簡易檢索 / 詳目顯示

研究生: 董佑萱
Tung, Yu-Hsuen
論文名稱: 探討青少年期捆綁處理對海馬迴功能之長期不良影響
Juvenile immobilization treatment elicits long-term adverse effects on the hippocampal function
指導教授: 呂國棟
Lu, Kwok-Tung
學位類別: 碩士
Master
系所名稱: 生命科學系
Department of Life Science
論文出版年: 2020
畢業學年度: 108
語文別: 英文
論文頁數: 115
中文關鍵詞: 青春期綑綁處理抑制迴避試驗開放空間試驗高架十字迷宮消減學習類焦慮行為海馬迴胞外記錄法長期增益效益鈉鉀氯共轉運蛋白環形利尿劑布美他尼呋塞米
英文關鍵詞: Juvenile, immobilization treatment, inhibitory avoidance, open field testing, elevated plus maze, extinction, anxiety-like behavior, hippocampus, extracellular recording, long-term potentiation, cation-chloride cotransporters, furosemide, bumetanide
DOI URL: http://doi.org/10.6345/NTNU202001278
論文種類: 學術論文
相關次數: 點閱:143下載:13
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 青春期遭遇性騷擾(sexual harassment)、學校霸凌(bullying)、情感虐待(psychological abuse)和家庭暴力(domestic violence)等創傷經驗會導致心理創傷(psychological trauma),已被證實會對心理健康產生長期的不良影響,並增加成年後罹患精神疾病的風險。不幸的是,針對其不良影響的發生,目前僅有少數研究與有限的治療策略。本研究之主要目的為利用青春期綑綁處理動物模式(juvenile immobilization treatment, J_IMO)模擬青少年期創傷經驗,以探究其對海馬迥功能(hippocampal function)的不良影響及其神經機。
    本研究採用C57BL / 6J品系雄性小鼠,在出生後第35天和第36天(postnatal day, PND)時進行每天一次2小時,共兩天的J_IMO處理。於第二次綑綁處理後一週時(PND-42)分別進行行為(behavioral)、電生理(electrophysiological)和生化(biochemical)實驗。行為實驗採用抑制迴避測試(inhibitory avoidance, IA) 來評估海馬迴(hippocampus)的學習記憶功能,IA為一與海馬迥功能高度相關的記憶試驗。我們也利用開放空間測試(open field test, OFT)和高架十字迷宮測試(elevated plus-maze test , EPM)來評估J_IMO的類焦慮行為(anxiety-like behavior)。部分動物則斷頭犧牲取腦,製成海馬迴腦切片用於離體胞外電生理記錄(in vitro extracellular recording),以高頻電刺激(high-frequency stimulation, HFS)誘發的長期增益效應(long-term potentiation, LTP),來評估動物海馬迴的神經可塑性(neuroplasticity)。為了防止行為實驗對生化分析所造成之干擾,我們以相同的方式準備了另外一批動物,於PND-42時斷頭犧牲,取出其海馬迴進行即時定量聚合酶連鎖反應(real-time polymerase chain reaction, qPCR)分析Grin2a (NR2A受體),Slc12a2 (鈉鉀氯共轉運蛋白2,又簡稱NKCC1),Slc12a5 (鈉鉀氯共轉運蛋白5,又簡稱KCC2), Gabra1(GABAA受體)和Fkbp5(co-chaperone with the heat shock protein 90 and steroid complex)等基因的表現,並且利用西方墨點法(Western blot)來確認NKCC1的蛋白質表現量。
    實驗結果顯示,經 J_IMO處理的小鼠,其抑制性迴避學習增強,合併出現削減學習(extinction learning)減弱的現象。而OFT的結果顯示J_IMO組的類焦慮行為有增加的情形,然而在EPM中並未獲得一致的結果。J_IMO組動物的海馬迴HFS-LTP有增強的現象,與對照組相較經J_IMO處理小鼠的輸入和輸出曲線比值(input/output curve ratio, I/O curve)中出現顯著增加的現象。而在配對脈衝促進(pair-pulse facilitation. PPF)的結果並無顯著差異。這些結果暗示海馬迴HFS-LTP的增強應是源自突觸後的機制(post-synaptic mechanism),例如受體表現量增加(up-regulation),或者信號傳遞活性增強。qPCR的結果顯示,經J_IMO處理小鼠海馬迴中Grin2a和Slc12a2的表現量顯著增加,可以證明J_IMO組海馬迴突觸後的機制改變。Fkbp5,Slc12a5和Gabra1的表現量未呈顯著差異。之後,我們以表面灌流(suprafusion)方式投予兩種環形利尿劑(loop diuretics),呋塞米(furosemide)或布美他尼(bumetanide)為NKCC1抑制劑,可將海馬迴HSF-LTP恢復至正常範圍。綜合各項實驗結果,經過J_IMO處理的小鼠表現出異常的行為表現,包括迴避學習之增強、消減學習(extinction learning)能力減弱,和類焦慮行為的增加。J_IMO組的海馬迴HSF-LTP增強海,馬迴功能相關基因如Grin2a,Slc12a2的表現量明顯改變。這些結果共同顯示出,給予J_IMO急性處理也可能對海馬迴功能產生長期影響。
    本研究結論為J_IMO處理成功地模擬了青少年的創傷經歷,並且產生長期的不良影響,這和前人之成年IMO處理的研究發現是一致的。有趣的是,我們發現NKCC1在J_IMO小鼠中的海馬迴表現量發生了變化,這解釋J_IMO治療處理不良反應的可能機制。我們建議NKCC1抑制劑如布美他尼可以作為治療藥物,以減輕青少年創傷事件引起的行為異常。

