1
|
Salama A, Elgohary R, M Amin M, Elwahab SA. Immunomodulatory effect of protocatechuic acid on cyclophosphamide induced brain injury in rat: Modulation of inflammosomes NLRP3 and SIRT1. Eur J Pharmacol 2022; 932:175217. [PMID: 36007603 DOI: 10.1016/j.ejphar.2022.175217] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022]
Abstract
Modulation of the inflammasome NLRP3 and SIRT1 are new combat strategy for brain injury protection. The inflammasome activates proinflammatory cytokines releasing interleukin-1β and interleukin-18 which in turn affect the toxins release from immune cells. In addition, SIRT1 controls many biological functions, such as immune response and oxidative stress. Protocatechuic has versatile biological activities and possesses antioxidant, anti-inflammatory and neuroprotective effects. So this work aims to study immunomodulatory effect of protocatechuic acid on cyclophosphamide chemotherapy drug-induced brain injury via modulation of inflammosomes NLRP3 and SIRT1. Rats were randomly assigned to four experimental groups. Normal control group was injected with a single i.p injection of saline. Cyclophosphamide group was injected with a single i.p injection of cyclophosphamide (200 mg/kg). Protocatechuic acid groups were orally administered (50 &100 mg/kg) once daily for 10 consecutive days after cyclophosphamide injection. Protocatechuic acid administration exhibited improvements of the cognition function and memory, a reduction in brain contents of MDA, NLRP3, IL-1 β, NF-κB, IKBKB and Galectin 3 and an elevation of GSH and SIRT1 compared to cyclophosphamide group. In addition, protocatechuic acid administration ameliorated the elevation of caspase 3 and iNOS gene expression and alleviated the neuron degeneration caused by cyclophosphamide. In conclusion, the therapeutic action of protocatechuic acid and its cellular and molecular mechanisms are new insights against various human ailments, especially, neuroprotective disease as brain injury induced by cyclophosphamide chemotherapy drug in rats through modulation of inflammosomes NLRP3 and SIRT1.
Collapse
Affiliation(s)
- Abeer Salama
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), 12622, Dokki, Cairo, Egypt
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), 12622, Dokki, Cairo, Egypt.
| | - Mohamed M Amin
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El Buhouth St. (Former El-Tahrir St.), 12622, Dokki, Cairo, Egypt
| | - Sahar Abd Elwahab
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
2
|
Sinomenine Attenuates Trimethyltin-Induced Cognitive Decline via Targeting Hippocampal Oxidative Stress and Neuroinflammation. J Mol Neurosci 2022; 72:1609-1621. [PMID: 35543800 DOI: 10.1007/s12031-022-02021-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Sinomenine is the main bioactive ingredient of the medicinal plant Sinomenium acutum with neuroprotective potential. This study was designed to assess beneficial effect of sinomenine in alleviation of trimethyltin (TMT)-induced cognitive dysfunction. TMT was administered i.p. (8 mg/kg, once) and sinomenine was daily given p.o. 1 h after TMT for 3 weeks at doses of 25 or 100 mg/kg. Cognitive performance was assessed in various behavioral tests. In addition, oxidative stress- and inflammation-associated factors were measured and histochemical evaluation of the hippocampus was conducted. Sinomenine at a dose of 100 mg/kg significantly and partially increased discrimination index in novel object recognition (NOR), improved alternation in short-term Y maze, increased step-through latency in passive avoidance paradigm, and also reduced probe trial errors and latency in the Barnes maze task. Moreover, sinomenine somewhat prevented inappropriate hippocampal changes of malondialdehyde (MDA), reactive oxygen species (ROS), protein carbonyl, nitrite, superoxide dismutase (SOD), tumor necrosis factor α (TNFα), interleukin 6 (IL 6), acetylcholinesterase (AChE) activity, beta secretase 1 (BACE 1) activity, and mitochondrial membrane potential (MMP) with no significant effect on glutathione (GSH), catalase, glutathione reductase, glutathione peroxidase, and myeloperoxidase (MPO). In addition, lower reactivity (IRA) for glial fibrillary acidic protein (GFAP) as an index of astrocyte activity was observed and loss of CA1 pyramidal neurons was attenuated following sinomenine treatment. This study demonstrated that sinomenine could lessen TMT-induced cognitive dysfunction which is partly due to its attenuation of hippocampal oxidative stress and neuroinflammation.
