1
|
Reich N, Hölscher C. Cholecystokinin (CCK): a neuromodulator with therapeutic potential in Alzheimer's and Parkinson's disease. Front Neuroendocrinol 2024; 73:101122. [PMID: 38346453 DOI: 10.1016/j.yfrne.2024.101122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Cholecystokinin (CCK) is a neuropeptide modulating digestion, glucose levels, neurotransmitters and memory. Recent studies suggest that CCK exhibits neuroprotective effects in Alzheimer's disease (AD) and Parkinson's disease (PD). Thus, we review the physiological function and therapeutic potential of CCK. The neuropeptide facilitates hippocampal glutamate release and gates GABAergic basket cell activity, which improves declarative memory acquisition, but inhibits consolidation. Cortical CCK alters recognition memory and enhances audio-visual processing. By stimulating CCK-1 receptors (CCK-1Rs), sulphated CCK-8 elicits dopamine release in the substantia nigra and striatum. In the mesolimbic pathway, CCK release is triggered by dopamine and terminates reward responses via CCK-2Rs. Importantly, activation of hippocampal and nigral CCK-2Rs is neuroprotective by evoking AMPK activation, expression of mitochondrial fusion modulators and autophagy. Other benefits include vagus nerve/CCK-1R-mediated expression of brain-derived neurotrophic factor, intestinal protection and suppression of inflammation. We also discuss caveats and the therapeutic combination of CCK with other peptide hormones.
Collapse
Affiliation(s)
- Niklas Reich
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK; Faculty of Health and Medicine, Biomedical & Life Sciences Division, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Christian Hölscher
- Second associated Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, China; Henan Academy of Innovations in Medical Science, Neurodegeneration research group, Xinzhen, Henan province, China
| |
Collapse
|
2
|
Akagi Y, Takayama Y, Nihashi Y, Yamashita A, Yoshida R, Miyamoto Y, Kida YS. Functional engineering of human iPSC-derived parasympathetic neurons enhances responsiveness to gastrointestinal hormones. FEBS Open Bio 2024; 14:63-78. [PMID: 38013211 PMCID: PMC10761937 DOI: 10.1002/2211-5463.13741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023] Open
Abstract
Food-derived biological signals are transmitted to the brain via peripheral nerves through the paracrine activity of gastrointestinal (GI) hormones. The signal transduction circuit of the brain-gut axis has been analyzed in animals; however, species-related differences and animal welfare concerns necessitate investigation using in vitro human experimental models. Here, we focused on the receptors of five GI hormones (CCK, GLP1, GLP2, PYY, and serotonin (5-HT)), and established human induced pluripotent stem cell (iPSC) lines that functionally expressed each receptor. Compared to the original iPSCs, iPSCs expressing one of the receptors did not show any differences in global mRNA expression, genomic stability, or differentiation capacities of the three germ layers. We induced parasympathetic neurons from these established iPSC lines to assess vagus nerve activity. We generated GI hormone receptor-expressing neurons (CCKAR, GLP1R, and NPY2R-neuron) and tested their responsiveness to each ligand using Ca2+ imaging and microelectrode array recording. GI hormone receptor-expressing neurons (GLP2R and HTR3A) were generated directly by gene induction into iPSC-derived peripheral nerve progenitors. These receptor-expressing neurons promise to contribute to a better understanding of how the body responds to GI hormones via the brain-gut axis, aid in drug development, and offer an alternative to animal studies.
Collapse
Affiliation(s)
- Yuka Akagi
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
- Tsukuba Life Science Innovation Program (T‐LSI), School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
| | - Yuzo Takayama
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Yuma Nihashi
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Azusa Yamashita
- Analytical Science Laboratories, Asahi Quality & Innovations, Ltd.MoriyaJapan
| | - Risa Yoshida
- Analytical Science Laboratories, Asahi Quality & Innovations, Ltd.MoriyaJapan
| | - Yasuhisa Miyamoto
- Analytical Science Laboratories, Asahi Quality & Innovations, Ltd.MoriyaJapan
| | - Yasuyuki S. Kida
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
- School of Integrative & Global Majors (SIGMA)University of TsukubaTsukubaJapan
| |
Collapse
|
3
|
Sheng N, Zheng H, Li M, Li M, Wang Z, Peng Y, Yu H, Zhang J. 4,5 caffeoylquinic acid and scutellarin, identified by integrated metabolomics and proteomics approach as the active ingredients of Dengzhan Shengmai, act against chronic cerebral hypoperfusion by regulating glutamatergic and GABAergic synapses. Pharmacol Res 2020; 152:104636. [PMID: 31926275 DOI: 10.1016/j.phrs.2020.104636] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/06/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Dengzhan Shengmai (DZSM) is a proprietary Chinese medicine for remarkable curative effect as a treatment of cerebrovascular diseases, such as chronic cerebral hypoperfusion (CCH) and dementia based on evidence-based medicine, which have been widely used in the recovery period of ischemic cerebrovascular diseases. The purpose of this study was to investigate the active substances and mechanism of DZSM against CCH. Integrative metabolomic and proteomic studies were performed to investigate the neuroprotective effect of DZSM based on CCH model rats. The exposed components of DZSM in target brain tissue were analysed by a high-sensitivity HPLC-MS/MS method, and the exposed components were tested on a glutamate-induced neuronal excitatory damage cell model for the verification of active ingredients and mechanism of DZSM. Upon proteomic and metabolomic analysis, we observed a significant response in DZSM therapy from the interconnected neurotransmitter transport pathways including glutamatergic and GABAergic synapses. Additionally, DZSM had a significant regulatory effect on glutamate and GABA-related proteins including vGluT1 and vIAAT, suggested that DZSM could be involved in the vesicle transport of excitatory and inhibitory neurotransmitters in the pre-synaptic membrane. DZSM could also regulated the metabolism of arachidonic acid (AA), phospholipids, lysophospholipids and the expression of phospholipase A2 in post-synaptic membrane. The results of glutamate-induced neuronal excitatory injury cell model experiment for verification of active ingredients and mechanism of DZSM showed that there are five active ingredients, and among them, 4,5 caffeoylquinic acid (4,5-CQA) and scutellarin (SG) could simultaneously affect the GABAergic and glutamatergic synaptic metabolism as well as the related receptors, the NR2b subunit of NMDA and the α1 subunit of GABAA. The active ingredients of DZSM could regulate the over-expression of the NMDA receptor, enhance the expression of the GABAA receptor, resist glutamate-induced neuronal excitatory damage, and finally maintain the balance of excitatory and inhibitory synaptic metabolism dominated by glutamate and GABA. Furtherly, we compared the efficacy of DZSM, 4,5-CQA, SG and the synergistic effect of 4,5-CQA and SG, and the results showed that all the groups significantly improved cell viability compared with the model group (p < 0.001). The western blot results showed that DZSM, 4,5-CQA, SG and 4,5-CQA/SG co-administration groups could significantly regulate the expression of receptors (GABAA α1 and NR2b subunit of NMDA) and synaptic-related proteins, such as Sv2a, Syp, Slc17a7, bin1 and Prkca, respectively. These results proved DZSM and its active ingredients (4,5-CQA and SG) had the effect of regulating glutamatergic and GABAergic synapses. Finally, membrane potential FLIPR assay of 4,5-CQA and SG was used for GABRA1 activity test, and it was found that the two compounds could increase GABA-induced activation of GABRA1 receptor (GABA 10 μM) in a dose-dependent manner with EC50 value of 48.74 μM and 29.77 μM, respectively. Manual patch clamp method was used to record NMDA NR1/NR2B subtype currents, and scutellarin could cause around 10 % blockade at 10 μM (p<0.05 compared with the control group). These studies provided definitive clues of the mechanism for the neuroprotective effect of DZSM for CCH treatment and the active compounds regulating glutamatergic and GABAergic synapses. Additionally, 4,5-CQA and SG might be potential drugs for the treatment of neurodegenerative disease related to CCH.
Collapse
Affiliation(s)
- Ning Sheng
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Hao Zheng
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Min Li
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Menglin Li
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Zhe Wang
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Ying Peng
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Haibo Yu
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Jinlan Zhang
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
4
|
Zeng Q, Ou L, Wang W, Guo DY. Gastrin, Cholecystokinin, Signaling, and Biological Activities in Cellular Processes. Front Endocrinol (Lausanne) 2020; 11:112. [PMID: 32210918 PMCID: PMC7067705 DOI: 10.3389/fendo.2020.00112] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022] Open
Abstract
The structurally-related peptides, gastrin and cholecystokinin (CCK), were originally discovered as humoral stimulants of gastric acid secretion and pancreatic enzyme release, respectively. With the aid of methodological advances in biochemistry, immunochemistry, and molecular biology in the past several decades, our concept of gastrin and CCK as simple gastrointestinal hormones has changed considerably. Extensive in vitro and in vivo studies have shown that gastrin and CCK play important roles in several cellular processes including maintenance of gastric mucosa and pancreatic islet integrity, neurogenesis, and neoplastic transformation. Indeed, gastrin and CCK, as well as their receptors, are expressed in a variety of tumor cell lines, animal models, and human samples, and might contribute to certain carcinogenesis. In this review, we will briefly introduce the gastrin and CCK system and highlight the effects of gastrin and CCK in the regulation of cell proliferation and apoptosis in both normal and abnormal conditions. The potential imaging and therapeutic use of these peptides and their derivatives are also summarized.