    Traumatic events during adolescence such as sexual molest, school bullying, emotional abuse, and domestic violence might result in psychological trauma and a long-term deleterious effect on mental health. They would eventually increase the risk of having psychiatry diseases in the adulthood. Unfortunately, the detailed related neural mechanism remains unclear, and limited therapeutic strategies are available to prevent the adverse consequences. Here, a modified juvenile immobilization treatment (J_IMO) animal model was applied to investigate the neural mechanism underlying the adverse effect of juvenile traumatic events on the function of the hippocampus. Briefly, C57BL/6J mice received J_IMO treatment at the postnatal day 35 and 36 (PND). One week later (PND-42), they were subjected to behavioral, electrophysiological, and biochemical experiments. We evaluated the hippocampal function by using an inhibitory avoidance test (IA), a well-known hippocampus-dependent memory task. The anxiety-like behaviors were examined by the open field test (OFT) and elevated plus-maze test (EPM). We also used the in vitro extracellular recording to study the high-frequency stimulation-induced long-term potentiation (HFS-LTP) of the hippocampus. Furthermore, the expression of anxiety-related genes such as Grin2a (NR2A receptor), Gabra1 (GABAA receptor), Slc12a2 (sodium potassium chloride cotransporter-1, NKCC1), Slc12a5 (potassium-chloride transporter member 5, KCC2), and Fkbp5 (co-chaperone with the heat shock protein 90 and steroid complex, FKBP5) of the six weeks old J_IMO mice was determined by using real-time polymerase chain reaction (qPCR) and western blot.
    Results showed that enhanced avoidance learning in the J_IMO treated mice. Also, the J_IMO-treated male mice displayed some degree of interference on the extinction of the IA task. Furthermore, an elevation of anxiety-like behavior was revealed in the test of OFT. The hippocampal HFS-LTP increased in the J_IMO treated group, which could explain the enhanced avoidance learning. A significant difference was found only in the input-output curves (I/O curve), but not in the pair-pulse facilitation (PPF), between control group and J_IMO treated mice. These results implied the enhanced hippocampal HFS-LTP was resulted from a postsynaptic mechanism such as increased receptor expression, or an increase of signal transduction activity. Our qPCR results showed that the expression of Grin2a and Slc12a2 were increased significantly, On the contrary, the expression of Fkbp5, Gabra1 and Slc12a5 were not significantly altered in the J_IMO treated mice. Administration of loop diuretics furosemide or bumetanide, NKCC1 inhibitors, restored the hippocampal HSF-LTP to the normal range.
