road, Jinan, Shandong 250012 China
| |
Collapse
|
33
|
Cheng X, Gao DX, Song JJ, Ren FZ, Mao XY. Casein glycomacropeptide hydrolysate exerts cytoprotection against H2O2-induced oxidative stress in RAW 264.7 macrophages via ROS-dependent heme oxygenase-1 expression. RSC Adv 2015. [DOI: 10.1039/c4ra10034d] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Casein glycomacropeptide hydrolysate had antioxidant activity and exerted protective actions against H2O2-induced oxidative stress via induction of Nrf2-mediated HO-1 expression in RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Xue Cheng
- Key Laboratory of Functional Dairy
- College of Food Science and Nutritional Engineering
- China Agricultural University
- Beijing
- P. R. China
| | - Dong-Xiao Gao
- Key Laboratory of Functional Dairy
- College of Food Science and Nutritional Engineering
- China Agricultural University
- Beijing
- P. R. China
| | - Jia-Jia Song
- Key Laboratory of Functional Dairy
- College of Food Science and Nutritional Engineering
- China Agricultural University
- Beijing
- P. R. China
| | - Fa-Zheng Ren
- Key Laboratory of Functional Dairy
- College of Food Science and Nutritional Engineering
- China Agricultural University
- Beijing
- P. R. China
| | - Xue-Ying Mao
- Key Laboratory of Functional Dairy
- College of Food Science and Nutritional Engineering
- China Agricultural University
- Beijing
- P. R. China
| |
Collapse
|
34
|
Pathania AS, Kumar S, Guru SK, Bhushan S, Sharma PR, Aithagani SK, Singh PP, Vishwakarma RA, Kumar A, Malik F. The synthetic tryptanthrin analogue suppresses STAT3 signaling and induces caspase dependent apoptosis via ERK up regulation in human leukemia HL-60 cells. PLoS One 2014; 9:e110411. [PMID: 25383546 PMCID: PMC4226462 DOI: 10.1371/journal.pone.0110411] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 09/20/2014] [Indexed: 01/05/2023] Open
Abstract
Tryptanthrin is a natural product which has been reported to have several medicinal properties. In this study, we tried to investigate the detailed molecular mechanism of its bromo analogue (TBr), a potent cytotoxic agent in the induction of cancer cell death. It was found that TBr primarily targets STAT3 and ERK signaling during the induction of apoptosis in several human leukemia cell lines. In HL-60 cells, TBr treatment caused early down regulation of p-STAT3 with concomitant up regulation of p-ERK which led to the activation of intrinsic and extrinsic pathways of apoptosis. The mechanism of TBr mediated inhibition of p-STAT3 was found to be due to the activation of ubiquitin dependent degradation of tyrosine 705 and serine 727 p-STAT3. As IL-6 is the main driver of the STAT3 pathway, the effect of TBr on cell death was subdued when treated in the combination with IL-6 in HL60 cells. Interestingly, PD98059 significantly reduced the apoptotic effects of TBr, thus showing the direct involvement of p-ERK in TBr mediated cell death. It was further shown that apoptotic protein Bax silencing in HL-60 cells resists TBr mediated ERK dependent apoptosis. In summary, for the first time we report the mechanism of TBr mediated cell death in human leukemia cell lines by targeting STAT3 and ERK pathways.