Collapse
Affiliation(s)
- Qiang Zeng
- Health Management Institute, People's Liberation Army General Hospital, Beijing, China
| | - Lei Ou
- Health Management Institute, People's Liberation Army General Hospital, Beijing, China
| | - Wei Wang
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
- *Correspondence: Wei Wang
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
- Dong-Yu Guo
| |
Collapse
|
5
|
Reisi P, Ghaedamini AR, Golbidi M, Shabrang M, Arabpoor Z, Rashidi B. Effect of cholecystokinin on learning and memory, neuronal proliferation and apoptosis in the rat hippocampus. Adv Biomed Res 2015; 4:227. [PMID: 26623402 PMCID: PMC4638054 DOI: 10.4103/2277-9175.166650] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 01/04/2015] [Indexed: 12/16/2022] Open
Abstract
Background: Cholecystokinin (CCK) has roles in learning and memory, but the cellular mechanism is poorly understood. This study investigated the effect of CCK on spatial learning and memory, neuronal proliferation and apoptosis in the hippocampus in rats. Materials and Methods: Experimental groups were control and CCK. The rats received CKK octapeptide sulfated (CCK-8S, 1.6 μg/kg, i.p.) for 14 days. Spatial learning and memory were tested by Morris water maze and finally immunohistochemical study was performed; neurogenesis by Ki-67 method and apoptosis by Terminal deoxynucleotidyl transferase mediated dUTP Nick End Labeling (TUNEL) assay in hippocampal dentate gyrus (DG). Results: Cholecystokinin increased Ki-67 positive cells and reduced TUNEL positive cells in the granular layer of hippocampal DG. CCK failed to have a significant effect on spatial learning and memory. Conclusion: Results indicate neuroprotective and proliferative effects of CCK in the hippocampus; however, other factors are probably involved until the newly born neurons achieve necessary integrity for behavioral changes.
Collapse
Affiliation(s)
- Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran ; Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran ; Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Reza Ghaedamini
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Golbidi
- School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moloud Shabrang
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohreh Arabpoor
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Rashidi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
6
|
Zhang LL, Wei XF, Zhang YH, Xu SJ, Chen XW, Wang C, Wang QW. CCK-8S increased the filopodia and spines density in cultured hippocampal neurons of APP/PS1 and wild-type mice. Neurosci Lett 2013; 542:47-52. [DOI: 10.1016/j.neulet.2013.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 03/19/2013] [Accepted: 03/20/2013] [Indexed: 11/17/2022]
|
7
|
Karlsson O, Kultima K, Wadensten H, Nilsson A, Roman E, Andrén PE, Brittebo EB. Neurotoxin-induced neuropeptide perturbations in striatum of neonatal rats. J Proteome Res 2013; 12:1678-90. [PMID: 23410195 DOI: 10.1021/pr3010265] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The cyanobacterial toxin β-N-methylamino-l-alanine (BMAA) is suggested to play a role in neurodegenerative disease. We have previously shown that although the selective uptake of BMAA in the rodent neonatal striatum does not cause neuronal cell death, exposure during the neonatal development leads to cognitive impairments in adult rats. The aim of the present study was to characterize the changes in the striatal neuropeptide systems of male and female rat pups treated neonatally (postnatal days 9-10) with BMAA (40-460 mg/kg). The label-free quantification of the relative levels of endogenous neuropeptides using mass spectrometry revealed that 25 peptides from 13 neuropeptide precursors were significantly changed in the rat neonatal striatum. The exposure to noncytotoxic doses of BMAA induced a dose-dependent increase of neurosecretory protein VGF-derived peptides, and changes in the relative levels of cholecystokinin, chromogranin, secretogranin, MCH, somatostatin and cortistatin-derived peptides were observed at the highest dose. In addition, the results revealed a sex-dependent increase in the relative level of peptides derived from the proenkephalin-A and protachykinin-1 precursors, including substance P and neurokinin A, in female pups. Because several of these peptides play a critical role in the development and survival of neurons, the observed neuropeptide changes might be possible mediators of BMAA-induced behavioral changes. Moreover, some neuropeptide changes suggest potential sex-related differences in susceptibility toward this neurotoxin. The present study also suggests that neuropeptide profiling might provide a sensitive characterization of the BMAA-induced noncytotoxic effects on the developing brain.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University , SE-751 24 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
8
|
Asano H, Horinouchi T, Mai Y, Sawada O, Fujii S, Nishiya T, Minami M, Katayama T, Iwanaga T, Terada K, Miwa S. Nicotine- and tar-free cigarette smoke induces cell damage through reactive oxygen species newly generated by PKC-dependent activation of NADPH oxidase. J Pharmacol Sci 2012; 118:275-87. [PMID: 22302021 DOI: 10.1254/jphs.11166fp] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
We examined cytotoxic effects of nicotine/tar-free cigarette smoke extract (CSE) on C6 glioma cells. The CSE induced plasma membrane damage (determined by lactate dehydrogenase leakage and propidium iodide uptake) and cell apoptosis {determined by MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] reduction activity and DNA fragmentation}. The cytotoxic activity decayed with a half-life of approximately 2 h at 37°C, and it was abolished by N-acetyl-L-cysteine and reduced glutathione. The membrane damage was prevented by catalase and edaravone (a scavenger of (•)OH) but not by superoxide dismutase, indicating involvement of (•)OH. In contrast, the CSE-induced cell apoptosis was resistant to edaravone and induced by authentic H(2)O(2) or O(2)(-) generated by the xanthine/xanthine oxidase system, indicating involvement of H(2)O(2) or O(2)(-) in cell apoptosis. Diphenyleneiodonium [NADPH oxidase (NOX) inhibitor] and bisindolylmaleimide I [BIS I, protein kinase C (PKC) inhibitor] abolished membrane damage, whereas they partially inhibited apoptosis. These results demonstrate that 1) a stable component(s) in the CSE activates PKC, which stimulates NOX to generate reactive oxygen species (ROS), causing membrane damage and apoptosis; 2) different ROS are responsible for membrane damage and apoptosis; and 3) part of the apoptosis is caused by oxidants independently of PKC and NOX.
Collapse
Affiliation(s)
- Hiroshi Asano
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Langmesser S, Cerezo-Guisado MI, Lorenzo MJ, Garcia-Marin LJ, Bragado MJ. CCK1 and 2 receptors are expressed in immortalized rat brain neuroblasts: Intracellular signals after cholecystokinin stimulation. J Cell Biochem 2007; 100:851-64. [PMID: 17226751 DOI: 10.1002/jcb.21193] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cholecystokinin (CCK) is one of the most abundant neuropeptides in the central nervous system (CNS) where it promotes important functions by activation of receptors CCK1 and CCK2. Our aim was to investigate CCK receptors expression and their downstream intracellular signaling in immortalized rat brain neuroblasts. Results show that CCK1 and CCK2 receptor mRNAs and CCK2 receptor protein are expressed in neuroblasts. CCK incubation of neuroblasts leads to stimulation in a time-dependent manner of several signaling pathways, such as tyrosine phosphorylation of adaptor proteins paxillin and p130(Cas), phosphorylation of p44/p42 ERKs as well as PKB (Ser473). Moreover, CCK-8 stimulates the DNA-binding activity of the transcription factor AP-1. The CCK2 receptor agonist gastrin stimulates ERK1/2 phosphorylation in a comparable degree as CCK does. ERK1/2 phosphorylation activated by CCK-8 was markedly inhibited by the CCK2 receptor antagonist CR2945. Incubation for 48 h with CCK-8 increases neuroblasts viability in a similar degree as EGF. In summary, our data clearly identify CCK1 and CCK2 receptor mRNAs and CCK2 receptor protein in brain neuroblasts and show that incubation with CCK promotes cell proliferation and activates the phosphorylation of survival transduction pathways. Stimulation of ERK1/2 phosphorylation by CCK is mainly mediated by the CCK2 receptor. Moreover, this work might provide a novel model of proliferating neuronal cells to further study the biochemical mechanisms by which the neuropeptide CCK exerts its actions in the CNS.
Collapse
Affiliation(s)
- Sonja Langmesser
- Departamento de Fisiología, Biología Molecular y Genética, Universidad de Extremadura, Cáceres, Spain
| | | | | | | | | |
Collapse
|
10
|
Tamura Y, Monden M, Shintani M, Kawai A, Shiomi H. Neuroprotective effects of hibernation-regulating substances against low-temperature-induced cell death in cultured hamster hippocampal neurons. Brain Res 2006; 1108:107-16. [PMID: 16854391 DOI: 10.1016/j.brainres.2006.06.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Revised: 06/01/2006] [Accepted: 06/05/2006] [Indexed: 01/20/2023]
Abstract
The neuroprotective effects of hibernation-regulating substances (HRS) such as adenosine (ADO), opioids, histamine and thyrotropin-releasing hormone (TRH) on low-temperature-induced cell death (LTCD) were examined using primary cultured hamster hippocampal neurons. LTCD was induced when cultures were maintained at <22 degrees C for 7 days. ADO (10-100 microM) protected cultured neurons from LTCD in a dose-dependent manner. The neuroprotective effects of ADO were reversed by both 8-cyclopenthyltheophilline (CPT; A(1) receptor antagonist) and 3,7-dimethyl-1-propargylxanthine (DMPX; A(2) receptor antagonist). Morphine (a non-selective opioid receptor agonist) was also effective in attenuating LTCD at an in vitro dose range of 10-100 muM. The neuroprotective effects of morphine were antagonized by naloxone (a non-selective opioid receptor antagonist). In addition, although [D-Ala(2), N-Me-Phe(4), Gly-ol(5)]-enkephalin (DAMGO; mu-opioid receptor agonist), [D-Pen(2,5)]-enkephalin (DPDPE; delta-opioid receptor agonist) and U-69593 (kappa-opioid receptor agonist) were also effective, LTCD of cultured hippocampal neurons was not affected by TRH. Furthermore, histamine produced hypothermia in Syrian hamsters and protected hippocampal neurons in vitro at 100 microM. The neuroprotective effect of histamine was reversed by pyrilamine (H(1) receptor antagonist). Apoptosis was probably involved in LTCD. These results suggest that ADO protected hippocampal neurons in vitro via its agonistic actions on both A(1) and A(2) receptors, whereas morphine probably elicited its neuroprotective effects via agonistic effects on the mu-, delta- and kappa-opioid receptors. In addition, histamine also protected hippocampal neurons via its agonistic action on the H(1) receptor. Thus, HRS-like adenosine-, opioid- and histamine-like hypothermic actions would most likely induce neuroprotective effects against LTCD in vitro.