    Based on our findings, the J_IMO treated mice actually revealed certain abnormal behavioral phenotypes, including a deficit in the extinction of avoidance learning, an increase in anxiety-like behavior, the enhancement of hippocampal HSF-LTP, and a significant upregulation of anxiety-related genes, Grin2a and Slc12a in hippocampus. These results collectively indicated that even an acute J_IMO treatment might elicit a long-term impact on hippocampal functions.
    Conclusively, the J_IMO treatment designed in this study has successfully simulated the juvenile traumatic experience, and has provoked a long-term deleterious impact consistent to the previous finding in adult IMO model studies. It is worth noting that the hippocampal expression of NKCC1 was altered in the J_IMO mice, and that might account for the long-term adverse effect of J_IMO treatment. Accordingly, the NKCC1 inhibitor bumetanide might become a therapeutic agent to relieve the behavioral abnormalities induced by juvenile traumatic events.

    INTRODUCTION 1 Adverse juvenile experiences and the associated long-term impact 1 Anxiety disorder and the dysfunction of the HPA axis 2 Brain structures correlated to the stress-induced behavioral abnormalities 5 Glucocorticoid effects on the function of the hippocampus 6 The related genes for stress-induced behavioral abnormalities related genes 8 The impact of stress on the excitatory neurotransmission 10 Stress-induced abnormality of cation-coupled chloride transporters (CCCs) 11 Research aim and significance 14 MATERIALS AND METHODS 15 Animals 15 Juvenile immobilization treatment (J_IMO) 15 Inhibitory avoidance test (IA) 16 Open field test (OFT) 17 Elevated plus-maze (EPM) 18 Brain slice extracellular recording 19 Administration of bumetanide and furosemide 20 Real-time polymerase chain reaction 21 Western blot 24 Statistics 25 RESULTS 27 Experiment-1: Examine the juvenile immobilization treatment effects on the inhibitory avoidance learning. 27 Experiment-2: Evaluation of the juvenile immobilization treatment induced anxiety-like behavior in the male mice by open field test and elevated plus-maze test. 30 Experiment-3: Determine the hippocampal long-term potentiation of the IMO animals using brain slice extracellular recording. 32 Experiment-4: Determine the input-output curve and paired-pulse facilitation of the brain slices of J_IMO treated animals. 34 Experiment-5: Examine the expression of HPA axis-related and neurotransmitter receptor genes of the hippocampal on the juvenile IMO animals by using real-time polymerase chain reaction. 36 Experiment-6: Analyze the hippocampal NKCC1 expression of J_IMO mice by Western blotting. 38 Experiment-7: The enhanced hippocampal HFS-LTP of the J_IMO animals were reversed by suprafusion of NKCC1 antagonist in a dose-dependent manner. 39 DISCUSSION 43 Summary of the results 43 The juvenile IMO treatment reveals an opposite effect on the acquisition and the extinction of inhibitory avoidance memory. 45 The high-frequency stimulation-induced hippocampal long-term potentiation is enhanced in juvenile IMO treated mice. 51 The involvement of NKCC1 in the J_IMO treatment effects on hippocampal function. 54 Conclusion 61 REFERENCES 63

    Adamcio, B., Sperling, S., Hagemeyer, N., Walkinshaw, G. & Ehrenreich, H. (2010) Hypoxia inducible factor stabilization leads to lasting improvement of hippocampal memory in healthy mice. Behav Brain Res, 208, 80-84.
    Afifi, T.O., Asmundson, G.J., Taylor, S. & Jang, K.L. (2010) The role of genes and environment on trauma exposure and posttraumatic stress disorder symptoms: a review of twin studies. Clin Psychol Rev, 30, 101-112.
