學術產出-Theses

Article View/Open

Publication Export

Google ScholarTM

政大圖書館

Citation Infomation

題名 愷他命和右旋愷他命異構物對大鼠行為及神經化學的影響效果:與右旋安非他命相比較
Behavioral and Neurochemical Effects of RS-ketamine and S-ketamine in Rats: Compared with d-Amphetamine
作者 王傳堯
Wang, Chuan-Yao
貢獻者 廖瑞銘<br>趙知章
Liao, Ruey-Ming<br>Chao, Chih-Chang
王傳堯
Wang, Chuan-Yao
關鍵詞 藥物酬賞和成癮
制約性場地偏好
行為致敏化
麩胺酸NMDA受體
多巴胺
大鼠
Drug reward and addiction
Conditioned place preference
Behavioral sensitization
Glutamate NMDA receptor
Dopamine
Rat
日期 2022
上傳時間 1-Aug-2022 18:14:28 (UTC+8)
摘要 愷他命是一種非競爭性氮-甲基天門冬胺酸(N-methyl-D-aspartate)受體拮抗劑,近年來被用於治療憂鬱症,但其神經行為機制仍然有待確證,其中一項問題是有關愷他命會致使精神迷幻和藥物上癮的效果。本研究實驗一利用一種同時測量制約性場地偏好和行為致敏化的藥癮行為動物模式,針對利用消旋性愷他命 (RS-KET)、右旋性愷他命異構物 [S(+)-KET]、和安非他命 (d-AMP),測試其藥物酬賞的行為藥理作用;實驗二以西方墨點法檢測五個腦區之麩胺酸與多巴胺相關的蛋白質、及大腦衍生神經滋養因子之表現量,分析藥物作用的神經化學機制。實驗一藉由七次間歇重複注射藥物的操弄,結果發現d-AMP組顯著引發場地偏好及行為致敏化的效果;RS-KET組及S-KET組沒有引發顯著的場地制約偏好,但有行為致敏化的反應,惟其藥效較d-AMP組弱。實驗二生化分析的結果顯示藥物影響效果包含:(1)依核和背側紋狀體之NMDA受體 (GluN1、GluN2A和GluN2B)蛋白質表現量顯著減少,(2)多巴胺相關蛋白質(D1受體、D2受體、和DAT)表現量依據腦區和藥物的不同而有差異,(3)BDNF的蛋白質表現量僅在杏仁核有增加的效應。綜合而言,相對於d-AMP引發制約性場地偏好及行為致敏化的行為藥理效果,RS-KET及S-KET的效果比較不明顯。這三種藥物之間的行為藥理差異,與不同腦區的特定蛋白質表現量變化有關。本研究顯示愷他命的藥物酬賞性質有別於心理興奮劑,這兩類藥物影響麩胺酸與多巴胺傳遞系統相關的蛋白質表現有差異,此結果可供後續探討這兩類藥物調控成癮行為的神經生物機制之用。
Ketamine is a non-competitive N-methyl-D-aspartate receptor antagonist that has recently been repurposed in depression therapy, although the underlying neurobehavioral mechanisms remain elusive. One of the key issues should be concerned is that ketamine may cause psychotic and addictive effects in its users. Thus, this study, in Experiment 1, was designed to investigate the effects of racemic ketamine [RS (+/-)-ketamine; RS-KET] and it S-isomer [S (+)-ketamine; S-KET] respectively, on a behavioral protocol combining tests of conditioned place preference (CPP) and behavioral sensitization (BS). Additionally, d-amphetamine (d-AMP) treatment was conducted as a positive control. In the Experiment 2, after completing behavioral test, the expression levels of NMDA receptor subunits, dopamine-related proteins, and brain-derived neurotrophic factor (BDNF) were analyzed by western blotting. The results of Experiment 1 showed that the d-AMP group significantly displayed CPP and BS effects following a drug treatment of 7-time intermittent repeated injections, while the RS-KET and S-KET groups did not have CPP but showed various degrees of BS. However, the magnitude of BS induced by RS-KET or S-KET was smaller than that of d-AMP. The results of Experiment 2 revealed drug effects significantly measured on (1) decreases of the protein expressions of NMDA receptor subunits (GluN1, GluN2A, and GluN2B) in both the nucleus accumbens and dorsal striatum, (2) region-dependent changes in dopamine-related proteins (D1 receptor, D2 receptor, and DAT), and (3) no change of BDNF except detecting an increase in the amygdala. Together, RS-KET and S-KET did not produce the effects of CPP and BS as profound as d-AMP on the present behavioral task. The disparity between drug treatments is related to the change of certain protein expression on specific brain area. These results indicate the differential characteristics of drug reward between ketamine and psychostimulant which could be associated with different patterns of protein expressions involved in the glutamate and dopamine transmission systems, highlighting the neurobiological mechanisms of drug reward and addiction underlying these drugs to be unfolded in future study.
參考文獻 Abiero A, Custodio RJP, Botanas CJ, Ortiz DM, Sayson VL, Kim M, Lee HJ, Yoon S, Lee YS, Cheong JH, Kim HJ (2021) 1-Phenylcyclohexan-1-amine hydrochloride (PCA HCl) alters mesolimbic dopamine system accompanied by neuroplastic changes: A neuropsychopharmacological evaluation in rodents. Neurochem Int 144:104962.
Arnsten AFT (1998) Catecholamine modulation of prefrontal cortical cognitive function. Trends Cogn Sci 2:435-447.
Bardo MT, Bevins RA (2000) Conditioned place preference: What does it add to our preclinical understanding of drug reward? Psychopharmacology (Berl.) 153:31-43.
Baudry M, Bi X, Gall C, Lynch G (2011) The biochemistry of memory: The 26year journey of a ‘new and specific hypotheses’. Neurobiol Learn Mem 95:125-133.
Brakatselos C, Delis F, Asprogerakas M-Z, Lekkas P, Tseti I, Tzimas PS, Petrakis EA, Halabalaki M, Skaltsounis LA, Antoniou K (2020) Cannabidiol modulates the motor profile and NMDA receptor-related alterations induced by ketamine. Neuroscience 454:105-115.
Beaulieu JM, Gainetdinov RR (2011) The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev 63:182–217.
Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH (2000) Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 47:351-354.
Berridge KC, Robinson TE (2016) Liking, wanting, and the incentive-sensitization theory of addiction. Am Psychol 71:670–679.
Buck SA, Torregrossa MM, Logan RW, Freyberg Z (2021) Roles of dopamine and glutamate co-release in the nucleus accumbens in mediating the actions of drugs of abuse. FEBS J 288:1462-1474.
Caffino L, Piva A, Giannotti G, Chio MD, Mottarlini F, Venniro M, Yew DT, Chiamulera C, Fumagalli F (2016) Ketamine self-administration reduces the homeostasis of the glutamate synapse in the rat brain. Mol Neurobiol 54:7186–7193.
Cahill E, Salery M, Vanhoutte P, Caboche J (2014) Convergence of dopamine and glutamate signaling onto striatal ERK activation in response to drugs of abuse. Front Pharmacol 4:172.
Calpe-López C, García-Pardo MP, Aguilar MA (2019) Cannabidiol treatment might promote resilience to cocaine and methamphetamine use disorders: A review of possible mechanisms. Molecules 24:2583.
Canuso CM, Singh JB, Fedgchin M, Alphs L, Lane R, Lim P, Pinter C, Hough D, Sanacora G, Manji H (2018) Efficacy and safety of intranasal esketamine for the rapid reduction of symptoms of depression and suicidality in patients at imminent risk for suicide: Results of a double-blind, randomized, placebo-controlled study. Am J Psychiatry 175:620-630.
Carlezon WA, Wise RA (1996) Rewarding actions of phencyclidine and related drugs in nucleus accumbens shell and frontal cortex. J Neurosci 16:3112–22.
Carlino D, De Vanna M, Tongiorgi E (2012) Is altered BDNF biosynthesis a general feature in patients with cognitive dysfunctions? Neuroscientist 19:345-353.
Castren E, Kojima M (2017) Brain-derived neurotrophic factor in mood disorders and antidepressant treatments. Neurobiol Dis 97:119–126.
Chang L, Zhang K, Pu Y, Qu Y, Wang SM, Xiong Z, Ren Q, Dong C, Fujita YK, Hashimoto K (2019) Comparison of antidepressant and side effects in mice after intranasal administration of (R,S)-ketamine, (R)-ketamine, and (S)-ketamine. Pharmacol, Biochem Behav 181:53-59.
Cheung ZH, Ip NY (2011) From understanding synaptic plasticity to the development of cognitive enhancers. Int J Neuropsychopharmacol 14:1247-1256.
Cooper S, Robison AJ, Mazei-Robison MS (2017) Reward circuitry in addiction. Neurotherapeutics 14:687-697.
Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, Thase ME, Winokur A, Van Nueten L, Manji H (2018) Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry 75:139–148.
Di Chiara G, Bassareo V (2005) Reward systems and addiction: What dopamine does and doesn’t do. Curr Opin Pharmacol 7:69–76.
Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, Kammerer WA, Quezado Z, Luckenbaugh DA, Salvadore G (2010) A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry 67:793–802.
Domino EF (2010) Taming the ketamine tiger. Anesthesiology 113:678–684.
Du Y, Du L, Cao J, Hölscher C, Feng Y, Su H, Wang Y, Yun KM (2017) Levo-tetrahydropalmatine inhibits the acquisition of ketamine-induced conditioned place preference by regulating the expression of ERK and CREB phosphorylation in rats. Behav Brain Res 317:367–373.
Dunham JS, Deakin JF, Miyajima F, Payton A, Toro CT (2009) Expression of hippocampal brain-derived neurotrophic factor and its receptors in Stanley consortium brains. J Psychiatr Res 43:1175–1184.
Gastaldon C, Papola D, Ostuzzi G, Barbui C (2019) Esketamine for treatment resistant depression: A trick of smoke and mirrors? Epidemiol Psychiatr Sci 29:1-4.
Ghitza UE, Zhai H,Wu P, Airavaara M, Shaham Y, Lu L (2010) Role of BDNF and GDNF in drug reward and relapse: A review. Neurosci Biobehav Rev 35:157-171.
Hashimoto K (2019) Rapid-acting antidepressant ketamine, its metabolites and other candidates: A historical overview and future perspective. Psychiatry Clin Neurosci 73:613-627.
Javoy-Agid F, Hirsch E, Dumas S, Duyckaerts C, Mallt J, Agid Y (1990) Decreased tyrosine hydroxylase messenger RNA in the surviving dopamine neurons of the substantia nigra in Parkinson’s disease: An in situ hybridization study. Neuroscience 38:245-253.
Jentsch JD, Roth RH (1999) The neuropsychopharmacology of phencyclidine: From NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia. Neuropsychopharmacology 20:201–225.
Jones S, Bonci A (2005) Synaptic plasticity and drug addiction. Curr Opin Pharmacol 5:20-25.
Kalivas PW, Stewart J (1991) Dopamine transmission in the initiation and expression of drug- and stress-induced sensitization of motor activity. Brain Res Rev 16:223–244.
Karege F, Vaudan G, Schwald M, Perroud N, La Harpe R (2005) Neurotrophin levels in postmortem brains of suicide victims and the effects of antemortem diagnosis and psychotropic drugs. Brain Res Mol Brain Res 136:29–37.
Kebabian JW, Calne DB (1979) Multiple receptors for dopamine. Nature 277:93–96.
Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB Jr, Charney DS (1994) Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 51:199–214.
Lett BT (1989) Repeated exposures intensify rather than diminish the rewarding effects of amphetamine, morphine, and cocaine. Psychopharmacology 98:357–362.
Li F, Fang Q, Liu Y, Zhao M, Li D, Wang J, Lu L (2008) Cannabinoid CB1 receptor antagonist rimonabant attenuates reinstatement of ketamine conditioned place preference in rats. Eur J Pharmacol 589:122–126.
Li X, Wolf ME (2015) Multiple faces of BDNF in cocaine addiction. Behav Brain Res 279:240–254.
Li CC, Wu ST, Cha TL, Sun GH, Yu DS, Meng E (2019) A survey for ketamine abuse and its relation to the lower urinary tract symptoms in Taiwan. Sci Rep 9:7240.
