101
|
Shi J, Sun Y, Hua J. Functional Genetic Variation in the 3'-UTRNTRK2 is Associated with Risk of Ischemic Stroke. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:577-584. [PMID: 33209049 PMCID: PMC7669521 DOI: 10.2147/pgpm.s270319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/30/2020] [Indexed: 12/05/2022]
Abstract
Background Stroke is a leading cause of death and disability worldwide. It remains difficult to treat brain injury and improve functional rehabilitation after cerebral ischemia. Brain-derived neurotrophic factor (BDNF) is involved in ischemic stroke (IS) through interactions in the CREB1-BDNF-NTRk2 pathway. In this study, we aimed to determine the association of NTRK2 gene polymorphisms and the effects of intergenetic interactions in the Chinese population. Materials and Methods A total of 400 patients diagnosed with IS and 400 healthy controls were enrolled for genotyping. Detailed sequence-based analysis was predicted through bioinformatical investigation. Polymorphisms associated with miRNA were analyzed by a dual-luciferase reporter assay system. Results Analysis of clinical characteristics revealed that IS was highly associated with exposure to cigarette smoking, alcohol intake, as well as metabolic diseases, such as diabetes, hypertension, and higher serum triglyceride concentration. Three polymorphisms in NTRK2 located in the 3ʹ-untranslated region (3ʹ-UTR) were genotyped. Logistic regression analysis showed that IS patients with rs11140793, rs7047042, and rs1221 polymorphisms had a higher risk of stroke and indicated a worse short-term recovery. The mRNA level of NTRK2 was suppressed in a mutant genotype compared with wild genotype. The suppression of NTRK2 was induced by the gain-of-binding ability of certain miRNAs through the direct binding of 3ʹ-UTR. Conclusion Our research indicated that, by influencing the expression of NTRK2, the SNPs rs11140793, rs7047042, and rs1221 in the 3′UTR of NTRK2 can be used as risk factors for IS patients.
Collapse
Affiliation(s)
- Jiajia Shi
- Department of Rehabilitation Medicine, Kunshan Rehabilitation Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Ying Sun
- Department of Rehabilitation Medicine, Kunshan Rehabilitation Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Jiajia Hua
- Department of Rehabilitation Medicine, The Sixth People's Hospital of Nantong, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
102
|
Functional validation of a human GLUD2 variant in a murine model of Parkinson's disease. Cell Death Dis 2020; 11:897. [PMID: 33093440 PMCID: PMC7582183 DOI: 10.1038/s41419-020-03043-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by Lewy body formation and progressive dopaminergic neuron death in the substantia nigra (SN). Genetic susceptibility is a strong risk factor for PD. Previously, a rare gain-of-function variant of GLUD2 glutamate dehydrogenase (T1492G) was reported to be associated with early onset in male PD patients; however, the function and underlying mechanism of this variant remains elusive. In the present study, we generated adeno-associated virus expressing GLUD2 and its mutant under the control of the glial fibrillary acidic protein promotor and injected the virus into the SN pars compacta of either untreated mice or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model mice. Our results demonstrate that GLUD2 mutation in MPTP-induced PD mice exacerbates movement deficits and nigral dopaminergic neuron death and reduces glutamate transporters expression and function. Using GC-Q-TOF/MS-based metabolomics, we determined that GLUD2 mutation damages mitochondrial function by decreasing succinate dehydrogenase activity to impede the tricarboxylic acid cycle in the SN of MPTP-induced PD mice. Accordingly, GLUD2 mutant mice had reduced energy metabolism and increased apoptosis, possibly due to downregulation of brain-derived neurotrophic factor/nuclear factor E2-related factor 2 signaling in in vitro and in vivo PD models. Collectively, our findings verify the function of GLUD2 in PD and unravel a mechanism by which a genetic variant in human GLUD2 may contribute to disease onset.
Collapse
|
103
|
Gharbi T, Zhang Z, Yang GY. The Function of Astrocyte Mediated Extracellular Vesicles in Central Nervous System Diseases. Front Cell Dev Biol 2020; 8:568889. [PMID: 33178687 PMCID: PMC7593543 DOI: 10.3389/fcell.2020.568889] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Astrocyte activation plays an important role during disease-induced inflammatory response in the brain. Exosomes in the brain could be released from bone marrow (BM)-derived stem cells, neuro stem cells (NSC), mesenchymal stem cells (MSC), etc. We summarized that exosomes release and transport signaling to the target cells, and then produce function. Furthermore, we discussed the pathological interactions between astrocytes and other brain cells, which are related to brain diseases such as stroke, Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) disease, multiple sclerosis (MS), psychiatric, traumatic brain injury (TBI), etc. We provide up-to-date, comprehensive and valuable information on the involvement of exosomes in brain diseases, which is beneficial for basic researchers and clinical physicians.
Collapse
Affiliation(s)
- Tahereh Gharbi
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
104
|
The Application of Ferroptosis in Diseases. Pharmacol Res 2020; 159:104919. [DOI: 10.1016/j.phrs.2020.104919] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/30/2020] [Accepted: 05/08/2020] [Indexed: 01/17/2023]
|
105
|
Wei YZ, Zhu GF, Zheng CQ, Li JJ, Sheng S, Li DD, Wang GQ, Zhang F. Ellagic acid protects dopamine neurons from rotenone-induced neurotoxicity via activation of Nrf2 signalling. J Cell Mol Med 2020; 24:9446-9456. [PMID: 32657027 PMCID: PMC7417702 DOI: 10.1111/jcmm.15616] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/29/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent central nervous system (CNS) degenerative disease. Oxidative stress is one of key contributors to PD. Nuclear factor erythroid-2-related factor 2 (Nrf2) is considered to be a master regulator of many genes involved in anti-oxidant stress to attenuate cell death. Therefore, activation of Nrf2 signalling provides an effective avenue to treat PD. Ellagic acid (EA), a natural polyphenolic contained in fruits and nuts, possesses amounts of pharmacological activities, such as anti-oxidant stress and anti-inflammation. Recent studies have confirmed EA could be used as a neuroprotective agent in neurodegenerative diseases. Here, mice subcutaneous injection of rotenone (ROT)-induced DA neuronal damage was performed to investigate EA-mediated neuroprotection. In addition, adult Nrf2 knockout mice and different cell cultures including MN9D-enciched, MN9D-BV-2 and MN9D-C6 cell co-cultures were applied to explore the underlying mechanisms. Results demonstrated EA conferred neuroprotection against ROT-induced DA neurotoxicity. Activation of Nrf2 signalling was involved in EA-mediated DA neuroprotection, as evidenced by the following observations. First, EA activated Nrf2 signalling in ROT-induced DA neuronal damage. Second, EA generated neuroprotection with the presence of astroglia and silence of Nrf2 in astroglia abolished EA-mediated neuroprotection. Third, EA failed to produce DA neuroprotection in Nrf2 knockout mice. In conclusion, this study identified EA protected against DA neuronal loss via an Nrf2-dependent manner.