    Ahmed, M.M., Lee, H., Clark, Z., Miranpuri, G.S., Nacht, C., Patel, K., Liu, L., Joslin, J., Kintner, D. & Resnick, D.K. (2014) Pathogenesis of spinal cord injury induced edema and neuropathic pain: expression of multiple isoforms of wnk1. Ann Neurosci, 21, 97-103.
    Albrecht, A., Muller, I., Ardi, Z., Caliskan, G., Gruber, D., Ivens, S., Segal, M., Behr, J., Heinemann, U., Stork, O. & Richter-Levin, G. (2017) Neurobiological consequences of juvenile stress: A GABAergic perspective on risk and resilience. Neurosci Biobehav Rev, 74, 21-43.
    Alves, N.D., Correia, J.S., Patricio, P., Mateus-Pinheiro, A., Machado-Santos, A.R., Loureiro-Campos, E., Morais, M., Bessa, J.M., Sousa, N. & Pinto, L. (2017) Adult hippocampal neuroplasticity triggers susceptibility to recurrent depression. Transl Psychiatry, 7, e1058.
    American Psychiatric Association (2013) Diagnostic and statistical manual of mental disorders (DSM-5®). American Psychiatric Pub, Arlington, VA, US,.
    Appel, K., Schwahn, C., Mahler, J., Schulz, A., Spitzer, C., Fenske, K., Stender, J., Barnow, S., John, U., Teumer, A., Biffar, R., Nauck, M., Volzke, H., Freyberger, H.J. & Grabe, H.J. (2011) Moderation of adult depression by a polymorphism in the FKBP5 gene and childhood physical abuse in the general population. Neuropsychopharmacology, 36, 1982-1991.
    Arion, D. & Lewis, D.A. (2011) Altered expression of regulators of the cortical chloride transporters NKCC1 and KCC2 in schizophrenia. Arch Gen Psychiatry, 68, 21-31.
    Arnett, J.J. (2000) Emerging adulthood. A theory of development from the late teens through the twenties. Am Psychol, 55, 469-480.
    Arp, J.M., Ter Horst, J.P., Loi, M., den Blaauwen, J., Bangert, E., Fernandez, G., Joels, M., Oitzl, M.S. & Krugers, H.J. (2016) Blocking glucocorticoid receptors at adolescent age prevents enhanced freezing between repeated cue-exposures after conditioned fear in adult mice raised under chronic early life stress. Neurobiol Learn Mem, 133, 30-38.
    Artola, A. & Singer, W. (1987) Long-term potentiation and NMDA receptors in rat visual cortex. Nature, 330, 649-652.
    Atucha, E. & Roozendaal, B. (2015) The inhibitory avoidance discrimination task to investigate accuracy of memory. Front Behav Neurosci, 9, 60.
    Autry, A.E. & Monteggia, L.M. (2012) Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev, 64, 238-258.
    Avoli, M. & Krnjevic, K. (2016) The Long and Winding Road to Gamma-Amino-Butyric Acid as Neurotransmitter. Can J Neurol Sci, 43, 219-226.
    Bartsch, T. (2012) The clinical neurobiology of the hippocampus : an integrative view. Oxford University Press, Oxford, United Kingdom.
    Battaglia, F.P., Benchenane, K., Sirota, A., Pennartz, C.M. & Wiener, S.I. (2011) The hippocampus: hub of brain network communication for memory. Trends Cogn Sci, 15, 310-318.
    Baudry, M., Zhu, G., Liu, Y., Wang, Y., Briz, V. & Bi, X. (2015) Multiple cellular cascades participate in long-term potentiation and in hippocampus-dependent learning. Brain Res, 1621, 73-81.
    Ben-Ari, Y. (2017) NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric Disorders. Trends Neurosci, 40, 536-554.
    Bergstrom, H.C., McDonald, C.G. & Johnson, L.R. (2011) Pavlovian fear conditioning activates a common pattern of neurons in the lateral amygdala of individual brains. PLoS One, 6, e15698.