Li XJ, Yu JH, Wu X, Zhu XM, Lv P, Du Z, Lu Y, Wu X, Yao J (2022) Ketamine enhances dopamine D1 receptor expression by modulating microRNAs in a ketamine-induced schizophrenia-like mouse model. Neurotoxicol Teratol 91:107079.
Liao RM, Chang YH, Wang SH (1998) Influence of SCH23390 and spiperone on the expression of conditioned place preference induced by d-amphetamine or cocaine in the rat. Chin J Physiol 41:85–92.
Liao RM, Chang YH, Wang SH, Lan CH (2000) Distinct accumbal subareas are involved in place conditioning of amphetamine and cocaine. Life Sci 67:2033-2043.
Liao RM, Lin HL (2008) Differential effects of lesions in the subareas of the medial prefrontal cortex on the development of behavioral sensitization to amphetamine: The role of environmental context. Chin J Physiol 51:394-401.
Lisek M, Ferenc B, Studzian M, Pulaski L, Guo F, Zylinska L, Boczek T (2017) Glutamate deregulation in ketamine-induced psychosis—A potential role of PSD95, NMDA receptor and PMCA interaction. Front Cell Neurosci 11:181.
Liu Y, Lin D, Wu B, Zhou W (2016) Ketamine abuse potential and use disorder. Brain Res Bull 126:68–73.
Luo Y, Yu Y, Zhang M, He H, Fan N (2021) Chronic administration of ketamine induces cognitive deterioration by restraining synaptic signaling. Mol Psychiatry 26:4702–4718.
Martinowich K, Manji H, Lu B (2007) New insights into BDNF function in depression and anxiety. Nat Neurosci 10:1089–1093.
Mascia P, Wang Q, Brown J, Nesbitt KM, Kennedy RT, Vezina P (2020) Maladaptive consequences of repeated intermittent exposure to uncertainty. Prog Neuropsychopharmacol Biol Psychiatry 99:109864.
Mattson BJ, Koya E, Simmons DE, Mitchell TB, Berkow A, Crombag HS, Hope BT (2008) Context-specific sensitization of cocaine-induced locomotor activity and associated neuronal ensembles in rat nucleus accumbens. Eur J Neurosci 27:202–212.
McKendrick G, Graziane NM (2020) Drug-induced conditioned place preference and its practical use in substance use disorder research. Front Behav Neurosci 14:582147.
McCarthy DM, Brown AN, Bhide PG (2012) Regulation of BDNF expression by cocaine. Yale J Biol Med 85:437-446.
Meredith GE, Callen S, Scheuer DA (2002) Brain-derived neurotrophic factor expression is increased in the rat amygdala, piriform cortex and hypothalamus following repeated amphetamine administration. Brain Res 949:218–227.
Parsons CG, Quack G, Bresink I, Baran L, Przegalinski E, Kostowski W, Krzascik P, Hartmann S, Danysz W (1995) Comparison of the potency, kinetics and voltage-dependency of a series of uncompetitive NMDA receptor antagonists in vitro with anticonvulsive and motor impairment activity in vivo. Neuropharmacology 34:1239–1258.
Piva A, Caffino L, Mottarlini F, Pintori N, Díaz FC, Fumagalli F, Chiamulera C (2021) Metaplastic effects of ketamine and MK-801 on glutamate receptors expression in rat medial prefrontal cortex and hippocampus. Mol Neurobiol 58:3443–3456.
Price RB, Nock MK, Charney DS, Mathew SJ (2009) Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry 66:522–526.
Qi XR, Zhao J, Liu J, Fang H, Swaab DF, Zhou JN (2015) Abnormal retinoid and TrkB signaling in the prefrontal cortex in mood disorders. Cereb Cortex 25:75–83.
Rademacher DJ, Kovacs B, Shen F, Napier C, Meredith GE (2006) The neural substrates of amphetamine conditioned place preference: Implications for the formation of conditioned stimulus-reward associations. Eur J Neurosci 24:2089–2097.
Robinson TE, Becker JB (1986 ) Enduring changes in brain and behavior produced by chronic amphetamine administration: A review and evaluation of animal models of amphetamine psychosis. Brain Res Rev 11:157-198.
Robinson TE, Berridge KC (1993) The neural basis of drug craving: An incentive-sensitization theory of addiction. Brain Res Rev 18:247–291.
Sanchis-Segura C, Spanagel R (2006) Behavioural assessment of drug reinforcement and addictive features in rodents: an overview. Addict Biol 11:2-38.
Schoepfer KJ, Strong CE, Saland SK, Wright KN, Kabbaj M (2019) Sex- and dose-dependent abuse liability of repeated subanesthetic ketamine in rats. Physiol Behav 203:60–69.
Shen YL, Chang TY, Chang YC, Tien HH, Yang FC, Wang PY, Liao RM (2014) Elevated BDNF mRNA expression in the medial prefrontal cortex after d-amphetamine reinstated conditioned place preference in rats. Neuroscience 263:88-95.
Shen F, Meredith GE, Napier TC (2006) Amphetamine-induced place preference and conditioned motor sensitization requires activation of tyrosine kinase receptors in the hippocampus. J Neurosci 26:11041–11051.
Shuto T, Kuroiwa M, Hamamura M, Yabuuchi K, Shimazoe T, Watanabe S, Nishi A, Yamamoto T (2006) Reversal of methamphetamine-induced behavioral sensitization by repeated administration of a dopamine D1 receptor agonist. Neuropharmacology 50:991-997.
Sial OK, Pariseb EM, Pariseb LF, Gneccoa T, Bolaños-Guzmán CA (2020) Ketamine: The final frontier or another depressing end? Behav Brain Res 383:112508.
Singh JB, Fedgchin M, Daly E, Xi L, Melman C, De Bruecker G, Tadic A, Sienaert P,
Wiegand F, Manji H (2016) Intravenous esketamine in adult treatment-resistant depression: A double-blind, double-randomization, placebo-controlled study. Biol Psychiatry 80:424–431.
Speranza L, di Porzio U, Viggiano Davide, de Donato A, Volpicelli F (2021) Dopamine: The neuromodulator of long-term synaptic plasticity, reward and movement control. Cells 10:735.