Collapse
Affiliation(s)
- Yi-Zheng Wei
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Guo-Fu Zhu
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Chang-Qing Zheng
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jing-Jie Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Shuo Sheng
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Dai-di Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Guo-Qing Wang
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
106
|
Wang G, Yang Y, Wang C, Huang J, Wang X, Liu Y, Wang H. Exploring the role and mechanisms of diallyl trisulfide and diallyl disulfide in chronic constriction-induced neuropathic pain in rats. Korean J Pain 2020; 33:216-225. [PMID: 32606266 PMCID: PMC7336342 DOI: 10.3344/kjp.2020.33.3.216] [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: 01/28/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 01/07/2023] Open
Abstract
Background Garlic oil is a rich source of organosulfur compounds including diallyl disulfide and diallyl trisulfide. There have been studies showing the neuroprotective actions of these organosulfur compounds. However, the potential of these organosulfur compounds in neuropathic pain has not been explored. The present study was aimed at investigating the pain attenuating potential of diallyl disulfide and diallyl trisulfide in chronic constriction injury (CCI)-induced neuropathic pain in rats. The study also explored their pain-attenuating mechanisms through modulation of H2S, brain-derived neurotrophin factor (BDNF) and nuclear factor erythroid 2-related factor 2 (Nrf2). Methods The rats were subjected to CCI injury by ligating the sciatic nerve in four places. The development of neuropathic pain was measured by assessing mechanical hyperalgesia (Randall–Selittotest), mechanical allodynia (Von Frey test), and cold allodynia (acetone drop test) on 14th day after surgery. Results Administration of diallyl disulfide (25 and 50 mg/kg) and diallyl trisulfide (20 and 40 mg/kg) for 14 days led to a significant reduction in pain in CCI-subjected rats. Moreover, treatment with these organosulfur compounds led to the restoration of H2S, BDNF and Nrf2 levels in the sciatic nerve and dorsal root ganglia. Co-administration of ANA-12 (BDNF blocker) abolished pain attenuating actions as well as BDNF and the Nrf2 restorative actions of diallyl disulfide and diallyl trisulfide, without modulating H2S levels. Conclusions Diallyl disulfide and diallyl trisulfide have the potential to attenuate neuropathic pain in CCI-subjected rats possibly through activation of H2S-BDNF-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Gang Wang
- Department of Anesthesiology, Tianjin First Central Hospital, Tianjin, China
| | - Yan Yang
- Department of Anesthesiology, First People's Hospital, Wuhan, Hubei, China
| | - Chunfeng Wang
- Department of Anesthesiology, The Second People's Hospital of Kunshan City, Kunshan, Jiangsu, China
| | - Jianzhong Huang
- Department of Anesthesiology, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, Fujian, China
| | - Xiao Wang
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ying Liu
- Department of Anesthesiology, Qinghai Women and Children Hospital, Xining, Qinghai, China
| | - Hao Wang
- Department of Anesthesiology, LinFen City Central Hospital of Shanxi Province, Linfen, Shanxi, China
| |
Collapse
|
107
|
Lananna BV, Musiek ES. The wrinkling of time: Aging, inflammation, oxidative stress, and the circadian clock in neurodegeneration. Neurobiol Dis 2020; 139:104832. [PMID: 32179175 PMCID: PMC7727873 DOI: 10.1016/j.nbd.2020.104832] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 01/17/2023] Open
Abstract
A substantial body of research now implicates the circadian clock in the regulation of an array of diverse biological processes including glial function, metabolism, peripheral immune responses, and redox homeostasis. Sleep abnormalities and other forms of circadian disruption are common symptoms of aging and neurodegeneration. Circadian clock disruption may also influence the aging processes and the pathogenesis of neurodegenerative diseases. The specific mechanisms governing the interaction between circadian systems, aging, and the immune system are still being uncovered. Here, we review the evidence supporting a bidirectional relationship between aging and the circadian system. Further, we explore the hypothesis that age-related circadian deterioration may exacerbate multiple pathogenic processes, priming the brain for neurodegeneration.
Collapse
Affiliation(s)
- Brian V Lananna
- Dept. of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erik S Musiek
- Dept. of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
108
|
Chen J, Yang X, Fang X, Wang F, Min J. [The role of ferroptosis in chronic diseases]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020; 49:44-57. [PMID: 32621416 DOI: 10.3785/j.issn.1008-9292.2020.02.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, ferroptosis, an iron-dependent novel type of cell death, has been characterized as an excessive accumulation of lipid peroxides and reactive oxygen species. Emerging studies demonstrate that ferroptosis not only plays an important role in the pathogenesis and progression of chronic diseases, but also functions differently in the different disease context. Notably, it is shown that activation of ferroptosis could potently inhibit tumor growth and increase sensitivity to chemotherapy and immunotherapy in various cancer settings. As a result, the development of more efficacious ferroptosis agonists remains the mainstay of ferroptosis-targeting strategy for cancer therapeutics. By contrast, in non-cancerous chronic diseases, including cardiovascular & cerebrovascular diseases and neurodegenerative diseases, ferroptosis functions as a risk factor to promote these diseases progression through triggering or accelerating tissue injury. As a matter of fact, blocking ferroptosis has been demonstrated to effectively prevent ischemia-reperfusion heart disease in preclinical animal models. Therefore, it is a promising field to develope potent ferroptosis inhibitors for preventing and treating cardiovascular & cerebrovascular diseases and neurodegenerative diseases. In this article, we summarize the most recent progress on ferroptosis in chronic diseases, and draw attention to the possible clinical impact of this recently emerged ferroptosis modalities.
Collapse
Affiliation(s)
- Junyi Chen
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiang Yang
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuexian Fang
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
109
|
Woo Y, Lim JS, Oh J, Lee JS, Kim JS. Neuroprotective Effects of Euonymus alatus Extract on Scopolamine-Induced Memory Deficits in Mice. Antioxidants (Basel) 2020; 9:antiox9050449. [PMID: 32456069 PMCID: PMC7278771 DOI: 10.3390/antiox9050449] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/09/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Euonymus alatus is considered to elicit various beneficial effects against cancer, hyperglycemia, menstrual discomfort, diabetic complications, and detoxification. The young leaves of this plant are exploited as food and also utilized for traditional medicine in East Asian countries, including Korea and China. Our preliminary study demonstrated that ethanolic extract from the Euonymus alatus leaf (EAE) exhibited the strongest antioxidant enzyme-inducing activity among more than 100 kinds of edible tree leaf extracts. This study investigated whether EAE could attenuate the cognitive deficits caused by oxidative stress in mice. Oral intubation of EAE at 100 mg/kg bw or higher resulted in significant improvements to the memory and behavioral impairment induced via i.p. injection of scopolamine. Furthermore, EAE enhanced the expression levels of hippocampal neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor in mice, activated the Nrf2, and the downstream heme oxygenase-1 (HO-1) a quintessential antioxidant enzyme. As rutin (quercetin-3-O-rutinose) was abundantly present in EAE and free quercetin was able to induce defensive antioxidant enzymes in an Nrf2-dependent manner, our findings suggested that quercetin derived from rutin via the intestinal microflora played a significant role in the protection of the mouse hippocampus from scopolamine-induced damage through BDNF-mediated Nrf2 activation, thereby dampening cognitive decline.