    Bhuiyan, M.I.H., Song, S., Yuan, H., Begum, G., Kofler, J., Kahle, K.T., Yang, S.S., Lin, S.H., Alper, S.L., Subramanya, A.R. & Sun, D. (2017) WNK-Cab39-NKCC1 signaling increases the susceptibility to ischemic brain damage in hypertensive rats. J Cereb Blood Flow Metab, 37, 2780-2794.
    Bialer, M., Johannessen, S.I., Levy, R.H., Perucca, E., Tomson, T. & White, H.S. (2017) Progress report on new antiepileptic drugs: A summary of the Thirteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIII). Epilepsia, 58, 181-221.
    Blaesse, P., Airaksinen, M.S., Rivera, C. & Kaila, K. (2009) Cation-chloride cotransporters and neuronal function. Neuron, 61, 820-838.
    Bonapersona, V., Damsteegt, R., Adams, M.L., van Weert, L., Meijer, O.C., Joels, M. & Sarabdjitsingh, R.A. (2019) Sex-Dependent Modulation of Acute Stress Reactivity After Early Life Stress in Mice: Relevance of Mineralocorticoid Receptor Expression. Front Behav Neurosci, 13, 181.
    Bremner, J.D. (2003) Long-term effects of childhood abuse on brain and neurobiology. Child Adolesc Psychiatr Clin N Am, 12, 271-292.
    Brown, G.W., Ban, M., Craig, T.K., Harris, T.O., Herbert, J. & Uher, R. (2013) Serotonin transporter length polymorphism, childhood maltreatment, and chronic depression: a specific gene-environment interaction. Depress Anxiety, 30, 5-13.
    Bruining, H., Passtoors, L., Goriounova, N., Jansen, F., Hakvoort, B., de Jonge, M. & Poil, S.S. (2015) Paradoxical Benzodiazepine Response: A Rationale for Bumetanide in Neurodevelopmental Disorders? Pediatrics, 136, e539-543.
    Brydges, N.M., Wood, E.R., Holmes, M.C. & Hall, J. (2014) Prepubertal stress and hippocampal function: sex-specific effects. Hippocampus, 24, 684-692.
    Buchmann, A.F., Holz, N., Boecker, R., Blomeyer, D., Rietschel, M., Witt, S.H., Schmidt, M.H., Esser, G., Banaschewski, T., Brandeis, D., Zimmermann, U.S. & Laucht, M. (2014) Moderating role of FKBP5 genotype in the impact of childhood adversity on cortisol stress response during adulthood. Eur Neuropsychopharmacol, 24, 837-845.
    Carmignoto, G. & Vicini, S. (1992) Activity-dependent decrease in NMDA receptor responses during development of the visual cortex. Science, 258, 1007-1011.
    Casey, B.J., Thomas, K.M., Davidson, M.C., Kunz, K. & Franzen, P.L. (2002) Dissociating striatal and hippocampal function developmentally with a stimulus-response compatibility task. J Neurosci, 22, 8647-8652.
    Caspi, A., Sugden, K., Moffitt, T.E., Taylor, A., Craig, I.W., Harrington, H., McClay, J., Mill, J., Martin, J., Braithwaite, A. & Poulton, R. (2003) Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science, 301, 386-389.
    Cazakoff, B.N., Johnson, K.J. & Howland, J.G. (2010) Converging effects of acute stress on spatial and recognition memory in rodents: a review of recent behavioural and pharmacological findings. Prog Neuropsychopharmacol Biol Psychiatry, 34, 733-741.
    Chattarji, S., Tomar, A., Suvrathan, A., Ghosh, S. & Rahman, M.M. (2015) Neighborhood matters: divergent patterns of stress-induced plasticity across the brain. Nat Neurosci, 18, 1364-1375.
    Chen, Y. & Baram, T.Z. (2016) Toward Understanding How Early-Life Stress Reprograms Cognitive and Emotional Brain Networks. Neuropsychopharmacology, 41, 197-206.
    Chen, Z.Y., Jing, D., Bath, K.G., Ieraci, A., Khan, T., Siao, C.J., Herrera, D.G., Toth, M., Yang, C., McEwen, B.S., Hempstead, B.L. & Lee, F.S. (2006) Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science, 314, 140-143.