Collapse
|
3
|
Nio-Kobayashi J, Itabashi T. Galectins and Their Ligand Glycoconjugates in the Central Nervous System Under Physiological and Pathological Conditions. Front Neuroanat 2021; 15:767330. [PMID: 34720894 PMCID: PMC8554236 DOI: 10.3389/fnana.2021.767330] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Galectins are β-galactoside-binding lectins consisting of 15 members in mammals. Galectin-1,-3,-4,-8, and -9 are predominantly expressed in the central nervous system (CNS) and regulate various physiological and pathological events. This review summarizes the current knowledge of the cellular expression and role of galectins in the CNS, and discusses their functions in neurite outgrowth, myelination, and neural stem/progenitor cell niches, as well as in ischemic/hypoxic/traumatic injuries and neurodegenerative diseases such as multiple sclerosis. Galectins are expressed in both neurons and glial cells. Galectin-1 is mainly expressed in motoneurons, whereas galectin-3-positive neurons are broadly distributed throughout the brain, especially in the hypothalamus, indicating its function in the regulation of homeostasis, stress response, and the endocrine/autonomic system. Astrocytes predominantly contain galectin-1, and galectin-3 and−9 are upregulated along with its activation. Activated, but not resting, microglia contain galectin-3, supporting its phagocytic activity. Galectin-1,−3, and -4 are characteristically expressed during oligodendrocyte differentiation. Galectin-3 from microglia promotes oligodendrocyte differentiation and myelination, while galectin-1 and axonal galectin-4 suppress its differentiation and myelination. Galectin-1- and- 3-positive cells are involved in neural stem cell niche formation in the subventricular zone and hippocampal dentate gyrus, and the migration of newly generated neurons and glial cells to the olfactory bulb or damaged lesions. In neurodegenerative diseases, galectin-1,-8, and -9 have neuroprotective and anti-inflammatory activities. Galectin-3 facilitates pro-inflammatory action; however, it also plays an important role during the recovery period. Several ligand glycoconjugates have been identified so far such as laminin, integrins, neural cell adhesion molecule L1, sulfatide, neuropilin-1/plexinA4 receptor complex, triggering receptor on myeloid cells 2, and T cell immunoglobulin and mucin domain. N-glycan branching on lymphocytes and oligodendroglial progenitors mediated by β1,6-N-acetylglucosaminyltransferase V (Mgat5/GnTV) influences galectin-binding, modulating inflammatory responses and remyelination in neurodegenerative diseases. De-sulfated galactosaminoglycans such as keratan sulfate are potential ligands for galectins, especially galectin-3, regulating neural regeneration. Galectins have multitudinous functions depending on cell type and context as well as post-translational modifications, including oxidization, phosphorylation, S-nitrosylation, and cleavage, but there should be certain rules in the expression patterns of galectins and their ligand glycoconjugates, possibly related to glucose metabolism in cells.
Collapse
Affiliation(s)
- Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tetsuya Itabashi
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
4
|
Liu M, Pi H, Xi Y, Wang L, Tian L, Chen M, Xie J, Deng P, Zhang T, Zhou C, Liang Y, Zhang L, He M, Lu Y, Chen C, Yu Z, Zhou Z. KIF5A-dependent axonal transport deficiency disrupts autophagic flux in trimethyltin chloride-induced neurotoxicity. Autophagy 2021; 17:903-924. [PMID: 32160081 PMCID: PMC8078766 DOI: 10.1080/15548627.2020.1739444] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 02/22/2020] [Accepted: 03/02/2020] [Indexed: 01/18/2023] Open
Abstract
Trimethyltin chloride (TMT) is widely used as a constituent of fungicides and plastic stabilizers in the industrial and agricultural fields, and is generally acknowledged to have potent neurotoxicity, especially in the hippocampus; however, the mechanism of induction of neurotoxicity by TMT remains elusive. Herein, we exposed Neuro-2a cells to different concentrations of TMT (2, 4, and 8 μM) for 24 h. Proteomic analysis, coupled with bioinformatics analysis, revealed the important role of macroautophagy/autophagy-lysosome machinery in TMT-induced neurotoxicity. Further analysis indicated significant impairment of autophagic flux by TMT via suppressed lysosomal function, such as by inhibiting lysosomal proteolysis and changing the lysosomal pH, thereby contributing to defects in autophagic clearance and subsequently leading to nerve cell death. Mechanistically, molecular interaction networks of Ingenuity Pathway Analysis identified a downregulated molecule, KIF5A (kinesin family member 5A), as a key target in TMT-impaired autophagic flux. TMT decreased KIF5A protein expression, disrupted the interaction between KIF5A and lysosome, and impaired lysosomal axonal transport. Moreover, Kif5a overexpression restored axonal transport, increased lysosomal dysfunction, and antagonized TMT-induced neurotoxicity in vitro. Importantly, in TMT-administered mice with seizure symptoms and histomorphological injury in the hippocampus, TMT inhibited KIF5A expression in the hippocampus. Gene transfer of Kif5a enhanced autophagic clearance in the hippocampus and alleviated TMT-induced neurotoxicity in vivo. Our results are the first to demonstrate KIF5A-dependent axonal transport deficiency to cause autophagic flux impairment via disturbance of lysosomal function in TMT-induced neurotoxicity; manipulation of KIF5A may be a therapeutic approach for antagonizing TMT-induced neurotoxicity.Abbreviations: 3-MA: 3-methyladenine; AAV: adeno-associated virus; ACTB: actin beta; AGC: automatic gain control; ATG: autophagy-related; ATP6V0D1: ATPase H+ transporting lysosomal V0 subunit D1; ATP6V1E1: ATPase H+ transporting lysosomal V1 subunit E1; CA: cornu ammonis; CQ: chloroquine; CTSB: cathepsin B; CTSD: cathepsin D; DCTN1: dynactin subunit 1; DG: dentate gyrus; DYNLL1: dynein light chain LC8-type 1; FBS: fetal bovine serum; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; GABARAPL2: GABA type A receptor associated protein like 2; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; IPA: Ingenuity Pathway Analysis; KEGG: Kyoto Encyclopedia of Genes and Genomes; KIF5A: kinesin family member 5A; LAMP: lysosomal-associated membrane protein; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PBS: phosphate-buffered saline; PFA: paraformaldehyde; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PRM: parallel reaction monitoring; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; SYP: synaptophysin; TAX1BP1: Tax1 binding protein 1; TMT: trimethyltin chloride; TUB: tubulin.