Collapse
Affiliation(s)
- Yutaka Tamura
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima 729-0292, Japan
| | | | | | | | | |
Collapse
|
11
|
Kuroki T, Etou K, Uchimura H. Ceruletide, an Analog of Cholecystokinin Octapeptide: New Perspectives on Its Efficacy. CNS DRUG REVIEWS 2006. [DOI: 10.1111/j.1527-3458.1999.tb00095.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
12
|
Bräuer AU, Savaskan NE, Plaschke M, Ninnemann O, Nitsch R. Cholecystokinin expression after hippocampal deafferentiation: molecular evidence revealed by differential display-reverse transcription-polymerase chain reaction. Neuroscience 2003; 121:111-21. [PMID: 12946704 DOI: 10.1016/s0306-4522(03)00336-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The cortical information flow via the perforant path represents a major excitatory projection to the hippocampus. Lesioning this projection leads to massive degeneration and subsequently to reorganization in its termination zones as well as in primary non-affected subfields of the hippocampus. The molecular mechanisms and factors which are involved in the postlesional events are poorly defined. Using a differential display reverse transcription-polymerase chain reaction (DDRT-PCR) strategy, we located one band which occurred only in control hippocampus lanes and almost disappeared in the lanes of lesioned hippocampi. By sequencing, we identified the corresponding gene as cholecystokinin (CCK). Northern blot analysis confirmed a decreased transcription of CCK after lesion. In situ hybridization analysis was performed for localization and quantification of altered CCK transcription. We noted a significant downregulation of CCK transcription in the hippocampus (20%) and in the contralateral cortex (12%) 1-day after lesion (dal) and an increased signal in the ipsilateral cortex (10.5%). This pattern was altered, showing upregulation of CCK mRNA expression, reaching its highest level of 70% above control levels at 5 dal. In the hippocampus, the control level was reached again at 21 dal, whereas the cortex reached the control level at 10 dal. In comparison, the mRNA transcripts of the receptors CCK(A) and CCK(B) remained unchanged. Since CCK-containing neurons are involved in the modulation of pyramidal and granule cell excitability, our data indicate a time course correlation between CCK mRNA expression and postlesional axonal sprouting response in the hippocampus.
Collapse
Affiliation(s)
- A U Bräuer
- Department of Cell and Neurobiology, Oskar-Hertwig House, Humboldt University Medical School Charité, Philippstrasse 12, Philippstrasse 12, D-10115 Berlin, Germany.
| | | | | | | | | |
Collapse
|
13
|
Ferraro G, Sardo P, Di Giovanni G, Di Maio R, La Grutta V. CCK-nitric oxide interaction in rat cortex, striatum and pallidum. Comp Biochem Physiol C Toxicol Pharmacol 2003; 135:425-33. [PMID: 12965187 DOI: 10.1016/s1532-0456(03)00144-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have chosen to study the effects of both nitric oxide (NO) and cholecystokinin neuromodulatory systems in some motor structures that are frequently involved in excitotoxic phenomena. In particular, 7-nitroindazole, a selective inhibitor of neuronal NO synthase, was administered in control and sulfated cholecystokinin octapeptide-treated rats. Cortical surface, striatal and pallidal depth bioelectric activities were examined through Fast Fourier Transform analysis. Cortical and pallidal recordings revealed an increase of rapid standard rhythms after the inhibition of neuronal NO synthase; in contrast, striatal depth recordings showed a marked increase of slow standard rhythms. All these effects were completely abolished by chronic pre-treatment with sulfated cholecystokinin octapeptide. The results suggest a functional co-operation between cholecystokinin and NO systems in the modulation of the bioelectric activity of all the motor structures examined, and the possibility of preventing excitotoxic damages induced by an anomalous balance between excitatory and inhibitory neurotransmitters in these areas.
Collapse
Affiliation(s)
- Giuseppe Ferraro
- Department of Experimental Medicine, Human Physiology Section G. Pagano--Corso Tuköry, 129, I-90134 Palermo, Italy
| | | | | | | | | |
Collapse
|
14
|
Petersson S, Lavebratt C, Schalling M, Hökfelt T. Expression of cholecystokinin, enkephalin, galanin and neuropeptide Y is markedly changed in the brain of the megencephaly mouse. Neuroscience 2001; 100:297-317. [PMID: 11008168 DOI: 10.1016/s0306-4522(00)00285-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Megencephaly, enlarged brain, is a major sign in several human neurological diseases. The mouse model for megencephaly (mceph/mceph) has an enlarged brain, presumably due to brain cell hypertrophy, and exhibits neurological and motor disturbances with seizure-like activity, as well as disturbances in the insulin-like growth factor system. Here, we report that expression of the neuropeptides cholecystokinin, enkephalin, galanin and neuropeptide Y is dramatically changed in mceph/mceph brains compared to wild type, as revealed by in situ hybridization and immunohistochemistry. The changes were confined to discrete brain regions and occurred in a parallel fashion for peptides and their transcripts. For cholecystokinin, mceph/mceph brains had region-specific up- and down-regulations in several layers of the hippocampal formation and increased levels in, especially ventral, cortical regions. Enkephalin messenger RNA expression was up-regulated in the dentate gyrus granular layer and in ventral cortices, but down-regulated in the CA1 pyramidal layer. Enkephalin-like immunoreactivity was elevated in mossy fibers of the hippocampus and the ventral cortices. Galanin expression was increased in several layers and interneurons of the hippocampal formation, as well as in ventral cortices. Galanin-like immunoreactivity was reduced in nerve terminals in the forebrain. Neuropeptide Y expression was increased in the hippocampal formation and ventral cortices. Whether the mainly increased peptide levels contribute to the excessive growth of the brain or represent a consequence of this growth and/or of the neurological and motor disturbances remains to be elucidated.
Collapse
Affiliation(s)
- S Petersson
- Neurogenetic Unit, Center for Molecular Medicine, Karolinska Institutet, 171 76, Stockholm, Sweden.
| | | | | | | |
Collapse
|
15
|
Kubota M, Iida Y, Magata Y, Kitamura Y, Kawashima H, Saji H. Mechanisms of [2,3-butanedione bis(N4-dimethylthiosemicarbazone)]zinc (Zn-ATSM2)-induced protection of cultured hippocampal neurons against N-methyl-D-aspartate receptor-mediated glutamate cytotoxicity. JAPANESE JOURNAL OF PHARMACOLOGY 2000; 84:334-8. [PMID: 11138735 DOI: 10.1254/jjp.84.334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hyperexcitation of glutamatergic neurons may play a key role in ischemia-related neurodegeneration. Recent studies have suggested that the zinc ion (Zn2+), which is present in the central nervous system, has a modulatory role in glutamatergic neuron activity. Zinc ions block glutamate-induced depolarizing currents and neuronal damage by binding with zinc sites on the NMDA subtypes. Therefore, we examined the usefulness of zinc as a therapeutic agent for the prevention of ischemic neuronal damage in the brain. In our previous study, 2,3-butanedione bis(N4-dimethylthiosemicarbazonato) zinc complex (Zn-ATSM2), with high brain uptake, showed significant neuroprotective effects against cerebral ischemia in rats when administered systemically. In this study, to elucidate the mechanism of the neuroprotective effect of Zn-ATSM2, we first examined its in vitro protective effects against glutamate-, NMDA- and kainite-induced neurotoxicity in primary cultures of hippocampal neurons. Zn-ATSM2 elicited protective effects against this glutamate- and NMDA-induced neurotoxicity, but did not affect kainite-induced cytotoxicity. In addition, we studied the effects of Zn-ATSM2 on influx of Ca2+, which undergoes modification subsequent to NMDA activation. Zn-ATSM2 significantly decreased glutamate-induced 45Ca2+ uptake. Thus, Zn-ATSM2 protected against glutamate-induced neurotoxicity and its protective effect was, at least in part, due to the blockage of NMDA receptor-mediated Ca2+ influx.
Collapse
Affiliation(s)
- M Kubota
- Department of Patho-functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Orphanin FQ (OFQ) is a heptadecapeptide that structurally resembles opioid peptides. It has been demonstrated that the moderate density of binding sites of OFQ were localized in the hippocampus and that the expression of OFQ receptor in the hippocampus have an important role in learning and memory. This study was designed to investigate whether activation of the OFQ receptor could induced hyperpolarization in the cultured hippocampus neurons in rats. In the current clamp mode, the application of OFQ (10(-8)-10(-5) M) hyperpolarized the membranes in cultured hippocampus neurons in a concentration-dependent manner. Moreover, in the voltage clamp mode, application of OFQ (10(-6) M) induced outward current in hippocampus CA3 pyramidal neurons. In the presence of TTX (3x10(-7) M), the average maximal amplitude of the outward current deflection induced by OFQ (10(-6) M) at -60 mV of a holding potential was 24.7+/-0.54 pA. The OFQ-induced current reversed at -99.06+/-3.80 mV (3 mM), which was quite close to the K(+) equilibrium potential as calculated by the Nernst equation (E(k)=-96.08 mV, 3 mM) for K(+) in our standard solution. This suggests that OFQ-induced current was mediated by K(+) ion. It has been demonstrated that [Phe(1)psi(CH(2)-NH)Gly(2)]Nociceptin(1-13)NH(2)) (a pseudopeptide analog of nociceptin), and nocistatin are selective antagonists of OFQ. OFQ (10(-6) M)-induced outward current was antagonized by application of [Phe(1)psi(CH(2)-NH)Gly(2)]Nociceptin(1-13)NH(2) (10(-5) M). In contrast, OFQ-induced outward current was not antagonized by application of nocistatin (10(-5) M). These results indicates that there is the physiological functioning receptor of OFQ in the hippocampus.
