1
|
Salari A, Roghani M, Khalili M. HMG-CoA reductase inhibitor simvastatin ameliorates trimethyltin neurotoxicity and cognitive impairment through reversal of Alzheimer's-associated markers. Metab Brain Dis 2024; 40:74. [PMID: 39704877 DOI: 10.1007/s11011-024-01515-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/18/2024] [Indexed: 12/21/2024]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder in elderly. The neurotoxicant trimethyltin (TMT) induces neurodegenerative changes, as observed in AD. The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor simvastatin (SV) has shown protective and promising therapeutic effects in neurological disorders such as AD and Parkinson's disease. The present study aimed to assess neuroprotective effect of simvastatin (SV) against trimethyltin (TMT) memory decline and hippocampal neurodegeneration. For inducing AD-like phenotype, rats were i.p. injected with TMT at 8 mg/kg and were treated with simvastatin daily for 3 weeks at 10 or 30 mg/kg. Our analysis of data indicated that simvastatin-treated TMT group has lower learning and memory deficits in behavioral tasks comprising Barnes maze, Y maze, and novel object discrimination (NOD). In addition, hippocampal inflammatory, oxidative, and apoptotic factors were attenuated besides reduction of acetylcholinesterase (AChE) activity and Alzheimer's pathology factors including presenilin-1 and hyperphorphorylated Tau (p-Tau) upon simvastatin. Moreover, simvastatin treatment of TMT group inverted hippocampal changes of Wnt, β-catenin, ERK, and Akt, ameliorated astrocytic and microglial reactivity, and also prevented injury of CA1 neurons. This study unraveled that simvastatin is capable to prevent TMT-induced Alzheimer's-like phenotype in association with Wnt/β-catenin/ERK/Akt as well as restraining hippocampal neurodegeneration.
Collapse
Affiliation(s)
- Adel Salari
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| | - Mohsen Khalili
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
2
|
Trans-Anethole Alleviates Trimethyltin Chloride-Induced Impairments in Long-Term Potentiation. Pharmaceutics 2022; 14:pharmaceutics14071422. [PMID: 35890317 PMCID: PMC9320999 DOI: 10.3390/pharmaceutics14071422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Trans-anethole is an aromatic compound that has been studied for its anti-inflammation, anticonvulsant, antinociceptive, and anticancer effects. A recent report found that trans-anethole exerted neuroprotective effects on the brain via multiple pathways. Since noxious stimuli may both induce neuronal cell injury and affect synaptic functions (e.g., synaptic transmission or plasticity), it is important to understand whether the neuroprotective effect of trans-anethole extends to synaptic plasticity. Here, the effects of trimethyltin (TMT), which is a neurotoxic organotin compound, was investigated using the field recording method on hippocampal slice of mice. The influence of trans-anethole on long-term potentiation (LTP) was also studied for both NMDA receptor-dependent and NMDA receptor–independent cases. The action of trans-anethole on TMT-induced LTP impairment was examined, too. These results revealed that trans-anethole enhances NMDA receptor-dependent and -independent LTP and alleviates TMT-induced LTP impairment. These results suggest that trans-anethole modulates hippocampal LTP induction, prompting us to speculate that it may be helpful for improving cognitive impairment arising from neurodegenerative diseases, including Alzheimer’s disease.
Collapse
|
3
|
Yang* J, Wang* L, Huang L, Che X, Zhang Z, Wang C, Bai L, Liu P, Zhao Y, Hu X, Shi B, Shen Y, Liang X, Wu C, Xue X. Receptor-targeting nanomaterials alleviate binge drinking-induced neurodegeneration as artificial neurotrophins. EXPLORATION (BEIJING, CHINA) 2021; 1:61-74. [PMID: 37366469 PMCID: PMC10291571 DOI: 10.1002/exp.20210004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/07/2021] [Indexed: 06/28/2023]
Abstract
The distinguished properties of nanomaterials promote us to explore whether their intrinsic activities would be beneficial to disease treatment. Furthermore, understanding the molecular mechanism is thereby crucial for biomedical applications. Here, we investigate the therapeutic effects of single-walled carbon nanotubes (SWNTs) in a rat model of binge alcohol-induced neurodegeneration. With selection from four types of SWNT structures, bundled SWNTs (bSWNTs) facilitated the recovery of learning and memory via enhancing neuroprotection and neuroregeneration. We screened the potential target for bSWNTs, and found that bSWNTs have the abilities to directly interact with neurotrophic receptors, especially tropomyosin-related kinase B (TrkB). Moreover, similar to the actions of endogenous neurotrophins, bSWNTs could trigger the dimerization and phosphorylation of TrkB, while these conformational changes resulted in activating their downstream signals involved in neuroprotection and neuroregeneration. With relatively clear mechanisms, these "artificial neurotrophins" provide a proof-of-concept example as an efficiently therapeutic strategy for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jingyu Yang*
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
| | - Lirong Wang*
- CAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and Technology of ChinaBeijingP. R. China
| | - Liwen Huang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjinP. R. China
| | - Xiaohang Che
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
| | - Zhen Zhang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
| | - Chunxiao Wang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjinP. R. China
| | - Lihuan Bai
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjinP. R. China
| | - Ping Liu
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
| | - Yanan Zhao
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
| | - Xiaomei Hu
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
- CAS Center for Excellence in NanoscienceCAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and Technology of ChinaBeijingP. R. China
| | - Bingyang Shi
- International Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenanP. R. China
| | - Yuequan Shen
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjinP. R. China
| | - Xing‐Jie Liang
- CAS Center for Excellence in NanoscienceCAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and Technology of ChinaBeijingP. R. China
| | - Chunfu Wu
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyangP. R. China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjinP. R. China
| |
Collapse
|
4
|
Rienecker KDA, Poston RG, Saha RN. Merits and Limitations of Studying Neuronal Depolarization-Dependent Processes Using Elevated External Potassium. ASN Neuro 2020; 12:1759091420974807. [PMID: 33256465 PMCID: PMC7711227 DOI: 10.1177/1759091420974807] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 01/24/2023] Open
Abstract
Elevated extracellular potassium chloride is widely used to achieve membrane depolarization of cultured neurons. This technique has illuminated mechanisms of calcium influx through L-type voltage sensitive calcium channels, activity-regulated signaling, downstream transcriptional events, and many other intracellular responses to depolarization. However, there is enormous variability in these treatments, including durations from seconds to days and concentrations from 3mM to 150 mM KCl. Differential effects of these variable protocols on neuronal activity and transcriptional programs are underexplored. Furthermore, potassium chloride treatments in vitro are criticized for being poor representatives of in vivo phenomena and are questioned for their effects on cell viability. In this review, we discuss the intracellular consequences of elevated extracellular potassium chloride treatment in vitro, the variability of such treatments in the literature, the strengths and limitations of this tool, and relevance of these studies to brain functions and dysfunctions.