Collapse
Affiliation(s)
- Mengyu Liu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing, China
- School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yu Xi
- Department of Environmental Medicine, and Department of Emergency Medicine of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Mengyan Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Tao Zhang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Chao Zhou
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yidan Liang
- Department of Cell Biology, School of Life Sciences and School of Medicine, Guangxi University, Nanning, China
| | - Lei Zhang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Mindi He
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhengping Yu
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Environmental Medicine, and Department of Emergency Medicine of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Lee S, Seo YH, Song JH, Kim WJ, Lee JH, Moon BC, Ang MJ, Kim SH, Moon C, Lee J, Kim JS. Neuroprotective Effect of Protaetia brevitarsis seulensis' Water Extract on Trimethyltin-Induced Seizures and Hippocampal Neurodegeneration. Int J Mol Sci 2021; 22:ijms22020679. [PMID: 33445535 PMCID: PMC7827571 DOI: 10.3390/ijms22020679] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/25/2020] [Accepted: 01/08/2021] [Indexed: 12/22/2022] Open
Abstract
This study aimed to investigate whether the Protaetia brevitarsis seulensis (PB)’ water extract (PBWE) ameliorates trimethyltin (TMT)-induced seizures and hippocampal neurodegeneration. To investigate the potential neuroprotective effect of the PBWE in vitro, a lactate dehydrogenase (LDH) assay was conducted in TMT-treated primary cultures of mouse hippocampal neurons. In TMT-treated adult C57BL/6 mice, behavioral and histopathological changes were evaluated by seizure scoring and Fluoro-Jade C staining, respectively. In our in vitro assay, we observed that pretreating mice hippocampal neuron cultures with the PBWE reduced TMT-induced cytotoxicity, as indicated by the decreased LDH release. Furthermore, pretreatment with the PBWE alleviated seizures and hippocampal neurodegeneration in TMT-treated mice. The antioxidant activity of the PBWE increased in a dose-dependent manner; moreover, pretreatment with the PBWE mitigated the TMT-induced Nrf2 stimulation. In addition, six major compounds, including adenine, hypoxanthine, uridine, adenosine, inosine, and benzoic acid, were isolated from the PBWE, and among them, inosine and benzoic acid have been confirmed to have an essential antioxidative activity. In conclusion, the PBWE ameliorated TMT-induced toxicity in hippocampal neurons in both in vitro and in vivo assays, through a potential antioxidative effect. Our findings suggest that the PBWE may have pharmacotherapeutic potential in neurodegenerative diseases such as seizures or epilepsy.
Collapse
Affiliation(s)
- Sueun Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
| | - Young Hye Seo
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
| | - Jun Ho Song
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
| | - Wook Jin Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
| | - Ji Hye Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
| | - Byeong Cheol Moon
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
| | - Mary Jasmin Ang
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, Korea; (M.J.A.); (S.H.K.); (C.M.)
| | - Sung Ho Kim
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, Korea; (M.J.A.); (S.H.K.); (C.M.)
| | - Changjong Moon
- College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, Korea; (M.J.A.); (S.H.K.); (C.M.)
| | - Jun Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
- Correspondence: (J.L.); (J.S.K.); Tel.: +82-61-338-7129 (J.L.); +82-61-338-7111 (J.S.K.)
| | - Joong Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Korea; (S.L.); (Y.H.S.); (J.H.S.); (W.J.K.); (J.H.L.); (B.C.M.)
- Correspondence: (J.L.); (J.S.K.); Tel.: +82-61-338-7129 (J.L.); +82-61-338-7111 (J.S.K.)
| |
Collapse
|
6
|
Ramírez Hernández E, Sánchez-Maldonado C, Mayoral Chávez MA, Hernández-Zimbrón LF, Patricio Martínez A, Zenteno E, Limón Pérez de León ID. The therapeutic potential of galectin-1 and galectin-3 in the treatment of neurodegenerative diseases. Expert Rev Neurother 2020; 20:439-448. [PMID: 32303136 DOI: 10.1080/14737175.2020.1750955] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Neuroinflammation has been proposed as a common factor and one of the main inducers of neuronal degeneration. Galectins are a group of β-galactoside-binding lectins, that play an important role in the immune response, adhesion, proliferation, differentiation, migration and cell growth. Up to 15 members of the galectin's family have been identified; however, the expression of galectin-1 and galectin-3 has been considered a key factor in neuronal regeneration and modulation of the inflammatory response. Galectin-1 is necessary to stimulate the secretion of neurotrophic factors in astrocytes and promoting neuronal regeneration. In contrast, galectin-3 fosters the proliferation of microglial cells and modulates cellular apoptosis, therefore these proteins are considered a useful alternative for the treatment of degenerative diseases.Areas covered: This review describes the roles of galectin-1 and galectin-3 in the modulation of neuroinflammation and their potential as therapeutic targets in the treatment for neurodegenerative diseases.Expert opinion: Although data in the literature vary, the effects of galectin-1 and galectin-3 on the activation and modulation of astrocytes and microglia has been described. Due to its anti-inflammatory effects, galectin-1 is proposed as a molecule with therapeutic potential, whereas the inhibition of galectin-3 could contribute to reduce the neuroinflammatory response in neurodegenerative diseases.