Collapse
Affiliation(s)
- T Amano
- Department of Pharmacology, Hiroshima University School of Medicine, Hiroshima, Japan
| | | | | | | |
Collapse
|
17
|
de Tullio P, Delarge J, Pirotte B. Therapeutic and chemical developments of cholecystokinin receptor ligands. Expert Opin Investig Drugs 2000; 9:129-46. [PMID: 11060666 DOI: 10.1517/13543784.9.1.129] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cholecystokinin (CCK) is an important 'brain-gut' hormone located both in the gastrointestinal (GI) system and in the CNS. At least two different G-coupled high affinity receptors have been identified: the CCK-A and the CCK-B receptors. Although the complex biological role of CCK is, as yet, not fully understood, its connection with many different physiological processes both at the GI level and at the CNS level is now well established. There is much potential for therapeutic use of CCK receptor ligands, however, clear investigations have yet to be completed. Several chemical families have been investigated over the last 20 years to find potent, subtype selective and stable CCK receptor agonists and antagonists. The main goal was to discover new therapeutic drugs acting on GI and/or on CNS diseases and also, to obtain powerful pharmacological tools that could permit a better understanding of the biological role of CCK. Despite promising results from investigations into medicinal chemistry of CCK receptor ligands, the therapeutical applications of these ligands still remains to be defined. This article reviews the main biological role of CCK, the therapeutic potential of CCK-A and CCK-B receptor agonists and antagonists and the common compounds from the different families of ligands.
Collapse
Affiliation(s)
- P de Tullio
- Department of Medicinal Chemistry, Université de Liège, CHU, Avenue de l'Hôpital 1, B-4000 Sart-Tilman (Liège), Belgium.
| | | | | |
Collapse
|
18
|
Cunha GM, Moraes RA, Moraes GA, França MC, Moraes MO, Viana GS. Nerve growth factor, ganglioside and vitamin E reverse glutamate cytotoxicity in hippocampal cells. Eur J Pharmacol 1999; 367:107-12. [PMID: 10082272 DOI: 10.1016/s0014-2999(98)00942-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The present work showed that glutamate decreased hippocampal cell viability in a dose-dependent manner. While no significant effect was observed after cell exposure to 0.1 mM glutamate, cell incubation for 0.5 h caused a progressive decrease of cell viability, which at 5 mM concentration reached 68% as compared to controls. No further effect was observed in the presence of 10 mM glutamate. While nerve growth factor (NGF) at the dose of 0.5 ng/ml presented no effect, it significantly reduced glutamate cytotoxicity at a higher dose (1 ng/ml) increasing the cell viability to 66%. Similarly, cell viabilities in the presence of the ganglioside GM, (5 and 10 ng/ml) after glutamate exposure were 19 and 73%, respectively. A dose-response relationship was observed after cell incubation with vitamin E (0.5 and 1 mM) which resulted in cell viability of the order of 34 and 70%, respectively. Surprisingly, a potentiation of the effect was observed after the association of NGF (0.5 ng/ml) plus ganglioside GM1 (5 ng/ml) or vitamin E (0.5 mM) plus ganglioside GM1 (5 ng/ml), after pre-incubation with glutamate. In these conditions, significantly higher viabilities were demonstrated (66 and 71% for the two associations, respectively) as compared to each one of the compounds alone (NGF 0.5 ng/ml--29.5%; ganglioside GM1 5 ng/ml--19.4%). However, no potentiation was seen after the association of NGF plus vitamin E on glutamate pre-exposed cells. These results showed a cytoprotective effect of ganglioside GM1, NGF and vitamin E on the glutamate-induced cytotoxicity in rat hippocampal cells.
Collapse
Affiliation(s)
- G M Cunha
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | | | | | |
Collapse
|
19
|
Arabia AM, Shen PJ, Gundlach AL. Increased striatal proenkephalin mRNA subsequent to production of spreading depression in rat cerebral cortex: activation of corticostriatal pathways? BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 61:195-202. [PMID: 9795215 DOI: 10.1016/s0169-328x(98)00189-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cortical Spreading Depression (CSD) is a slowly propagating wave of depolarization and negative interstitial DC potential, that when induced in the rat brain extends across the entire homolateral hemisphere. Despite evidence that CSD does not penetrate into subcortical regions, neurochemical changes in areas anatomically connected to cortex have been reported. In this study in situ hybridization histochemistry was used to examine the levels of cholecystokinin (CCK), proenkephalin (ENK) and prodynorphin (DYN) mRNA in cortex and forebrain basal ganglia following KCl-induced CSD. Unilateral CSD was induced by topical application of 3 M KCl ( approximately 10 microliter) onto the right parietal cortex for 10 min and rats were then killed 1-6 h and 1-28 days later. CCK mRNA levels were increased (P<0.01) in the ipsilateral neocortex 3 h after CSD (13% above levels in contralateral side), reached a peak at 2 days ( approximately 70%) and were still elevated at 7 (30%) but not, 14 or 28 days later. Unilateral CSD also produced a rapid and sustained increase (P<0.05) in ENK mRNA in ipsilateral piriform cortex (from 3 h to 2 days; 70-250% above contralateral), and a delayed increase in caudate putamen and olfactory tubercle at 1 and 2 days ( approximately 25% in both regions), but levels were again equivalent to control at 7 days and beyond. In contrast, no marked changes in neocortical ENK mRNA, or DYN mRNA in both cortex and basal ganglia, were observed under these conditions. These findings demonstrate that CSD has specific, rapid and long-lasting effects on neuropeptide expression in neocortex and subcortical areas. CSD-induced changes in mesostriatal ENK mRNA are proposed to reflect synaptic activation of local neurons via cortical afferent projections.
Collapse
Affiliation(s)
- A M Arabia
- The University of Melbourne, Clinical Pharmacology and Therapeutics Unit, Department of Medicine, Austin and Repatriation Medical Centre, Heidelberg, Victoria, 3084, Australia
| | | | | |
Collapse
|
20
|
Kihara T, Shimohama S, Urushitani M, Sawada H, Kimura J, Kume T, Maeda T, Akaike A. Stimulation of alpha4beta2 nicotinic acetylcholine receptors inhibits beta-amyloid toxicity. Brain Res 1998; 792:331-4. [PMID: 9593977 DOI: 10.1016/s0006-8993(98)00138-3] [Citation(s) in RCA: 164] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We examined the effects of nicotinic receptor agonists against beta amyloid (Abeta) cytotoxicity to rat cortical neurons. Administration of nicotine protected against Abeta-induced neuronal death. This neuroprotection was blocked by dihydro-beta-erythroidine, an alpha4beta2 nicotinic receptor antagonist. Furthermore, incubation with cytisine, a selective alpha4beta2 nicotinic receptor agonist, inhibited Abeta cytotoxicity. These results suggest that alpha4beta2 nicotinic receptor activation plays an important role in neuroprotection against Abeta cytotoxicity.
Collapse
Affiliation(s)
- T Kihara
- Department of Neurology, Faculty of Medicine, Kyoto University, Kyoto 606, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Müller W, Heinemann U, Berlin K. Cholecystokinin activates CCKB-receptor-mediated Ca-signaling in hippocampal astrocytes. J Neurophysiol 1997; 78:1997-2001. [PMID: 9325368 DOI: 10.1152/jn.1997.78.4.1997] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cholecystokinin-8S (CCK-8S) is the most abundant neuropeptide in the mammalian cortex and the limbic system; however, its physiological functions remained largely obscure. We studied effects of CCK on astrocytic Ca signaling, which has met considerable interest as a second messenger in astrocytic-neuronal signaling, by digital ratio-imaging of Fura-2/AM loaded rat and mouse hippocampal astrocytes in dissociated culture. Superfusion of CCK-8S (5-50 nM for 2 min) evoked repetitive Ca increases of several hundred nanomolar in a subpopulation of astrocytes. Mouse astrocytes appeared to be more responsive to CCK than rat cells with respect to the fraction of cells responding as well as to the amplitudes of Ca increases. The Ca responses persisted in the absence of extracellular Ca, indicating that release of Ca from intracellular stores is the primary source of these Ca increases. The CCK-8S-induced Ca increases were blocked by the CCKB receptor antagonist PD135158 (100 nM) but not by the CCKA antagonist lorglumide (100 nM). We surmise that astrocytes might be a major primary target for CCK in the CNS.
Collapse
Affiliation(s)
- W Müller
- Institut für Physiologie der Charité, Abteilung Neurophysiologie, Arbeitsgruppe Molekulare Zellphysiologie, D-10117 Berlin, Germany
| | | | | |
Collapse
|
22
|
Kaneko S, Maeda T, Kume T, Kochiyama H, Akaike A, Shimohama S, Kimura J. Nicotine protects cultured cortical neurons against glutamate-induced cytotoxicity via alpha7-neuronal receptors and neuronal CNS receptors. Brain Res 1997; 765:135-40. [PMID: 9310404 DOI: 10.1016/s0006-8993(97)00556-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We examined the effects of nicotine on glutamate-induced cytotoxicity using primary cultures of rat cortical neurons. The cell viability decreased significantly when cultures were exposed to glutamate for 10 min and then incubated with glutamate-free medium for 1 h. The exposure of cultures to nicotine (10 microM) for 8-24 h prior to glutamate application ameliorated the glutamate-induced cytotoxicity, with no significant effect of nicotine alone on the cell viability. Neuroprotection by nicotine was dependent on the incubation period. alpha-bungarotoxin (alpha-BTX) and methyllycaconitine (MLA), both of which are alpha7-neuronal receptor antagonists, and dihydro-beta-erythroidine (DHbetaE), a neuronal central nervous system (CNS) receptor antagonist, each significantly antagonized the protection by nicotine against glutamate-induced cytotoxicity. Ionomycin, a calcium ionophore, and S-nitrosocysteine (SNOC), a nitric oxide (NO) donor, also induced cytotoxicity in a manner similar to glutamate. Nicotine protected cultures against ionomycin-induced cytotoxicity, but not against SNOC-induced cytotoxicity. These results suggest that nicotine protects cultured cortical neurons against glutamate-induced cytotoxicity via alpha7-neuronal receptors and neuronal CNS receptors by reducing NO-formation triggered by Ca2+ influx.