Collapse
Affiliation(s)
- Kira D. A. Rienecker
- Department of Molecular and Cell Biology,
School of Natural Sciences, University of California, Merced, United
States
| | - Robert G. Poston
- Department of Molecular and Cell Biology,
School of Natural Sciences, University of California, Merced, United
States
| | - Ramendra N. Saha
- Department of Molecular and Cell Biology,
School of Natural Sciences, University of California, Merced, United
States
| |
Collapse
|
5
|
The effect of crocin on apoptotic, inflammatory, BDNF, Pt, and Aβ40 indicators and neuronal density of CA1, CA2, and CA3 regions of hippocampus in the model of Alzheimer suffering rats induced with trimethyltin chloride. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s00580-019-02981-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
6
|
Tu THT, Sharma N, Shin EJ, Tran HQ, Lee YJ, Jeong JH, Jeong JH, Nah SY, Tran HYP, Byun JK, Ko SK, Kim HC. Ginsenoside Re Protects Trimethyltin-Induced Neurotoxicity via Activation of IL-6-Mediated Phosphoinositol 3-Kinase/Akt Signaling in Mice. Neurochem Res 2017; 42:3125-3139. [PMID: 28884396 DOI: 10.1007/s11064-017-2349-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/19/2017] [Accepted: 07/01/2017] [Indexed: 12/23/2022]
Abstract
Ginseng (Panax ginseng), an herbal medicine, has been used to prevent neurodegenerative disorders. Ginsenosides (e.g., Re, Rb1, or Rg1) were obtained from Korean mountain cultivated ginseng. The anticonvulsant activity of ginsenoside Re (20 mg/kg/day × 3) against trimethyltin (TMT) insult was the most pronounced out of ginsenosides (e.g., Re, Rb1, and Rg1). Re itself did not significantly alter tumor necrosis factor-α (TNF-α), interferon-ϒ (IFN-ϒ), and interleukin-1β (IL-1β) expression, however, it significantly increases the interleukin-6 (IL-6) expression. In addition, Re attenuated the TMT-induced decreases in IL-6 protein level. Therefore, IL-6 knockout (-/-) mice were employed to investigate whether Re requires IL-6-dependent neuroprotective activity against TMT toxicity. Re significantly attenuated TMT-induced lipid peroxidation, protein peroxidation, and reactive oxygen species in the hippocampus. Re-mediated antioxidant effects were more pronounced in IL-6 (-/-) mice than in WT mice. Consistently, TMT-induced increase in c-Fos-immunoreactivity (c-Fos-IR), TUNEL-positive cells, and nuclear chromatin clumping in the dentate gyrus of the hippocampus were significantly attenuated by Re. Furthermore, Re attenuated TMT-induced proapoptotic changes. Protective potentials by Re were comparable to those by recombinant IL-6 protein (rIL-6) against TMT-insult in IL-6 (-/-) mice. Moreover, treatment with a phosphoinositol 3-kinase (PI3K) inhibitor, LY294002 (1.6 µg, i.c.v) counteracted the protective potential mediated by Re or rIL-6 against TMT insult. The results suggest that ginsenoside Re requires IL-6-dependent PI3K/Akt signaling for its protective potential against TMT-induced neurotoxicity.
Collapse
Affiliation(s)
- Thu-Hien Thi Tu
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Yu Jeung Lee
- Clinical Pharmacy, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Pharmacology, College of Medicine, Chug-Ang University, Seoul, Republic of Korea
| | - Jung Hwan Jeong
- Headquarters of Forestry Support, Korea Forestry Promotion Institute, Seoul, 07570, Republic of Korea
| | - Seung Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Hoang-Yen Phi Tran
- Physical Chemistry Department, University of Medicine and Pharmacy, Ho Chi Minh City, 760000, Vietnam
| | - Jae Kyung Byun
- Korean Society of Forest Environment Research, Namyangju, 12014, Republic of Korea
| | - Sung Kwon Ko
- Department of Oriental Medical Food & Nutrition, Semyung University, Jecheon, 27136, Republic of Korea.
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
7
|
Kim J, Lee S, Kang S, Kim SH, Kim JC, Yang M, Moon C. Brain-derived neurotropic factor and GABAergic transmission in neurodegeneration and neuroregeneration. Neural Regen Res 2017; 12:1733-1741. [PMID: 29171440 PMCID: PMC5696856 DOI: 10.4103/1673-5374.217353] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurotoxicity induced by stress, radiation, chemicals, or metabolic diseases, is commonly associated with excitotoxicity, oxidative stress, and neuroinflammation. The pathological process of neurotoxicity induces neuronal death, interrupts synaptic plasticity in the brain, and is similar to that of diverse neurodegenerative diseases. Animal models of neurotoxicity have revealed that clinical symptoms and brain lesions can recover over time via neuroregenerative processes. Specifically, brain-derived neurotropic factor (BDNF) and gamma-aminobutyric acid (GABA)-ergic transmission are related to both neurodegeneration and neuroregeneration. This review summarizes the accumulating evidences that suggest a pathogenic role of BDNF and GABAergic transmission, their underlying mechanisms, and the relationship between BDNF and GABA in neurodegeneration and neuroregeneration. This review will provide a comprehensive overview of the underlying mechanisms of neuroregeneration that may help in developing potential strategies for pharmacotherapeutic approaches to treat neurotoxicity and neurodegenerative disease.
