1
|
Wang X, Sun X, Zhang X, Shen N, Xia J, Wang L, Ye S. AMPK-YAP signaling pathway-mediated mitochondrial dynamics and mitophagy participate in the protective effect of silibinin on HaCaT cells under high glucose conditions. Arch Biochem Biophys 2025; 769:110433. [PMID: 40268264 DOI: 10.1016/j.abb.2025.110433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/23/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
UVB irradiation and diabetes lead to skin injury. However, UVB irradiation has rarely been studied in the field of diabetes. Silibinin has a positive therapeutic effect on many diseases. Nevertheless, the inhibitory effects of silibinin on UVB-induced damage to epidermal cells under high glucose (HG) conditions have been infrequently investigated. Consequently, this study examined the protective efficacy and mechanisms of silibinin in mitigating UVB-induced apoptosis in epidermal cells cultured under HG conditions. The effects of combination of HG and UVB on mitochondrial apoptosis and pro-inflammatory factors production in human immortalized keratinocytes (HaCaT) were mitigated by silibinin. Meantime, silibinin reversed the UVB-induced imbalance of fission/fusion in HG-cultured HaCaT cells. Furthermore, UVB exposure increased ROS levels and reduced mitophagy in HaCaT cells under HG conditions; however, these effects were subsequently reversed by silibinin treatment. AMPK preserves energy balance by negatively regulating YAP. Silibinin increased the levels of p-AMPK and cytoplasmic YAP proteins in HaCaT cells subjected to HG and UVB treatment. Moreover, silibinin improved the dysfunction of mitochondrial dynamics, increased mitophagy levels, the viability and the expression of cytoplasmic YAP protein, and these effects were reversed via the application of an AMPK inhibitor (compound C). In summary, silibinin safeguarded epidermal cells from UVB-induced apoptosis under HG conditions by modulating mitochondrial dynamics and mitophagy through the AMPK-YAP signaling pathway.
Collapse
Affiliation(s)
- Xianshi Wang
- Department of Pharmacy, Nuclear Industry 215 Hospital of Shaanxi Province, Xianyang, 712000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xueying Sun
- Central Sterile Supply Department, The Second Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaodi Zhang
- Department of Pharmacy, Nuclear Industry 215 Hospital of Shaanxi Province, Xianyang, 712000, China
| | - Naiying Shen
- Department of Pharmacy, Nuclear Industry 215 Hospital of Shaanxi Province, Xianyang, 712000, China
| | - Junlong Xia
- Department of Pharmacy, Nuclear Industry 215 Hospital of Shaanxi Province, Xianyang, 712000, China
| | - Lu Wang
- Department of Pharmacy, Nuclear Industry 215 Hospital of Shaanxi Province, Xianyang, 712000, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Ashique S, Mohanto S, Kumar N, Nag S, Mishra A, Biswas A, Rihan M, Srivastava S, Bhowmick M, Taghizadeh-Hesary F. Unlocking the possibilities of therapeutic potential of silymarin and silibinin against neurodegenerative Diseases-A mechanistic overview. Eur J Pharmacol 2024; 981:176906. [PMID: 39154829 DOI: 10.1016/j.ejphar.2024.176906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/28/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Silymarin, a bioflavonoid derived from the Silybum marianum plant, was discovered in 1960. It contains C25 and has been extensively used as a therapeutic agent against liver-related diseases caused by alcohol addiction, acute viral hepatitis, and toxins-inducing liver failure. Its efficacy stems from its role as a potent anti-oxidant and scavenger of free radicals, employed through various mechanisms. Additionally, silymarin or silybin possesses immunomodulatory characteristics, impacting immune-enhancing and immune-suppressive functions. Recently, silymarin has been recognized as a potential neuroprotective therapy for various neurological conditions, including Parkinson's and Alzheimer's diseases, along with conditions related to cerebral ischemia. Its hepatoprotective qualities, primarily due to its anti-oxidant and tissue-regenerating properties, are well-established. Silymarin also enhances health by modifying processes such as inflammation, β-amyloid accumulation, cellular estrogenic receptor mediation, and apoptotic machinery. While believed to reduce oxidative stress and support neuroprotective mechanisms, these effects represent just one aspect of the compound's multifaceted protective action. This review article further delves into the possibilities of potential therapeutic advancement of silymarin and silibinin for the management of neurodegenerative disorders via mechanics modules.
Collapse
Affiliation(s)
- Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India.