Collapse
Affiliation(s)
- S Kaneko
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Kihara T, Shimohama S, Sawada H, Kimura J, Kume T, Kochiyama H, Maeda T, Akaike A. Nicotinic receptor stimulation protects neurons against beta-amyloid toxicity. Ann Neurol 1997; 42:159-63. [PMID: 9266724 DOI: 10.1002/ana.410420205] [Citation(s) in RCA: 238] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
beta-Amyloid (A beta), a major constituent of senile plaques in Alzheimer's disease (AD), is thought to contribute to the neurodegeneration. We examined the effects of nicotinic receptor agonists on A beta cytotoxicity in cultured rat cortical neurons. The number of viable neurons decreased significantly when cultures were exposed to synthetic A beta peptides (25-35). Concomitant administration of nicotine with A beta markedly reduced the number of dead cells. This nicotine-induced neuroprotection was dependent on the concentration. When hexamethonium or mecamylamine, nicotinic antagonist, was added, neuroprotective effect of nicotine was blocked, which indicates that effect of nicotine was mediated by nicotinic receptors. In addition, a selective alpha7-receptor antagonist, alpha-bungarotoxin (alpha-BTX), blocked the neuroprotective effect of nicotine. Furthermore, incubation with 3-(2,4)-dimethoxybenzylidene anabaseine (DMXB), a selective alpha7-receptor agonist, protected against A beta-induced neuronal death. These results suggest that alpha7-receptor activation plays an important role in neuroprotection against A beta cytotoxicity. This study suggests that nicotinic receptor stimulation, especially alpha7-receptor activation, may be able to protect neurons from degeneration induced by A beta and may have effects that counter the progress of AD.
Collapse
Affiliation(s)
- T Kihara
- Department of Neurology, Faculty of Medicine, Kyoto University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Kaneda K, Kikuchi M, Kashii S, Honda Y, Maeda T, Kaneko S, Akaike A. Effects of B vitamins on glutamate-induced neurotoxicity in retinal cultures. Eur J Pharmacol 1997; 322:259-64. [PMID: 9098696 DOI: 10.1016/s0014-2999(96)00997-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The effects of B vitamins on glutamate-induced neurotoxicity were examined using primary cultures obtained from the rat retina. Cell viability was markedly reduced by a brief exposure to glutamate followed by incubation with glutamate-free media for 1 h. Glutamate cytotoxicity was reduced in the cultures that had been maintained in thiamine-, pyridoxine- or nicotinamide-containing medium before the exposure to glutamate. Glutamate cytotoxicity was also reduced by chronic application of thiamine pyrophosphate and pyridoxal phosphate, which are active coenzyme forms of thiamine and pyridoxine, respectively. By contrast, chronic application of riboflavin, pantothenate, biotin, folic acid and inositol did not affect glutamate cytotoxicity. None of the B vitamins tested had any effect on glutamate cytotoxicity when added only during the exposure to glutamate. These findings suggest that chronically applied thiamine, pyridoxine and nicotinamide protect retinal neurons against glutamate cytotoxicity.
Collapse
Affiliation(s)
- K Kaneda
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Bachus SE, Hyde TM, Herman MM, Egan MF, Kleinman JE. Abnormal cholecystokinin mRNA levels in entorhinal cortex of schizophrenics. J Psychiatr Res 1997; 31:233-56. [PMID: 9278188 DOI: 10.1016/s0022-3956(96)00041-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Limbic cortical regions, including anterior cingulate cortex (ACC), prefrontal cortex (PFC) and entorhinal cortex (ERC), have been implicated in the neuropathology of schizophrenia. Glutamate projection neurons connect these limbic cortical regions to each other, as well as to the terminal fields of the striatal/accumbens dopamine neurons. Subsets of these glutamate projection neurons, and of the GABA interneurons in cortex, contain the neuropeptide cholecystokinin (CCK). In an effort to study the limbic cortical glutamate projection neurons and GABA interneurons in schizophrenia, we have measured CCK mRNA with in situ hybridization histochemistry in postmortem samples of dorsolateral (DL)PFC, ACC and ERC of seven schizophrenics, nine non-psychotic suicides and seven normal controls. CCK mRNA is decreased in ERC (especially layers iii vi) and subiculum in schizophrenics relative to controls. Cellular analysis indicates that there is a decrease in density of CCK mRNA in labelled neurons. In so far as ERC CCK mRNA is not reduced in rats treated chronically with haloperidol, this decrease in schizophrenics does not appear to be related to neuroleptic treatment. In contrast, in DLPFC, where schizophrenics do not differ from normals, the suicide victims have elevated CCK mRNA (especially in layers v and vi), and increased cellular density of CCK mRNA, relative to both normals and schizophrenics. These results lend further support for the involvement of ERC and hippocampus in schizophrenia, suggesting that neurons that utilize CCK may be particularly important. Similarly, an increase in CCK mRNA levels in the PFC of suicides adds to a growing body of evidence implicating this structure in this pathological state. In so far as CCK is co-localized with GABA or glutamate in cortical neurons, both of these neuronal populations need to be studied further in schizophrenia and suicide.
Collapse
Affiliation(s)
- S E Bachus
- Clinical Brain Disorders Branch, IRP/NIMH/NIH, Neuroscience Center at St. Elizabeths Hospital, Washington, DC 20032, USA.
| | | | | | | | | |
Collapse
|
26
|
Cheng N, Maeda T, Kume T, Kaneko S, Kochiyama H, Akaike A, Goshima Y, Misu Y. Differential neurotoxicity induced by L-DOPA and dopamine in cultured striatal neurons. Brain Res 1996; 743:278-83. [PMID: 9017256 DOI: 10.1016/s0006-8993(96)01056-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The neurotoxicity of L-DOPA and dopamine (DA) on striatal neurons was examined by using primary cultures of rat striatum. Exposure to L-DOPA and DA at concentrations of 30-300 microM induced dose-dependent cell death in both younger cultures (3 days in culture, 3 DIC) and elder cultures (10 days in culture, 10 DIC). The cytotoxicity of L-DOPA and DA was also dependent on the exposure time (6-24 h). Ascorbic acid (200 microM) inhibited both L-DOPA- and DA-induced cytotoxicity in 3 DIC cultures, whereas it provided significant protection against DA- but not L-DOPA-induced cytotoxicity in 10 DIC cultures. The L-DOPA cytotoxicity in 10 DIC cultures was prevented by a non-NMDA receptor antagonist, 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX), and by an NMDA receptor antagonist, MK-801. Neither antagonist prevented DA cytotoxicity. D-DOPA did not affect the viability of 10 DIC cultures, though it elicited marked toxicity in 3 DIC cultures. These results suggest that there are two components in the mechanisms that mediate the L-DOPA neurotoxicity on striatal neurons: one is autoxidation-relevant and the other is autoxidation-irrelevant. With respect to the latter, glutamate receptor stimulation may be involved. In contrast, autoxidation plays an important role in DA neurotoxicity.
Collapse
Affiliation(s)
- N Cheng
- Department of Pharmacology, Faculty of Phannaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Löfberg C, Harro J, Gottfries CG, Oreland L. Cholecystokinin peptides and receptor binding in Alzheimer's disease. J Neural Transm (Vienna) 1996; 103:851-60. [PMID: 8872869 DOI: 10.1007/bf01273363] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cholecystokinin (CCK) is a peptide that can be found in the cerebral cortex in high concentrations and is involved in learning and memory as well as neurodegenerative processes. Cortical brain samples from 9 patients with Alzheimer's disease and 9 matched control cases were studied with respect to the concentrations of various molecular forms of CCK and the CCK receptor binding characteristics. No differences were found between patients and controls in any of these measures. Significant correlations were found between the concentrations of CCK-8 sulphated and the three nonsulphated CCK peptides measured. In addition, the concentrations of CCK-4 and CCK-5 showed a highly significant and positive correlation.
Collapse
Affiliation(s)
- C Löfberg
- Department of Medical Pharmacology, University of Uppsala, Sweden
| | | | | | | |
Collapse
|
28
|
Gronier B, Debonnel G. Electrophysiological evidence for the implication of cholecystokinin in the modulation of the N-methyl-D-aspartate response by sigma ligands in the rat CA3 dorsal hippocampus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 1996; 353:382-90. [PMID: 8935704 DOI: 10.1007/bf00261434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Previous studies from our laboratory have demonstrated that low doses of selective sigma (sigma) ligands potentiate the response of pyramidal neurones to N-methyl-D-aspartate (NMDA) in the CA3 region of the rat dorsal hippocampus. It has also been found that the neuropeptide cholecystokinin (CCK) is involved in the effects induced by sigma ligands on colonic motility. The present experiments were undertaken to determine if this interaction is also present in the rat dorsal hippocampus. Using microiontophoresis and in vivo extracellular recordings of rat CA3 dorsal hippocampus pyramidal neurones, we assessed the effects of CCKA and CCKB receptor antagonists on the potentiation of the NMDA response, induced by the intravenous administration of low doses of the sigma ligands 1,3-di(2-tolyl)guanidine (DTG), (+)-pentazocine and JO-1784. The potentiation of the NMDA response induced by these sigma ligands was abolished by the selective CCKA receptor antagonist SR 27897, but not by the CCKB antagonist Cl-988. CCK-8S, applied with a low current, insufficient to induce by itself an increase of the firing activity, markedly potentiated the response of NMDA without affecting significantly that of quisqualate. SR 27897, but not Cl-988, significantly reduced the potentiation of the NMDA response by CCK-8S. These results suggest the existence of a functional interaction between CCK and sigma receptor-mediated effects in the dorsal hippocampus.