Collapse
Affiliation(s)
- Jinwook Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Sueun Lee
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Sohi Kang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Sung-Ho Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Jong-Choon Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Jeonbuk, South Korea
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 PLUS Project Team, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
8
|
More SV, Kumar H, Cho DY, Yun YS, Choi DK. Toxin-Induced Experimental Models of Learning and Memory Impairment. Int J Mol Sci 2016; 17:E1447. [PMID: 27598124 PMCID: PMC5037726 DOI: 10.3390/ijms17091447] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 02/07/2023] Open
Abstract
Animal models for learning and memory have significantly contributed to novel strategies for drug development and hence are an imperative part in the assessment of therapeutics. Learning and memory involve different stages including acquisition, consolidation, and retrieval and each stage can be characterized using specific toxin. Recent studies have postulated the molecular basis of these processes and have also demonstrated many signaling molecules that are involved in several stages of memory. Most insights into learning and memory impairment and to develop a novel compound stems from the investigations performed in experimental models, especially those produced by neurotoxins models. Several toxins have been utilized based on their mechanism of action for learning and memory impairment such as scopolamine, streptozotocin, quinolinic acid, and domoic acid. Further, some toxins like 6-hydroxy dopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and amyloid-β are known to cause specific learning and memory impairment which imitate the disease pathology of Parkinson's disease dementia and Alzheimer's disease dementia. Apart from these toxins, several other toxins come under a miscellaneous category like an environmental pollutant, snake venoms, botulinum, and lipopolysaccharide. This review will focus on the various classes of neurotoxin models for learning and memory impairment with their specific mechanism of action that could assist the process of drug discovery and development for dementia and cognitive disorders.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Hemant Kumar
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Duk-Yeon Cho
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Yo-Sep Yun
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea.
| |
Collapse
|
9
|
Khalin I, Alyautdin R, Wong TW, Gnanou J, Kocherga G, Kreuter J. Brain-derived neurotrophic factor delivered to the brain using poly (lactide-co-glycolide) nanoparticles improves neurological and cognitive outcome in mice with traumatic brain injury. Drug Deliv 2016; 23:3520-3528. [PMID: 27278330 DOI: 10.1080/10717544.2016.1199609] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Currently, traumatic brain injury (TBI) is the leading cause of death or disabilities in young individuals worldwide. The multi-complexity of its pathogenesis as well as impermeability of the blood-brain barrier (BBB) makes the drug choice and delivery very challenging. The brain-derived neurotrophic factor (BDNF) regulates neuronal plasticity, neuronal cell growth, proliferation, cell survival and long-term memory. However, its short half-life and low BBB permeability are the main hurdles to be an effective therapeutic for TBI. Poly (lactic-co-glycolic acid) (PLGA) nanoparticles coated by surfactant can enable the delivery of a variety of molecules across the BBB by receptor-mediated transcytosis. This study examines the ability of PLGA nanoparticles coated with poloxamer 188 (PX) to deliver BDNF into the brain and neuroprotective effects of BNDF in mice with TBI. C57bl/6 mice were subjected to weight-drop closed head injuries under anesthesia. Using enzyme-linked immunosorbent assay, we demonstrated that the intravenous (IV) injection of nanoparticle-bound BDNF coated by PX (NP-BDNF-PX) significantly increased BDNF levels in the brain of sham-operated mice (p < 0.001) and in both ipsi- (p < 0.001) and contralateral (p < 0.001) parts of brain in TBI mice compared to controls. This study also showed using the passive avoidance (PA) test, that IV injection of NP-BDNF-PX 3 h post-injury prolonged the latent time in mice with TBI thereby reversing cognitive deficits caused by brain trauma. Finally, neurological severity score test demonstrated that our compound efficiently reduced the scores at day 7 after the injury indicating the improvement of neurological deficit in animals with TBI. This study shows that PLGA nanoparticles coated with PX effectively delivered BDNF into the brain, and improved neurological and cognitive deficits in TBI mice, thereby providing a neuroprotective effect.
Collapse
Affiliation(s)
- Igor Khalin
- a Faculty of Medicine and Defence Health , National Defence University of Malaysia , Kuala Lumpur , Malaysia
| | - Renad Alyautdin
- b Scientific Centre for Expertise of Medical Application Products , Moscow , Russia
| | - Tin Wui Wong
- c iPROMISE, Non-Destructive Biomedical and Pharmaceutical Research Centre, Universiti Teknologi MARA , Selangor , Malaysia
| | - Justin Gnanou
- a Faculty of Medicine and Defence Health , National Defence University of Malaysia , Kuala Lumpur , Malaysia
| | - Ganna Kocherga
- d Ophthalmic Microsurgery Department, International Medical Center Oftalmika , Kharkiv , Ukraine , and
| | - Jörg Kreuter
- e Institute of Pharmaceutical Technology, Goethe University , Frankfurt , Germany
| |
Collapse
|
10
|
Lycium barbarum Polysaccharides Protect against Trimethyltin Chloride-Induced Apoptosis via Sonic Hedgehog and PI3K/Akt Signaling Pathways in Mouse Neuro-2a Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9826726. [PMID: 27143997 PMCID: PMC4838808 DOI: 10.1155/2016/9826726] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/19/2016] [Accepted: 03/10/2016] [Indexed: 02/04/2023]
Abstract
Trimethyltin chloride (TMT) is a classic neurotoxicant that can cause severe neurodegenerative diseases. Some signaling pathways involving cell death play pivotal roles in the central nervous system. In this study, the role of Sonic Hedgehog (Shh) and PI3K/Akt pathways in TMT-induced apoptosis and protective effect of Lycium barbarum polysaccharides (LBP) on mouse neuro-2a (N2a) cells were investigated. Results showed that TMT treatment significantly enhanced apoptosis, upregulated proapoptotic Bax, downregulated antiapoptotic Bcl-2 expression, and increased caspase-3 activity in a dose-dependent manner in N2a cells. TMT induced oxidative stress in cells, performing reactive oxygen species (ROS) and malondialdehyde (MDA) excessive generation, and superoxide dismutase (SOD) activity reduction. TMT significantly decreased phosphorylated glycogen synthase kinase-3β (GSK-3β) and inhibited Shh and PI3K/Akt pathways. However, the addition of LBP upregulated GSK-3β phosphorylation, activated Shh and PI3K/Akt pathways, and eventually reduced apoptosis and oxidative stress caused by TMT. The interaction between Shh and PI3K/Akt pathways was clarified by specific PI3K inhibitor LY294002 or Shh inhibitor GDC-0449. Moreover, LY294002 and GDC-0449 pretreatment both induced phosphorylated GSK-3β downregulation and significantly promoted apoptosis induced by TMT. These results suggest that LBP could reduce TMT-induced N2a cells apoptosis by regulating GSK-3β phosphorylation, Shh, and PI3K/Akt signaling pathways.