Collapse
Affiliation(s)
- Yunju Woo
- School of Food Science and Biotechnology (BK21 plus) and Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea
| | - Ji Sun Lim
- School of Food Science and Biotechnology (BK21 plus) and Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea
| | - Jisun Oh
- School of Food Science and Biotechnology (BK21 plus) and Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea
| | - Jeong Soon Lee
- Forest Resources Development Institute of Gyeongsangbuk-do, Andong 36605, Korea
| | - Jong-Sang Kim
- School of Food Science and Biotechnology (BK21 plus) and Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
110
|
Shi L, Huang C, Luo Q, Xia Y, Liu W, Zeng W, Cheng A, Shi R, Zhengli C. Clioquinol improves motor and non-motor deficits in MPTP-induced monkey model of Parkinson's disease through AKT/mTOR pathway. Aging (Albany NY) 2020; 12:9515-9533. [PMID: 32424108 PMCID: PMC7288933 DOI: 10.18632/aging.103225] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/20/2020] [Indexed: 01/05/2023]
Abstract
Despite decades of research into the pathology mechanisms of Parkinson’s disease (PD), disease-modifying therapy of PD is scarce. Thus, searching for new drugs or more effective neurosurgical treatments has elicited much interest. Clioquinol (CQ) has been shown to have therapeutic benefits in rodent models of neurodegenerative disorders. However, it’s neuroprotective role and mechanisms in PD primate models and PD patients, especially in the advanced stages, are not fully understood. Furthermore, issues such as spontaneous recovery of motor function and high symptom variability in different monkeys after the same toxic protocol, has not been resolved before the present study. In this study, we designed a chronic and long-term progressive protocol to generate a stabilized PD monkey model showed with classic motor and non-motor deficits, followed by treatment analysis of CQ. We found that CQ could remarkably improve the motor and non-motor deficits, which were based on the reduction of iron content and ROS level in the SN and further improvement in pathology. Meanwhile, we also showed that ferroptosis was probably involved in the pathogenesis of PD. In addition, the study shows a positive effect of CQ on AKT/mTOR survival pathway and a blocking effect on p53 medicated cell death in vivo and in vitro.
Collapse
Affiliation(s)
- Liangqin Shi
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu Sichuan, China
| | - Chao Huang
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu Sichuan, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Qihui Luo
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu Sichuan, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Yu Xia
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu Sichuan, China
| | - Wentao Liu
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu Sichuan, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Wen Zeng
- Sichuan Primed Biological Technology Co., Ltd, National Experimental Macaque Reproduce Laboratory, Ya'an, Sichuan, China
| | - Anchun Cheng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Chen Zhengli
- Laboratory of Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu Sichuan, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, Sichuan, China
| |
Collapse
|
111
|
Hisaoka-Nakashima K, Azuma H, Ishikawa F, Nakamura Y, Wang D, Liu K, Wake H, Nishibori M, Nakata Y, Morioka N. Corticosterone Induces HMGB1 Release in Primary Cultured Rat Cortical Astrocytes: Involvement of Pannexin-1 and P2X7 Receptor-Dependent Mechanisms. Cells 2020; 9:cells9051068. [PMID: 32344830 PMCID: PMC7290518 DOI: 10.3390/cells9051068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
A major risk factor for major depressive disorder (MDD) is stress. Stress leads to the release of high-mobility group box-1 (HMGB1), which in turn leads to neuroinflammation, a potential pathophysiological basis of MDD. The mechanism underlying stress-induced HMGB1 release is not known, but stress-associated glucocorticoids could be involved. To test this, rat primary cultured cortical astrocytes, the most abundant cell type in the central nervous system (CNS), were treated with corticosterone and HMGB1 release was assessed by Western blotting and ELISA. Significant HMGB1 was released with treatment with either corticosterone or dexamethasone, a synthetic glucocorticoid. HMGB1 translocated from the nucleus to the cytoplasm following corticosterone treatment. HMGB1 release was significantly attenuated with glucocorticoid receptor blocking. In addition, inhibition of pannexin-1, and P2X7 receptors led to a significant decrease in corticosterone-induced HMGB1 release. Taken together, corticosterone stimulates astrocytic glucocorticoid receptors and triggers cytoplasmic translocation and extracellular release of nuclear HMGB1 through a mechanism involving pannexin-1 and P2X7 receptors. Thus, under conditions of stress, glucocorticoids induce astrocytic HMGB1 release, leading to a neuroinflammatory state that could mediate neurological disorders such as MDD.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan; (K.H.-N.); (H.A.); (F.I.); (Y.N.); (Y.N.)
| | - Honami Azuma
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan; (K.H.-N.); (H.A.); (F.I.); (Y.N.); (Y.N.)
| | - Fumina Ishikawa
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan; (K.H.-N.); (H.A.); (F.I.); (Y.N.); (Y.N.)
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan; (K.H.-N.); (H.A.); (F.I.); (Y.N.); (Y.N.)
| | - Dengli Wang
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama 700-8558, Japan; (D.W.); (K.L.); (H.W.); (M.N.)
| | - Keyue Liu
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama 700-8558, Japan; (D.W.); (K.L.); (H.W.); (M.N.)
| | - Hidenori Wake
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama 700-8558, Japan; (D.W.); (K.L.); (H.W.); (M.N.)
| | - Masahiro Nishibori
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama 700-8558, Japan; (D.W.); (K.L.); (H.W.); (M.N.)
| | - Yoshihiro Nakata
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan; (K.H.-N.); (H.A.); (F.I.); (Y.N.); (Y.N.)
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan; (K.H.-N.); (H.A.); (F.I.); (Y.N.); (Y.N.)
- Correspondence: ; Tel.: +81-082-257-5310
| |
Collapse
|
112
|
Song X, Long D. Nrf2 and Ferroptosis: A New Research Direction for Neurodegenerative Diseases. Front Neurosci 2020; 14:267. [PMID: 32372896 PMCID: PMC7186402 DOI: 10.3389/fnins.2020.00267] [Citation(s) in RCA: 371] [Impact Index Per Article: 74.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Ferroptosis is a kind of regulated cell death (RCD) caused by the redox state disorder of intracellular microenvironment controlled by glutathione (GSH) peroxidase 4 (GPX4), which is inhibited by iron chelators and lipophilic antioxidants. In addition to classical regulatory mechanisms, new regulatory factors for ferroptosis have been discovered in recent years, such as the P53 pathway, the activating transcription factor (ATF)3/4 pathway, Beclin 1 (BECN1) pathway, and some non-coding RNA. Ferroptosis is closely related to cancer treatment, neurodegenerative diseases, ischemia–reperfusion of organ, neurotoxicity, and others, in particular, in the field of neurodegenerative diseases treatment has aroused people’s interest. The nuclear factor E2 related factor 2 (Nrf2/NFE2L2) has been proved to play a key role in neurodegenerative disease treatment and ferroptosis regulation. Ferroptosis promotes the progression of neurodegenerative diseases, while the expression of Nrf2 and its target genes (Ho-1, Nqo-1, and Trx) has been declined with aging; therefore, there is still insufficient evidence for ferroptosis and Nrf2 regulatory networks in the field of neurodegenerative diseases. In this review, we will provide a brief overview of ferroptosis regulatory mechanisms, as well as an emphasis on the mechanism of Nrf2 regulating ferroptosis. We also highlight the role of ferroptosis and Nrf2 during the process of neurodegenerative diseases and investigate a theoretical basis for further research on the relationship between Nrf2 and ferroptosis in the process of neurodegenerative diseases treatment.