Collapse
Affiliation(s)
- Anup S. Pathania
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Suresh Kumar
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Santosh K. Guru
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
| | - Shashi Bhushan
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
| | - Parduman R. Sharma
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
| | - Sravan K. Aithagani
- Medicinal chemistry division, Indian institute of Integrative Medicine, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Parvinder P. Singh
- Medicinal chemistry division, Indian institute of Integrative Medicine, Jammu and Kashmir, India
| | - Ram A. Vishwakarma
- Medicinal chemistry division, Indian institute of Integrative Medicine, Jammu and Kashmir, India
| | - Ajay Kumar
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
- * E-mail: (AK); (FM)
| | - Fayaz Malik
- Department of Cancer Pharmacology, Indian Institute of Integrative Medicine, Jammu and Kashmir, India
- Experimental Breast Cancer Research Laboratory, University of Michigan North Campus Research Complex, Ann Arbor, Michigan, United States of America
- * E-mail: (AK); (FM)
| |
Collapse
|
35
|
Ke K, Sul OJ, Choi EK, Safdar AM, Kim ES, Choi HS. Reactive oxygen species induce the association of SHP-1 with c-Src and the oxidation of both to enhance osteoclast survival. Am J Physiol Endocrinol Metab 2014; 307:E61-70. [PMID: 24824657 DOI: 10.1152/ajpendo.00044.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Loss of ovarian function causes oxidative stress as well as bone loss. We hypothesized that reactive oxygen species (ROS) induced by the failure of ovarian function are responsible for the bone loss by increasing the number of osteoclasts (OC). We found that ROS enhanced OC survival via Src homology 2 domain-containing phosphatase-1 (SHP-1), c-Src, Akt, and ERK. ROS induced the association of SHP-1 with c-Src as well as the oxidation of c-Src and SHP-1. This resulted in inactivation of SHP-1 and activation of c-Src via phosphorylation of Tyr(416). Knockdown of c-Src or SHP-1 abolished the effect of ROS on OC survival. Moreover, downregulation of SHP-1 upregulated activation of c-Src, Akt, and ERK in the absence of any stimulus, suggesting that inactivation of SHP-1 is required for OC survival. We demonstrated that the association and oxidation of c-Src and SHP-1 by ROS are key steps in enhancing OC survival, which are responsible for increased bone loss when ovarian function ceases.
Collapse
Affiliation(s)
- Ke Ke
- Department of Biological Sciences, University of Ulsan, Ulsan, South Korea; and
| | - Ok-Joo Sul
- Department of Biological Sciences, University of Ulsan, Ulsan, South Korea; and
| | - Eun-Kyung Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, South Korea; and
| | - Ali M Safdar
- Department of Biological Sciences, University of Ulsan, Ulsan, South Korea; and
| | - Eun-Sook Kim
- Department of Endocrinology, Ulsan University Hospital, Ulsan, South Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, South Korea; and
| |
Collapse
|
36
|
Abstract
SIGNIFICANCE Oxidative (reactive oxygen species [ROS]) and nitrosative (reactive nitrogen species [RNS]) stress affects many physiological processes, including survival and death. Although high levels of ROS/RNS mainly causes cell death, low levels of free radicals directly modulate the activities of transcriptional factors, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), p53, and nuclear factor (erythroid-derived) 2-like (Nrf2), and regulate numerous protein kinase cascades that participate in the regulation of the cross talk between autophagy and apoptosis. RECENT ADVANCES Low levels of ROS modify Atg4 and high mobility group box 1 (HMGB1) proteins, activate AMP-activated protein kinase (AMPK) and apoptosis signal-regulating kinase/c-Jun N-terminal kinase (JNK) pathways, or transactivate various proteins that could upregulate autophagy, leading to reductions in apoptosis. Transactivation of antioxidant genes blocks apoptosis and serves as a feedback loop to reduce autophagy. Free radicals could also activate protein kinase B (PKB, or Akt), preventing both autophagy and apoptosis. Stimulation of nitric oxide formation causes S-nitrosylation of several kinases, including JNK1 and IκB kinase β, which blocks autophagy and could promote apoptosis. However, S-nitrosylation of some proapoptotic proteins could block apoptosis. CRITICAL ISSUES Endoplasmic reticulum and mitochondria are the main sources of free radicals, which play an essential role in the regulation of apoptosis and autophagy. Oxidation of cardiolipin promotes cytochrome c release and apoptosis that potentially could be inhibited by autophagic clearance of damaged mitochondria. Elimination of damaged mitochondria reduces ROS accumulation, creating a feedback loop that causes inhibition of autophagy. Low levels of RNS could inhibit fission of mitochondria, which would block their degradation by autophagy and spare cells from apoptosis. FUTURE DIRECTIONS Understanding of mechanisms that regulate the cross talk between cell fates is essential for discovery of therapeutic tools in the strenuous fight against various disorders, including neurodegeneration and cancer.