Spyraki C, Fibiger HC, Phillips AG (1982) Dopaminergic substrates of amphetamine-induced place preference conditioning. Brain Res 253:185–193.
Steketee JD, Kalivas PW (2011) Drug wanting: Behavioral sensitization and relapse to drug-seeking behavior. Pharmacol Rev 63:348–365.
Strong CE, Schoepfer KJ, Dossat AM, Saland SK, Wright KN, Kabbaj M (2017) Locomotor sensitization to intermittent ketamine administration is associated with nucleus accumbens plasticity in male and female rats. Neuropharmacology 121:195-203.
Sulzer D (2011) How addictive drugs disruptpresynaptic dopamine neurotransmission. Neuron 69:628–649.
Sun Y, Chen G, Zhou K, Zhu Y (2018) A conditioned place preference protocol for measuring incubation of craving in rats. J Vis Exp 141:e58384.
Sun Z, Ma Y, Xie L, Huang J, Duan S, Guo R, Xie Y, Lv J, Lin Z, Ma Shuhua (2019) Behavioral changes and neuronal damage in rhesus monkeys after 10 weeks of ketamine administration involve prefrontal cortex dopamine D2 receptor and dopamine transporter. Neuroscience 415:97-106.
Suzuki T, Katob H, Aokib T, Tsudab M, Naritaa M, Misawa M (2000) Effects of the non-competitive NMDA receptor antagonist ketamine on morphine-induced place preference in mice. Life Sciences 67:383–389.
Traynelis SF , Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R (2010) Glutamate receptor ion channels: Structure, regulation, and function. Pharmacol Rev 62:405-96.
Tritsch NX, Sabatini BL (2012) Dopaminergic modulation of synaptic transmission in cortex and striatum. Neuron 76:33–50.
Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden W, Ritz L, Norquist G, Howland RH, Lebowitz B, McGrath PJ, Shores-Wilson K, Biggs MM, Balasubramani GK, Fava M (2006) Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: Implications for clinical practice. Am J Psychiatry 163:28-40.
Trujillo KA, Heller CY (2020) Ketamine sensitization: Influence of dose, environment, social isolation, and treatment interval. Behav Brain Res 378:112-271.
Trujillo KA, Zamora JJ, Warmoth KP (2008) Increased response to ketamine following treatment at long intervals: Implications for intermittent use. Biol Psychiatry 63:178-183.
Turner EH (2019) Esketamine for treatment-resistant depression: Seven concerns about efficacy and FDA approval. Lancet Psychiatry 6:977–979.
Tzschentke TM (2007) Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol 12:227-462.
Uchihashi Y, Kuribara H, Morita T, Fujita T (1993) The repeated administration of ketamine induces an enhancement of its stimulant action in mice. Jpn J Pharmacol 61:149-151.
Wise RA (2004) Dopamine, learning and motivation. Nat Neuro Rev 5:483–494.
Wise RA (2009) Roles for nigrostriatal – not just mesocoticolimbic – dopamine in reward and addiction. Cell 32:517–524.
Xu DD, Moa ZX, Yung KKL, Yang Y, Leung AWN (2008) Individual and Combined Effects of Methamphetamine and Ketamine on Conditioned Place Preference and NR1 Receptor Phosphorylation in Rats. Neurosignals 15:322–331.
Yamamoto T, Nakayama T, Yamaguchi J, Matsuzawa M, Mishina M, Ikeda K,
Yamamoto H (2016) Role of the NMDA receptor GluN2D subunit in the expression of ketamine-induced behavioral sensitization and region-specific activation of neuronal nitric oxide synthase. Neurosci Lett 610:48-53.
Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, Dong C, Hashimoto K (2015) R-ketamine: A rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry 5: e632.
Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, Charney DS, Manji HK (2006) A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 63:856–864.
Zarate CA Jr, Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A, Selter J, Marquardt CA, Liberty V, Luckenbaugh DA (2012) Replication of ketamine’s antidepressant efficacy in bipolar depression: A randomized controlled add-on trial. Biol Psychiatry 71:939–946.
Zorumski CF, Izumi Y, Mennerick S (2016) Ketamine: NMDA receptors and beyond. J Neurosci 36:11158–11164.
描述 碩士
國立政治大學
神經科學研究所
108754003
資料來源 http://thesis.lib.nccu.edu.tw/record/#G0108754003
資料類型 thesis
dc.contributor.advisor 廖瑞銘<br>趙知章zh_TW
dc.contributor.advisor Liao, Ruey-Ming<br>Chao, Chih-Changen_US
dc.contributor.author (Authors) 王傳堯zh_TW
dc.contributor.author (Authors) Wang, Chuan-Yaoen_US
dc.creator (作者) 王傳堯zh_TW
dc.creator (作者) Wang, Chuan-Yaoen_US
dc.date (日期) 2022en_US
dc.date.accessioned 1-Aug-2022 18:14:28 (UTC+8)-
dc.date.available 1-Aug-2022 18:14:28 (UTC+8)-
dc.date.issued (上傳時間) 1-Aug-2022 18:14:28 (UTC+8)-
dc.identifier (Other Identifiers) G0108754003en_US
dc.identifier.uri (URI) http://nccur.lib.nccu.edu.tw/handle/140.119/141188-
dc.description (描述) 碩士zh_TW
dc.description (描述) 國立政治大學zh_TW
dc.description (描述) 神經科學研究所zh_TW
dc.description (描述) 108754003zh_TW