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Sagnik Nag
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Anuradha Mishra
- Amity Institute of Pharmacy, Amity University Lucknow Campus, Uttar Pradesh, 226010, India
| | - Aritra Biswas
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Rahara Akhil Mukherjee Road, Khardaha, West Bengal, 700118, India; UNESCO Regional Centre for Biotechnology, Department of Biotechnology, Government of India, NCR Biotech Science Cluster, Faridabad, 121001, Haryana, India.
| | - Mohd Rihan
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Shriyansh Srivastava
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, 203201, India; Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi, 110017, India
| | - Mithun Bhowmick
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Ashique S, Mohanto S, Kumar N, Nag S, Mishra A, Biswas A, Rihan M, Srivastava S, Bhowmick M, Taghizadeh-Hesary F. Unlocking the possibilities of therapeutic potential of silymarin and silibinin against neurodegenerative Diseases-A mechanistic overview. Eur J Pharmacol 2024; 981:176906. [DOI: https:/doi.org/10.1016/j.ejphar.2024.176906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
|
4
|
Kanungo J, Sorkin BC, Krzykwa J, Mitchell CA, Embry M, Spencer P, Harry GJ, Cannon J, Liu F, McPherson CA, Gafner S, Westerink RH. Screening tools to evaluate the neurotoxic potential of botanicals: building a strategy to assess safety. Expert Opin Drug Metab Toxicol 2024; 20:629-646. [PMID: 38984683 PMCID: PMC11542175 DOI: 10.1080/17425255.2024.2378895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/08/2024] [Indexed: 07/11/2024]
Abstract
AREAS COVERED This paper outlines the selection of NAMs, including in vitro assays using primary rat cortical neurons, zebrafish embryos, and Caenorhabditis elegans. These assays aim to assess neurotoxic endpoints such as neuronal activity and behavioral responses. Microelectrode array recordings of rat cortical neurons provide insights into the impact of botanical extracts on neuronal function, while the zebrafish embryos and C. elegans assays evaluate neurobehavioral responses. The paper also provides an account of the selection of botanical case studies based on expert judgment and existing neuroactivity/toxicity information. The proposed battery of assays will be tested with these case studies to evaluate their utility for neurotoxicity screening. EXPERT OPINION The complexity of botanicals necessitates the use of multiple NAMs for effective neurotoxicity screening. This paper discusses the evaluation of methodologies to develop a robust framework for evaluating botanical safety, including complex neuronal models and key neurodevelopmental process assays. It aims to establish a comprehensive screening framework.
Collapse
Affiliation(s)
- Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079
| | - Barbara C. Sorkin
- Office of Dietary Supplements, Division of Program Coordination, Planning, and Strategic Initiatives, U.S. National Institutes of Health, Bethesda, MD
| | - Julie Krzykwa
- Health and Environmental Sciences Institute, Washington, DC, USA
| | | | - Michelle Embry
- Health and Environmental Sciences Institute, Washington, DC, USA
| | - Peter Spencer
- Department of Neurology, School of Medicine, Oregon Health & Science University
| | - G. Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Jason Cannon
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Fang Liu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079
| | - Christopher A. McPherson
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Stefan Gafner
- American Botanical Council, 6200 Manor Road, Austin, Texas 78723, United States
| | - Remco H.S. Westerink
- Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
5
|
Fan G, Liu M, Liu J, Huang Y. The initiator of neuroexcitotoxicity and ferroptosis in ischemic stroke: Glutamate accumulation. Front Mol Neurosci 2023; 16:1113081. [PMID: 37033381 PMCID: PMC10076579 DOI: 10.3389/fnmol.2023.1113081] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Glutamate plays an important role in excitotoxicity and ferroptosis. Excitotoxicity occurs through over-stimulation of glutamate receptors, specifically NMDAR, while in the non-receptor-mediated pathway, high glutamate concentrations reduce cystine uptake by inhibiting the System Xc-, leading to intracellular glutathione depletion and resulting in ROS accumulation, which contributes to increased lipid peroxidation, mitochondrial damage, and ultimately ferroptosis. Oxidative stress appears to crosstalk between excitotoxicity and ferroptosis, and it is essential to maintain glutamate homeostasis and inhibit oxidative stress responses in vivo. As researchers work to develop natural compounds to further investigate the complex mechanisms and regulatory functions of ferroptosis and excitotoxicity, new avenues will be available for the effective treatment of ischaemic stroke. Therefore, this paper provides a review of the molecular mechanisms and treatment of glutamate-mediated excitotoxicity and ferroptosis.