Collapse
Affiliation(s)
- Xiaohua Song
- School of Public Health, University of South China, Hengyang, China
| | - Dingxin Long
- School of Public Health, University of South China, Hengyang, China
| |
Collapse
|
113
|
Ratan RR. The Chemical Biology of Ferroptosis in the Central Nervous System. Cell Chem Biol 2020; 27:479-498. [PMID: 32243811 DOI: 10.1016/j.chembiol.2020.03.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/04/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Over the past five decades, thanatology has come to include the study of how individual cells in our bodies die appropriately and inappropriately in response to physiological and pathological stimuli. Morphological and biochemical criteria have been painstakingly established to create clarity around definitions of distinct types of cell death and mechanisms for their activation. Among these, ferroptosis has emerged as a unique, oxidative stress-induced cell death pathway with implications for diseases as diverse as traumatic brain injury, hemorrhagic stroke, Alzheimer's disease, cancer, renal ischemia, and heat stress in plants. In this review, I highlight some of the formative studies that fostered its recognition in the nervous system and describe how chemical biological tools have been essential in defining events necessary for its execution. Finally, I discuss emerging opportunities for antiferroptotic agents as therapeutic agents in neurological diseases.
Collapse
Affiliation(s)
- Rajiv R Ratan
- Burke Neurological Institute at Weill Cornell Medicine, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| |
Collapse
|
114
|
Wen C, Huang C, Yang M, Fan C, Li Q, Zhao J, Gan D, Li A, Zhu L, Lu D. The Secretion from Bone Marrow Mesenchymal Stem Cells Pretreated with Berberine Rescues Neurons with Oxidative Damage Through Activation of the Keap1-Nrf2-HO-1 Signaling Pathway. Neurotox Res 2020; 38:59-73. [PMID: 32108297 DOI: 10.1007/s12640-020-00178-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/17/2019] [Accepted: 02/06/2020] [Indexed: 12/14/2022]
Abstract
Oxidative stress is a potential pathological mechanism of Alzheimer's disease (AD). Berberine (BBR) can improve antioxidative capacity and inhibit Aβ protein aggregation and tau protein hyperphosphorylation in AD, and stem cell therapy is also increasingly recognized as a therapy for AD. Bone marrow mesenchymal stem cells (BMSCs) have many advantages, as they exhibit antioxidant and anti-inflammatory activity and secrete a variety of neurotrophic factors, and play important roles in neurodegenerative disease treatment. In this study, we investigated the antioxidant effects of secretions from BMSCs pretreated with BBR on tert-butyl hydroperoxide (t-BHP)-damaged neurons. We demonstrated that BBR can enhance BMSC viability and the secretion of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), both of which are vital neurotrophic factors that maintain neuronal growth. Moreover, conditioned medium from BBR-treated BMSCs (BBR-BMSC-CM) reduced reactive oxygen species (ROS) production, attenuated a decrease in the mitochondrial membrane potential, and ameliorated neuronal apoptosis by decreasing levels of the apoptotic proteins Bax/Bcl-2, cytochrome c, and cleaved caspase-3/caspase-3. In addition, increased synaptophysin (SYP) and postsynaptic density protein 95 (PSD95) levels indicated that neuronal synaptic function was restored. Further study revealed that BBR-BMSC-CM activated the antioxidant proteins Keap1, Nrf2, and HO-1. In conclusion, our results showed that BBR-BMSC-CM attenuated apoptosis and oxidative damage in neurons by activating the Keap1-Nrf2-HO-1 signaling pathway. Taken together, these results also suggest BBR as a drug to stimulate the secretion of nutritional cytokines with the potential to treat AD.
Collapse
Affiliation(s)
- Caiyan Wen
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Cuiqin Huang
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Mei Yang
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Chongzhu Fan
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Qin Li
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Jiayi Zhao
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Danhui Gan
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - An Li
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Lihong Zhu
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Daxiang Lu
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
115
|
Exosomal Nrf2: From anti-oxidant and anti-inflammation response to wound healing and tissue regeneration in aged-related diseases. Biochimie 2020; 171-172:103-109. [PMID: 32109502 DOI: 10.1016/j.biochi.2020.02.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
Accumulation of oxidative stress in cells is an essential feature of cellular senescence and aging. This phenomenon is involved in different age-related diseases through dysregulation of homeostasis and impairing repair and regeneration (wound healing) capacity, which can suppress antioxidant responses such as the activity of antioxidant enzymes and damaged protein clearance system. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor which regulates basal and inducible expression pattern of specific genes (antioxidants and detoxifications) through antioxidant element response (ARE) sites in the stress condition, specifically in chronic and age-related stresses. Nrf2 maintains cellular redox hemostasis and promotes rejuvenation. Exosomes are nanoscale vesicles that are released by various cells to actively regulate the complex cellular signaling networks. Exosomal-Nrf2 and exosomal-Nrf2-mediated products can modulate oxidative hemostasis in target cells to induce tissue repairing with therapeutic proposes, and regeneration capability. In this study, we summarized the role of exosomal-Nrf2 in different age-related diseases, including diabetic foot ulcers, atherosclerosis, chronic heart failure, reproductive cell failures, and neurodegenerative diseases. In addition, we briefly explained the crosstalk between plant exosomes and mammalian cell metabolism in the benefit of cellular stress suppression.
Collapse
|
116
|
Li H, Kilgallen AB, Münzel T, Wolf E, Lecour S, Schulz R, Daiber A, Van Laake LW. Influence of mental stress and environmental toxins on circadian clocks: Implications for redox regulation of the heart and cardioprotection. Br J Pharmacol 2020; 177:5393-5412. [PMID: 31833063 PMCID: PMC7680009 DOI: 10.1111/bph.14949] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023] Open
Abstract
Risk factors in the environment such as air pollution and mental stress contribute to the development of chronic non-communicable disease. Air pollution was identified as the leading health risk factor in the physical environment, followed by water pollution, soil pollution/heavy metals/chemicals and occupational exposures, however neglecting the non-chemical environmental health risk factors (e.g. mental stress and noise). Epidemiological data suggest that environmental risk factors are associated with higher risk for cardiovascular, metabolic and mental diseases, including hypertension, heart failure, myocardial infarction, diabetes, arrhythmia, stroke, depression and anxiety disorders. We provide an overview on the impact of the external exposome comprising risk factors/exposures on cardiovascular health with a focus on dysregulation of stress hormones, mitochondrial function, redox balance and inflammation with special emphasis on the circadian clock. Finally, we assess the impact of circadian clock dysregulation on cardiovascular health and the potential of environment-specific preventive strategies or "chrono" therapy for cardioprotection. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.