Collapse
Affiliation(s)
- Vitaliy O Kaminskyy
- 1 Division of Toxicology, Institute of Environmental Medicine , Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
37
|
Zhu Y, Zhou J, Ao R, Yu B. A-769662 protects osteoblasts from hydrogen dioxide-induced apoptosis through activating of AMP-activated protein kinase (AMPK). Int J Mol Sci 2014; 15:11190-203. [PMID: 24960362 PMCID: PMC4100207 DOI: 10.3390/ijms150611190] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/14/2014] [Accepted: 05/04/2014] [Indexed: 12/17/2022] Open
Abstract
Here we report that 5'-monophosphate (AMP)-activated protein kinase (AMPK) agonist A-769662 inhibited hydrogen peroxide (H2O2)-induced viability loss and apoptosis of human and mouse osteoblast cells. H2O2-induced moderate AMPK activation in osteoblast cells, which was enhanced by A-769662. Inactivation of AMPK by its inhibitor compound C, or by target shRNA-mediated silencing and kinase dead (KD) mutation exacerbated H2O2-induced cytotoxicity in osteoblast cells. A-769662-mediated protective effect against H2O2 was also blocked by AMPK inhibition or depletion. A-769662 inhibited reactive oxygen species (ROS) accumulation by H2O2 in osteoblast cells. Meanwhile, H2O2-induced ATP depletion was inhibited by A-769662, but was aggravated by compound C. Further, H2O2 induced AMPK-dependent and pro-survival autophagy in cultured osteoblast cells, which was enhanced by A-769662. Our results suggested that activation of AMPK by H2O2 is anti-apoptosis and pro-survival in osteoblast cells, probably due to its anti-oxidant, pro-autophagy and ATP preservation abilities, and A-769662-mediated cell-protective effect in osteoblast cells requires AMPK activation. Our study suggests that A-769662 might be further investigated as a novel anti-osteonecrosis agent.
Collapse
Affiliation(s)
- Yalong Zhu
- Orthopedics Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, China.
| | - Jianhua Zhou
- Orthopedics Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, China.
| | - Rongguang Ao
- Orthopedics Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, China.
| | - Baoqing Yu
- Orthopedics Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, China.
| |
Collapse
|
38
|
Takano-Yamamoto T. Osteocyte function under compressive mechanical force. JAPANESE DENTAL SCIENCE REVIEW 2014. [DOI: 10.1016/j.jdsr.2013.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
39
|
Hoshi K, Kawaki H, Takahashi I, Takeshita N, Seiryu M, Murshid SA, Masuda T, Anada T, Kato R, Kitaura H, Suzuki O, Takano-Yamamoto T. Compressive force-produced CCN2 induces osteocyte apoptosis through ERK1/2 pathway. J Bone Miner Res 2014; 29:1244-57. [PMID: 24155087 DOI: 10.1002/jbmr.2115] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 09/27/2013] [Accepted: 10/08/2013] [Indexed: 11/06/2022]
Abstract
Osteocytes produce various factors that mediate the onset of bone formation and resorption and play roles in maintaining bone homeostasis and remodeling in response to mechanical stimuli. One such factor, CCN2, is thought to play a significant role in osteocyte responses to mechanical stimuli, but its function in osteocytes is not well understood. Here, we showed that CCN2 induces apoptosis in osteocytes under compressive force loading. Compressive force increased CCN2 gene expression and production, and induced apoptosis in osteocytes. Application of exogenous CCN2 protein induced apoptosis, and a neutralizing CCN2 antibody blocked loading-induced apoptosis. We further examined how CCN2 induces loaded osteocyte apoptosis. In loaded osteocytes, extracellular signal-regulated kinase 1/2 (ERK1/2) was activated, and an ERK1/2 inhibitor blocked loading-induced apoptosis. Furthermore, application of exogenous CCN2 protein caused ERK1/2 activation, and the neutralizing CCN2 antibody inhibited loading-induced ERK1/2 activation. Therefore, this study demonstrated for the first time to our knowledge that enhanced production of CCN2 in osteocytes under compressive force loading induces apoptosis through activation of ERK1/2 pathway.