Collapse
Affiliation(s)
- Genhao Fan
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Menglin Liu
- Graduate School, Tianjin University of Chinese Medicine, Tianjin, China
| | - Jia Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
| | - Yuhong Huang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Tianjin University of Chinese Medicine, Tianjin, China
- *Correspondence: Yuhong Huang,
| |
Collapse
|
6
|
Zhang H, Yan L. Solasonine relieves sevoflurane-induced neurotoxicity via activating the AMP-activated protein kinase/FoxO3a pathway. Hum Exp Toxicol 2022; 41:9603271211069984. [PMID: 35350913 DOI: 10.1177/09603271211069984] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Solasonine (SS), the main active ingredient of Solanumnigrum L, has been reported to boast extensive anti-tumor, anti-oxidant, and anti-inflammatory properties. This study is committed to exploring whether solasonine can alleviate neurotoxicity resulting from sevoflurane. MATERIALS AND METHODS The mouse hippocampal neuron cell line HT22 was treated with sevoflurane and/or solasonine of different doses. The proliferation, inflammation, oxidative stress response, and apoptosis of HT22 cells were examined. The AMP-activated protein kinase (AMPK)/FoxO3a signaling pathway was ascertained through Western blot and cellular immunofluorescence. In in-vivo experiments, Morris water maze figured out the changes in learning and memory abilities of mice treated with 8 mg/kg solasonine and exposed to SEV. RESULTS Sevoflurane induced apoptosis and hampered proliferation in HT22 cells. It also aggravated the release of inflammatory factors and oxidative stress mediators. Solasonine weakened neuron damage mediated by sevoflurane in a concentration-dependent pattern. Mechanically, sevoflurane clogged AMPK/FoxO3a signaling pathway activation, which was strengthened by solasonine. AMPK inhibition greatly influenced solasonine's protective effect on HT22 cells. Invivo, solasonine prominently ameliorated learning and memory disorders and nerve damage in mice exposed to sevoflurane. CONCLUSIONS Solasonine alleviates sevoflurane-induced neurotoxicity through activating the AMPK/FoxO3a signaling pathway.
Collapse
Affiliation(s)
- Huifang Zhang
- Department of Anesthesiology, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Lei Yan
- Department of Anesthesiology, 159426People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
7
|
Evaluation of anti-cancer potency of silibinin on murine renal carcinoma RenCa cells in an animal model with an intact immune system. Anticancer Drugs 2021; 31:785-791. [PMID: 32209841 DOI: 10.1097/cad.0000000000000931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Silibinin is a flavonoid extract isolated from milk thistle and has been proved to be a promising chemotherapeutic drug for cancer. However, most of those studies were performed on the human cancer cells, where the effects of silibinin could only be observed on an animal model with a deficient immune system. RenCa cells were isolated from a murine spontaneous renal cell carcinoma, which resembles many features of human renal cell carcinoma, and have been used to establish animal models with a sound immune response. Herein, we investigated the anti-cancer effects of silibinin on RenCa cells, revealing that it inhibited cell viability in both dose- and time-dependent manners. Silibinin slightly triggered apoptosis and significantly induced G2-M cell cycle arrest by downregulating cyclin B1 and CDK1 and increasing expression of p21. Furthermore, silibinin significantly inhibited the growth of RenCa cell xenografts in vivo. In addition, we found that silibinin reduced programmed cell death 1 ligand 1 expression of RenCa cells in vivo and in vitro. Our findings demonstrate that silibinin can inhibit the growth of mouse tumor cells in an animal model with an intact immune system, and silibinin may decrease the immunosuppression effect of tumor cells. Our results provide new evidence for evaluation of Silibinin application in cancer therapy.
Collapse
|
8
|
Haddadi R, Shahidi Z, Eyvari-Brooshghalan S. Silymarin and neurodegenerative diseases: Therapeutic potential and basic molecular mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 79:153320. [PMID: 32920285 DOI: 10.1016/j.phymed.2020.153320] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/20/2020] [Accepted: 07/01/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Neurodegenerative diseases (NDDs) are primarily characterized by selective neuronal loss in the brain. Alzheimer's disease as the most common NDDs and the most prevalent cause of dementia is characterized by Amyloid-beta deposition, which leads to cognitive and memory impairment. Parkinson's disease is a progressive neurodegenerative disease characterized by the dramatic death of dopaminergic neuronal cells, especially in the SNc and caused alpha-synuclein accumulation in the neurons. Silymarin, an extract from seeds of Silybum marianum, administered mostly for liver disorders and also had anti-oxidant and anti-carcinogenic activities. PURPOSE The present comprehensive review summarizes the beneficial effects of Silymarin in-vivo and in-vitro and even in animal models for these NDDs. METHODS A diagram model for systematic review is utilized for this search. The research is conducted in the following databases: PubMed, Web of Science, Scopus, and Science Direct. RESULTS Based on the inclusion criteria, 83 studies were selected and discussed in this review. CONCLUSION Lastly, we review the latest experimental evidences supporting the potential effects of Silymarin, as a neuroprotective agent in NDDs.