Collapse
Affiliation(s)
- B Gronier
- Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | | |
Collapse
|
29
|
Shimohama S, Akaike A, Kimura J. Nicotine-induced protection against glutamate cytotoxicity. Nicotinic cholinergic receptor-mediated inhibition of nitric oxide formation. Ann N Y Acad Sci 1996; 777:356-61. [PMID: 8624112 DOI: 10.1111/j.1749-6632.1996.tb34445.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cortical neurodegeneration in Alzheimer's disease (AD) is suggested to be attributable not only to beta-protein but also to glutamate. Although degeneration of cholinergic projection to the cerebral cortex is recognized to be one of the most prominent pathological changes in AD, there is only limited information concerning the cholinergic interaction with the cortical neurodegeneration. This study was performed to examine the protective effect of nicotine against glutamate-induced cytotoxicity using rat cultured cortical neurons. The cell viability was significantly reduced when cultures were briefly exposed to glutamate or N-methyl-D-aspartate (NMDA). The simultaneous addition of nicotine did not reduce glutamate cytotoxicity. In contrast, the simultaneous application of NMDA receptor antagonists such as MK-801 reduced glutamate cytotoxicity. Incubating the cultures with nicotine (10 microM) for 0.5-24 h prior to glutamate exposure reduced its cytotoxicity. Neuroprotection by nicotine was dependent on both the concentration and incubation period. In contrast to nicotine, muscarine (10 microM) weakly potentiated glutamate cytotoxicity. The neuroprotective effect of nicotine against glutamate cytotoxicity was antagonized by hexamethonium but not by artopine. Nicotine prevented NMDA cytotoxicity but did not affect cytotoxicity induced by either kainate or alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA). Cell viability was significantly reduced by a brief exposure of cultures to ionomycin, a calcium ionophore. Ionomycin-induced cytotoxicity was abolished by removing Ca2+ from incubating medium. S-nitrosocysteine (SNOC), which spontaneously releases nitric oxide (NO), also induced delayed cell death. Nicotine prevented ionomycin-induced cytotoxicity without affecting SNOC-induced cytotoxicity. These results suggest that nicotinic cholinergic receptor stimulation induces neuroprotection against glutamate cytotoxicity by its inhibitory action on NO-formation. Therefore, we propose that acetylcholine, acting through nicotinic cholinergic receptors, can function as a putative neuroprotective factor against neurodegeneration caused by the excessive release of glutamate and/or NMDA receptor activation.
Collapse
Affiliation(s)
- S Shimohama
- Department of Neurology, Faculty of Medicine, Kyoto University, Japan.
| | | | | |
Collapse
|
30
|
Yoon KW, Mitchell HL, Broder LD, Brooker RW, Delisle RK. Trauma-induced neurotoxicity in rat hippocampal neurons. Stroke 1996; 27:122-6. [PMID: 8553387 DOI: 10.1161/01.str.27.1.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND PURPOSE We have previously shown that traumatic injury of hippocampal cells triggers release of a soluble neurotoxin that can be transferred to an uninjured culture. The mechanism of this trauma-induced neurotoxicity is independent of glutamate receptor activation. We extended this observation to study the mechanism of this neurotoxicity. METHODS Dissociated rat hippocampal neurons were traumatized by disrupting the culture by scratching the plate. The toxicity expressed by the injured culture was studied by transferring the medium to an uninjured culture and assessing the death rate by trypan blue exclusion. RESULTS This neurotoxin is stable in the medium at room temperature for several hours and withstands boiling. The molecular weight is between 100 and 500. The release and the effect of this toxin seem to be independent of glutamate receptor activation. The toxicity is unaffected by removal of extracellular calcium. However, dantrolene dose-dependently blocked the toxicity in the recipient culture, suggesting that the release of intracellular stores of calcium is involved in the toxic effect. This release of calcium is likely to be followed by an activation of nitric oxide synthase because competitive nitric oxide synthase inhibitors attenuated this toxicity. Consistent with this result, cholecystokinin octapeptide significantly reduced cell death when combined with this toxic medium. CONCLUSIONS Traumatic injury of dissociated cells can propagate neurotoxicity in uninjured cells by a soluble toxin released into the extracellular space. This toxin causes a rise in cytosolic calcium that activates nitric oxide synthase that can be blocked by cholecystokinin.
Collapse
Affiliation(s)
- K W Yoon
- Department of Surgery, St Louis, Mo University Health Science Center, MO 63110-0250, USA
| | | | | | | | | |
Collapse
|
31
|
Gabriel S, Grützmann R, Lemke M, Gabriel HJ, Henklein P, Davidowa H. Interaction of cholecystokinin and glutamate agonists within the dLGN, the dentate gyrus, and the hippocampus. Brain Res Bull 1996; 39:381-9. [PMID: 9138748 DOI: 10.1016/0361-9230(96)00030-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The interaction of sulfated cholecystokinin (CCK-8S) with excitatory amino acids (EAA) was studied on single units of the dorsal lateral geniculate nucleus (dLGN), the dentate gyrus, and the hippocampal CA3 region in rats anaesthetized with urethane. lontophoretic co-administration of small, individually ineffective currents of CCK-8S and kainic acid or N-methyl-D-aspartate repeatedly elicited an increase of the discharge rate in nearly all geniculate and half of the dentate neurons but not in those of the CA3 region. The effect could be reduced by the CCKB receptor antagonist PD 135,158 more often than by the CCKA antagonist KL 1001. The increased firing due to co-administration of CCK and kainate could also be suppressed by the non-NMDA antagonist CNQX but not by the NMDA antagonists CPP or AP-5, which were otherwise able to prevent the neuron from responding to co-administration of CCK and NMDA. It is suggested that in distinct brain regions the effectivity of the "low level" EAA transmission may be enhanced by small amounts of CCK-8S. This is thought to be mediated by a coactivation of CCK and EEA receptors.
Collapse
Affiliation(s)
- S Gabriel
- Department of Neurophysiology, Institute of Physiology, Humboldt University Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Tsuji K, Nakamura Y, Ogata T, Mitani A, Kataoka K, Shibata T, Ishida M, Shinozaki H. Neurotoxicity of acromelic acid in cultured neurons from rat spinal cord. Neuroscience 1995; 68:585-91. [PMID: 7477968 DOI: 10.1016/0306-4522(95)00149-d] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Acromelic acid A, which contains the kainic acid structure in its molecule, is known to cause selective damage of interneurons in the rat lower spinal cord. In the present study, the potent neurotoxicity of acromelic acid A was demonstrated in cultured rat spinal neurons in terms of the activity of lactate dehydrogenase that was released from degenerated neurons into the culture medium. Acromelic acid A increased the lactate dehydrogenase activity in time- and concentration-dependent manners, and its EC50 was about 2.5 microM, which was much lower than that of kainic acid (70 microM) and (RS)-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (EC50; 11 microM). The maximum level of lactate dehydrogenase released by acromelic acid A was quite similar to that by kainic acid, but was about twice the level produced by (RS)-alpha-3-hydroxy-5-methyl-4-isoxazolepropionic acid. Exposure to acromelic acid A caused release of L-glutamate from the cells into the medium; however, the concentration of L-glutamate released was far below the level for inducing the neurotoxic effects. The neurotoxicity of 10 microM acromelic acid A was almost completely inhibited by 30 microM 6-nitro-7-sulphamoylbenzo(F)quinoxaline-2,3-dione and 6-cyano-7-nitroquinoxaline-2,3-dione, potent antagonists for non-N-methyl-D-aspartate receptors, but was partly (35%) reduced by 30 microM dizocilpine maleate. In cultured hippocampal neurons, the toxicity of acromelic acid A was significantly less effective (EC50: 18 microM) than that in spinal neurons, whereas the toxicity of kainic acid was almost the same in both neurons. These results suggest that acromelic acid A directly activates non-N-methyl-D-aspartate receptors on the cultured spinal neurons to induce neuronal death. A new type of non-N-methyl-D-aspartate receptors which is specific to acromelic acid A is suggested to be present at least in spinal neurons.
Collapse
Affiliation(s)
- K Tsuji
- Department of Physiology, Ehime University, School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Shimohama S, Akaike A, Tamura Y, Matsushima H, Kume T, Fujimoto S, Takenawa T, Kimura J. Glutamate-induced antigenic changes of phospholipase C-delta in cultured cortical neurons. J Neurosci Res 1995; 41:418-26. [PMID: 7563235 DOI: 10.1002/jnr.490410315] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Phosphoinositide-specific phospholipase C (PLC) is a key enzyme in signal transduction. It was previously demonstrated that an antibody to an isozyme of PLC, PLC-delta, produces intense staining of neurofibrillary tangles (NFT), the neurites surrounding senile plaque (SP) cores and neuropil threads in the brains of patients with Alzheimer's disease (AD). Although the etiology of neuronal degeneration in AD is still to be defined, excitotoxic glutamate might be a candidate. In the present study, an anti-PLC-delta antibody was used to examine the influence of glutamate on PLC-delta immunoreactivity in cultured rat cortical neurons. Exposure to glutamate caused the death of cultured cortical neurons and exhibited increased immunostaining with the anti-PLC-delta antibody. Subtoxic doses of glutamate also increased PLC-delta immunoreactivity in a dose-dependent manner. Both glutamate-induced neuronal degeneration and the increases in PLC-delta immunoreactivity were prevented by removal of extracellular Ca2+ or the application of an N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801. The glutamate-induced increase in PLC-delta immunoreactivity was also prevented by N omega-nitro-L-arginine, a nitric oxide (NO) synthase inhibitor. These results suggest that NO formation secondary to Ca2+ influx by NMDA receptor activation leads to similar modifications of PLC-delta to those seen in AD.
Collapse
Affiliation(s)
- S Shimohama
- Department of Neurology, Faculty of Medicine, Kyoto University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Yasui M, Kawasaki K. CCKB receptor activation protects CA1 neurons from ischemia-induced dysfunction in stroke-prone spontaneously hypertensive rats hippocampal slices. Neurosci Lett 1995; 191:99-102. [PMID: 7659302 DOI: 10.1016/0304-3940(95)11570-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We examined the effect of cholecystokinin octapeptide sulfated type (CCK-8S) on dysfunction of CA1 pyramidal neurons induced by in vitro ischemic insult in hippocampal slices of stroke-prone spontaneously hypertensive rats (SHRSP). CCK-8S shortened the time required for partial recovery from block of a population spike produced by ischemia. Furthermore, CCK-8S reduced ischemic insult-induced accumulation of K+ in extracellular space. Suppression of the K+ conductance by the CCKB receptor activation is suggested to contribute to neuroprotection by CCK-8S.