Collapse
Affiliation(s)
- Kenji Hoshi
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ahn SH, Lee SH, Kim BJ, Lim KH, Bae SJ, Kim EH, Kim HK, Choe JW, Koh JM, Kim GS. Higher serum uric acid is associated with higher bone mass, lower bone turnover, and lower prevalence of vertebral fracture in healthy postmenopausal women. Osteoporos Int 2013; 24:2961-70. [PMID: 23644878 DOI: 10.1007/s00198-013-2377-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/19/2013] [Indexed: 12/13/2022]
Abstract
UNLABELLED Higher serum uric acid (UA) was associated with higher bone mass, lower bone turnover, and lower prevalence of vertebral fracture in postmenopausal women. Furthermore, UA suppressed osteoclastogenesis and decreased production of reactive oxygen species in osteoclast precursors, indicating UA may have beneficial effects on bone metabolism as an antioxidant. INTRODUCTION UA is known to play a physiological role as an antioxidant, and oxidative stress has detrimental effects on bone metabolism. In the present study, we investigated the association of serum UA level with the osteoporosis-related phenotypes and its direct effect on bone-resorbing osteoclasts using in vitro systems. METHODS This is a large cross-sectional study, including 7,502 healthy postmenopausal women. Bone mineral density (BMD) and serum UA concentrations were obtained from all subjects. Data on bone turnover markers and lateral thoracolumbar radiographs were available for 1,023 and 6,918 subjects, respectively. An in vitro study investigated osteoclastogenesis and reactive oxygen species (ROS) levels according to UA treatment. RESULTS After adjusting for multiple confounders, serum UA levels were positively associated with BMD at all sites (all p < 0.001). Compared with the participants in the highest UA quartile, the odds for osteoporosis were 40 % higher in those in the lowest quartile. The serum UA levels were inversely related to both serum C-terminal telopeptide of type I collagen and osteocalcin levels (p < 0.001 and p = 0.004, respectively). Consistently, subjects with vertebral fracture had lower serum UA levels, compared with those without it (p = 0.009). An in vitro study showed that UA decreased osteoclastogenesis in a dose-dependent manner and reduced the production of ROS in osteoclast precursors. CONCLUSION These results provide epidemiological and experimental evidence that serum UA may have a beneficial effect on bone metabolism as an antioxidant in postmenopausal women.