Collapse
Affiliation(s)
- Rasool Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal plant and natural products Research Center, Hamadan University of Medical Sciences, Hamadan 6517838678, Iran.
| | - Zahra Shahidi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahla Eyvari-Brooshghalan
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
9
|
Barialai L, Strecker MI, Luger AL, Jäger M, Bruns I, Sittig ACM, Mildenberger IC, Heller SM, Delaidelli A, Lorenz NI, Voss M, Ronellenfitsch MW, Steinbach JP, Burger MC. AMPK activation protects astrocytes from hypoxia‑induced cell death. Int J Mol Med 2020; 45:1385-1396. [PMID: 32323755 PMCID: PMC7138264 DOI: 10.3892/ijmm.2020.4528] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 02/10/2020] [Indexed: 01/20/2023] Open
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a major cellular energy sensor that is activated by an increase in the AMP/adenosine triphosphate (ATP) ratio. This causes the initiation of adaptive cellular programs, leading to the inhibition of anabolic pathways and increasing ATP synthesis. AMPK indirectly inhibits mammalian target of rapamycin (mTOR) complex 1 (mTORC1), a serine/threonine kinase and central regulator of cell growth and metabolism, which integrates various growth inhibitory signals, such as the depletion of glucose, amino acids, ATP and oxygen. While neuroprotective approaches by definition focus on neurons, that are more sensitive under cell stress conditions, astrocytes play an important role in the cerebral energy homeostasis during ischemia. Therefore, the protection of astrocytic cells or other glial cells may contribute to the preservation of neuronal integrity and function. In the present study, it was thus hypothesized that a preventive induction of energy deprivation-activated signaling pathways via AMPK may protect astrocytes from hypoxia and glucose deprivation. Hypoxia-induced cell death was measured in a paradigm of hypoxia and partial glucose deprivation in vitro in the immortalized human astrocytic cell line SVG. Both the glycolysis inhibitor 2-deoxy-d-glucose (2DG) and the AMPK activator A-769662 induced the phosphorylation of AMPK, resulting in mTORC1 inhibition, as evidenced by a decrease in the phosphorylation of the target ribosomal protein S6 (RPS6). Treatment with both 2DG and A-769662 also decreased glucose consumption and lactate production. Furthermore, A-769662, but not 2DG induced an increase in oxygen consumption, possibly indicating a more efficient glucose utilization through oxidative phosphorylation. Hypoxia-induced cell death was profoundly reduced by treatment with 2DG or A-769662. On the whole, the findings of the present study demonstrate, that AMPK activation via 2DG or A-769662 protects astrocytes under hypoxic and glucose-depleted conditions.
Collapse
Affiliation(s)
- Leli Barialai
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Maja I Strecker
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Anna-Luisa Luger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Manuel Jäger
- Department of Dermatology, Venerology and Allergology, University Hospital Frankfurt, Goethe University, D-60590 Frankfurt am Main
| | - Ines Bruns
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Alina C M Sittig
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Iris C Mildenberger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Sonja M Heller
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Alberto Delaidelli
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Nadja I Lorenz
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Martin Voss
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Michael W Ronellenfitsch
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| | - Michael C Burger
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, D-60528 Frankfurt am Main, Germany
| |
Collapse
|
10
|
Li J, Wang J, Liu W, Hayashi T, Itoh K, Onodera S, Ikejima T. Metformin protects Escherichia coli from bleomycin-induced bactericide via enhanced generation of hydrogen peroxide. Free Radic Res 2020; 54:64-75. [PMID: 31905044 DOI: 10.1080/10715762.2019.1703968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Bleomycin is a glycopeptide antibiotic that is widely employed in the therapy of a range of lymphomas and germ cell tumours. But the therapeutic efficacy of bleomycin is limited by development of lung fibrosis. The cytotoxicity of bleomycin is mostly ascribed to mitochondrial DNA (mtDNA) damage, while a protective effect of metformin against bleomycin-induced lung fibrosis results from the inhibition of mitochondrial complex I. Since mitochondria and bacteria have certain similarities in structure and function, we used Escherichia coli for simplification in the present work to investigate the relationship between metformin and bleomycin with apparently opposite effects on mitochondrial DNA damage. Bleomycin lethality to E. coli was ameliorated by metformin treatment accompanying further increase of the level of reactive oxygen species. Catalase but not superoxide dismutases attenuated the protective effect of metformin. Meanwhile, treatment with hydrogen peroxide enhanced the protection, indicating that metformin may protect E. coli from bleomycin-induced bactericide via enhanced generation of hydrogen peroxide. Moreover, silibinin, a hepatoprotective polyphenolic flavonoid attenuates the cytotoxicity of bleomycin to E. coli via enhanced generation of hydrogen peroxide as well. This bacterial model in place of mitochondria can provide us with easier screening for the molecules with capability of reducing the bleomycin side effect.