dc.description.abstract (摘要) 愷他命是一種非競爭性氮-甲基天門冬胺酸(N-methyl-D-aspartate)受體拮抗劑,近年來被用於治療憂鬱症,但其神經行為機制仍然有待確證,其中一項問題是有關愷他命會致使精神迷幻和藥物上癮的效果。本研究實驗一利用一種同時測量制約性場地偏好和行為致敏化的藥癮行為動物模式,針對利用消旋性愷他命 (RS-KET)、右旋性愷他命異構物 [S(+)-KET]、和安非他命 (d-AMP),測試其藥物酬賞的行為藥理作用;實驗二以西方墨點法檢測五個腦區之麩胺酸與多巴胺相關的蛋白質、及大腦衍生神經滋養因子之表現量,分析藥物作用的神經化學機制。實驗一藉由七次間歇重複注射藥物的操弄,結果發現d-AMP組顯著引發場地偏好及行為致敏化的效果;RS-KET組及S-KET組沒有引發顯著的場地制約偏好,但有行為致敏化的反應,惟其藥效較d-AMP組弱。實驗二生化分析的結果顯示藥物影響效果包含:(1)依核和背側紋狀體之NMDA受體 (GluN1、GluN2A和GluN2B)蛋白質表現量顯著減少,(2)多巴胺相關蛋白質(D1受體、D2受體、和DAT)表現量依據腦區和藥物的不同而有差異,(3)BDNF的蛋白質表現量僅在杏仁核有增加的效應。綜合而言,相對於d-AMP引發制約性場地偏好及行為致敏化的行為藥理效果,RS-KET及S-KET的效果比較不明顯。這三種藥物之間的行為藥理差異,與不同腦區的特定蛋白質表現量變化有關。本研究顯示愷他命的藥物酬賞性質有別於心理興奮劑,這兩類藥物影響麩胺酸與多巴胺傳遞系統相關的蛋白質表現有差異,此結果可供後續探討這兩類藥物調控成癮行為的神經生物機制之用。zh_TW
dc.description.abstract (摘要) Ketamine is a non-competitive N-methyl-D-aspartate receptor antagonist that has recently been repurposed in depression therapy, although the underlying neurobehavioral mechanisms remain elusive. One of the key issues should be concerned is that ketamine may cause psychotic and addictive effects in its users. Thus, this study, in Experiment 1, was designed to investigate the effects of racemic ketamine [RS (+/-)-ketamine; RS-KET] and it S-isomer [S (+)-ketamine; S-KET] respectively, on a behavioral protocol combining tests of conditioned place preference (CPP) and behavioral sensitization (BS). Additionally, d-amphetamine (d-AMP) treatment was conducted as a positive control. In the Experiment 2, after completing behavioral test, the expression levels of NMDA receptor subunits, dopamine-related proteins, and brain-derived neurotrophic factor (BDNF) were analyzed by western blotting. The results of Experiment 1 showed that the d-AMP group significantly displayed CPP and BS effects following a drug treatment of 7-time intermittent repeated injections, while the RS-KET and S-KET groups did not have CPP but showed various degrees of BS. However, the magnitude of BS induced by RS-KET or S-KET was smaller than that of d-AMP. The results of Experiment 2 revealed drug effects significantly measured on (1) decreases of the protein expressions of NMDA receptor subunits (GluN1, GluN2A, and GluN2B) in both the nucleus accumbens and dorsal striatum, (2) region-dependent changes in dopamine-related proteins (D1 receptor, D2 receptor, and DAT), and (3) no change of BDNF except detecting an increase in the amygdala. Together, RS-KET and S-KET did not produce the effects of CPP and BS as profound as d-AMP on the present behavioral task. The disparity between drug treatments is related to the change of certain protein expression on specific brain area. These results indicate the differential characteristics of drug reward between ketamine and psychostimulant which could be associated with different patterns of protein expressions involved in the glutamate and dopamine transmission systems, highlighting the neurobiological mechanisms of drug reward and addiction underlying these drugs to be unfolded in future study.en_US
dc.description.tableofcontents 中文摘要 I
英文摘要 II
表次 VI
圖次 VII
第一章、緒論 1
一、前言 1
二、愷他命的臨床運用 1
三、藥物酬賞相關的動物行為模式 2
(一)制約性場地偏好 (CPP) 2
(二)行為致敏化 (BS) 4
四、愷他命相關的神經化學反應 4
(一)酬賞動機相關的腦區 5
(二)麩胺酸 (Glutamate; Glu) 受體及功能 5
(三)多巴胺 (Dopamine; DA) 受體及功能 6
(四)大腦衍生神經滋養因子 (brain-dervied neurotrophic factor; BDNF) 7
五、研究目的 8
第二章、材料與方法 9
一、實驗動物 9
二、藥物 9
三、行為實驗測試儀器 9
(一)場地制約箱 9
(二)行為致敏箱 10
四、行為藥理實驗程序 10
(一)制約性場地偏好 (Conditioned place preference; CPP) 10
(二)行為致敏化 (Behavioral sensitization; BS) 11
五、行為藥理學實驗設計 11
六、西方墨點法 (Western blot) 12
七、統計分析 15
第三章、結果 16
ㄧ、藥物引發之成癮相關行為變化 16
二、藥物引發蛋白質表現量變化 17
(一)GluN1、GluN2及GluN2B蛋白質表現量之檢測結果 17
(二)D1受體、D2受體及DAT蛋白質表現量之檢測結果 18
(三)BDNF蛋白質表現量之檢測結果 19
三、行為數據與蛋白質含量的統計相關分析結果 19
第四章、討論 21
一、藥物引發CPP與BS之行為反應 21
二、藥物對NMDA受體之次單元的影響 23
三、藥物對DA相關蛋白質的影響 25
四、藥物對BDNF的影響 26
五、行為數據及生化分析結果的相關分析 26
六、結論 27
參考文獻 29
附表 37
附圖 44
zh_TW
dc.format.extent 12488915 bytes-
dc.format.mimetype application/pdf-
dc.source.uri (資料來源) http://thesis.lib.nccu.edu.tw/record/#G0108754003en_US
dc.subject (關鍵詞) 藥物酬賞和成癮zh_TW
dc.subject (關鍵詞) 制約性場地偏好zh_TW
dc.subject (關鍵詞) 行為致敏化zh_TW
dc.subject (關鍵詞) 麩胺酸NMDA受體zh_TW
dc.subject (關鍵詞) 多巴胺zh_TW
dc.subject (關鍵詞) 大鼠zh_TW
dc.subject (關鍵詞) Drug reward and addictionen_US
dc.subject (關鍵詞) Conditioned place preferenceen_US
dc.subject (關鍵詞) Behavioral sensitizationen_US
dc.subject (關鍵詞) Glutamate NMDA receptoren_US
dc.subject (關鍵詞) Dopamineen_US
dc.subject (關鍵詞) Raten_US
dc.title (題名) 愷他命和右旋愷他命異構物對大鼠行為及神經化學的影響效果:與右旋安非他命相比較zh_TW
dc.title (題名) Behavioral and Neurochemical Effects of RS-ketamine and S-ketamine in Rats: Compared with d-Amphetamineen_US
dc.type (資料類型) thesisen_US
dc.relation.reference (參考文獻) Abiero A, Custodio RJP, Botanas CJ, Ortiz DM, Sayson VL, Kim M, Lee HJ, Yoon S, Lee YS, Cheong JH, Kim HJ (2021) 1-Phenylcyclohexan-1-amine hydrochloride (PCA HCl) alters mesolimbic dopamine system accompanied by neuroplastic changes: A neuropsychopharmacological evaluation in rodents. Neurochem Int 144:104962.