Collapse
Affiliation(s)
- Huige Li
- Department of Pharmacology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Aoife B Kilgallen
- Division Heart and Lungs and Regenerative Medicine Centre, University Medical Centre Utrecht and Utrecht University, Utrecht, Netherlands
| | - Thomas Münzel
- Center of Cardiology 1, Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Eva Wolf
- Structural Chronobiology, Institute of Molecular Physiology, Johannes Gutenberg University, Mainz, Germany.,Structural Chronobiology, Institute of Molecular Biology, Mainz, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Daiber
- Center of Cardiology 1, Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Linda W Van Laake
- Division Heart and Lungs and Regenerative Medicine Centre, University Medical Centre Utrecht and Utrecht University, Utrecht, Netherlands
| |
Collapse
|
117
|
Ren YZ, Zhang BZ, Zhao XJ, Zhang ZY. Resolvin D1 ameliorates cognitive impairment following traumatic brain injury via protecting astrocytic mitochondria. J Neurochem 2020; 154:530-546. [PMID: 31951012 DOI: 10.1111/jnc.14962] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/29/2019] [Accepted: 01/02/2020] [Indexed: 12/18/2022]
Abstract
Cognitive impairment is one of the most common and devastating neuropsychiatric sequelae after traumatic brain injury (TBI), and hippocampal neuronal survival plays a causal role in this pathological process. Resolvin D1 (RvD1), an important endogenous specialized pro-resolving mediator, has recently been reported to exert a potent protective effect on mitochondria. This suggests that RvD1 may suppress neuroinflammation and protect astrocytic mitochondria at the same time to play further neuroprotective roles. C57BL/6 mice subjected to TBI using a controlled cortical impact device were used for in vivo experiments. Cultured primary mouse astrocytes and an N2a mouse neuroblastoma cell line were used for in vitro experiments. In TBI mice, RvD1 significantly ameliorated cognitive impairment, suppressed gliosis and alleviated neuronal loss in the hippocampus. To explore the mechanism underlying this activity, we verified that RvD1 can induce a higher level of mitophagy to remove damaged mitochondria and eliminate extra mitochondria-derived reactive oxygen species (mitoROS) by activating ALX4/FPR2 receptors in astrocytes. In an in vitro model, we further confirmed that RvD1 can protect mitochondrial morphology and membrane potential in astrocytes and thereby enhance the survival of neurons. Meanwhile, RvD1 was also shown to increase the expression of brain-derived neurotrophic factor and glutamate aspartate transporter in the hippocampus following TBI, which indicates a possible way by which RvD1 increases the supportive function of astrocytes. These findings suggest that RvD1 may be a potent therapeutic option for ameliorating cognitive impairment following TBI by controlling neuroinflammation and protecting astrocytic mitochondria.
Collapse
Affiliation(s)
- Yi-Zhi Ren
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ben-Zheng Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xiao-Jing Zhao
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Zhi-Yuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China.,The Key Laboratory of Antibody Technique of the Ministry of Health, Nanjing Medical University, Nanjing, China.,Department of Neurology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
118
|
Brain-Derived Neurotrophic Factor and Its Potential Therapeutic Role in Stroke Comorbidities. Neural Plast 2020; 2020:1969482. [PMID: 32399020 PMCID: PMC7204205 DOI: 10.1155/2020/1969482] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/14/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
With the rise in the aging global population, stroke comorbidities have become a serious health threat and a tremendous economic burden on human society. Current therapeutic strategies mainly focus on protecting neurons from cytotoxic damage at the acute phase upon stroke onset, which not only is a difficult way to ameliorate stroke symptoms but also presents a challenge for the patients to receive effective treatment in time. The brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain, which possesses a remarkable capability to repair brain damage. Recent promising preclinical outcomes have made BDNF a popular late-stage target in the development of novel stroke treatments. In this review, we aim to summarize the latest progress in the understanding of the cellular/molecular mechanisms underlying stroke pathogenesis, current strategies and difficulties in drug development, the mechanism of BDNF action in poststroke neurorehabilitation and neuroplasticity, and recent updates in novel therapeutic methods.
Collapse
|
119
|
Pretreatment with Roxadustat (FG-4592) Attenuates Folic Acid-Induced Kidney Injury through Antiferroptosis via Akt/GSK-3 β/Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6286984. [PMID: 32051732 PMCID: PMC6995323 DOI: 10.1155/2020/6286984] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/29/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022]
Abstract
Folic acid- (FA-) induced kidney injury is characterized by the tubule damage due to the disturbance of the antioxidant system and subsequent interstitial fibrosis. FG-4592 is an inhibitor of prolyl hydroxylase of hypoxia-inducible factor (HIF), an antioxidant factor. The present study investigated the protective role of FG-4592 pretreatment at the early stage of the kidney injury and long-term impact on the progression of renal fibrosis. FG-4592 was administrated two days before FA injection in mice. On the second day after FA injection, the mice with FG-4592 pretreatment showed an improved renal function, compared with those without FG-4592 pretreatment, indicated by biochemical and histological parameters; meanwhile, the cellular content of iron, malondialdehyde, and 4-hydroxynonenal histologically decreased, implying the suppression of iron accumulation and lipid peroxidation. Simultaneously, upregulation of HIF-1α was found, along with Nrf2 activation, which was reflected by increased nuclear translocation and high-expression of downstream proteins, including heme-oxygenase1, glutathione peroxidase4, and cystine/glutamate transporter, as well as ferroportin. Correspondingly, the elevated levels of antioxidative enzymes and glutathione, as well as reduced iron accumulation, were observed, suggesting a lower risk of occurrence of ferroptosis with FG-4592 pretreatment. This was confirmed by reversed pathological parameters and improved renal function in FA-treated mice with the administration of ferrostatin-1, a specific ferroptosis inhibitor. Furthermore, a signal pathway study indicated that Nrf2 activation was associated with increased phosphorylation of Akt and GSK-3β, verified by the use of an inhibitor of the PI3K that phosphorylates Akt. Moreover, FG-4592 pretreatment also decreased macrophage infiltration and expression of inflammatory factors TNF-α and IL-1β. On the 14th day after FA injection, FG-4592 pretreatment decreased collagen deposition and expression of fibrosis biomarkers. These findings suggest that the protective role of FG-4592 pretreatment is achieved mainly by decreasing ferroptosis at the early stage of FA-induced kidney injury via Akt/GSK-3β-mediated Nrf2 activation, which retards the fibrosis progression.
Collapse
|
120
|
Li LH, Peng WN, Deng Y, Li JJ, Tian XR. Action of trichostatin A on Alzheimer's disease-like pathological changes in SH-SY5Y neuroblastoma cells. Neural Regen Res 2020; 15:293-301. [PMID: 31552902 PMCID: PMC6905323 DOI: 10.4103/1673-5374.265564] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The histone deacetylase inhibitor, trichostatin A, is used to treat Alzheimer's disease and can improve learning and memory but its underlying mechanism of action is unknown. To determine whether the therapeutic effect of trichostatin A on Alzheimer's disease is associated with the nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like epichlorohydrin-related protein-1 (Keap1) signaling pathway, amyloid β-peptide 25-35 (Aβ25-35) was used to induce Alzheimer's disease-like pathological changes in SH-SY5Y neuroblastoma cells. Cells were then treated with trichostatin A. The effects of trichostatin A on the expression of Keap1 and Nrf2 were detected by real-time quantitative polymerase chain reaction, western blot assays and immunofluorescence. Total antioxidant capacity and autophagy activity were evaluated by total antioxidant capacity assay kit and light chain 3-I/II levels, respectively. We found that trichostatin A increased cell viability and Nrf2 expression, and decreased Keap1 expression in SH-SY5Y cells. Furthermore, trichostatin A increased the expression of Nrf2-related target genes, such as superoxide dismutase, NAD(P)H quinone dehydrogenase 1 and glutathione S-transferase, thereby increasing the total antioxidant capacity of SH-SY5Y cells and inhibiting amyloid β-peptide-induced autophagy. Knockdown of Keap1 in SH-SY5Y cells further increased trichostatin A-induced Nrf2 expression. These results indicate that the therapeutic effect of trichostatin A on Alzheimer's disease is associated with the Keap1-Nrf2 pathway. The mechanism for this action may be that trichostatin A increases cell viability and the antioxidant capacity of SH-SY5Y cells by alleviating Keap1-mediated inhibition Nrf2 signaling, thereby alleviating amyloid β-peptide-induced cell damage.