Collapse
Affiliation(s)
- Jian Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Jiaojiao Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China.,Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Hachioji, Japan
| | | | | | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, P. R. China.,Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang, P. R. China
| |
Collapse
|
11
|
Young Park S, Jin Kim Y, Park G, Kim HH. Neuroprotective effect of Dictyopteris divaricata extract-capped gold nanoparticles against oxygen and glucose deprivation/reoxygenation. Colloids Surf B Biointerfaces 2019; 179:421-428. [PMID: 31003168 DOI: 10.1016/j.colsurfb.2019.03.066] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
Abstract
Combination therapy remains a promising approach to ameliorate cerebral ischemia injury. Nevertheless, the primary mechanism of the neuroprotective properties of Dictyopteris divaricata extract-capped gold nanoparticles (DD-GNPs) is not completely understood. DD-GNPs displayed maximum absorption at 525 nm and a diameter of 62.6 ± 1.2 nm, with a zeta potential value of -26.1 ± 0.6 mV. High resolution-transmission electron microscopy confirmed the spherical shape and average diameter (28.01 ± 2.03 nm). Crystalline structure and gold nanoparticle synthesis of DD-GNPs were determined by X-ray powder diffraction, and the presence of elemental gold was confirmed by energy-dispersive X-ray spectroscopy and Fourier transform-infrared spectroscopy. We examined the neuroprotective properties of DD-GNPs and explored their potential mechanisms in human SH-SY5Y neuroblastoma cells treated with oxygen and glucose deprivation/reoxygenation (OGD/R). We found that DD-GNPs inhibited OGD/R-induced release of lactate dehydrogenase (LDH), loss of cell viability, and production of reactive oxygen species. This neuroprotection was accompanied by regulation of apoptosis-related proteins, as indicated by decreased levels of cleaved-caspase-3, cleaved-PARP, cleaved-caspase-9, p53, p21, and Bax, as well as an increased level of Bcl-2. Notably, the neuroprotective effects of DD-GNPs were partially abolished by HO-1, NQO1, Nrf2, and AMPK knockdown. Our results established that DD-GNPs effectively attenuated OGD/R-stimulated neuronal injury, as evidenced by reduced neuronal injury. Even though the accumulating evidence has indicated the low toxicity and minimal side effects of GNPs, experimental clinical trials of DD-GNPs are still limited because of the lack of knowledge regarding the effects of DD-GNPs as neuroprotective agents against neurodegenerative diseases.
Collapse
Affiliation(s)
- Sun Young Park
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Republic of Korea
| | - Yeong Jin Kim
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Geuntae Park
- Department of Nanomaterials Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Hyung-Hoi Kim
- Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea; Department of Laboratory Medicine, Pusan National University Hospital, Busan 49241, Republic of Korea
| |
Collapse
|
12
|
Silibinin Alleviates the Learning and Memory Defects in Overtrained Rats Accompanying Reduced Neuronal Apoptosis and Senescence. Neurochem Res 2019; 44:1818-1829. [PMID: 31102026 DOI: 10.1007/s11064-019-02816-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 12/21/2022]
Abstract
Excessive physical exercise (overtraining; OT) increases oxidative stress and induces damage in multiple organs including the brain, especially the hippocampus that plays an important role in learning and memory. Silibinin, a natural flavonoid derived from milk thistle of Silybum marianum, has been reported to exert neuroprotective effect. In this study, rats were subjected to overtraining exercise, and the protective effects of silibinin were investigated in these models. Morris water maze and novel object recognition tests showed that silibinin significantly attenuated memory defects in overtrained rats. At the same time, the results of Nissl, TUNEL and SA-β-gal staining showed that silibinin reversed neuronal loss caused by apoptosis, and delayed cell senescence of the hippocampus in the overtrained rats, respectively. In addition, silibinin decreased malondialdehyde (MDA) levels which is associated with reactive oxygen species (ROS) generation. Silibinin prevented impairment of learning and memory caused by excessive physical exercise in rats, accompanied by reduced apoptosis and senescence in hippocampus cells.
Collapse
|
13
|
Ahlawat J, Deemer EM, Narayan M. Chitosan Nanoparticles Rescue Rotenone-Mediated Cell Death. MATERIALS 2019; 12:ma12071176. [PMID: 30978909 PMCID: PMC6480189 DOI: 10.3390/ma12071176] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 12/20/2022]
Abstract
The aim of the present investigation was to study the anti-oxidant effect of chitosan nanoparticles on a human SH-SY5Y neuroblastoma cell line using a rotenone model to generate reactive oxygen species. Chitosan nanoparticles were synthesized using an ionotropic gelation method. The obtained nanoparticles were characterized using various analytical techniques such as Dynamic Light Scattering, Scanning Electron Microscopy, Transmission Electron Microscopy, Fourier Transmission Infrared spectroscopy and Atomic Force Microscopy. Incubation of SH-SY5Y cells with 50 µM rotenone resulted in 35-50% cell death within 24 h of incubation time. Annexin V/Propidium iodide dual staining verified that the majority of neuronal cell death occurred via the apoptotic pathway. The incubation of cells with chitosan nanoparticles reduced rotenone-initiated cytotoxicity and apoptotic cell death. Given that rotenone insult to cells causes oxidative stress, our results suggest that Chitosan nanoparticles have antioxidant and anti-apoptotic properties. Chitosan can not only serve as a novel therapeutic drug in the near future but also as a carrier for combo-therapy.