Collapse
Affiliation(s)
- Eleazar Ramírez Hernández
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, México.,Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Claudia Sánchez-Maldonado
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Miguel A Mayoral Chávez
- Centro de Investigaciones Médicas UNAM-UABJO, Facultad de Medicina, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| | - Luis F Hernández-Zimbrón
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México.,Departamento de Investigación, Asociación Para Evitar la Ceguera en México, "Hospital Dr. Luis Sánchez Bulnes", Ciudad de México, México
| | - Aleidy Patricio Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, México.,Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - I Daniel Limón Pérez de León
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, México
| |
Collapse
|
7
|
Protective Effects of Scolopendra Water Extract on Trimethyltin-Induced Hippocampal Neurodegeneration and Seizures in Mice. Brain Sci 2019; 9:brainsci9120369. [PMID: 31842431 PMCID: PMC6955677 DOI: 10.3390/brainsci9120369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/15/2023] Open
Abstract
Trimethyltin (TMT) is an organotin compound with potent neurotoxic action characterized by neuronal degeneration in the hippocampus. This study evaluated the protective effects of a Scolopendra water extract (SWE) against TMT intoxication in hippocampal neurons, using both in vitro and in vivo model systems. Specifically, we examined the actions of SWE on TMT- (5 mM) induced cytotoxicity in primary cultures of mouse hippocampal neurons (7 days in vitro) and the effects of SWE on hippocampal degeneration in adult TMT- (2.6 mg/kg, intraperitoneal) treated C57BL/6 mice. We found that SWE pretreatment (0–100 μg/mL) significantly reduced TMT-induced cytotoxicity in cultured hippocampal neurons in a dose-dependent manner, as determined by lactate dehydrogenase and 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assays. Additionally, this study showed that perioral administration of SWE (5 mg/kg), from −6 to 0 days before TMT injection, significantly attenuated hippocampal cell degeneration and seizures in adult mice. Furthermore, quantitative analysis of Iba-1 (Allograft inflammatory factor 1)- and GFAP (Glial fibrillary acidic protein)-immunostained cells revealed a significant reduction in the levels of Iba-1- and GFAP-positive cell bodies in the dentate gyrus (DG) of mice treated with SWE prior to TMT injection. These data indicated that SWE pretreatment significantly protected the hippocampus against the massive activation of microglia and astrocytes elicited by TMT. In addition, our data showed that the SWE-induced reduction of immune cell activation was linked to a significant reduction in cell death and a significant improvement in TMT-induced seizure behavior. Thus, we conclude that SWE ameliorated the detrimental effects of TMT toxicity on hippocampal neurons, both in vivo and in vitro. Altogether, our findings hint at a promising pharmacotherapeutic use of SWE in hippocampal degeneration and dysfunction.
Collapse
|
8
|
Xue J, Yu Y, Zhang X, Zhang C, Zhao Y, Liu B, Zhang L, Wang L, Chen R, Gao X, Jiao P, Song G, Jiang XC, Qin S. Sphingomyelin Synthase 2 Inhibition Ameliorates Cerebral Ischemic Reperfusion Injury Through Reducing the Recruitment of Toll-Like Receptor 4 to Lipid Rafts. J Am Heart Assoc 2019; 8:e012885. [PMID: 31718447 PMCID: PMC6915272 DOI: 10.1161/jaha.119.012885] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Inflammation is recognized as an important contributor of ischemia/reperfusion (I/R) damage after ischemic stroke. Sphingomyelin synthase 2 (SMS2), the key enzyme for the biosynthesis of sphingomyelin, can function as a critical mediator of inflammation. In the present study, we investigated the role of SMS2 in a mouse model of cerebral I/R. Methods and Results Cerebral I/R was induced by 60‐minute transient middle cerebral artery occlusion in SMS2 knockout (SMS2‐/‐) mice and wild‐type mice. Brain injury was determined by neurological deficits and infarct volume at 24 and 72 hours after transient middle cerebral artery occlusion. Microglia activation and inflammatory factors were detected by immunofluorescence staining, flow cytometry, western blot, and RT‐PCR. SMS2 deficiency significantly improved neurological function and minimized infarct volume at 72 hours after transient middle cerebral artery occlusion. The neuroprotective effects of SMS2 deficiency were associated with (1) suppression of microglia activation through Toll‐like receptor 4/nuclear factor kappa‐light‐chain‐enhancer of activated B cells pathway and (2) downregulation of the level of galactin‐3 and other proinflammatory cytokines. The mechanisms underlying the beneficial effects of SMS2 deficiency may include altering sphingomyelin components in lipid raft fractions, thus impairing the recruitment of Toll‐like receptor 4 to lipid rafts and subsequently reducing Toll‐like receptor 4/myeloid differentiation factor 2 complex formation on the surface of microglia. Conclusions SMS2 deficiency ameliorated inflammatory injury after cerebral I/R in mice, and SMS2 may be a key modulator of Toll‐like receptor 4/nuclear factor kappa‐light‐chain‐enhancer of activated B cells activation by disturbing the membrane component homeostasis during cerebral I/R.