Collapse
Affiliation(s)
- Li-Hua Li
- College of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Wen-Na Peng
- Department of Rehabilitation, Second Xiangya Hospital, Changsha, Hunan Province, China
| | - Yu Deng
- College of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Jing-Jing Li
- College of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Xiang-Rong Tian
- College of Biology and Environmental Science, Jishou University, Jishou, Hunan Province, China
| |
Collapse
|
121
|
Zhou C, Chen H, Zheng JF, Guo ZD, Huang ZJ, Wu Y, Zhong JJ, Sun XC, Cheng CJ. Pentraxin 3 contributes to neurogenesis after traumatic brain injury in mice. Neural Regen Res 2020; 15:2318-2326. [PMID: 32594056 PMCID: PMC7749468 DOI: 10.4103/1673-5374.285001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence indicates that pentraxin 3 is an acute-phase protein that is linked with the immune response to inflammation. It is also a newly discovered marker of anti-inflammatory A2 reactive astrocytes, and potentially has multiple protective effects in stroke; however, its role in the adult brain after traumatic brain injury is unknown. In the present study, a moderate model of traumatic brain injury in mice was established using controlled cortical impact. The models were intraventricularly injected with recombinant pentraxin 3 (the recombinant pentraxin 3 group) or an equal volume of vehicle (the control group). The sham-operated mice underwent craniotomy, but did not undergo the controlled cortical impact. The potential neuroprotective and neuroregenerative roles of pentraxin 3 were investigated on days 14 and 21 after traumatic brain injury. Western blot assay showed that the expression of endogenous pentraxin 3 was increased after traumatic brain injury in mice. Furthermore, the neurological severity test and wire grip test revealed that recombinant pentraxin 3 treatment reduced the neurological severity score and increased the wire grip score, suggesting an improved recovery of sensory-motor functions. The Morris water maze results demonstrated that recombinant pentraxin 3 treatment reduced the latency to the platform, increased the time spent in the correct quadrant, and increased the number of times traveled across the platform, thus suggesting an improved recovery of cognitive function. In addition, to investigate the effects of pentraxin 3 on astrocytes, specific markers of A2 astrocytes were detected in primary astrocyte cultures in vitro using western blot assay. The results demonstrated that pentraxin 3 administration activates A2 astrocytes. To explore the protective mechanisms of pentraxin 3, immunofluorescence staining was used. Intraventricular injection of recombinant pentraxin 3 increased neuronal maintenance in the peri-injured cortex and ipsilateral hippocampus, increased the number of doublecortin-positive neural progenitor cells in the subventricular and subgranular zones, and increased the number of bromodeoxyuridine (proliferation) and neuronal nuclear antigen (mature neuron) double-labeled cells in the hippocampus and peri-injured cortex. Pentraxin 3 administration also increased the number of neurospheres and the number of bromodeoxyuridine and doublecortin double-labeled cells in neurospheres, and enhanced the proliferation of neural progenitor cells in primary neural progenitor cell cultures in vitro. In conclusion, recombinant pentraxin 3 administration activated A2 astrocytes, and consequently improved the recovery of neural function by increasing neuronal survival and enhancing neurogenesis. All experiments were approved by the Animal Ethics Committee of the First Affiliated Hospital of Chongqing Medical University, China on March 1, 2016.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Feng Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zong-Duo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Jian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Jun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Chuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chong-Jie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
122
|
Haddad M, Perrotte M, Khedher MRB, Demongin C, Lepage A, Fülöp T, Ramassamy C. Methylglyoxal and Glyoxal as Potential Peripheral Markers for MCI Diagnosis and Their Effects on the Expression of Neurotrophic, Inflammatory and Neurodegenerative Factors in Neurons and in Neuronal Derived-Extracellular Vesicles. Int J Mol Sci 2019; 20:ijms20194906. [PMID: 31623327 PMCID: PMC6801730 DOI: 10.3390/ijms20194906] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/14/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022] Open
Abstract
Methylglyoxal (MG) and glyoxal (GO) are suggested to be associated with the development of neurodegenerative pathologies. However, their peripheral levels in relation to cognitive decline and their effects on key factors in neuronal cells are poorly investigated. The aim of this study was to determine their serum levels in MCI (mild cognitive impairment) and Alzheimer’s disease (AD) patients, to analyze their effects on the neurotrophic and inflammatory factors, on neurodegenerative markers in neuronal cells and in neuronal derived-extracellular vesicles (nEVs). Our results show that MG and GO levels in serum, determined by HPLC, were higher in MCI. ROC (receiver-operating characteristic curves) analysis showed that the levels of MG in serum have higher sensitivity to differentiate MCI from controls but not from AD. Meanwhile, serum GO levels differentiate MCI from control and AD groups. Cells and nEVs levels of BDNF, PRGN, NSE, APP, MMP-9, ANGPTL-4, LCN2, PTX2, S100B, RAGE, Aβ peptide, pTau T181 and alpha-synuclein were quantified by luminex assay. Treatment of neuronal cells with MG or GO reduced the cellular levels of NSE, PRGN, APP, MMP-9 and ANGPTL-4 and the nEVs levels of BDNF, PRGN and LCN2. Our findings suggest that targeting MG and GO may be a promising therapeutic strategy to prevent or delay the progression of AD.
Collapse
Affiliation(s)
- Mohamed Haddad
- Institut National de Recherche Scientifique - Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada.
- Institute on Nutrition and Functional Foods, Laval University, Quebec City, QC G1V 0A6, Canada.
| | - Morgane Perrotte
- Institut National de Recherche Scientifique - Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada.
- Institute on Nutrition and Functional Foods, Laval University, Quebec City, QC G1V 0A6, Canada.
| | - Mohamed Raâfet Ben Khedher
- Institut National de Recherche Scientifique - Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada.
- Institute on Nutrition and Functional Foods, Laval University, Quebec City, QC G1V 0A6, Canada.
| | - Clément Demongin
- Institut National de Recherche Scientifique - Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada.
| | - Aurélie Lepage
- Department of Medicine, Geriatric Division, Research Center on Aging, Sherbrooke University, Sherbrooke, QC J1H 4C4, Canada.
| | - Tamás Fülöp
- Department of Medicine, Geriatric Division, Research Center on Aging, Sherbrooke University, Sherbrooke, QC J1H 4C4, Canada.
| | - Charles Ramassamy
- Institut National de Recherche Scientifique - Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada.
- Institute on Nutrition and Functional Foods, Laval University, Quebec City, QC G1V 0A6, Canada.
| |
Collapse
|
123
|
Mishra PK, Adameova A, Hill JA, Baines CP, Kang PM, Downey JM, Narula J, Takahashi M, Abbate A, Piristine HC, Kar S, Su S, Higa JK, Kawasaki NK, Matsui T. Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol 2019; 317:H891-H922. [PMID: 31418596 DOI: 10.1152/ajpheart.00259.2019] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell death is a fundamental process in cardiac pathologies. Recent studies have revealed multiple forms of cell death, and several of them have been demonstrated to underlie adverse cardiac remodeling and heart failure. With the expansion in the area of myocardial cell death and increasing concerns over rigor and reproducibility, it is important and timely to set a guideline for the best practices of evaluating myocardial cell death. There are six major forms of regulated cell death observed in cardiac pathologies, namely apoptosis, necroptosis, mitochondrial-mediated necrosis, pyroptosis, ferroptosis, and autophagic cell death. In this article, we describe the best methods to identify, measure, and evaluate these modes of myocardial cell death. In addition, we discuss the limitations of currently practiced myocardial cell death mechanisms.