Collapse
Affiliation(s)
- Jyoti Ahlawat
- Department of Chemistry & Biochemistry, The University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Eva M Deemer
- Material Science & Engineering department, The University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Mahesh Narayan
- Department of Chemistry & Biochemistry, The University of Texas at El Paso, El Paso, TX 79968, USA.
| |
Collapse
|
14
|
Sharifpanah F, Ali EH, Wartenberg M, Sauer H. The milk thistle (Silybum marianum) compound Silibinin stimulates leukopoiesis from mouse embryonic stem cells. Phytother Res 2019; 33:452-460. [PMID: 30548344 DOI: 10.1002/ptr.6241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 01/02/2023]
Abstract
The milk thistle compound Silibinin (i.e., a 1:1 mixture of Silybin A and Silybin B) stimulates vasculogenesis of mouse embryonic stem (ES) cells. Because vasculogenesis and leukopoiesis are interrelated, the effect of Silibinin on leukopoiesis of ES cells was investigated. Treatment of differentiating ES cells with hydrosoluble Silibinin-C-2',3-dihydrogen succinate dose-dependent increased the number of CD18+ , CD45+ , and CD68+ cells, indicating leukocyte/macrophage differentiation. Silibinin treatment activated phosphoinositide 3-kinase (PI3K), AKT (protein kinase B), signal transducer and activator of transcription 3 (STAT3), stimulated hypoxia-induced factor-1α (HIF-1α), and vascular endothelial growth factor receptor 2 (VEGFR2) expression and raised intracellular nitric oxide (NO). Western blot experiments showed that upon coincubation with either the PI3K inhibitor LY294002, the STAT3 inhibitor Stattic, the AKT antagonist AKT inhibitor VIII, or the NO inhibitor L-NAME, the Silibinin-induced expression of CD18, CD45, and CD68 was abolished. Moreover, the stimulation of HIF-1α and VEGFR2 expression was blunted upon STAT3 and PI3K/AKT inhibition. Treatment of differentiating ES cells with L-NAME abolished the stimulation of VEGFR2 and VE-cadherin expression achieved with Silibinin, indicating that NO is involved in vasculogenesis and leukocyte differentiation pathways. In summary, the data of the present study demonstrate that Silibinin stimulates leukocyte differentiation of ES cells, which is associated to vasculogenesis and regulated by PI3K/AKT-, STAT3-, and NO-mediated signaling.
Collapse
Affiliation(s)
- Fatemeh Sharifpanah
- Department of Physiology, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Enas Hussein Ali
- Department of Physiology, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Maria Wartenberg
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
15
|
Shiri S, Abbasi N, Alizadeh K, Karimi E. Novel and green synthesis of a nanopolymer and its use as a drug delivery system of silibinin and silymarin extracts in the olfactory ensheathing cells of rats in normal and high-glucose conditions. RSC Adv 2019; 9:38912-38927. [PMID: 35540667 PMCID: PMC9075964 DOI: 10.1039/c9ra05608d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/27/2019] [Indexed: 01/03/2023] Open
Abstract
Drug delivery systems have been of interest to researchers. The effects of synthesized nano-polymers as silibinin and silymarin extract drug delivery systems on olfactory ensheathing cells under normal and high-glucose conditions were studied. The structure of the nanopolymer was characterized by IR, HNMR, GPC, DLS, and AFM. The toxicity was evaluated by an MTT assay. The production of ROS and the generation of NO were evaluated by a probe of fluorescein diacetate and Griess methods, respectively. The expressions of the protein levels of ILK, VEGF, BDNF, and NGF were investigated by western blotting. The polymer size was between 50 and 150 nm. The loading capacities for silibinin and silymarin were 68.5% and 56.4%, respectively, and the drug release for them was estimated at 54.1% and 50.8%, respectively. In high-glucose conditions, the cells were protected (EC50 = 4.88 ± 0.5 μM) by silibinin and nanopolymer in low concentrations by reducing the amount of ROS and NO, maintaining ILK, reducing VEGF and increasing NGF and BDNF. Incubation with silibinin and nanopolymer at high concentrations increased cell death with LC50 = 57.36 ± 2.5 and 43.18 ± 1.8 μM, respectively, in high-glucose states. Thus, the cells were protected by silibinin and nanopolymer in protective concentrations by reducing the amount of ROS and NO, maintaining ILK, reducing VEGF, and increasing BDNF and NGF. The mentioned mechanisms were totally reversed at high concentrations. A schematic of a new synthesized nanopolymer (CGONP) and its use as a drug delivery system of silibinin and silymarin extract in the olfactory ensheathing cells (OECs) of rats in normal and high-glucose conditions.![]()
Collapse
Affiliation(s)
- Sabah Shiri
- Department of Chemistry
- Lorestan University
- Khorramabad
- Iran
- Biotechnology and Medicinal Plants Research Center
| | - Naser Abbasi
- Biotechnology and Medicinal Plants Research Center
- Ilam University of Medical Sciences
- Ilam
- Iran
- Department of Pharmacology
| | - Kamal Alizadeh
- Department of Chemistry
- Lorestan University
- Khorramabad
- Iran
| | - Elahe Karimi