    Chiba, S., Numakawa, T., Ninomiya, M., Richards, M.C., Wakabayashi, C. & Kunugi, H. (2012) Chronic restraint stress causes anxiety- and depression-like behaviors, downregulates glucocorticoid receptor expression, and attenuates glutamate release induced by brain-derived neurotrophic factor in the prefrontal cortex. Prog Neuropsychopharmacol Biol Psychiatry, 39, 112-119.
    Collingridge, G.L., Kehl, S.J. & McLennan, H. (1983) Excitatory amino acids in synaptic transmission in the Schaffer collateral-commissural pathway of the rat hippocampus. J Physiol, 334, 33-46.
    Cooke, S.F. & Bliss, T.V. (2006) Plasticity in the human central nervous system. Brain, 129, 1659-1673.
    Corcoran, C., Walker, E., Huot, R., Mittal, V., Tessner, K., Kestler, L. & Malaspina, D. (2003) The stress cascade and schizophrenia: etiology and onset. Schizophr Bull, 29, 671-692.
    Cordella, A., Krashia, P., Nobili, A., Pignataro, A., La Barbera, L., Viscomi, M.T., Valzania, A., Keller, F., Ammassari-Teule, M., Mercuri, N.B., Berretta, N. & D'Amelio, M. (2018) Dopamine loss alters the hippocampus-nucleus accumbens synaptic transmission in the Tg2576 mouse model of Alzheimer's disease. Neurobiol Dis, 116, 142-154.
    Cormier, R.J., Greenwood, A.C. & Connor, J.A. (2001) Bidirectional synaptic plasticity correlated with the magnitude of dendritic calcium transients above a threshold. J Neurophysiol, 85, 399-406.
    Cusmano, D.M. & Mong, J.A. (2014) In utero exposure to valproic acid changes sleep in juvenile rats: a model for sleep disturbances in autism. Sleep, 37, 1489-1499.
    Dalle, E. & Mabandla, M.V. (2018) Early Life Stress, Depression And Parkinson's Disease: A New Approach. Mol Brain, 11, 18.
    Danese, A. & Baldwin, J.R. (2017) Hidden Wounds? Inflammatory Links Between Childhood Trauma and Psychopathology. Annu Rev Psychol, 68, 517-544.
    Danese, A., Moffitt, T.E., Harrington, H., Milne, B.J., Polanczyk, G., Pariante, C.M., Poulton, R. & Caspi, A. (2009) Adverse childhood experiences and adult risk factors for age-related disease: depression, inflammation, and clustering of metabolic risk markers. Arch Pediatr Adolesc Med, 163, 1135-1143.
    de Los Heros, P., Alessi, D.R., Gourlay, R., Campbell, D.G., Deak, M., Macartney, T.J., Kahle, K.T. & Zhang, J. (2014) The WNK-regulated SPAK/OSR1 kinases directly phosphorylate and inhibit the K+-Cl- co-transporters. Biochem J, 458, 559-573.
    de Quervain, D., Schwabe, L. & Roozendaal, B. (2017) Stress, glucocorticoids and memory: implications for treating fear-related disorders. Nat Rev Neurosci, 18, 7-19.
    de Quervain, D.J., Roozendaal, B. & McGaugh, J.L. (1998) Stress and glucocorticoids impair retrieval of long-term spatial memory. Nature, 394, 787-790.
    Dean, B., Keriakous, D., Scarr, E. & Thomas, E.A. (2007) Gene expression profiling in Brodmann's area 46 from subjects with schizophrenia. Aust N Z J Psychiatry, 41, 308-320.
    Delawary, M., Tezuka, T., Kiyama, Y., Yokoyama, K., Inoue, T., Hattori, S., Hashimoto, R., Umemori, H., Manabe, T., Yamamoto, T. & Nakazawa, T. (2010) NMDAR2B tyrosine phosphorylation regulates anxiety-like behavior and CRF expression in the amygdala. Mol Brain, 3, 37.