Collapse
Affiliation(s)
- Jing Xue
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Yang Yu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis Shandong First Medical University & Shandong Academy of Medical Sciences Taian China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Xiangjian Zhang
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Cong Zhang
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Yanan Zhao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis Shandong First Medical University & Shandong Academy of Medical Sciences Taian China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Boyan Liu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis Shandong First Medical University & Shandong Academy of Medical Sciences Taian China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Lan Zhang
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Lina Wang
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Rong Chen
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Xuan Gao
- Department of Neurology Second Hospital of Hebei Medical University Shijiazhuang China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis Shandong First Medical University & Shandong Academy of Medical Sciences Taian China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Guohua Song
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis Shandong First Medical University & Shandong Academy of Medical Sciences Taian China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| | - Xian-Cheng Jiang
- Department of Anatomy and Cell Biology SUNY Downstate Medical Center Brooklyn NY
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis Shandong First Medical University & Shandong Academy of Medical Sciences Taian China.,Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis Shijiazhuang China
| |
Collapse
|
9
|
Chemotherapy-induced neuroinflammation is associated with disrupted colonic and bacterial homeostasis in female mice. Sci Rep 2019; 9:16490. [PMID: 31712703 PMCID: PMC6848141 DOI: 10.1038/s41598-019-52893-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy treatment negatively affects the nervous and immune systems and alters gastrointestinal function and microbial composition. Outside of the cancer field, alterations in commensal bacteria and immune function have been implicated in behavioral deficits; however, the extent to which intestinal changes are related to chemotherapy-associated behavioral comorbidities is not yet known. Thus, this study identified concurrent changes in behavior, central and peripheral immune activation, colon histology, and bacterial community structure in mice treated with paclitaxel chemotherapy. In paclitaxel-treated mice, increased fatigue and decreased cognitive performance occurred in parallel with reduced microglia immunoreactivity, increased circulating chemokine expression (CXCL1), as well as transient increases in pro-inflammatory cytokine/chemokine (Il-1β, Tnfα, Il-6, and Cxcl1) gene expression in the brain. Furthermore, mice treated with paclitaxel had altered colonic bacterial community composition and increased crypt depth. Relative abundances of multiple bacterial taxa were associated with paclitaxel-induced increases in colon mass, spleen mass, and microglia activation. Although microbial community composition was not directly related to available brain or behavioral measures, structural differences in colonic tissue were strongly related to microglia activation in the dentate gyrus and the prefrontal cortex. These data indicate that the chemotherapeutic paclitaxel concurrently affects the gut microbiome, colonic tissue integrity, microglia activation, and fatigue in female mice, thus identifying a novel relationship between colonic tissue integrity and behavioral responses that is not often assessed in studies of the brain-gut-microbiota axis.
Collapse
|
10
|
Ramírez E, Sánchez-Maldonado C, Mayoral MA, Mendieta L, Alatriste V, Patricio-Martínez A, Limón ID. Neuroinflammation induced by the peptide amyloid-β (25-35) increase the presence of galectin-3 in astrocytes and microglia and impairs spatial memory. Neuropeptides 2019; 74:11-23. [PMID: 30795916 DOI: 10.1016/j.npep.2019.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 01/05/2023]
Abstract
Galectins are animal lectins that bind to β-galactosides, such as lactose and N-acetyllactosamine, contained in glycoproteins or glycolipids. Galectin-1 (Gal-1) and Galectin-3 (Gal-3) are involved in pathologies associated with the inflammatory process, cell proliferation, adhesion, migration, and apoptosis. Recent evidence has shown that the administration of Amyloid-β 25-35 (Aβ25-35) into the hippocampus of rats increases the inflammatory response that is associated with memory impairment and neurodegeneration. Galectins could participate in the modulation of the neuroinflammation induced by the Aβ25-35. The aim of this study was to evaluate the presence of Gal-1 and Gal-3 in the neuroinflammation induced by administration of Aβ25-35 into the hippocampus and to examine spatial memory in the Morris water maze. After the administration of Aβ25-35, animals were tested for learning and spatial memory in the Morris water maze. Behavioral performance showed that Aβ25-35 didn't affect spatial learning but did impair memory, with animals taking longer to find the platform. On the day 32, hippocampus was examined for astrocytes (GFAP), microglia (Iba1), Gal-1 and Gal-3 via immunohistochemical analysis, and the cytokines IL-1β, TNF-α, IFN-γ by ELISA. This study's results showed a significant increase in the expression of Gal-3 in the microglia and astrocytes, while Gal-1 didn't increase in the dorsal hippocampus. The expression of galectins is associated with increased cytokines in the hippocampal formation of Aβ25-35 treated rats. These findings suggest that Gal-3 could participate in the inflammation induced by administration of Aβ25-35 and could be involved in the neurodegeneration progress and memory impairment.
Collapse
Affiliation(s)
- Eleazar Ramírez
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico
| | | | | | - Liliana Mendieta
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico
| | | | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico; Facultad de Ciencias Biológicas, BUAP, Puebla, Mexico
| | - I Daniel Limón
- Laboratorio de Neurofarmacología, 105 C-FCQ BUAP, Puebla, Mexico.