Collapse
Affiliation(s)
- M Yasui
- Division of Pharmacology, Shionogi & Co., Ltd., Osaka, Japan
| | | |
Collapse
|
35
|
Akaike A, Kaneko S, Tamura Y, Nakata N, Shiomi H, Ushikubi F, Narumiya S. Prostaglandin E2 protects cultured cortical neurons against N-methyl-D-aspartate receptor-mediated glutamate cytotoxicity. Brain Res 1994; 663:237-43. [PMID: 7874506 DOI: 10.1016/0006-8993(94)91268-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effects of prostaglandin (PG) E2 on glutamate-induced cytotoxicity were examined using primary cultures of rat cortical neurons. The cell viability was significantly reduced when cultures were briefly exposed to either glutamate or N-methyl-D-aspartate (NMDA) then incubated with normal medium for 1 h. Similar cytotoxicity was observed with the brief application of ionomycin, a calcium ionophore, and S-nitrosocysteine, a nitric oxide (NO)-generating agent. PGE2 at concentrations of 0.01-1 microM dose-dependently ameliorated the glutamate-induced cytotoxicity. PGE1, butaprost, an EP2 receptor agonist, and 8-bromo-cAMP were also effective in protecting cultures against glutamate cytotoxicity. By contrast, neither 17-phenyl-omega-trinor-PGE2, an EP1 receptor agonist, nor M&B 28767, an EP3 receptor agonist, affected glutamate-induced cytotoxicity. NMDA-induced cytotoxicity was ameliorated by PGE2, butaprost, MK-801, N-omega-nitro-L-arginine, a NO synthase inhibitor, and hemoglobin, which binds NO. These agents excluding MK-801 ameliorated the ionomycin-induced cytotoxicity. The cytotoxicity induced by S-nitrosocysteine was prevented only by hemoglobin but not by the other agents including PGE2. These findings indicate that PGE2 protects cultured cortical neurons against NMDA receptor-mediated glutamate neurotoxicity via EP2 receptors. EP2 receptor stimulation may suppress a step in NO formation triggered by Ca(2+)-influx through NMDA receptors.
Collapse
Affiliation(s)
- A Akaike
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Amano T, Ujihara H, Matsubayashi H, Sasa M, Yokota T, Tamura Y, Akaike A. Dopamine-induced protection of striatal neurons against kainate receptor-mediated glutamate cytotoxicity in vitro. Brain Res 1994; 655:61-9. [PMID: 7812790 DOI: 10.1016/0006-8993(94)91597-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effects of dopamine on glutamate-induced cytotoxicity were examined using the primary cultures of rat striatal neurons. Cell viability was significantly reduced by exposure of cultures to glutamate or kainate for 24 h. In contrast, similar application of N-methyl-D-aspartate (NMDA) or alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionate (AMPA) did not induce cytotoxicity. Kainate-induced cytotoxicity was significantly inhibited by kynurenate but not by MK-801. Dopamine at concentrations of 1-100 microM dose-dependently reduced kainate-induced cytotoxicity. Forskolin also significantly reduced kainate cytotoxicity. The neuroprotective effect of dopamine was antagonized by SCH 23390, a D1 receptor antagonist, but not by domperidone, a D2 receptor antagonist. Moreover, kainate-induced cytotoxicity was prevented by SKF 38393, a D1 receptor agonist, or forskolin but not by quinpirole, a D2 receptor agonist. The patch clamp study revealed that the same striatal neurons responded to both kainate and NMDA. During voltage clamp recording, neither kainate-induced currents nor NMDA-induced currents were affected by dopamine. Moreover, dopamine did not affect glutamate- or kainate-induced Ca2+ influx measured with fura-2. These findings indicate that dopamine prevents kainate receptor-mediated cytotoxicity without affecting the kainate receptor activities and intracellular Ca2+ movement. Dopamine-induced neuroprotection may be mediated by an increased intracellular cAMP formed following activation of D1 receptors.
Collapse
Affiliation(s)
- T Amano
- Department of Pharmacology, Hiroshima University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Akaike A, Tamura Y, Yokota T, Shimohama S, Kimura J. Nicotine-induced protection of cultured cortical neurons against N-methyl-D-aspartate receptor-mediated glutamate cytotoxicity. Brain Res 1994; 644:181-7. [PMID: 7519524 DOI: 10.1016/0006-8993(94)91678-0] [Citation(s) in RCA: 160] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The effects of nicotine on glutamate-induced cytotoxicity were examined using primary cultures of rat cortical neurons. The cell viability was significantly reduced when cultures were briefly exposed to glutamate or N-methyl-D-aspartate (NMDA) then incubated with normal medium for 1 h. A 1-h exposure of the cultures to kainate or alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) reduced cell viability. Incubating cultures with nicotine for 1-24 h protected cortical neurons against glutamate cytotoxicity. Maximum protection against glutamate cytotoxicity was induced with a 2-h nicotine incubation. Exposure to nicotine for up to 2 h did not affect cell viability by itself although cell viability was reduced in a time-dependent manner when the exposure exceeded 4 h. Neuroprotection by nicotine was dependent on both the concentration and incubation period. Nicotine reduced the NMDA cytotoxicity but did not attenuate that of kainate and AMPA. The neuroprotective effects of nicotine against glutamate cytotoxicity were antagonized by mecamylamine and hexamethonium but not by atropine. These results indicate that nicotinic receptor stimulation induces neuroprotection against glutamate cytotoxicity mediated by NMDA receptors.
Collapse
Affiliation(s)
- A Akaike
- Department of Neuropharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Japan
| | | | | | | | | |
Collapse
|
38
|
Minamoto Y, Tanaka T, Shibata S, Watanabe S. Neuroprotective effect of cholecystokininB receptor antagonist on ischemia-induced decrease in CA1 presynaptic fiber spikes in rat hippocampal slices. Neurosci Lett 1994; 167:81-4. [PMID: 8177534 DOI: 10.1016/0304-3940(94)91032-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The effects of cholecystokinin (CCK) receptor agonists and antagonists on hypoxia/hypoglycemia (ischemia)-induced decrease in CA1 presynaptic fiber spikes elicited by the stimulation of Schaffer collaterals were investigated using rat hippocampal slices. Treatment with the CCKB receptor agonist CCK tetrapeptide (CCK4, 0.01-10 microM) exacerbated the ischemia-induced decrease in the CA1 presynaptic potential in a concentration-dependent manner. Whereas, treatment with the CCKB receptor antagonist [(3R(+)-N-(2,3-dihydro-1-methyl-2-oxo-5-phenyl-1H-1,4- benzodiazepin-3-yl)-N1-(3-methylphenyl)-urea] (L365260), and not with CCKA receptor antagonist [(3S(-)-N-(2,3-dihydro-1-methyl-2-oxo-5-phenyl-1H-1,4- benzodiazepin-3-yl)-1H-indole-2-carboxamide] (L364718), produced a concentration-dependent attenuation of the ischemia-induced decrease. The magnitude of recovery of the CA1 field potentials in L365260-treated groups at 10 and 100 nM was 34% and 45%, respectively. The neuroprotective effect of L365260 (0.01 and 0.1 microM) was completely blocked by co-treatment with CCK4 (0.1 microM), a concentration that did not affect the decreased presynaptic potential induced by ischemia. These results demonstrated that the stimulation of the CCKB receptor played a detrimental role in the development of ischemic damage, whereas the blockade of CCKB receptors played a neuroprotective role in ischemic damage, suggesting a facilitatory role of CCK receptor-operated function in ischemia-induced neuronal deficits.
Collapse
Affiliation(s)
- Y Minamoto
- Research Laboratory, Yoshitomi Pharmaceutical Industries Ltd., Fukuoka, Japan
| | | | | | | |
Collapse
|
39
|
Abstract
CCK was first identified and characterized in the digestive tract where it is known to be a factor involved in the control of gut motility. Later, CCK and CCK receptors were identified in regions of the central nervous system that are associated with the control of emotion, motivation and sensory processing. The recent discovery and development of CCK-receptor antagonists having selective affinity for either CCKA or CCKB receptors has led to a better understanding of the functional role of CCK and its binding sites in the brain and periphery. Some of these compounds are being examined in man for their therapeutic usefulness in mental as well as in digestive disorders. This review will highlight the results from both basic and clinical investigations that have examined the effects of selective CCK receptor ligands. The focus will be on the central nervous system pharmacology of CCK antagonists and the involvement of CCK in gastrointestinal and colonic motility.
Collapse
|
40
|
Akaike A, Tamura Y, Terada K, Nakata N. Regulation by neuroprotective factors of NMDA receptor mediated nitric oxide synthesis in the brain and retina. PROGRESS IN BRAIN RESEARCH 1994; 103:391-403. [PMID: 7886220 DOI: 10.1016/s0079-6123(08)61153-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- A Akaike
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Kyoto University, Japan
| | | | | | | |
Collapse
|
41
|
Abstract
Cholecystokinin (CCK) has emerged as an important mammalian neuropeptide, localized in peripheral organs and in the central nervous system. This review presents an overview of the molecular aspects of CCK peptides and CCK receptors, the anatomical distribution of CCK, the neurophysiological actions of CCK, release of CCK and effects of CCK on release of other neurotransmitters, and the actions of CCK on digestion, feeding, cardiovascular function, respiratory function, neurotoxicity and seizures, cancer cell proliferation, analgesia, sleep, sexual and reproductive behaviors, memory, anxiety, and dopamine-mediated exploratory and rewarded behaviors. Human clinical studies of CCK in feeding disorders and panic disorders are described. New findings are presented on potent, nonpeptide CCK antagonists, selective for the two CCK receptor subtypes, which demonstrate that endogenous CCK has biologically important effects on physiology and behavior.