Arnsten AFT (1998) Catecholamine modulation of prefrontal cortical cognitive function. Trends Cogn Sci 2:435-447.
Bardo MT, Bevins RA (2000) Conditioned place preference: What does it add to our preclinical understanding of drug reward? Psychopharmacology (Berl.) 153:31-43.
Baudry M, Bi X, Gall C, Lynch G (2011) The biochemistry of memory: The 26year journey of a ‘new and specific hypotheses’. Neurobiol Learn Mem 95:125-133.
Brakatselos C, Delis F, Asprogerakas M-Z, Lekkas P, Tseti I, Tzimas PS, Petrakis EA, Halabalaki M, Skaltsounis LA, Antoniou K (2020) Cannabidiol modulates the motor profile and NMDA receptor-related alterations induced by ketamine. Neuroscience 454:105-115.
Beaulieu JM, Gainetdinov RR (2011) The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev 63:182–217.
Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH (2000) Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 47:351-354.
Berridge KC, Robinson TE (2016) Liking, wanting, and the incentive-sensitization theory of addiction. Am Psychol 71:670–679.
Buck SA, Torregrossa MM, Logan RW, Freyberg Z (2021) Roles of dopamine and glutamate co-release in the nucleus accumbens in mediating the actions of drugs of abuse. FEBS J 288:1462-1474.
Caffino L, Piva A, Giannotti G, Chio MD, Mottarlini F, Venniro M, Yew DT, Chiamulera C, Fumagalli F (2016) Ketamine self-administration reduces the homeostasis of the glutamate synapse in the rat brain. Mol Neurobiol 54:7186–7193.
Cahill E, Salery M, Vanhoutte P, Caboche J (2014) Convergence of dopamine and glutamate signaling onto striatal ERK activation in response to drugs of abuse. Front Pharmacol 4:172.
Calpe-López C, García-Pardo MP, Aguilar MA (2019) Cannabidiol treatment might promote resilience to cocaine and methamphetamine use disorders: A review of possible mechanisms. Molecules 24:2583.
Canuso CM, Singh JB, Fedgchin M, Alphs L, Lane R, Lim P, Pinter C, Hough D, Sanacora G, Manji H (2018) Efficacy and safety of intranasal esketamine for the rapid reduction of symptoms of depression and suicidality in patients at imminent risk for suicide: Results of a double-blind, randomized, placebo-controlled study. Am J Psychiatry 175:620-630.
Carlezon WA, Wise RA (1996) Rewarding actions of phencyclidine and related drugs in nucleus accumbens shell and frontal cortex. J Neurosci 16:3112–22.
Carlino D, De Vanna M, Tongiorgi E (2012) Is altered BDNF biosynthesis a general feature in patients with cognitive dysfunctions? Neuroscientist 19:345-353.
Castren E, Kojima M (2017) Brain-derived neurotrophic factor in mood disorders and antidepressant treatments. Neurobiol Dis 97:119–126.
Chang L, Zhang K, Pu Y, Qu Y, Wang SM, Xiong Z, Ren Q, Dong C, Fujita YK, Hashimoto K (2019) Comparison of antidepressant and side effects in mice after intranasal administration of (R,S)-ketamine, (R)-ketamine, and (S)-ketamine. Pharmacol, Biochem Behav 181:53-59.
Cheung ZH, Ip NY (2011) From understanding synaptic plasticity to the development of cognitive enhancers. Int J Neuropsychopharmacol 14:1247-1256.
Cooper S, Robison AJ, Mazei-Robison MS (2017) Reward circuitry in addiction. Neurotherapeutics 14:687-697.
Daly EJ, Singh JB, Fedgchin M, Cooper K, Lim P, Shelton RC, Thase ME, Winokur A, Van Nueten L, Manji H (2018) Efficacy and safety of intranasal esketamine adjunctive to oral antidepressant therapy in treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry 75:139–148.
Di Chiara G, Bassareo V (2005) Reward systems and addiction: What dopamine does and doesn’t do. Curr Opin Pharmacol 7:69–76.
Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein P, Khalife S, Kammerer WA, Quezado Z, Luckenbaugh DA, Salvadore G (2010) A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch Gen Psychiatry 67:793–802.
Domino EF (2010) Taming the ketamine tiger. Anesthesiology 113:678–684.
Du Y, Du L, Cao J, Hölscher C, Feng Y, Su H, Wang Y, Yun KM (2017) Levo-tetrahydropalmatine inhibits the acquisition of ketamine-induced conditioned place preference by regulating the expression of ERK and CREB phosphorylation in rats. Behav Brain Res 317:367–373.
Dunham JS, Deakin JF, Miyajima F, Payton A, Toro CT (2009) Expression of hippocampal brain-derived neurotrophic factor and its receptors in Stanley consortium brains. J Psychiatr Res 43:1175–1184.
Gastaldon C, Papola D, Ostuzzi G, Barbui C (2019) Esketamine for treatment resistant depression: A trick of smoke and mirrors? Epidemiol Psychiatr Sci 29:1-4.
Ghitza UE, Zhai H,Wu P, Airavaara M, Shaham Y, Lu L (2010) Role of BDNF and GDNF in drug reward and relapse: A review. Neurosci Biobehav Rev 35:157-171.
Hashimoto K (2019) Rapid-acting antidepressant ketamine, its metabolites and other candidates: A historical overview and future perspective. Psychiatry Clin Neurosci 73:613-627.