- Biotechnology and Medicinal Plants Research Center
- Ilam University of Medical Sciences
- Ilam
- Iran
| |
Collapse
|
16
|
Low Molecular Weight Sulfated Chitosan: Neuroprotective Effect on Rotenone-Induced In Vitro Parkinson’s Disease. Neurotox Res 2018; 35:505-515. [DOI: 10.1007/s12640-018-9978-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/25/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022]
|
17
|
Li XX, Jiang ZH, Zhou B, Chen C, Zhang XY. Hepatoprotective effect of gastrodin against alcohol-induced liver injury in mice. J Physiol Biochem 2018; 75:29-37. [PMID: 30242628 DOI: 10.1007/s13105-018-0647-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 09/02/2018] [Indexed: 12/18/2022]
Abstract
Alcoholic liver disease (ALD) is a common and serious threat to human health worldwide. In this study, the hepatoprotective effect of gastrodin against alcohol-induced liver injury in mice was examined. Mice with alcohol-induced hepatotoxicity were treated intragastrically with gastrodin (50, 80, or 100 mg/kg). The mice treated with gastrodin experienced better outcomes than those who received only one dose of alcohol (50%, 10 mL/kg b.w.). Gastrodin treatment reduced the activities of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST), decreased hepatic malondialdehyde (MDA) content, and increased hepatic superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) activities in a dose-dependent manner. Gastrodin also alleviated histopathological changes induced by alcohol. Gastrodin protected against alcohol-induced increases in expression levels of the cytochrome P450 2E1 (CYP2E1) and mRNA levels of chemokine (C-X-C motif) ligand 1 (CXCL-1), interferon-γ (IFN-γ), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), vascular cell adhesion molecule 1 (VCAM-1), nuclear factor-kappa B (NF-κB), Toll-like receptor 4 (TLR-4), and activator of transcription 3 (STAT-3). Moreover, gastrodin-increased nuclear transcription factor 2 (Nrf2) translocates to the nucleus and enhanced the activity of anti-oxidant enzymes, and could thereby ameliorate alcohol-induced liver injury in mice. This study demonstrated that gastrodin may be an effective therapeutic agent against alcohol-induced liver injury.
Collapse
Affiliation(s)
- Xin-Xin Li
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qingling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, East on the 1st Ring Road, Hanzhong, 723000, Shaanxi Province, China
| | - Zhi-Hui Jiang
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qingling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, East on the 1st Ring Road, Hanzhong, 723000, Shaanxi Province, China
| | - Bo Zhou
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qingling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, East on the 1st Ring Road, Hanzhong, 723000, Shaanxi Province, China
| | - Chen Chen
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qingling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, East on the 1st Ring Road, Hanzhong, 723000, Shaanxi Province, China.
| | - Xiao-Ying Zhang
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qingling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, East on the 1st Ring Road, Hanzhong, 723000, Shaanxi Province, China. .,College of Veterinary Medicine, Northwest A&F University (North Campus), Xinong Rd. 22, Post Box 19, Yangling, 712100, Shaanxi Province, China. .,Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
18
|
Park SY, Choi YW, Park G. Nrf2-mediated neuroprotection against oxygen-glucose deprivation/reperfusion injury by emodin via AMPK-dependent inhibition of GSK-3β. J Pharm Pharmacol 2018; 70:525-535. [DOI: 10.1111/jphp.12885] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/03/2018] [Indexed: 01/10/2023]
Abstract
Abstract
Objectives
Our study verified the neuroprotective properties of emodin against oxygen-glucose deprivation/reoxygenation (OGD/R) and demonstrated its mechanism.
Methods
Human neuronal SH-SY5Y cells were investigated by analysing cell viability, lactate dehydrogenase levels, expression of molecules related to apoptotic cell death, and using biochemical techniques, flow cytometry and Western blot assays.
Key findings
Emodin reduced OGD/R-lead to neurotoxicity in SH-SY5Y cells. OGD/R significantly increased levels of cleaved poly ADP ribose polymerase, cleaved caspase-3, cleaved caspase-9, p53, p21 and Bax protein. However, emodin treatment effectively inhibited these OGD/R-induced changes. Emodin treatment also increased HO-1 and NQO1 expression in a concentration- and time-dependent manner and caused antioxidant response element (ARE) transcription activity and nuclear Nrf2 accumulation. Emodin phosphorylated AMPK and GSK3β, and pretreatment of cells with an AMPK inhibitor suppressed emodin-induced nuclear Nrf2 accumulation and HO-1 and NQO1 expression. AMPK inhibitor treatment decreased GSK3β phosphorylation, suggesting that AMPK is upstream of GSK3β, Nrf2, HO-1 and NQO1. Emodin's neuroprotective effect was completely blocked by HO-1, NQO1 and Nrf2 knock-down and an AMPK inhibitor, indicating the action of AMPK/GSK3β/Nrf2/ARE in the neuroprotective effect of emodin subjected to OGD/R.