    Dinkel, K., Ogle, W.O. & Sapolsky, R.M. (2002) Glucocorticoids and central nervous system inflammation. J Neurovirol, 8, 513-528.
    Dohring, J., Schmuck, A. & Bartsch, T. (2014) Stress-related factors in the emergence of transient global amnesia with hippocampal lesions. Front Behav Neurosci, 8, 287.
    Donato, F., Rompani, S.B. & Caroni, P. (2013) Parvalbumin-expressing basket-cell network plasticity induced by experience regulates adult learning. Nature, 504, 272-276.
    Dumas, T.C. (2005) Developmental regulation of cognitive abilities: modified composition of a molecular switch turns on associative learning. Prog Neurobiol, 76, 189-211.
    Dzhala, V.I., Kuchibhotla, K.V., Glykys, J.C., Kahle, K.T., Swiercz, W.B., Feng, G., Kuner, T., Augustine, G.J., Bacskai, B.J. & Staley, K.J. (2010) Progressive NKCC1-dependent neuronal chloride accumulation during neonatal seizures. J Neurosci, 30, 11745-11761.
    Ebrahimi-Ghiri, M., Rostampour, M., Jamshidi-Mehr, M., Nasehi, M. & Zarrindast, M.R. (2018) Role of CA1 GABAA and GABAB receptors on learning deficit induced by D-AP5 in passive avoidance step-through task. Brain Res, 1678, 164-173.
    Fan, J.M., Chen, X.Q., Jin, H. & Du, J.Z. (2009) Gestational hypoxia alone or combined with restraint sensitizes the hypothalamic-pituitary-adrenal axis and induces anxiety-like behavior in adult male rat offspring. Neuroscience, 159, 1363-1373.
    Feit, P.W. (1981) Bumetanide--the way to its chemical structure. J Clin Pharmacol, 21, 531-536.
    Finke, C., Bruehl, H., Duzel, E., Heekeren, H.R. & Ploner, C.J. (2013) Neural correlates of short-term memory reorganization in humans with hippocampal damage. J Neurosci, 33, 11061-11069.
    Fu, P., Tang, R., Yu, Z., Huang, S., Xie, M., Luo, X. & Wang, W. (2015) Bumetanide-induced NKCC1 inhibition attenuates oxygen-glucose deprivation-induced decrease in proliferative activity and cell cycle progression arrest in cultured OPCs via p-38 MAPKs. Brain Res, 1613, 110-119.
    Gao, C., Gill, M.B., Tronson, N.C., Guedea, A.L., Guzman, Y.F., Huh, K.H., Corcoran, K.A., Swanson, G.T. & Radulovic, J. (2010) Hippocampal NMDA receptor subunits differentially regulate fear memory formation and neuronal signal propagation. Hippocampus, 20, 1072-1082.
    Garin-Aguilar, M.E., Diaz-Cintra, S., Quirarte, G.L., Aguilar-Vazquez, A., Medina, A.C. & Prado-Alcala, R.A. (2012) Extinction procedure induces pruning of dendritic spines in CA1 hippocampal field depending on strength of training in rats. Front Behav Neurosci, 6, 12.
    Garza, K. & Jovanovic, T. (2017) Impact of Gender on Child and Adolescent PTSD. Curr Psychiatry Rep, 19, 87.
    Gold, P.E. (1986) The use of avoidance training in studies of modulation of memory storage. Behav Neural Biol, 46, 87-98.
    Gonzalez, M.C., Kramar, C.P., Tomaiuolo, M., Katche, C., Weisstaub, N., Cammarota, M. & Medina, J.H. (2014) Medial prefrontal cortex dopamine controls the persistent storage of aversive memories. Front Behav Neurosci, 8, 408.
    Grandgeorge, M., Lemonnier, E., Degrez, C. & Jallot, N. (2014) The effect of bumetanide treatment on the sensory behaviours of a young girl with Asperger syndrome. BMJ Case Rep, 2014.
    Gruart, A., Munoz, M.D. & Delgado-Garcia, J.M. (2006) Involvement of the CA3-CA1 synapse in the acquisition of associative learning in behaving mice. J Neurosci, 26, 1077-1087.