Collapse
Affiliation(s)
- S H Ahn
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 388-1 Poongnap2-Dong, Songpa-Gu, Seoul, 138-736, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
SHE FEI, WANG WENBO, WANG YAN, TANG PEIFU, WEI JUNQIANG, CHEN HUA, ZHANG BOXUN. Melatonin protects MG63 osteoblast-like cells from hydrogen peroxide-induced cytotoxicity by maintaining mitochondrial function. Mol Med Rep 2013; 9:493-8. [DOI: 10.3892/mmr.2013.1832] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 11/18/2013] [Indexed: 11/06/2022] Open
|
42
|
The transcription factor NFAT1 induces apoptosis through cooperation with Ras/Raf/MEK/ERK pathway and upregulation of TNF-α expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2016-28. [DOI: 10.1016/j.bbamcr.2013.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 03/20/2013] [Accepted: 04/02/2013] [Indexed: 12/26/2022]
|
43
|
Casati L, Sendra R, Poletti A, Negri-Cesi P, Celotti F. Androgen receptor activation by polychlorinated biphenyls: epigenetic effects mediated by the histone demethylase Jarid1b. Epigenetics 2013; 8:1061-8. [PMID: 23907094 PMCID: PMC3891687 DOI: 10.4161/epi.25811] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The exposure to environmental endocrine disrupting compounds (EDC), as polychlorinated biphenyls (PCBs), widely diffused in the environment may produce epigenetic changes that affect the endocrine system. We found that PCBs activate AR transcriptional activity and that this effect is potentiated by the demethylase Jarid1b, a histone demethylase that catalyzes the removal of trimethylation of lysine 4 on histone H3 (H3K4me3), induced by PCB. The aim of the present study was to investigate the effect of the treatment of cultured cells (HEK293) with a mixture of the most diffused environmental PCBs and, also with dihydrotestosterone (DHT), on the functional interaction between AR and Jarid1b. Although the effect induced by DHT on the AR transactivation was considerably higher, the PCB mixture produced an AR-mediated transactivation in a dose-dependent manner. Cotransfection with plasmids expressing Jarid1b and various AR isoforms containing polyglutamine tracts (polyQ tracts) of different lengths showed that Jarid1b potentiates the AR transcriptional activity induced by PCBs but only with the shortest AR isoform. The potentiating effect of Jarid1b on the AR is mediated by a direct interaction of the enzyme with the AR promoter. In fact, utilizing constructs containing AR promoters with a different length and a luciferase reporter gene, we showed that the effect of PCBs, but not of DHT, needs the presence of Jarid1b and of at least two DNA binding sites for Jarid1b.
Collapse
Affiliation(s)
- Lavinia Casati
- Department of Pharmacological and Biomolecular Sciences; University of Milan; Milano, Italy
| | - Ramon Sendra
- Departament de Bioquímica i Biologia Molecular; Universitat de València; Valencia, Spain
| | - Angelo Poletti
- Department of Pharmacological and Biomolecular Sciences; University of Milan; Milano, Italy
| | - Paola Negri-Cesi
- Department of Pharmacological and Biomolecular Sciences; University of Milan; Milano, Italy
| | - Fabio Celotti
- Department of Pharmacological and Biomolecular Sciences; University of Milan; Milano, Italy
| |
Collapse
|
44
|
Lin T, Chen Y, Ding Z, Luo G, Liu J, Shen J. Novel insights into the synergistic interaction of a thioredoxin reductase inhibitor and TRAIL: the activation of the ASK1-ERK-Sp1 pathway. PLoS One 2013; 8:e63966. [PMID: 23696862 PMCID: PMC3655947 DOI: 10.1371/journal.pone.0063966] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 04/09/2013] [Indexed: 01/17/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces cell death in various types of cancer cells but has little or no effects on normal cells. Unfortunately, not all cancer cells respond to TRAIL; therefore, TRAIL sensitizing agents are currently being explored. Here, we reported that 6-(4-N,N-dimethylaminophenyltelluro)-6-deoxy-β-cyclodextrin (DTCD), a cyclodextrin-derived diorganyl telluride which has been identified as an excellent inhibitor of thioredoxin reductase (TrxR), could sensitize TRAIL resistant human ovarian cancer cells to undergo apoptosis. In vitro, DTCD enhanced TRAIL-induced cytotoxicity in human ovarian cancer cells through up-regulation of DR5. Luciferase analysis and CHIP assays showed that DTCD increased DR5 promoter activity via Sp1 activation. Additionally, DTCD stimulated extracellular signal-regulated kinase (ERK) activation, while the ERK inhibitor PD98059 blocked DTCD-induced DR5 expression and suppressed binding of Sp1 to the DR5 promoter. We further demonstrated that DTCD could induce the release of ASK1 from its complex with Trx-1, and recovered its kinase activity. Meanwhile, suppression of ASK1 by RNA interference led to decreased ERK phosphorylation induced by DTCD. The underlying mechanisms reveal that Trx-1 is heavily oxidized in response to DTCD treatment, in accordance with the fact that DTCD could inhibit the activity of TrxR that reduces oxidized Trx-1. Moreover, using an A2780 xenograft model, DTCD plus TRAIL significantly inhibited the growth of tumor in vivo. Our results suggest that Trx/TrxR system inhibition may play a critical role in apoptosis by combined treatment with DTCD and TRAIL, and raise the possibility that their combination may be a promising strategy for ovarian carcinoma treatment.