Collapse
Affiliation(s)
- Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University of Bratislava, Bratislava, Slovakia
| | - Joseph A Hill
- Departments of Medicine (Cardiology) and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christopher P Baines
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Peter M Kang
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - James M Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - Masafumi Takahashi
- Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Antonio Abbate
- Virginia Commonwealth University, Pauley Heart Center, Richmond, Virginia
| | - Hande C Piristine
- Department of Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sumit Kar
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Shi Su
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jason K Higa
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Nicholas K Kawasaki
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Takashi Matsui
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| |
Collapse
|
124
|
Luo LL, Li YF, Shan HM, Wang LP, Yuan F, Ma YY, Li WL, He TT, Wang YY, Qu MJ, Liang HB, Zhang ZJ, Yang GY, Tang YH, Wang YT. L-glutamine protects mouse brain from ischemic injury via up-regulating heat shock protein 70. CNS Neurosci Ther 2019; 25:1030-1041. [PMID: 31218845 PMCID: PMC6698979 DOI: 10.1111/cns.13184] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/17/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION L-glutamine is an antioxidant that plays a role in a variety of biochemical processes. Given that oxidative stress is a key component of stroke pathology, the potential of L-glutamine in the treatment of ischemic stroke is worth exploring. AIMS In this study, we investigated the effect and mechanisms of action of L-glutamine after cerebral ischemic injury. RESULTS L-glutamine reduced brain infarct volume and promoted neurobehavioral recovery in mice. L-glutamine administration increased the expression of heat-shock protein 70 (HSP70) in astrocytes and endothelial cells. Such effects were abolished by the coadministration of Apoptozole, an inhibitor of the ATPase activity of HSP70. L-glutamine also reduced oxidative stress and neuronal apoptosis, and increased the level of superoxide dismutase, glutathione, and brain-derived neurotrophic factor. Cotreatment with Apoptozole abolished these effects. Cell culture study further revealed that the conditioned medium from astrocytes cultured with L-glutamine reduced the apoptosis of neurons after oxygen-glucose deprivation. CONCLUSION L-glutamine attenuated ischemic brain injury and promoted functional recovery via HSP70, suggesting its potential in ischemic stroke therapy.
Collapse
Affiliation(s)
- Long-Long Luo
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Fang Li
- Department of Neurology, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Min Shan
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Ping Wang
- Department of Neurology, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Yuan
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan-Yuan Ma
- Department of Neurology, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wan-Lu Li
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ting-Ting He
- Department of Neurology, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Yang Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Mei-Jie Qu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Huai-Bin Liang
- Department of Neurology, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Jun Zhang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, School of Medicine, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yao-Hui Tang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Ting Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
125
|
Chiu KM, Lin TY, Lee MY, Lu CW, Wang MJ, Wang SJ. Dexmedetomidine protects neurons from kainic acid-induced excitotoxicity by activating BDNF signaling. Neurochem Int 2019; 129:104493. [PMID: 31220473 DOI: 10.1016/j.neuint.2019.104493] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
Glutamatergic excitotoxicity is crucial in the pathogenesis of epileptic seizures. Dexmedetomidine, a potent and highly selective α2 adrenoceptor agonist, inhibits glutamate release from nerve terminals in rat cerebrocortical nerve terminals. However, the ability of dexmedetomidine to affect glutamate-induced brain injury is still unknown. Therefore, the present study evaluated the protective effect of dexmedetomidine against brain damage by using a kainic acid (KA) rat model, a frequently used model for temporal lobe epilepsy. Rats were treated with dexmedetomidine (1 or 5 μg/kg, intraperitoneally) 30 min before the KA (15 mg/kg) intraperitoneal injection. KA-induced seizure score and elevations of glutamate release in rat hippocampi were inhibited by pretreatment with dexmedetomidine. Histopathological and TUNEL staining analyzes showed that dexmedetomidine attenuated KA-induced neuronal death in the hippocampus. Dexmedetomidine ameliorated KA-induced apoptosis, and this neuroprotective effect was accompanied by inhibited the KA-induced caspase-3 expression as well as MAPKs phosphorylation, and reversed Bcl-2 down-expression, coupled with increased Nrf2, BDNF and TrkB expression in KA-treated rats. The results suggest that dexmedetomidine protected rat brains from KA-induced excitotoxic damage by reducing glutamate levels, suppressing caspase-3 activation and MAPKs phosphorylation, and enhancing Bcl-2, Nrf2, BDNF and TrkB expression in the hippocampus. Therefore, dexmedetomidine may be beneficial for preventing or treating brain disorders associated with excitotoxic neuronal damage. In conclusion, these data suggest that dexmedetomidine has the therapeutic potential for treating epilepsy.
Collapse
Affiliation(s)
- Kuan-Ming Chiu
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Nursing, Oriental Institute of Technology, New Taipei City, Taiwan; Department of Photonics Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Tzu-Yu Lin
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Ming-Yi Lee
- Division of Cardiovascular Surgery, Cardiovascular Center, Far-Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Cheng-Wei Lu
- Department of Anesthesiology, Far-Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Mechanical Engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Ming-Jiuh Wang
- Department of Anesthesiology, National Taiwan University Hospital, Taipei City, Taiwan
| | - Su-Jane Wang
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
126
|
Spiers JG, Breda C, Robinson S, Giorgini F, Steinert JR. Drosophila Nrf2/Keap1 Mediated Redox Signaling Supports Synaptic Function and Longevity and Impacts on Circadian Activity. Front Mol Neurosci 2019; 12:86. [PMID: 31040766 PMCID: PMC6476960 DOI: 10.3389/fnmol.2019.00086] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022] Open
Abstract
Many neurodegenerative conditions and age-related neuropathologies are associated with increased levels of reactive oxygen species (ROS). The cap "n" collar (CncC) family of transcription factors is one of the major cellular system that fights oxidative insults, becoming activated in response to oxidative stress. This transcription factor signaling is conserved from metazoans to human and has a major developmental and disease-associated relevance. An important mammalian member of the CncC family is nuclear factor erythroid 2-related factor 2 (Nrf2) which has been studied in numerous cellular systems and represents an important target for drug discovery in different diseases. CncC is negatively regulated by Kelch-like ECH associated protein 1 (Keap1) and this interaction provides the basis for a homeostatic control of cellular antioxidant defense. We have utilized the Drosophila model system to investigate the roles of CncC signaling on longevity, neuronal function and circadian rhythm. Furthermore, we assessed the effects of CncC function on larvae and adult flies following exposure to stress. Our data reveal that constitutive overexpression of CncC modifies synaptic mechanisms that positively impact on neuronal function, and suppression of CncC inhibitor, Keap1, shows beneficial phenotypes on synaptic function and longevity. Moreover, supplementation of antioxidants mimics the effects of augmenting CncC signaling. Under stress conditions, lack of CncC signaling worsens survival rates and neuronal function whilst silencing Keap1 protects against stress-induced neuronal decline. Interestingly, overexpression and RNAi-mediated downregulation of CncC have differential effects on sleep patterns possibly via interactions with redox-sensitive circadian cycles. Thus, our data illustrate the important regulatory potential of CncC signaling in neuronal function and synaptic release affecting multiple aspects within the nervous system.