Collapse
Affiliation(s)
- J N Crawley
- Section on Behavioral Neuropharmacology, National Institute of Mental Health, Bethesda, MD 20892
| | | |
Collapse
|
42
|
Shimohama S, Ogawa N, Tamura Y, Akaike A, Tsukahara T, Iwata H, Kimura J. Protective effect of nerve growth factor against glutamate-induced neurotoxicity in cultured cortical neurons. Brain Res 1993; 632:296-302. [PMID: 7908599 DOI: 10.1016/0006-8993(93)91164-n] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effect of recombinant human nerve growth factor (hNGF) and mouse NGF on cultured rat cortical neurons was examined. The DNA fragment coding the human NGF gene was isolated and inserted downstream from the SV40 promoter in a plasmid containing the dihydrofolate reductase cDNA, and this plasmid was introduced into Chinese hamster ovary (CHO) cells to establish cells producing recombinant hNGF. The recombinant hNGF protein secreted by CHO cells was confirmed to be biologically active in an assay using PC12 cells. Brief exposure of cortical cells to glutamate followed by incubation with glutamate-free medium reduced cell viability by 60-70% when compared with the control culture. Simultaneous addition of recombinant hNGF or mouse NGF to rat cortical cultures with glutamate did not affect this reduction of cell viability. However, 24 h pretreatment of rat cortical cultures with recombinant hNGF or mouse NGF resulted in a significant reduction of glutamate-induced neuronal damage. Mouse NGF also protected cortical neurons against N-methyl-D-aspartate (NMDA)- and kainate-induced neuronal damage. These findings suggest that NGF can protect cortical neurons against glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- S Shimohama
- Department of Neurology, Faculty of Medicine, Kyoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Shimohama S, Tamura Y, Akaike A, Tsukahara T, Ohara O, Watanabe S, Kimura J. Brain-derived neurotrophic factor pretreatment exerts a partially protective effect against glutamate-induced neurotoxicity in cultured rat cortical neurons. Neurosci Lett 1993; 164:55-8. [PMID: 7908729 DOI: 10.1016/0304-3940(93)90856-g] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effect of recombinant brain-derived neurotrophic factor (BDNF) on cultured rat cortical neurons was examined. Brief exposure of cortical neurons to glutamate followed by incubation with glutamate-free medium reduced cell viability by 60-70% when compared with the control value. Simultaneous addition of recombinant BDNF to rat cortical cultures with glutamate did not affect this reduction of cell viability. However, 24 h pretreatment of rat cortical cultures with recombinant BDNF resulted in a significant reduction of glutamate-induced neuronal damage. These findings suggest that BDNF can protect cortical neurons against glutamate-induced neurotoxicity.
Collapse
Affiliation(s)
- S Shimohama
- Department of Neurology, Faculty of Medicine, Kyoto University, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Yamamoto Y, Itano T, Miyamoto O, Tokuda M, Matsui H, Janjua NA, Suwaki H, Okada Y, Negi T, Murakami TH. Effect of ceruletide on epileptogenesis in amygdaloid kindled rats. Brain Res 1993; 630:353-6. [PMID: 8118705 DOI: 10.1016/0006-8993(93)90678-g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The inhibitory effects of ceruletide (CLT), a cholecystokinin-8 (CCK)-like peptide, were investigated in the epileptogenesis in the amygdaloid kindled rats. Lower doses of CLT (20-80 micrograms/kg) inhibited the progression of kindling process. After acquiring C5 stage, a higher dose (160 micrograms/kg) was required to suppress the seizure susceptibility. These results, in light of several previous studies showing no serious side effects, suggest that CLT might be useful as an anti-epileptogenic agent for clinical usage.
Collapse
Affiliation(s)
- Y Yamamoto
- Department of Physiology, Kagawa Medical School, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Akaike A, Tamura Y, Sato Y, Yokota T. Protective effects of a vitamin B12 analog, methylcobalamin, against glutamate cytotoxicity in cultured cortical neurons. Eur J Pharmacol 1993; 241:1-6. [PMID: 7901032 DOI: 10.1016/0014-2999(93)90925-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effects of methylcobalamin, a vitamin B12 analog, on glutamate-induced neurotoxicity were examined using cultured rat cortical neurons. Cell viability was markedly reduced by a brief exposure to glutamate followed by incubation with glutamate-free medium for 1 h. Glutamate cytotoxicity was prevented when the cultures were maintained in methylcobalamin-containing medium. Glutamate cytotoxicity was also prevented by chronic exposure to S-adenosylmethionine, which is formed in the metabolic pathway of methylcobalamin. Chronic exposure to methylcobalamin and S-adenosylmethionine also inhibited the cytotoxicity induced by N-methyl-D-aspartate or sodium nitroprusside that releases nitric oxide. In cultures maintained in a standard medium, glutamate cytotoxicity was not affected by adding methylcobalamin to the glutamate-containing medium. In contrast, acute exposure to MK-801, a NMDA receptor antagonist, prevented glutamate cytotoxicity. These results indicate that chronic exposure to methylcobalamin protects cortical neurons against NMDA receptor-mediated glutamate cytotoxicity.
Collapse
Affiliation(s)
- A Akaike
- Department of Neuropharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Japan
| | | | | | | |
Collapse
|
46
|
Ogata T, Nakamura Y, Tsuji K, Shibata T, Kataoka K. Steroid hormones protect spinal cord neurons from glutamate toxicity. Neuroscience 1993; 55:445-9. [PMID: 8104320 DOI: 10.1016/0306-4522(93)90513-f] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The effects of steroid hormones on glutamate neurotoxicity were examined in cultured spinal cord neurons. The extent of neuronal damage, produced by glutamate exposure for 15 min, was estimated based on the activity of lactate dehydrogenase released from degenerated neurons to the media during 24 h of post-exposure incubation. This damage was dependent on the glutamate concentrations used. The addition of dexamethasone, a synthetic steroid, in post-exposure media remarkably reduced the extent of damage in a dose-dependent manner. The half effective concentration for the steroid was approximately 0.7 microM, which was in the range of pharmacological concentration. Dexamethasone was effective even when it was added 2 h after glutamate exposure. Some endogenous steroid hormones--aldosterone, progesterone and testosterone--also showed similar neuroprotective effects. However, cholesterol, a precursor of these steroid hormones, had no effect on glutamate neurotoxicity. This direct protective effect on neurons against glutamate neurotoxicity may explain, at least partly, the mechanisms of beneficial effects of steroid hormones on in vivo spinal cord injury.
Collapse
Affiliation(s)
- T Ogata
- Department of Physiology, Ehime University, School of Medicine, Japan
| | | | | | | | | |
Collapse
|
47
|
Harro J, Westerling P, Oreland L. CCKB receptor activation reduces glutamate-induced depolarization in slices of rat cerebral cortex. J Neural Transm (Vienna) 1993; 93:61-6. [PMID: 8103993 DOI: 10.1007/bf01244938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Recent studies on cell cultures have indicated that the neuropeptide cholecystokinin (CCK) can prevent glutamate-induced cytotoxicity. In a preparation of rat cortical tissue placed into a two-compartment bath, the cortical tissue could be depolarized, relative to the corpus callosum, by superfusions of KCl or glutamate (1.25-10 mM). Caerulein (1-100 nM), a CCK receptor agonist, caused a rightward shift of the glutamate dose-response curve. The effect of caerulein was abolished by adding L365,260 (1 microM), a selective CCKB receptor antagonist. These findings suggest that CCK may be a physiological antagonist of glutamate-mediated neurotransmission in the rat brain.
Collapse
Affiliation(s)
- J Harro
- Department of Medical Pharmacology, Uppsala University, Sweden
| | | | | |
Collapse
|
48
|
Tamura Y, Sato Y, Akaike A, Shiomi H. Mechanisms of cholecystokinin-induced protection of cultured cortical neurons against N-methyl-d-aspartate receptor-mediated glutamate cytotoxicity. Brain Res 1992; 592:317-25. [PMID: 1360313 DOI: 10.1016/0006-8993(92)91691-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The protective effects of cholecystokinin (CCK) against glutamate-induced cytotoxicity were examined using cultured neurons obtained from the rat cerebral cortex. Cell viability was significantly reduced when the cultures were briefly exposed to glutamate or N-methyl-D-aspartate (NMDA) and then incubated with normal medium for 60 min. A 60-min exposure to kainate also reduced cell viability. CCK protected cortical neurons against glutamate-, NMDA- and kainate-induced cytotoxicity. Glutamate- and NMDA-induced cytotoxicity was also reduced by N omega-nitro-L-arginine, a nitric oxide (NO) synthase inhibitor. However, CCK did not prevent the cytotoxic effects of sodium nitroprusside (SNP) which spontaneously releases NO. Moreover, CCK did not affect NMDA-induced Ca2+ influx measured with rhod-2, a fluorescent Ca2+ indicator. Therefore, release of a NO-like factor from the cerebral cortex was assayed using the thoracic artery in vitro. When the artery was incubated with minced cerebral tissues, glutamate elicited marked relaxation. SNP also elicited relaxation of the smooth muscle. CCK inhibited glutamate-induced relaxation but did not affect that induced by SNP. These results indicate that CCK prevents NMDA receptor-mediated cytotoxicity without reducing the Ca2+ influx. It is suggested that CCK inhibits NO-formation triggered by NMDA receptor activation.
Collapse
Affiliation(s)
- Y Tamura
- 2nd Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Japan
| | | | | | | |
Collapse
|
49
|
Ogata T, Nakamura Y, Shibata T, Kataoka K. Release of excitatory amino acids from cultured hippocampal astrocytes induced by a hypoxic-hypoglycemic stimulation. J Neurochem 1992; 58:1957-9. [PMID: 1560245 DOI: 10.1111/j.1471-4159.1992.tb10075.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
An excess release of excitatory amino acids (EAA) is an important factor for postischemic brain damage. In the present communication, we demonstrate that cultured hippocampal cells release EAA after hypoxic-hypoglycemic treatment. The amounts of EAA released from astrocytes were appreciably above those released from neurons. Furthermore, the amount of aspartate released from astrocytes was comparable to that of glutamate, although the endogenous content of aspartate was one-fifth that of glutamate. The endogenous content of aspartate in astrocytes increased even after hypoxic-hypoglycemic treatment. These results suggests that ischemic neuronal death is due, at least in part, to the excitotoxicity of aspartate and glutamate derived from surrounding astrocytes.
Collapse
Affiliation(s)
- T Ogata
- Department of Physiology, Ehime University, School of Medicine, Japan
| | | | | | | |
Collapse
|