Javoy-Agid F, Hirsch E, Dumas S, Duyckaerts C, Mallt J, Agid Y (1990) Decreased tyrosine hydroxylase messenger RNA in the surviving dopamine neurons of the substantia nigra in Parkinson’s disease: An in situ hybridization study. Neuroscience 38:245-253.
Jentsch JD, Roth RH (1999) The neuropsychopharmacology of phencyclidine: From NMDA receptor hypofunction to the dopamine hypothesis of schizophrenia. Neuropsychopharmacology 20:201–225.
Jones S, Bonci A (2005) Synaptic plasticity and drug addiction. Curr Opin Pharmacol 5:20-25.
Kalivas PW, Stewart J (1991) Dopamine transmission in the initiation and expression of drug- and stress-induced sensitization of motor activity. Brain Res Rev 16:223–244.
Karege F, Vaudan G, Schwald M, Perroud N, La Harpe R (2005) Neurotrophin levels in postmortem brains of suicide victims and the effects of antemortem diagnosis and psychotropic drugs. Brain Res Mol Brain Res 136:29–37.
Kebabian JW, Calne DB (1979) Multiple receptors for dopamine. Nature 277:93–96.
Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB Jr, Charney DS (1994) Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 51:199–214.
Lett BT (1989) Repeated exposures intensify rather than diminish the rewarding effects of amphetamine, morphine, and cocaine. Psychopharmacology 98:357–362.
Li F, Fang Q, Liu Y, Zhao M, Li D, Wang J, Lu L (2008) Cannabinoid CB1 receptor antagonist rimonabant attenuates reinstatement of ketamine conditioned place preference in rats. Eur J Pharmacol 589:122–126.
Li X, Wolf ME (2015) Multiple faces of BDNF in cocaine addiction. Behav Brain Res 279:240–254.
Li CC, Wu ST, Cha TL, Sun GH, Yu DS, Meng E (2019) A survey for ketamine abuse and its relation to the lower urinary tract symptoms in Taiwan. Sci Rep 9:7240.
Li XJ, Yu JH, Wu X, Zhu XM, Lv P, Du Z, Lu Y, Wu X, Yao J (2022) Ketamine enhances dopamine D1 receptor expression by modulating microRNAs in a ketamine-induced schizophrenia-like mouse model. Neurotoxicol Teratol 91:107079.
Liao RM, Chang YH, Wang SH (1998) Influence of SCH23390 and spiperone on the expression of conditioned place preference induced by d-amphetamine or cocaine in the rat. Chin J Physiol 41:85–92.
Liao RM, Chang YH, Wang SH, Lan CH (2000) Distinct accumbal subareas are involved in place conditioning of amphetamine and cocaine. Life Sci 67:2033-2043.
Liao RM, Lin HL (2008) Differential effects of lesions in the subareas of the medial prefrontal cortex on the development of behavioral sensitization to amphetamine: The role of environmental context. Chin J Physiol 51:394-401.
Lisek M, Ferenc B, Studzian M, Pulaski L, Guo F, Zylinska L, Boczek T (2017) Glutamate deregulation in ketamine-induced psychosis—A potential role of PSD95, NMDA receptor and PMCA interaction. Front Cell Neurosci 11:181.
Liu Y, Lin D, Wu B, Zhou W (2016) Ketamine abuse potential and use disorder. Brain Res Bull 126:68–73.
Luo Y, Yu Y, Zhang M, He H, Fan N (2021) Chronic administration of ketamine induces cognitive deterioration by restraining synaptic signaling. Mol Psychiatry 26:4702–4718.
Martinowich K, Manji H, Lu B (2007) New insights into BDNF function in depression and anxiety. Nat Neurosci 10:1089–1093.
Mascia P, Wang Q, Brown J, Nesbitt KM, Kennedy RT, Vezina P (2020) Maladaptive consequences of repeated intermittent exposure to uncertainty. Prog Neuropsychopharmacol Biol Psychiatry 99:109864.
Mattson BJ, Koya E, Simmons DE, Mitchell TB, Berkow A, Crombag HS, Hope BT (2008) Context-specific sensitization of cocaine-induced locomotor activity and associated neuronal ensembles in rat nucleus accumbens. Eur J Neurosci 27:202–212.
McKendrick G, Graziane NM (2020) Drug-induced conditioned place preference and its practical use in substance use disorder research. Front Behav Neurosci 14:582147.
McCarthy DM, Brown AN, Bhide PG (2012) Regulation of BDNF expression by cocaine. Yale J Biol Med 85:437-446.
Meredith GE, Callen S, Scheuer DA (2002) Brain-derived neurotrophic factor expression is increased in the rat amygdala, piriform cortex and hypothalamus following repeated amphetamine administration. Brain Res 949:218–227.
Parsons CG, Quack G, Bresink I, Baran L, Przegalinski E, Kostowski W, Krzascik P, Hartmann S, Danysz W (1995) Comparison of the potency, kinetics and voltage-dependency of a series of uncompetitive NMDA receptor antagonists in vitro with anticonvulsive and motor impairment activity in vivo. Neuropharmacology 34:1239–1258.
Piva A, Caffino L, Mottarlini F, Pintori N, Díaz FC, Fumagalli F, Chiamulera C (2021) Metaplastic effects of ketamine and MK-801 on glutamate receptors expression in rat medial prefrontal cortex and hippocampus. Mol Neurobiol 58:3443–3456.
Price RB, Nock MK, Charney DS, Mathew SJ (2009) Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry 66:522–526.
Qi XR, Zhao J, Liu J, Fang H, Swaab DF, Zhou JN (2015) Abnormal retinoid and TrkB signaling in the prefrontal cortex in mood disorders. Cereb Cortex 25:75–83.
Rademacher DJ, Kovacs B, Shen F, Napier C, Meredith GE (2006) The neural substrates of amphetamine conditioned place preference: Implications for the formation of conditioned stimulus-reward associations. Eur J Neurosci 24:2089–2097.
Robinson TE, Becker JB (1986 ) Enduring changes in brain and behavior produced by chronic amphetamine administration: A review and evaluation of animal models of amphetamine psychosis. Brain Res Rev 11:157-198.
Robinson TE, Berridge KC (1993) The neural basis of drug craving: An incentive-sensitization theory of addiction. Brain Res Rev 18:247–291.