Collapse
Affiliation(s)
- Jereme G Spiers
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Sue Robinson
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Joern R Steinert
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
127
|
Ishii T, Warabi E. Mechanism of Rapid Nuclear Factor-E2-Related Factor 2 (Nrf2) Activation via Membrane-Associated Estrogen Receptors: Roles of NADPH Oxidase 1, Neutral Sphingomyelinase 2 and Epidermal Growth Factor Receptor (EGFR). Antioxidants (Basel) 2019; 8:antiox8030069. [PMID: 30889865 PMCID: PMC6466580 DOI: 10.3390/antiox8030069] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Membrane-associated estrogen receptors (ER)-α36 and G protein-coupled estrogen receptor (GPER) play important roles in the estrogen’s rapid non-genomic actions including stimulation of cell proliferation. Estrogen via these receptors induces rapid activation of transcription factor nuclear factor-E2-related factor 2 (Nrf2), a master regulator of detoxification and antioxidant systems, playing a key role in the metabolic reprogramming to support cell proliferation. This review highlights the possible mechanism underlying rapid Nrf2 activation via membrane-associated estrogen receptors by estrogen and phytoestrogens. Stimulation of ER-α36-GPER signaling complex rapidly induces Src-mediated transactivation of epidermal growth factor receptor (EGFR) leading to a kinase-mediated signaling cascade. We propose a novel hypothesis that ER-α36-GPER signaling initially induces rapid and temporal activation of NADPH oxidase 1 to generate superoxide, which subsequently activates redox-sensitive neutral sphingomyelinase 2 generating the lipid signaling mediator ceramide. Generation of ceramide is required for Ras activation and ceramide-protein kinase C ζ-casein kinase 2 (CK2) signaling. Notably, CK2 enhances chaperone activity of the Cdc37-Hsp90 complex supporting activation of various signaling kinases including Src, Raf and Akt (protein kinase B). Activation of Nrf2 may be induced by cooperation of two signaling pathways, (i) Nrf2 stabilization by direct phosphorylation by CK2 and (ii) EGFR-Ras-PI 3 kinase (PI3K)-Akt axis which inhibits glycogen synthase kinase 3β leading to enhanced nuclear transport and stability of Nrf2.
Collapse
Affiliation(s)
- Tetsuro Ishii
- Faculty of Medicine, University of Tsukuba, Tsukuba Ibaraki 305-8575, Japan.
| | - Eiji Warabi
- Faculty of Medicine, University of Tsukuba, Tsukuba Ibaraki 305-8575, Japan.
| |
Collapse
|
128
|
Harigae H, Hino K, Toyokuni S. Iron as Soul of Life on Earth Revisited: From Chemical Reaction, Ferroptosis to Therapeutics. Free Radic Biol Med 2019; 133:1-2. [PMID: 30736912 DOI: 10.1016/j.freeradbiomed.2019.01.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan.
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
129
|
Núñez MT, Hidalgo C. Noxious Iron-Calcium Connections in Neurodegeneration. Front Neurosci 2019; 13:48. [PMID: 30809110 PMCID: PMC6379295 DOI: 10.3389/fnins.2019.00048] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/18/2019] [Indexed: 12/26/2022] Open
Abstract
Iron and calcium share the common feature of being essential for normal neuronal function. Iron is required for mitochondrial function, synaptic plasticity, and the development of cognitive functions whereas cellular calcium signals mediate neurotransmitter exocytosis, axonal growth and synaptic plasticity, and control the expression of genes involved in learning and memory processes. Recent studies have revealed that cellular iron stimulates calcium signaling, leading to downstream activation of kinase cascades engaged in synaptic plasticity. The relationship between calcium and iron is Janus-faced, however. While under physiological conditions iron-mediated reactive oxygen species generation boosts normal calcium-dependent signaling pathways, excessive iron levels promote oxidative stress leading to the upsurge of unrestrained calcium signals that damage mitochondrial function, among other downstream targets. Similarly, increases in mitochondrial calcium to non-physiological levels result in mitochondrial dysfunction and a predicted loss of iron homeostasis. Hence, if uncontrolled, the iron/calcium self-feeding cycle becomes deleterious to neuronal function, leading eventually to neuronal death. Here, we review the multiple cell-damaging responses generated by the unregulated iron/calcium self-feeding cycle, such as excitotoxicity, free radical-mediated lipid peroxidation, and the oxidative modification of crucial components of iron and calcium homeostasis/signaling: the iron transporter DMT1, plasma membrane, and intracellular calcium channels and pumps. We discuss also how iron-induced dysregulation of mitochondrial calcium contributes to the generation of neurodegenerative conditions, including Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Collapse
Affiliation(s)
- Marco Tulio Núñez
- Iron and Neuroregeneration Laboratory, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Calcium Signaling Laboratory, Biomedical Research Institute, CEMC, Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
130
|
Uric Acid Protects against Focal Cerebral Ischemia/Reperfusion-Induced Oxidative Stress via Activating Nrf2 and Regulating Neurotrophic Factor Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6069150. [PMID: 30581534 PMCID: PMC6276484 DOI: 10.1155/2018/6069150] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022]
Abstract
The aim of this study was to investigate whether uric acid (UA) might exert neuroprotection via activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway and regulating neurotrophic factors in the cerebral cortices after transient focal cerebral ischemia/reperfusion (FCI/R) in rats. UA was intravenously injected through the tail vein (16 mg/kg) 30 min after the onset of reperfusion in rats subjected to middle cerebral artery occlusion for 2 h. Neurological deficit score was performed to analyze neurological function at 24 h after reperfusion. Terminal deoxynucleotidyl transferase-mediated dNTP nick end labeling (TUNEL) staining and hematoxylin and eosin (HE) staining were used to detect histological injury of the cerebral cortex. Malondialdehyde (MDA), the carbonyl groups, and 8-hydroxyl-2′-deoxyguanosine (8-OHdG) levels were employed to evaluate oxidative stress. Nrf2 and its downstream antioxidant protein, heme oxygenase- (HO-) 1,were detected by western blot. Nrf2 DNA-binding activity was observed using an ELISA-based measurement. Expressions of BDNF and NGF were analyzed by immunohistochemistry. Our results showed that UA treatment significantly suppressed FCI/R-induced oxidative stress, accompanied by attenuating neuronal damage, which subsequently decreased the infarct volume and neurological deficit. Further, the treatment of UA activated Nrf2 signaling pathway and upregulated BDNF and NGF expression levels. Interestingly, the aforementioned effects of UA were markedly inhibited by administration of brusatol, an inhibitor of Nrf2. Taken together, the antioxidant and neuroprotective effects afforded by UA treatment involved the modulation of Nrf2-mediated oxidative stress and regulation of BDNF and NGF expression levels. Thus, UA treatment could be of interest to prevent FCI/R injury.
Collapse
|
131
|
Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, Wang J. Ferroptosis and Its Role in Diverse Brain Diseases. Mol Neurobiol 2018; 56:4880-4893. [PMID: 30406908 DOI: 10.1007/s12035-018-1403-3] [Citation(s) in RCA: 344] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/18/2018] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a recently identified, iron-regulated, non-apoptotic form of cell death. It is characterized by cellular accumulation of lipid reactive oxygen species that ultimately leads to oxidative stress and cell death. Although first identified in cancer cells, ferroptosis has been shown to have significant implications in several neurologic diseases, such as ischemic and hemorrhagic stroke, Alzheimer's disease, and Parkinson's disease. This review summarizes current research on ferroptosis, its underlying mechanisms, and its role in the progression of different neurologic diseases. Understanding the role of ferroptosis could provide valuable information regarding treatment and prevention of these devastating diseases.
Collapse
Affiliation(s)
- Abigail Weiland
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yamei Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Weihua Wu
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Qian Li
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Advanced Innovation Center for Human Brain Protection, Captical Medical University, Beijing, 100069, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|