Collapse
Affiliation(s)
- Tingting Lin
- College of Instrumentation and Electrical Engineering, Jilin University, Changchun, PR China.
| | | | | | | | | | | |
Collapse
|
45
|
Pan LL, Liu XH, Jia YL, Wu D, Xiong QH, Gong QH, Wang Y, Zhu YZ. A novel compound derived from danshensu inhibits apoptosis via upregulation of heme oxygenase-1 expression in SH-SY5Y cells. Biochim Biophys Acta Gen Subj 2013; 1830:2861-71. [DOI: 10.1016/j.bbagen.2013.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 12/16/2012] [Accepted: 01/08/2013] [Indexed: 01/06/2023]
|
46
|
Sun Y, Shuang F, Chen DM, Zhou RB. Treatment of hydrogen molecule abates oxidative stress and alleviates bone loss induced by modeled microgravity in rats. Osteoporos Int 2013; 24:969-78. [PMID: 22648000 DOI: 10.1007/s00198-012-2028-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/14/2012] [Indexed: 10/28/2022]
Abstract
UNLABELLED Treatment with molecular hydrogen alleviates microgravity-induced bone loss through abating oxidative stress, restoring osteoblastic differentiation, and suppressing osteoclast differentiation and osteoclastogenesis. INTRODUCTION Recently, it has been suggested that hydrogen gas exerts a therapeutic antioxidant activity by selectively reducing cytotoxic reactive oxygen species (ROS). The aim of the present study was to elucidate whether treatment with molecular hydrogen alleviated bone loss induced by modeled microgravity in rats. METHODS Hindlimb suspension (HLS) and rotary wall vessel bioreactor were used to model microgravity in vivo and in vitro, respectively. Sprague-Dawley rats were exposed to HLS for 6 weeks to induced bone loss and simultaneously administrated with hydrogen water (HW). Then, we investigated the effects of incubation with hydrogen-rich medium (HRM) on MC3T3-E1 and RAW264.7 cells exposed to modeled microgravity. RESULTS Treatment with HW alleviated HLS-induced reduction of bone mineral density, ultimate load, stiffness, and energy in femur and lumbar vertebra. Treatment with HW alleviated HLS-induced augmentation of malondialdehyde content and peroxynitrite content and reduction of total sulfhydryl content in femur and lumbar vertebra. In cultured MC3T3-E1 cells, incubation with HRM inhibited modeled microgravity-induced ROS formation, reduction of osteoblastic differentiation, increase of ratio of receptor activator of nuclear factor kappa B ligand to osteoprotegerin, inducible nitric oxide synthetase upregulation, and Erk1/2 phosphorylation. In cultured RAW264.7, incubation with HRM aggravated modeled microgravity-induced ROS formation, osteoclastic differentiation, and osteoclastogenesis. CONCLUSION Treatment with molecular hydrogen alleviates microgravity-induced bone loss in rats. Molecular hydrogen could thus be envisaged as a nutritional countermeasure for spaceflight but remains to be tested in humans.