Collapse
|
11
|
Weig BC, Richardson JR, Lowndes HE, Reuhl KR. Trimethyltin intoxication induces the migration of ventricular/subventricular zone cells to the injured murine hippocampus. Neurotoxicology 2016; 54:72-80. [PMID: 27045884 DOI: 10.1016/j.neuro.2016.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
Abstract
Following the postnatal decline of cell proliferation in the mammalian central nervous system, the adult brain retains progenitor cells with stem cell-like properties in the subventricular zone (SVZ) and the subgranular zone (SGZ) of the hippocampus. Brain injury can stimulate proliferation and redirect the migration pattern of SVZ precursor cells to the injury site. Sublethal exposure to the neurotoxicant trimethyltin (TMT) causes dose-dependent necrosis and apoptosis in the hippocampus dentate gyrus and increases SGZ stem cell proliferation to generate new granule cells. To determine whether SVZ cells also contribute to the repopulation of the TMT-damaged dentate gyrus, 6-8 week old male C3H mice were injected with the carbocyanine dye spDiI and bromodeoxyuridine (80mg/kg; ip.) to label ventricular cells prior to TMT exposure. The presence of labeled cells in hippocampus was determined 7 and 28days after TMT exposure. No significant change in the number of BrdU(+) and spDiI(+) cells was observed in the dentate gyrus 7days after TMT treatment. However, 28days after TMT treatment there was a 3-4 fold increase in the number of spDiI-labeled cells in the hippocampal hilus and dentate gyrus. Few spDiI(+) cells stained positive for the mature phenotypic markers NeuN or GFAP, suggesting they may represent undifferentiated cells. A small percentage of migrating cells were BrdU(+)/spDiI(+), indicating some newly produced, SVZ- derived precursors migrated to the hippocampus. Taken together, these data suggest that TMT-induced injury of the hippocampus can stimulate the migration of ventricular zone-derived cells to injured dentate gyrus.
Collapse
Affiliation(s)
- Blair C Weig
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy and Joint Program in Toxicology, Rutgers Biomedical Health Sciences, Piscataway, NJ, United States
| | - Jason R Richardson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy and Joint Program in Toxicology, Rutgers Biomedical Health Sciences, Piscataway, NJ, United States; Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Herbert E Lowndes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy and Joint Program in Toxicology, Rutgers Biomedical Health Sciences, Piscataway, NJ, United States
| | - Kenneth R Reuhl
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy and Joint Program in Toxicology, Rutgers Biomedical Health Sciences, Piscataway, NJ, United States.
| |
Collapse
|
12
|
Lee S, Yang M, Kim J, Son Y, Kim J, Kang S, Ahn W, Kim SH, Kim JC, Shin T, Wang H, Moon C. Involvement of BDNF/ERK signaling in spontaneous recovery from trimethyltin-induced hippocampal neurotoxicity in mice. Brain Res Bull 2016; 121:48-58. [PMID: 26772626 DOI: 10.1016/j.brainresbull.2016.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/28/2015] [Accepted: 01/04/2016] [Indexed: 11/17/2022]
Abstract
Trimethyltin (TMT) toxicity causes histopathological damage in the hippocampus and induces seizure behaviors in mice. The lesions and symptoms recover spontaneously over time; however, little is known about the precise mechanisms underlying this recovery from TMT toxicity. We investigated changes in the brain-derived neurotrophic factor/extracellular signal-regulated kinases (BDNF/ERK) signaling pathways in the mouse hippocampus following TMT toxicity. Mice (7 weeks old, C57BL/6) administered TMT (2.6 mg/kg intraperitoneally) showed acute and severe neurodegeneration with increased TUNEL-positive cells in the dentate gyrus (DG) of the hippocampus. The mRNA and protein levels of BDNF in the hippocampus were elevated by TMT treatment. Immunohistochemical analysis showed that TMT treatment markedly increased phosphorylated ERK1/2 expression in the mouse hippocampus 1-4 days after TMT treatment, although the intensity of ERK immunoreactivity in mossy fiber decreased at 1-8 days post-treatment. In addition, ERK-immunopositive cells were localized predominantly in doublecortin-positive immature progenitor neurons in the DG. In primary cultured immature hippocampal neurons (4 days in vitro), BDNF treatment alleviated TMT-induced neurotoxicity, via activation of the ERK signaling pathway. Thus, we suggest that BDNF/ERK signaling pathways may be associated with cell differentiation and survival of immature progenitor neurons, and will eventually lead to spontaneous recovery in TMT-induced hippocampal neurodegeneration.