Conclusions
Emodin treatment protected against OGD/R-lead to neurotoxicity by potentiating Nrf2/ARE-regulated neuroprotection through the AMPK/GSK3β pathway, indicating that emodin may be useful for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Sun Young Park
- Bio-IT Fusion Technology Research Institute, Pusan National University, Busan, Korea
| | - Young-Whan Choi
- Department of Horticultural Bioscience, Pusan National University, Miryang, Korea
| | - Geuntae Park
- Department of Nanomaterials Engineering, Pusan National University, Busan, Korea
| |
Collapse
|
19
|
A Novel Phenylchromane Derivative Increases the Rate of Glucose Uptake in L6 Myotubes and Augments Insulin Secretion from Pancreatic Beta-Cells by Activating AMPK. Pharm Res 2017; 34:2873-2890. [DOI: 10.1007/s11095-017-2271-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/24/2017] [Indexed: 01/04/2023]
|
20
|
Antioxidative and Anti-Apoptotic Roles of Silibinin in Reversing Learning and Memory Deficits in APP/PS1 Mice. Neurochem Res 2017; 42:3439-3445. [DOI: 10.1007/s11064-017-2389-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 08/08/2017] [Accepted: 08/16/2017] [Indexed: 11/27/2022]
|
21
|
Ran QQ, Chen HL, Liu YL, Yu HX, Shi F, Wang MS. Electroacupuncture preconditioning attenuates ischemic brain injury by activation of the adenosine monophosphate-activated protein kinase signaling pathway. Neural Regen Res 2015; 10:1069-75. [PMID: 26330828 PMCID: PMC4541236 DOI: 10.4103/1673-5374.160095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2015] [Indexed: 01/16/2023] Open
Abstract
Electroacupuncture has therapeutic effects on ischemic brain injury, but its mechanism is still poorly understood. In this study, mice were stimulated by electroacupuncture at the Baihui (GV20) acupoint for 30 minutes at 1 mA and 2/15 Hz for 5 consecutive days. A cerebral ischemia model was established by ligating the bilateral common carotid artery for 15 minutes. At 72 hours after injury, neuronal injury in the mouse hippocampus had lessened, and the number of terminal deoxynucleotide transferase-mediated dUTP nick-end labeling-positive cells reduced after electroacupuncture treatment. Moreover, expression of adenosine monophosphate-activated protein kinase α (AMPKα) and phosphorylated AMPKα was up-regulated. Intraperitoneal injection of the AMPK antagonist, compound C, suppressed this phenomenon. Our findings suggest that electroacupuncture preconditioning alleviates ischemic brain injury via AMPK activation.
Collapse
Affiliation(s)
- Qiang-Qiang Ran
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Huai-Long Chen
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Yan-Li Liu
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Hai-Xia Yu
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Fei Shi
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Ming-Shan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
22
|
Cross-validation of item selection and scoring for the SF-12 Health Survey in nine countries: results from the IQOLA Project. International Quality of Life Assessment. PPAR Res 1998; 2016:9282087. [PMID: 27051413 PMCID: PMC4802016 DOI: 10.1155/2016/9282087] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/11/2016] [Indexed: 02/08/2023] Open
Abstract
Activated AMPK protects the heart from cardiac ischemia-reperfusion (IR) injury and is associated with inhibition of mitochondrial permeability transition pore (PTP) opening. On the other hand, pharmacological inhibition of the PTP reduces infarct size and improves cardiac function. However, it is unclear whether beneficial effects of AMPK are mediated through the PTP and, if they are not, whether simultaneous activation of AMPK and inhibition of the PTP exert synergistic protective effects against cardiac IR injury. Here, we examined the effects of the AMPK activator, A-769662 in combination with the PTP inhibitor, sanglifehrin A (SfA) on in vivo cardiac IR. Cardiac dysfunction following IR injury was associated with decreased activity of the mitochondrial electron transport chain (ETC) and increased mitochondrial ROS and PTP opening. Administration of A-769662 or SfA individually upon reperfusion improved cardiac function, reduced infarction size, and inhibited ROS production and PTP opening. However, simultaneous administration of SfA and A-769662 did not provide synergistic improvement of postischemic recovery of cardiac and mitochondrial function, though both compounds disrupted IR-induced interaction between PPARα and CyP-D. In conclusion, A-769662 or SfA prevents PPARα interaction with CyP-D, improving cardiac outcomes and increasing mitochondrial function, and simultaneous administration of the drugs does not provide synergistic effects.
Collapse
|