| |
Collapse
|
11
|
Lee S, Kang S, Kim J, Yoon S, Kim SH, Moon C. Enhanced expression of immediate-early genes in mouse hippocampus after trimethyltin treatment. Acta Histochem 2016; 118:679-684. [PMID: 27614947 DOI: 10.1016/j.acthis.2016.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/27/2022]
Abstract
Immediate-early genes (IEGs) are transiently and rapidly activated in response to various cellular stimuli. IEGs mediate diverse functions during pathophysiologic events by regulating cellular signal transduction. We investigated the temporal expression of several IEGs, including c-fos, early growth response protein-1 (Egr-1), and activity-regulated cytoskeleton-associated protein (Arc), in trimethyltin (TMT)-induced hippocampal neurodegeneration. Mice (7 weeks old, C57BL/6) administered TMT (2.6mg/kg intraperitoneally) presented severe neurodegenerative lesions in the dentate gyrus (DG) and showed behavioral seizure activity on days 1-4 post-treatment, after which the lesions and behavior recovered spontaneously over time. c-fos, Egr-1, and Arc mRNA and protein levels significantly increased in the mouse hippocampus after TMT treatment. Immunohistochemical analysis showed that nuclear c-fos expression increased mainly in the DG, whereas nuclear Egr-1 expression was increased extensively in cornu ammonis (CA) 1, CA3, and the DG after TMT treatment. Increased Arc levels were detected in the cellular somata/dendrites of the hippocampal subregions after TMT treatment. Therefore, we suggest that increased IEGs are associated with TMT-induced pathological events in mouse hippocampus.
Collapse
Affiliation(s)
- Sueun Lee
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Juhwan Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Seongwook Yoon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Sung-Ho Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Animal Medical Institute, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
12
|
Lee S, Yang M, Kim J, Kang S, Kim J, Kim JC, Jung C, Shin T, Kim SH, Moon C. Trimethyltin-induced hippocampal neurodegeneration: A mechanism-based review. Brain Res Bull 2016; 125:187-99. [PMID: 27450702 DOI: 10.1016/j.brainresbull.2016.07.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
Trimethyltin (TMT), a toxic organotin compound, induces neurodegeneration selectively involving the limbic system and especially prominent in the hippocampus. Neurodegeneration-associated behavioral abnormalities, such as hyperactivity, aggression, cognitive deficits, and epileptic seizures, occur in both exposed humans and experimental animal models. Previously, TMT had been used generally in industry and agriculture, but the use of TMT has been limited because of its dangers to people. TMT has also been used to make a promising in vivo rodent model of neurodegeneration because of its region-specific characteristics. Several studies have demonstrated that TMT-treated animal models of epileptic seizures can be used as tools for researching hippocampus-specific neurotoxicity as well as the molecular mechanisms leading to hippocampal neurodegeneration. This review summarizes the in vivo and in vitro underlying mechanisms of TMT-induced hippocampal neurodegeneration (oxidative stress, inflammatory responses, and neuronal death/survival). Thus, the present review may be helpful to provide general insights into TMT-induced neurodegeneration and approaches to therapeutic interventions for neurodegenerative diseases, including temporal lobe epilepsy.
Collapse
Affiliation(s)
- Sueun Lee
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Jeonbuk 54538, South Korea
| | - Jinwook Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Juhwan Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, 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 61186, South Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, South Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju 63243, 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 61186, South Korea
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
13
|
Lee B, Sur B, Cho SG, Yeom M, Shim I, Lee H, Hahm DH. Wogonin Attenuates Hippocampal Neuronal Loss and Cognitive Dysfunction in Trimethyltin-Intoxicated Rats. Biomol Ther (Seoul) 2016; 24:328-37. [PMID: 27133262 PMCID: PMC4859797 DOI: 10.4062/biomolther.2015.152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/06/2016] [Accepted: 02/12/2016] [Indexed: 11/11/2022] Open
Abstract
We examined whether wogonin (WO) improved hippocampal neuronal activity, behavioral alterations and cognitive impairment, in rats induced by administration of trimethyltin (TMT), an organotin compound that is neurotoxic to these animals. The ability of WO to improve cognitive efficacy in the TMT-induced neurodegenerative rats was investigated using a passive avoidance test, and the Morris water maze test, and using immunohistochemistry to detect components of the acetylcholinergic system, brain-derived neurotrophic factor (BDNF), and cAMP-response element-binding protein (CREB) expression. Rats injected with TMT showed impairments in learning and memory and daily administration of WO improved memory function, and reduced aggressive behavior. Administration of WO significantly alleviated the TMT-induced loss of cholinergic immunoreactivity and restored the hippocampal expression levels of BDNF and CREB proteins and their encoding mRNAs to normal levels. These findings suggest that WO might be useful as a new therapy for treatment of various neurodegenerative diseases.
Collapse
Affiliation(s)
- Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bongjun Sur
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seong-Guk Cho
- The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Republic of Korea.,The Graduate School of Basic Science of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
14
|
Magnolol protects against trimethyltin-induced neuronal damage and glial activation in vitro and in vivo. Neurotoxicology 2016; 53:173-185. [DOI: 10.1016/j.neuro.2016.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/05/2015] [Accepted: 01/01/2016] [Indexed: 02/07/2023]
|
15
|
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: 3.6] [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
|
16
|
Kim J, Son Y, Kim J, Lee S, Kang S, Park K, Kim SH, Kim JC, Kim J, Takayama C, Im HI, Yang M, Shin T, Moon C. Developmental and degenerative modulation of GABAergic transmission in the mouse hippocampus. Int J Dev Neurosci 2015; 47:320-32. [PMID: 26394279 DOI: 10.1016/j.ijdevneu.2015.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/05/2015] [Accepted: 08/21/2015] [Indexed: 11/30/2022] Open
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter involved in synaptic plasticity. GABAergic transmission is also implicated in developmental and degenerative processes in the brain. The goal of the present study was to understand the developmental and degenerative regulation of GABAergic transmission in the mouse hippocampus by examining changes in GABA receptor subunit mRNA levels and GABA-related protein expression during postnatal development of the hippocampus and trimethyltin (TMT)-induced neurodegeneration in the juvenile (postnatal day [PD] 24) and adult hippocampus (PD 56). During postnatal development, the mRNA levels of GABA A receptor (GABAAR) subunits, including α1, α4, β1, β2, and δ; GABA B receptor (GABABR) subunit 2; and the expression of GABA-related proteins, including glutamic acid decarboxylase, vesicular GABA transporter (VGAT), and potassium chloride cotransporter 2 increased gradually in the mouse hippocampus. The results of seizure scoring and histopathological findings in the hippocampus revealed a more pronounced response to the same administered TMT dose in juvenile mice, compared with that in adult mice. The mRNA levels of most GABA receptor subunits in the juvenile hippocampus, excluding GABAAR subunit β3, were dynamically altered after TMT treatment. The mRNA levels of GABAAR subunits γ2 and δ decreased significantly in the adult hippocampus following TMT treatment, whereas the level of GABABR subunit 1 mRNA increased significantly. Among the GABA-related proteins, only VGAT decreased significantly in the juvenile and adult mouse hippocampus after TMT treatment. In conclusion, regulation of GABAergic signaling in the mouse hippocampus may be related to maturation of the central nervous system and the degree of neurodegeneration during postnatal development and TMT-induced neurodegeneration in the experimental animals.