Sanchis-Segura C, Spanagel R (2006) Behavioural assessment of drug reinforcement and addictive features in rodents: an overview. Addict Biol 11:2-38.
Schoepfer KJ, Strong CE, Saland SK, Wright KN, Kabbaj M (2019) Sex- and dose-dependent abuse liability of repeated subanesthetic ketamine in rats. Physiol Behav 203:60–69.
Shen YL, Chang TY, Chang YC, Tien HH, Yang FC, Wang PY, Liao RM (2014) Elevated BDNF mRNA expression in the medial prefrontal cortex after d-amphetamine reinstated conditioned place preference in rats. Neuroscience 263:88-95.
Shen F, Meredith GE, Napier TC (2006) Amphetamine-induced place preference and conditioned motor sensitization requires activation of tyrosine kinase receptors in the hippocampus. J Neurosci 26:11041–11051.
Shuto T, Kuroiwa M, Hamamura M, Yabuuchi K, Shimazoe T, Watanabe S, Nishi A, Yamamoto T (2006) Reversal of methamphetamine-induced behavioral sensitization by repeated administration of a dopamine D1 receptor agonist. Neuropharmacology 50:991-997.
Sial OK, Pariseb EM, Pariseb LF, Gneccoa T, Bolaños-Guzmán CA (2020) Ketamine: The final frontier or another depressing end? Behav Brain Res 383:112508.
Singh JB, Fedgchin M, Daly E, Xi L, Melman C, De Bruecker G, Tadic A, Sienaert P,
Wiegand F, Manji H (2016) Intravenous esketamine in adult treatment-resistant depression: A double-blind, double-randomization, placebo-controlled study. Biol Psychiatry 80:424–431.
Speranza L, di Porzio U, Viggiano Davide, de Donato A, Volpicelli F (2021) Dopamine: The neuromodulator of long-term synaptic plasticity, reward and movement control. Cells 10:735.
Spyraki C, Fibiger HC, Phillips AG (1982) Dopaminergic substrates of amphetamine-induced place preference conditioning. Brain Res 253:185–193.
Steketee JD, Kalivas PW (2011) Drug wanting: Behavioral sensitization and relapse to drug-seeking behavior. Pharmacol Rev 63:348–365.
Strong CE, Schoepfer KJ, Dossat AM, Saland SK, Wright KN, Kabbaj M (2017) Locomotor sensitization to intermittent ketamine administration is associated with nucleus accumbens plasticity in male and female rats. Neuropharmacology 121:195-203.
Sulzer D (2011) How addictive drugs disruptpresynaptic dopamine neurotransmission. Neuron 69:628–649.
Sun Y, Chen G, Zhou K, Zhu Y (2018) A conditioned place preference protocol for measuring incubation of craving in rats. J Vis Exp 141:e58384.
Sun Z, Ma Y, Xie L, Huang J, Duan S, Guo R, Xie Y, Lv J, Lin Z, Ma Shuhua (2019) Behavioral changes and neuronal damage in rhesus monkeys after 10 weeks of ketamine administration involve prefrontal cortex dopamine D2 receptor and dopamine transporter. Neuroscience 415:97-106.
Suzuki T, Katob H, Aokib T, Tsudab M, Naritaa M, Misawa M (2000) Effects of the non-competitive NMDA receptor antagonist ketamine on morphine-induced place preference in mice. Life Sciences 67:383–389.
Traynelis SF , Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R (2010) Glutamate receptor ion channels: Structure, regulation, and function. Pharmacol Rev 62:405-96.
Tritsch NX, Sabatini BL (2012) Dopaminergic modulation of synaptic transmission in cortex and striatum. Neuron 76:33–50.
Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden W, Ritz L, Norquist G, Howland RH, Lebowitz B, McGrath PJ, Shores-Wilson K, Biggs MM, Balasubramani GK, Fava M (2006) Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: Implications for clinical practice. Am J Psychiatry 163:28-40.
Trujillo KA, Heller CY (2020) Ketamine sensitization: Influence of dose, environment, social isolation, and treatment interval. Behav Brain Res 378:112-271.
Trujillo KA, Zamora JJ, Warmoth KP (2008) Increased response to ketamine following treatment at long intervals: Implications for intermittent use. Biol Psychiatry 63:178-183.
Turner EH (2019) Esketamine for treatment-resistant depression: Seven concerns about efficacy and FDA approval. Lancet Psychiatry 6:977–979.
Tzschentke TM (2007) Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol 12:227-462.
Uchihashi Y, Kuribara H, Morita T, Fujita T (1993) The repeated administration of ketamine induces an enhancement of its stimulant action in mice. Jpn J Pharmacol 61:149-151.
Wise RA (2004) Dopamine, learning and motivation. Nat Neuro Rev 5:483–494.
Wise RA (2009) Roles for nigrostriatal – not just mesocoticolimbic – dopamine in reward and addiction. Cell 32:517–524.
Xu DD, Moa ZX, Yung KKL, Yang Y, Leung AWN (2008) Individual and Combined Effects of Methamphetamine and Ketamine on Conditioned Place Preference and NR1 Receptor Phosphorylation in Rats. Neurosignals 15:322–331.
Yamamoto T, Nakayama T, Yamaguchi J, Matsuzawa M, Mishina M, Ikeda K,
Yamamoto H (2016) Role of the NMDA receptor GluN2D subunit in the expression of ketamine-induced behavioral sensitization and region-specific activation of neuronal nitric oxide synthase. Neurosci Lett 610:48-53.
Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, Dong C, Hashimoto K (2015) R-ketamine: A rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry 5: e632.
Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, Charney DS, Manji HK (2006) A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 63:856–864.
Zarate CA Jr, Brutsche NE, Ibrahim L, Franco-Chaves J, Diazgranados N, Cravchik A, Selter J, Marquardt CA, Liberty V, Luckenbaugh DA (2012) Replication of ketamine’s antidepressant efficacy in bipolar depression: A randomized controlled add-on trial. Biol Psychiatry 71:939–946.
Zorumski CF, Izumi Y, Mennerick S (2016) Ketamine: NMDA receptors and beyond. J Neurosci 36:11158–11164.
zh_TW
dc.identifier.doi (DOI) 10.6814/NCCU202200873en_US