Collapse
Affiliation(s)
- Sueun Lee
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine, Wonkwang University, Iksan, Jeonbuk 570-740, South Korea
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Juhwan Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Yeonghoon Son
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Jinwook Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Sohi Kang
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Wooseok Ahn
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Sung-Ho Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| | - Taekyun Shin
- College of Veterinary Medicine, Jeju National University, Jeju 690-756, South Korea
| | - Hongbing Wang
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Changjong Moon
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 500-757, South Korea
| |
Collapse
|
13
|
Abud EM, Blurton-Jones M. Could Stem Cells Be Used to Treat or Model Alzheimer’s Disease? Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
14
|
Hachem LD, Mothe AJ, Tator CH. Effect of BDNF and Other Potential Survival Factors in Models of In Vitro Oxidative Stress on Adult Spinal Cord-Derived Neural Stem/Progenitor Cells. Biores Open Access 2015; 4:146-59. [PMID: 26309791 PMCID: PMC4497651 DOI: 10.1089/biores.2014.0058] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Transplantation of neural stem/progenitor cells (NSPCs) is a promising strategy in spinal cord injury (SCI). However, poor survival of transplanted stem cells remains a major limitation of this therapy due to the hostile environment of the injured cord. Oxidative stress is a hallmark in the pathogenesis of SCI; however, its effects on NSPCs from the adult spinal cord have yet to be examined. We therefore developed in vitro models of mild and severe oxidative stress of adult spinal cord-derived NSPCs and used these models to examine potential cell survival factors. NSPCs harvested from the adult rat spinal cord were treated with hydrogen peroxide (H2O2) in vitro to induce oxidative stress. A mild 4 h exposure to H2O2 (500 μM) significantly increased the level of intracellular reactive oxygen species with minimal effect on viability. In contrast, 24 h of oxidative stress led to a marked reduction in cell survival. Pretreatment with brain-derived neurotrophic factor (BDNF) for 48 h attenuated the increase in intracellular reactive oxygen species and enhanced survival. This survival effect was associated with a significant reduction in the number of apoptotic cells and a significant increase in the activity of the antioxidant enzymes glutathione reductase and superoxide dismutase. BDNF treatment had no effect on NSPC differentiation or proliferation. In contrast, cyclosporin A and thyrotropin-releasing hormone had minimal or no effect on NSPC survival. Thus, these models of in vitro oxidative stress may be useful for screening neuroprotective factors administered prior to transplantation to enhance survival of stem cell transplants.
Collapse
Affiliation(s)
- Laureen D Hachem
- Division of Genetics and Development, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network , Ontario, Canada
| | - Andrea J Mothe
- Division of Genetics and Development, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network , Ontario, Canada
| | - Charles H Tator
- Division of Genetics and Development, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network , Ontario, Canada . ; Department of Surgery, Division of Neurosurgery, University of Toronto , Ontario, Canada
| |
Collapse
|
15
|
Shu X, Zhang Y, Xu H, Kang K, Cai D. Brain-derived neurotrophic factor inhibits glucose intolerance after cerebral ischemia. Neural Regen Res 2014; 8:2370-8. [PMID: 25206547 PMCID: PMC4146044 DOI: 10.3969/j.issn.1673-5374.2013.25.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 07/20/2013] [Indexed: 11/18/2022] Open
Abstract
Brain-derived neurotrophic factor is associated with the insulin signaling pathway and glucose tabolism. We hypothesized that expression of brain-derived neurotrophic factor and its receptor may be involved in glucose intolerance following ischemic stress. To verify this hypothesis, this study aimed to observe the changes in brain-derived neurotrophic factor and tyrosine kinase B receptor expression in glucose metabolism-associated regions following cerebral ischemic stress in mice. At day 1 after middle cerebral artery occlusion, the expression levels of brain-derived neurotrophic factor were significantly decreased in the ischemic cortex, hypothalamus, liver, skeletal muscle, and pancreas. The expression levels of tyrosine kinase B receptor were decreased in the hypothalamus and liver, and increased in the skeletal muscle and pancreas, but remained unchanged in the cortex. Intrahypothalamic administration of brain-derived neurotrophic factor (40 ng) suppressed the decrease in insulin receptor and tyrosine-phosphorylated insulin receptor expression in the liver and skeletal muscle, and inhibited the overexpression of gluconeogenesis-associated phosphoenolpyruvate carboxykinase and glucose-6-phosphatase in the liver of cerebral ischemic mice. However, serum insulin levels remained unchanged. Our experimental findings indicate that brain-derived neurotrophic factor can promote glucose metabolism, reduce gluconeogenesis, and decrease blood glucose levels after cerebral ischemic stress. The low expression of brain-derived neurotrophic factor following cerebral ischemia may be involved in the development of glucose intolerance.
Collapse
Affiliation(s)
- Xiaoliang Shu
- Department of Nutrition, Affiliated Dongfang Hospital of Tongji University, Shanghai 200120, China
| | - Yongsheng Zhang
- Department of Nutrition, First Affiliated Hospital of Guangxi Medical University, Nanning 530027, Guangxi Zhuang Autonomous Region, China
| | - Han Xu
- Department of Nutrition, Affiliated Dongfang Hospital of Tongji University, Shanghai 200120, China
| | - Kai Kang
- Department of Nutrition, Affiliated Dongfang Hospital of Tongji University, Shanghai 200120, China
| | - Donglian Cai
- Department of Nutrition, Affiliated Changhai Hospital of the Second Military Medical University of Chinese PLA, Shanghai 200433, China
| |
Collapse
|
16
|
Ma F, Xiao Z, Chen B, Hou X, Han J, Zhao Y, Dai J, Xu R. Accelerating proliferation of neural stem/progenitor cells in collagen sponges immobilized with engineered basic fibroblast growth factor for nervous system tissue engineering. Biomacromolecules 2014; 15:1062-8. [PMID: 24527809 DOI: 10.1021/bm500062n] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neural stem/progenitor cells (NS/PCs) play a therapeutic role in nervous system diseases and contribute to functional recovery. However, their efficacy is limited as the majority of cells die post-transplantation. In this study, collagen sponges were utilized as carriers for NS/PCs. Basic fibroblast growth factor (bFGF), a mitogen for NS/PCs, was incorporated into the collagen sponges to stimulate NS/PC proliferation. However, the effect of native bFGF is limited because it diffuses into the culture medium and is lost following medium exchange. To overcome this problem, a collagen-binding polypeptide domain, which has high affinity to collagen, was fused with bFGF to sustain the exposure of NS/PCs within the collagen sponges to bFGF. The results indicated that the number of NS/PCs was significantly higher in collagen sponges incorporating engineered bFGF than in those with native bFGF or the PBS control after 7 days in culture. Here, we designed a natural biological neural scaffold consisting of collagen sponges, engineered bFGF, and NS/PCs. In addition to the effect of proliferated NS/PCs, the engineered bFGF retained in the natural biological neural scaffolds could have a direct effect on nervous system reconstruction. The two aspects of the natural biological neural scaffolds may produce synergistic effects, and so they represent a promising candidate for nervous system repair.