Collapse
Affiliation(s)
- Jinwook Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Yeonghoon Son
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Juhwan Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea; Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 136-791, South Korea
| | - Sueun Lee
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Sohi Kang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Kyunghwan Park
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Sung-Ho Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Jong-Choon Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea
| | - Jeongtae Kim
- Department of Molecular Anatomy, Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Chitoshi Takayama
- Department of Molecular Anatomy, Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Heh-In Im
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 136-791, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, South Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju 690-756, South Korea.
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, South Korea.
| |
Collapse
|
17
|
Neuropharmacological Potential of Gastrodia elata Blume and Its Components. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:309261. [PMID: 26543487 PMCID: PMC4620291 DOI: 10.1155/2015/309261] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/06/2015] [Accepted: 09/15/2015] [Indexed: 02/07/2023]
Abstract
Research has been conducted in various fields in an attempt to develop new therapeutic agents for incurable neurodegenerative diseases. Gastrodia elata Blume (GE), a traditional herbal medicine, has been used in neurological disorders as an anticonvulsant, analgesic, and sedative medication. Several neurodegenerative models are characterized by oxidative stress and inflammation in the brain, which lead to cell death via multiple extracellular and intracellular signaling pathways. The blockade of certain signaling cascades may represent a compensatory therapy for injured brain tissue. Antioxidative and anti-inflammatory compounds isolated from natural resources have been investigated, as have various synthetic chemicals. Specifically, GE rhizome extract and its components have been shown to protect neuronal cells and recover brain function in various preclinical brain injury models by inhibiting oxidative stress and inflammatory responses. The present review discusses the neuroprotective potential of GE and its components and the related mechanisms; we also provide possible preventive and therapeutic strategies for neurodegenerative disorders using herbal resources.
Collapse
|
18
|
Kim J, Yang M, Son Y, Jang H, Kim D, Kim JC, Kim SH, Kang MJ, Im HI, Shin T, Moon C. Glial activation with concurrent up-regulation of inflammatory mediators in trimethyltin-induced neurotoxicity in mice. Acta Histochem 2014; 116:1490-500. [PMID: 25265880 DOI: 10.1016/j.acthis.2014.09.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trimethyltin (TMT), a potent neurotoxic chemical, causes dysfunction and neuroinflammation in the brain, particularly in the hippocampus. The present study assessed TMT-induced glial cell activation and inflammatory cytokine alterations in the mouse hippocampus, BV-2 microglia, and primary cultured astrocytes. In the mouse hippocampus, TMT treatment significantly increased the expression of glial cell markers, including the microglial marker ionized calcium-binding adapter molecule 1 and the astroglial marker glial fibrillary acidic protein. The expression of M1 and M2 microglial markers (inducible nitric oxide synthase [iNOS] and CD206, respectively) and pro-inflammatory cytokines (interleukin [IL]-1β, IL-6 and tumor necrosis factor [TNF]-α) were significantly increased in the mouse hippocampus following TMT treatment. In BV-2 microglia, iNOS, IL-1β, TNF-α, and IL-6 expression increased significantly, whereas arginase-1 and CD206 expression decreased significantly after TMT treatment in a time- and concentration-dependent manner. In primary cultured astrocytes, iNOS, arginase-1, IL-1β, TNF-α, and IL-6 expression increased significantly, whereas IL-10 expression decreased significantly after TMT treatment in a time- and concentration-dependent manner. These results indicate that significant up-regulation of pro-inflammatory signals in TMT-induced neurotoxicity may be associated with pathological processing of TMT-induced neurodegeneration.