Collapse
Affiliation(s)
- Y Sun
- Department of Emergency, The Military General Hospital of Beijing PLA, Beijing 100700, China
| | | | | | | |
Collapse
|
47
|
Kang SM, Cha SH, Ko JY, Kang MC, Kim D, Heo SJ, Kim JS, Heu MS, Kim YT, Jung WK, Jeon YJ. Neuroprotective effects of phlorotannins isolated from a brown alga, Ecklonia cava, against H2O2-induced oxidative stress in murine hippocampal HT22 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 34:96-105. [PMID: 22465981 DOI: 10.1016/j.etap.2012.03.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 02/06/2012] [Accepted: 03/03/2012] [Indexed: 05/31/2023]
Abstract
Exposure of neurons to hydrogen peroxide (H(2)O(2)) results in oxidative stress and the activation of a cascade of intracellular toxic events resulting in oxidation, lipid peroxidation, and Ca(2+) elevation, ultimately resulting in cell death. In this study, we attempted to characterize the neuroprotective effects of phlorotannins isolated from Ecklonia cava, including phloroglucinol, eckol, triphloroethol A, eckstolonol, and dieckol, against H(2)O(2)-induced cell damage in murine hippocampus neuronal (HT22) cells. We measured the reactive oxygen species (ROS) and lipid peroxidation levels and evaluated the resultant cell death and alterations in Ca(2+)-concentrations. All phlorotannins were to scavenge intracellular ROS and repress ROS accumulation, thus preventing lipid peroxidation. Consquently, all phlorotannins reduced H(2)O(2)-induced cell death in HT22 cells. Moreover, phlorotannins inhibited H(2)O(2)-induced Ca(2+) release. This study provides a new useful strategy for preventing neuronal H(2)O(2)-induced oxidative stress.
Collapse
Affiliation(s)
- Sung-Myung Kang
- Department of Marine Life Science, Jeju National University, Jeju 690-756, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Palanichamy K, Sreejayan N, Ontko AC. Overcoming cisplatin resistance using gold(III) mimics: anticancer activity of novel gold(III) polypyridyl complexes. J Inorg Biochem 2011; 106:32-42. [PMID: 22112837 DOI: 10.1016/j.jinorgbio.2011.08.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 11/27/2022]
Abstract
Gold(III) compounds have been recognized as anticancer agents due to their structural and electronic similarities with currently employed platinum(II) species. An added benefit to gold(III) agents is the ability to overcome cisplatin resistance. This work identified four gold(III) compounds, [Au(Phen)Cl(2)]PF(6), [Au(DPQ)Cl(2)]PF(6), [Au(DPPZ)Cl(2)]PF(6), and [Au(DPQC)Cl(2)]PF(6), (Phen = 1,10-phenanthroline, DPQ = dipyrido[3,2-d:2',3'-f]quinoxaline, DPPZ = dipyrido[3,2-a:2',3'-c] phenazine, DPQC = dipyrido[3,2-d:2',3'-f] cyclohexyl quinoxaline) that exhibited anticancer activity in both cisplatin sensitive and cisplatin resistant ovarian cancer cells. Two of these compounds, [Au(DPQ)Cl(2)]PF(6) (AQ) and [Au(DPPZ)Cl(2)]PF(6) (AZ), displayed exceptional anticancer activity and were the focus of more intensive mechanistic study. At the molecular level, AQ and AZ formed DNA adducts, generated free radicals, and upregulated pro-apoptotic signaling molecules (p53, caspases, PARP, death effectors). Taken together, these two novel gold(III) polypyridyl complexes exhibit potent antitumor activity in cisplatin resistant cancer cells. These activities may be mediated, in part, by the activation of apoptotic signaling.