Collapse
Affiliation(s)
- Fukai Ma
- The Affiliated Bayi Brain Hospital, Bayi Clinical College, Southern Medical University , No. 1838, North of Guangzhou Avenue, Guangzhou 510515, Guangdong, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Wang M, Li P, Liu M, Song W, Wu Q, Fan Y. Potential protective effect of biphasic electrical stimulation against growth factor-deprived apoptosis on olfactory bulb neural progenitor cells through the brain-derived neurotrophic factor-phosphatidylinositol 3'-kinase/Akt pathway. Exp Biol Med (Maywood) 2014; 238:951-9. [PMID: 23970410 DOI: 10.1177/1535370213494635] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stem cell therapy may provide a therapeutic method for the replacement and regeneration of damaged neurons of the central nervous system. However, neural stem cells (NSCs) and neural precursor cells (NPCs) are especially vulnerable after transplantation due to a lack of sufficient growth factors at the transplant site. Electrical stimulation (ES) has recently been found to participate in the regulation of cell proliferation, growth, differentiation, and migration, but its underlying anti-apoptotic effects remain unclear. This study investigated the protective effects of biphasic electrical stimulation (BES) on olfactory bulb NPCs against growth factor-deprived apoptosis, examining the survival and apoptotic features of the cells. Differentiation was assessed by neuronal and glial markers. Brain-derived neurotrophic factor-phosphatidylinositol 3'-kinase (BDNF)-PI3K/Akt pathway activation was determined by enzyme-linked immunosorbent assay and Western blot. The chemical inhibitor wortmannin was used to inhibit the PI3K/Akt pathway. BES exerts a protective effect against growth factor-deprived apoptosis in the NPCs. BES enhanced cell survival and decreased the apoptotic/necrotic rate. Expression of phosphorylated Akt and BDNF secretion increased with BES for 12 h. Furthermore, the protective effects of BES were inhibited by blocking PI3K/AKT signalling. These results suggest that BES prevents growth factor-deprived apoptosis through the BDNF-PI3K/Akt signalling. This work strengthens the opinion that BES may be used as an auxiliary strategy for improving cell survival and preventing cell apoptosis in stem cell-based transplantation therapy.
Collapse
Affiliation(s)
- Menghang Wang
- School of Biological Science and Medical Engineering, Beihang University, Hai Dian District 100191, Beijing, China
| | | | | | | | | | | |
Collapse
|
18
|
Marei HES, Althani A, Afifi N, Abd-Elmaksoud A, Bernardini C, Michetti F, Barba M, Pescatori M, Maira G, Paldino E, Manni L, Casalbore P, Cenciarelli C. Over-expression of hNGF in adult human olfactory bulb neural stem cells promotes cell growth and oligodendrocytic differentiation. PLoS One 2013; 8:e82206. [PMID: 24367504 PMCID: PMC3868548 DOI: 10.1371/journal.pone.0082206] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/21/2013] [Indexed: 12/22/2022] Open
Abstract
The adult human olfactory bulb neural stem/progenitor cells (OBNC/PC) are promising candidate for cell-based therapy for traumatic and neurodegenerative insults. Exogenous application of NGF was suggested as a promising therapeutic strategy for traumatic and neurodegenerative diseases, however effective delivery of NGF into the CNS parenchyma is still challenging due mainly to its limited ability to cross the blood-brain barrier, and intolerable side effects if administered into the brain ventricular system. An effective method to ensure delivery of NGF into the parenchyma of CNS is the genetic modification of NSC to overexpress NGF gene. Overexpression of NGF in adult human OBNSC is expected to alter their proliferation and differentiation nature, and thus might enhance their therapeutic potential. In this study, we genetically modified adult human OBNS/PC to overexpress human NGF (hNGF) and green fluorescent protein (GFP) genes to provide insight about the effects of hNGF and GFP genes overexpression in adult human OBNS/PC on their in vitro multipotentiality using DNA microarray, immunophenotyping, and Western blot (WB) protocols. Our analysis revealed that OBNS/PC-GFP and OBNS/PC-GFP-hNGF differentiation is a multifaceted process involving changes in major biological processes as reflected in alteration of the gene expression levels of crucial markers such as cell cycle and survival markers, stemness markers, and differentiation markers. The differentiation of both cell classes was also associated with modulations of key signaling pathways such MAPK signaling pathway, ErbB signaling pathway, and neuroactive ligand-receptor interaction pathway for OBNS/PC-GFP, and axon guidance, calcium channel, voltage-dependent, gamma subunit 7 for OBNS/PC-GFP-hNGF as revealed by GO and KEGG. Differentiated OBNS/PC-GFP-hNGF displayed extensively branched cytoplasmic processes, a significant faster growth rate and up modulated the expression of oligodendroglia precursor cells markers (PDGFRα, NG2 and CNPase) respect to OBNS/PC-GFP counterparts. These findings suggest an enhanced proliferation and oligodendrocytic differentiation potential for OBNS/PC-GFP-hNGF as compared to OBNS/PC-GFP.