Collapse
|
19
|
Lee S, Yang M, Kim J, Kim J, Son Y, Kwon S, Kim SH, Kim JC, Kang SS, Wang H, Shin T, Moon C. Nestin expression and glial response in the hippocampus of mice after trimethyltin treatment. Acta Histochem 2014; 116:1276-88. [PMID: 25139577 DOI: 10.1016/j.acthis.2014.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/21/2014] [Accepted: 07/22/2014] [Indexed: 11/29/2022]
Abstract
Nestin is a protein of embryonic intermediate filaments expressed by multipotent neural stem cells. In the present study, the nestin expression pattern in the mouse hippocampus 1, 2, 3, 4, and 8 days after treatment with trimethyltin (TMT) was examined to explore the possible role played by nestin in chemically induced hippocampal injury. TMT treatment (2.5mg/kg, intraperitoneally) selectively injured the dentate gyrus (DG) of the mouse hippocampus. The level of hippocampal mRNA encoding nestin increased significantly 2 and 3 days post-treatment and thereafter decreased (at 4 and 8 days post-treatment). The level of nestin protein significantly increased 2 - 4 days post-treatment, particularly in the injured region of the DG, and predominantly in glial fibrillary acidic protein-positive astrocytes in the hippocampal DG. Ki67-positive proliferating cells were increased following TMT treatment and co-localized with nestin-positive reactive astrocytes. Thus, we suggest that nestin contributes to remodeling of the chemically injured DG via glial scar formation and the alteration of neurogenesis.
Collapse
Affiliation(s)
- Sueun Lee
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Miyoung Yang
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea; Department of Physiology and Neuroscience Program, Michigan State University, MI 48824, USA
| | - Jinwook Kim
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Juhwan Kim
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Yeonghoon Son
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Seungjoo Kwon
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Sung-Ho Kim
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Seong Soo Kang
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Hongbing Wang
- Department of Physiology and Neuroscience Program, Michigan State University, MI 48824, USA
| | - Taekyun Shin
- College of Veterinary Medicine, Jeju National University, Jeju 690-756, Republic of Korea.
| | - Changjong Moon
- College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 500-757, Republic of Korea.
| |
Collapse
|
20
|
Developmental and degenerative modulation of brain-derived neurotrophic factor transcript variants in the mouse hippocampus. Int J Dev Neurosci 2014; 38:68-73. [PMID: 25124374 DOI: 10.1016/j.ijdevneu.2014.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/01/2014] [Accepted: 08/03/2014] [Indexed: 02/07/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is regarded as an important factor for neurogenesis, synaptic plasticity, and neuronal network organization in brain circuits. However, little is known about the regulation of BDNF transcript variants in the hippocampus during postnatal development and following chemically induced neurotoxicity. In the present study, we examined the expression of individual BDNF transcript variants in the mouse hippocampus on postnatal day (PD) 3, 7, 14, 21, and 56, as well as in the adult hippocampus 1, 2, 4, and 8 days after trimethyltin (TMT) treatment. During postnatal development, the expression levels of common BDNF-coding transcripts and BDNF transcript variants increased gradually in the hippocampus, but the temporal patterns of each exon transcript showed significant differences. In the TMT-treated hippocampus, the levels of common BDNF-coding transcripts and exon I, IIC, III, VII, VIII, and IXA transcripts were significantly increased 1 day post-treatment. These observations suggest that the differential regulation of BDNF exon transcripts may be associated with neuronal and synaptic maturation during postnatal development, and neuronal survival and synaptic plasticity in chemically induced neurodegeneration.
Collapse
|
21
|
Lee JW, Norden AD, Ligon KL, Golby AJ, Beroukhim R, Quackenbush J, Wells W, Oelschlager K, Maetzold D, Wen PY. Tumor associated seizures in glioblastomas are influenced by survival gene expression in a region-specific manner: a gene expression imaging study. Epilepsy Res 2014; 108:843-52. [PMID: 24690158 DOI: 10.1016/j.eplepsyres.2014.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/31/2014] [Accepted: 02/28/2014] [Indexed: 11/15/2022]
Abstract
Tumor associated seizures (TAS) are common and cause significant morbidity. Both imaging and gene expression features play significant roles in determining TAS, with strong interactions between them. We describe gene expression imaging tools which allow mapping of brain regions where gene expression has significant influence on TAS, and apply these methods to study 77 patients who underwent surgical evaluation for supratentorial glioblastomas. Tumor size and location were measured from MRI scans. A 9-set gene expression profile predicting long-term survivors was obtained from RNA derived from formalin-fixed paraffin embedded tissue. A total of 32 patients (42%) experienced preoperative TAS. Tumor volume was smaller (31.1 vs. 58.8 cubic cm, p<0.001) and there was a trend toward median survival being higher (48.4 vs. 32.7 months, p=0.055) in patients with TAS. Although the expression of only OLIG2 was significantly lower in patients with TAS in a groupwise analysis, gene expression imaging analysis revealed regions with significantly lower expression of OLIG2 and RTN1 in patients with TAS. Gene expression imaging is a powerful technique that demonstrates that the influence of gene expression on TAS is highly region specific. Regional variability should be evaluated with any genomic or molecular markers of solid brain lesions.
Collapse
Affiliation(s)
- Jong Woo Lee
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States.
| | - Andrew D Norden
- Center for Neuro-oncology, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, United States
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States; Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02115, United States
| | - Alexandra J Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, United States
| | - Rameen Beroukhim
- Center for Neuro-oncology, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, United States
| | - John Quackenbush
- Department of Biostatistics, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, United States
| | - William Wells
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, United States
| | - Kristen Oelschlager
- Castle Biosciences Inc., 3330N 2nd Street, Suite 207, Phoenix, AZ 85012, United States
| | - Derek Maetzold
- Castle Biosciences Inc., 3330N 2nd Street, Suite 207, Phoenix, AZ 85012, United States
| | - Patrick Y Wen
- Center for Neuro-oncology, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02115, United States
| |
Collapse
|
22
|
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: 2.8] [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
|