Collapse
Affiliation(s)
- Kamalakannan Palanichamy
- Department of Radiation Oncology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | |
Collapse
|
49
|
Xu ZS, Wang XY, Xiao DM, Hu LF, Lu M, Wu ZY, Bian JS. Hydrogen sulfide protects MC3T3-E1 osteoblastic cells against H2O2-induced oxidative damage-implications for the treatment of osteoporosis. Free Radic Biol Med 2011; 50:1314-23. [PMID: 21354302 DOI: 10.1016/j.freeradbiomed.2011.02.016] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 02/15/2011] [Accepted: 02/16/2011] [Indexed: 01/16/2023]
Abstract
Osteoporosis is a bone disease that leads to an increased risk of fracture. Oxidative damage is an important contributor to the morphological and functional changes in the development of osteoporosis. We found in this study that hydrogen sulfide (H2S), a novel endogenous gaseous mediator, protected MC3T3-E1 osteoblastic cells against hydrogen peroxide (H2O2)-induced oxidative injury. NaHS, an H2S donor, increased cell viability and reduced cell apoptosis caused by H2O2. NaHS also stimulated osteoblast proliferation by enhancing both transcription and activity of alkaline phosphatase in MC3T3-E1 osteoblastic cells. Moreover, treatment with NaHS stimulated the transcriptional level of osteocalcin, the main bone matrix protein, and the protein expression of collagen, a major constituent of bone tissue. The above effects were mediated by the antioxidant effect of H2S. NaHS reversed the reduced superoxide dismutase activity, decreased reactive oxygen species production, and suppressed NADPH oxidase activity in H2O2-treated osteoblasts. In addition, NaHS treatment also produced anti-inflammatory effects via inhibition of the production of nitric oxide and TNF-α, suggesting an anti-inflammatory effect of H2S. Cell viability and Western blotting analysis demonstrated that the protective effects of H2S were mediated by p38 and ERK1/2 MAPKs. In conclusion, H2S protects osteoblastic cells against oxidative stress-induced cell injury and suppression of proliferation and differentiation via a MAPK (p38 and ERK1/2)-dependent mechanism. Our findings suggest that H2S may have a potentially therapeutic value for osteoporosis.
Collapse
Affiliation(s)
- Zhong-Shi Xu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore
| | | | | | | | | | | | | |
Collapse
|
50
|
Ishihara Y, Ito F, Shimamoto N. Increased expression of c-Fos by extracellular signal-regulated kinase activation under sustained oxidative stress elicits BimEL upregulation and hepatocyte apoptosis. FEBS J 2011; 278:1873-81. [PMID: 21439021 DOI: 10.1111/j.1742-4658.2011.08105.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We previously reported that the inhibition of catalase and glutathione peroxidase activities by treatment with 3-amino-1,2,4-triazole (ATZ) and mercaptosuccinic acid evoked sustained increases in the levels of reactive oxygen species and apoptosis in rat primary hepatocytes. Apoptosis was accompanied by increased expression of BimEL, following activation of extracellular signal-regulated kinase. The aim of this study was to characterize the mechanism underlying hepatocyte apoptosis by identifying the transcription factor that induces BimEL expression. The bim promoter region was cloned into a promoterless-luc vector, and promoter activity was monitored by a luciferase assay. The luciferase activity increased in the presence of ATZ + mercaptosuccinic acid. Pretreatment with a MEK inhibitor, U0126, or an antioxidant, vitamin C, suppressed the promoter activity. Furthermore, ATZ + mercaptosuccinic acid-induced luciferase activity was attenuated by mutation of the activator protein-1 binding site in the bim promoter region. The amounts of total and phosphorylated c-Fos increased over time in the presence of ATZ + mercaptosuccinic acid, whereas the amounts of total and phosphorylated c-Jun remained unchanged. Chromatin immunoprecipitation revealed that both c-Fos and c-Jun localized to the activator protein-1-binding site in the bim promoter region. BimEL expression and hepatocyte apoptosis were suppressed by knockdown of c-Fos and c-Jun, respectively. These results indicate that increases in c-Fos following extracellular signal-regulated kinase activation are critical for BimEL upregulation and apoptosis.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, Japan
| | | | | |
Collapse
|