Collapse
Affiliation(s)
- Hany E. S. Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Asmaa Althani
- College of Arts & Sciences, Health Sciences Department, Qatar University, Doha, Qatar
| | - Nahla Afifi
- Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Abd-Elmaksoud
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Camilla Bernardini
- Institute of Anatomy and Cell Biology, Università Cattolica del S. Cuore, Roma, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del S. Cuore, Roma, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del S. Cuore, Roma, Italy
| | - Mario Pescatori
- Department of Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Giulio Maira
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Emanuela Paldino
- Institute of Cell Biology and Neurobiology, National Research Council of Italy, Roma, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy, Roma, Italy
| | - Patrizia Casalbore
- Institute of Cell Biology and Neurobiology, National Research Council of Italy, Roma, Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council of Italy, Roma, Italy
| |
Collapse
|
19
|
Chen A, Xiong LJ, Tong Y, Mao M. Neuroprotective effect of brain-derived neurotrophic factor mediated by autophagy through the PI3K/Akt/mTOR pathway. Mol Med Rep 2013; 8:1011-6. [PMID: 23942837 DOI: 10.3892/mmr.2013.1628] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 07/03/2013] [Indexed: 11/05/2022] Open
Abstract
Brain‑derived neurotrophic factor (BDNF) has been demonstrated to be a potent growth factor that is beneficial in neuronal functions following hypoxia‑ischemia (HI). Mature BDNF triggers three enzymes, mitogen‑activated protein kinase (MAPK), phosphatidylinositol 3‑kinase (PI3K) and phosphoinositide phospholipase C-γ (PLCγ), which are its predominant downstream regulators. The PI3K‑Akt signaling pathway is upstream of the mammalian target of rapamycin (mTOR), which is important in the induction of autophagy. However, whether the neuroprotective effect of BDNF is mediated by autophagy through the PI3K/Akt/mTOR pathway remains to be elucidated. Cortical neurons were cultured following isolation from pregnant rats (gestational days 16‑18). The induction of autophagy following BDNF treatment was analyzed by microtubule‑associated protein light chain 3 (LC3) conversion and autophagosome formation. The phosphorylation of Akt, mTOR and ribosomal protein S6 kinase (p70S6K) was analyzed in cultured cells with or without BDNF treatment. Cell viability was determined by a Cell Counting Kit‑8 for estimating the protective effect of BDNF. Results demonstrated that autophagy was induced in cells with oxygen deprivation. BDNF promoted cell viability via the upregulation of autophagy. Moreover, LC3 upregulation was related to Akt/mTOR/p70S6K inhibition by BDNF. In conclusion, the results suggested that the neuroprotective effect of BDNF was mediated by autophagy through the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ai Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | | | | | |
Collapse
|
20
|
Kim J, Yang M, Kim SH, Kim JC, Wang H, Shin T, Moon C. Possible role of the glycogen synthase kinase-3 signaling pathway in trimethyltin-induced hippocampal neurodegeneration in mice. PLoS One 2013; 8:e70356. [PMID: 23940567 PMCID: PMC3734066 DOI: 10.1371/journal.pone.0070356] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 06/13/2013] [Indexed: 11/18/2022] Open
Abstract
Trimethyltin (TMT) is an organotin compound with potent neurotoxic effects characterized by neuronal destruction in selective regions, including the hippocampus. Glycogen synthase kinase-3 (GSK-3) regulates many cellular processes, and is implicated in several neurodegenerative disorders. In this study, we evaluated the therapeutic effect of lithium, a selective GSK-3 inhibitor, on the hippocampus of adult C57BL/6 mice with TMT treatment (2.6 mg/kg, intraperitoneal [i.p.]) and on cultured hippocampal neurons (12 days in vitro) with TMT treatment (5 µM). Lithium (50 mg/kg, i.p., 0 and 24 h after TMT injection) significantly attenuated TMT-induced hippocampal cell degeneration, seizure, and memory deficits in mice. In cultured hippocampal neurons, lithium treatment (0–10 mM; 1 h before TMT application) significantly reduced TMT-induced cytotoxicity in a dose-dependent manner. Additionally, the dynamic changes in GSK-3/β-catenin signaling were observed in the mouse hippocampus and cultured hippocampal neurons after TMT treatment with or without lithium. Therefore, lithium inhibited the detrimental effects of TMT on the hippocampal neurons in vivo and in vitro, suggesting involvement of the GSK-3/β-catenin signaling pathway in TMT-induced hippocampal cell degeneration and dysfunction.
Collapse
Affiliation(s)
- Juhwan Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju, Republic of Korea
| | - Miyoung Yang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju, Republic of Korea
- Department of Physiology and Neurosceince Program, Michigan State University, East Lansing, Michigan, United States of America
| | - Sung-Ho Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju, Republic of Korea
| | - Jong-Choon Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju, Republic of Korea
| | - Hongbing Wang
- Department of Physiology and Neurosceince Program, Michigan State University, East Lansing, Michigan, United States of America
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju, Republic of Korea
- * E-mail: (TS); (CM)
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju, Republic of Korea
- * E-mail: (TS); (CM)
| |
Collapse
|
21
|
Ishihara Y, Kawami T, Ishida A, Yamazaki T. Allopregnanolone-mediated protective effects of progesterone on tributyltin-induced neuronal injury in rat hippocampal slices. J Steroid Biochem Mol Biol 2013; 135:1-6. [PMID: 23280249 DOI: 10.1016/j.jsbmb.2012.12.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 12/20/2012] [Accepted: 12/20/2012] [Indexed: 01/12/2023]
Abstract
Increasing evidence shows that progesterone, a neuroactive steroid, has protective actions in central nervous system, but there is little evidence to show the protective mechanism of progesterone on neurotoxicity induced by environmental chemicals. In this study, we examined the effects of progesterone on neuronal injury induced by tributyltin (TBT) in rat hippocampal slices. Treatment with progesterone dose-dependently suppressed hippocampal neuronal injury induced by TBT. The neuroprotective action of progesterone was completely canceled with pretreatment by finasteride, a 5α-reductase inhibitor, but it was not affected by mifepristone, a progesterone receptor antagonist, or by SU-10603, a cytochrome P450 17α inhibitor. The content of allopregnanolone in the slices was significantly increased by treatment with progesterone, and this increment was greatly suppressed with a pretreatment of finasteride. Treatment with allopregnanolone attenuated neuronal injury induced by TBT in a dose-dependent manner. The neuroprotective effects not only of progesterone but also of allopregnanolone were canceled by bicuculline, a potent gamma-aminobutyric acid A (GABAA) receptor antagonist. Pretreatment with muscimol, a GABAA receptor agonist, attenuated hippocampal neuronal injury elicited by TBT. Taken together, allopregnanolone converted from progesterone in hippocampal slices could protect neurons from TBT-induced neurotoxicity due to a GABAA receptor-dependent mechanism. One of the physiological roles of neuroactive steroids might be neuroprotection from environmental chemicals.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | | | | | | |
Collapse
|
22
|
Shukla A, Mohapatra TM, Agrawal AK, Parmar D, Seth K. Salsolinol induced apoptotic changes in neural stem cells: amelioration by neurotrophin support. Neurotoxicology 2013; 35:50-61. [PMID: 23261589 DOI: 10.1016/j.neuro.2012.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 12/14/2012] [Accepted: 12/14/2012] [Indexed: 11/20/2022]
Abstract
Salsolinol (SAL), a catechol isoquinoline has invited considerable attention due to its structural similarity with dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Its high endogenous presence in Parkinsonian brain implicated its possible association with the disease process. SAL is also present in alcohol beverages and certain food materials and can get access to brain especially in conditions of immature or impaired BBB. Besides this, the effect of SAL on neural stem cells (NSCs) which are potential candidates for adult neurogenesis and transplantation mediated rejuvenating attempts for Parkinson's disease (PD) brain has not been known so far. NSCs in both the cases have to overcome suppressive cues of diseased brain for their survival and function. In this study we explored the toxicity of SAL toward NSCs focusing on apoptosis and status of PI3K survival signaling. NSCs cultured from embryonic day 11 rat fetal brain including those differentiated to TH(+ve) colonies, when challenged with SAL (1-100μM), elicited a concentration and time dependent cell death/loss of mitochondrial viability. 10μM SAL on which significant mitochondrial impairment initiated was further used to study mechanism of toxicity. Morphological impairment, enhanced TUNEL positivity, cleaved caspase-3 and decreased Bcl-2:Bax suggested apoptosis. Sal toxicity coincided with reduced pAkt level and its downstream effectors: pCREB, pGSK-3β, Bcl-2 and neurotrophins GDNF, BDNF suggesting repressed PI3K/Akt signaling. Multiple neurotrophic factor support in the form of Olfactory Ensheathing Cell's Conditioned Media (OEC CM) potentially protected NSCs against SAL through activating PI3K/Akt pathway. This was confirmed on adding LY294002 the PI3K inhibitor which abolished the protection. We inferred that SAL exerts substantial toxicity toward NSCs. These findings will lead to better understanding of endogenous threats that might affect the fate of transplanted NSCs and their probable antidotes.
Collapse
Affiliation(s)
- A Shukla
- Indian Institute of Toxicology Research (CSIR), Developmental Toxicology Division, Mahatma Gandhi Marg, Post Box 80, Lucknow 226 001, India; Department of Microbiology, IMS, Banaras Hindu University, Varanasi 221 005, India
| | | | | | | | | |
Collapse
|
23
|
Chen A, Xiong LJ, Tong Y, Mao M. The neuroprotective roles of BDNF in hypoxic ischemic brain injury. Biomed Rep 2012; 1:167-176. [PMID: 24648914 DOI: 10.3892/br.2012.48] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 10/16/2012] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-ischemia (H/I) brain injury results in various degrees of damage to the body, and the immature brain is particularly fragile to oxygen deprivation. Hypothermia and erythropoietin (EPO) have long been known to be neuroprotective in ischemic brain injury. Brain-derived neurotrophic factor (BDNF) has recently been recognized as a potent modulator capable of regulating a wide repertoire of neuronal functions. This review was based on studies concerning the involvement of BDNF in the protection of H/I brain injury following a search in PubMed between 1995 and December, 2011. We initially examined the background of BDNF, and then focused on its neuroprotective mechanisms against ischemic brain injury, including its involvement in promoting neural regeneration/cognition/memory rehabilitation, angiogenesis within ischemic penumbra and the inhibition of the inflammatory process, neurotoxicity, epilepsy and apoptosis. We also provided a literature overview of experimental studies, discussing the safety and the potential clinical application of BDNF as a neuroprotective agent in the ischemic brain injury.
Collapse
Affiliation(s)
- Ai Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li-Jing Xiong
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Tong
- Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Chengdu, Sichuan 610041, P.R. China ; ; Laboratory of Early Developmental and Injuries, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Meng Mao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
24
|
Neuroprotective strategies in hippocampal neurodegeneration induced by the neurotoxicant trimethyltin. Neurochem Res 2012. [PMID: 23179590 DOI: 10.1007/s11064-012-0932-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The selective vulnerability of specific neuronal subpopulations to trimethyltin (TMT), an organotin compound with neurotoxicant effects selectively involving the limbic system and especially marked in the hippocampus, makes it useful to obtain in vivo models of neurodegeneration associated with behavioural alterations, such as hyperactivity and aggression, cognitive impairment as well as temporal lobe epilepsy. TMT has been widely used to study neuronal and glial factors involved in selective neuronal death, as well as the molecular mechanisms leading to hippocampal neurodegeneration (including neuroinflammation, excitotoxicity, intracellular calcium overload, mitochondrial dysfunction and oxidative stress). It also offers a valuable instrument to study the cell-cell interactions and signalling pathways that modulate injury-induced neurogenesis, including the involvement of newly generated neurons in the possible repair processes. Since TMT appears to be a useful tool to damage the brain and study the various responses to damage, this review summarises current data from in vivo and in vitro studies on neuroprotective strategies to counteract TMT-induced neuronal death, that may be useful to elucidate the role of putative candidates for translational medical research on neurodegenerative diseases.
Collapse
|
25
|
Chen BY, Wang X, Wang ZY, Wang YZ, Chen LW, Luo ZJ. Brain-derived neurotrophic factor stimulates proliferation and differentiation of neural stem cells, possibly by triggering the Wnt/β-catenin signaling pathway. J Neurosci Res 2012; 91:30-41. [DOI: 10.1002/jnr.23138] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 07/27/2012] [Accepted: 08/01/2012] [Indexed: 12/11/2022]
|