    Gulyaeva, N.V. (2016) Studies on stress-induced modulation of long term potentiation in rodent hippocampus: what can we learn about pathogenesis of depression? Translat. Brain Rhythm, 1.
    Gutschmidt, K.U., Stenkamp, K., Buchheim, K., Heinemann, U. & Meierkord, H. (1999) Anticonvulsant actions of furosemide in vitro. Neuroscience, 91, 1471-1481.
    Haas, M. (1994) The Na-K-Cl cotransporters. Am J Physiol, 267, C869-885.
    Hammoud, M.Z., Foa, E.B. & Milad, M.R. (2020) Oestradiol, threat conditioning and extinction, post-traumatic stress disorder, and prolonged exposure therapy: A common link. J Neuroendocrinol, 32, e12800.
    Hartley, C.A. & Phelps, E.A. (2012) Extinction Learning. Seel N.M. (eds) Encyclopedia of the Sciences of Learning. Springer, Boston, MA. https://doi.org/10.1007/978-1-4419-1428-6_306.
    Hayashi, Y., Yokota, A., Harada, H. & Huang, G. (2019) Hypoxia/pseudohypoxia-mediated activation of hypoxia-inducible factor-1alpha in cancer. Cancer Sci, 110, 1510-1517.
    Heim, C. & Nemeroff, C.B. (2001) The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biol Psychiatry, 49, 1023-1039.
    Holmes, G.L., Tian, C., Hernan, A.E., Flynn, S., Camp, D. & Barry, J. (2015) Alterations in sociability and functional brain connectivity caused by early-life seizures are prevented by bumetanide. Neurobiol Dis, 77, 204-219.
    Howland, J.G. & Wang, Y.T. (2008) Synaptic plasticity in learning and memory: stress effects in the hippocampus. Prog Brain Res, 169, 145-158.
    Hsiao, Y.M., Tsai, T.C., Lin, Y.T., Chen, C.C., Huang, C.C. & Hsu, K.S. (2016) Early life stress dampens stress responsiveness in adolescence: Evaluation of neuroendocrine reactivity and coping behavior. Psychoneuroendocrinology, 67, 86-99.
    Huang, E.J. & Reichardt, L.F. (2001) Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci, 24, 677-736.
    Huang, H., Bhuiyan, M.I.H., Jiang, T., Song, S., Shankar, S., Taheri, T., Li, E., Schreppel, P., Hintersteininger, M., Yang, S.S., Lin, S.H., Molyneaux, B.J., Zhang, Z., Erker, T. & Sun, D. (2019a) A Novel Na(+)-K(+)-Cl(-) Cotransporter 1 Inhibitor STS66* Reduces Brain Damage in Mice After Ischemic Stroke. Stroke, 50, 1021-1025.
    Huang, H., Song, S., Banerjee, S., Jiang, T., Zhang, J., Kahle, K.T., Sun, D. & Zhang, Z. (2019b) The WNK-SPAK/OSR1 Kinases and the Cation-Chloride Cotransporters as Therapeutic Targets for Neurological Diseases. Aging Dis, 10, 626-636.
    Hueston, C.M., Cryan, J.F. & Nolan, Y.M. (2017) Stress and adolescent hippocampal neurogenesis: diet and exercise as cognitive modulators. Transl Psychiatry, 7, e1081.
    Hui, H., Rao, W., Zhang, L., Xie, Z., Peng, C., Su, N., Wang, K., Wang, L., Luo, P., Hao, Y.L., Zhang, S. & Fei, Z. (2016) Inhibition of Na(+)-K(+)-2Cl(-) Cotransporter-1 attenuates traumatic brain injury-induced neuronal apoptosis via regulation of Erk signaling. Neurochem Int, 94, 23-31.
    Hyde, T.M., Lipska, B.K., Ali, T., Mathew, S.V., Law, A.J., Metitiri, O.E., Straub, R.E., Ye, T., Colantuoni, C., Herman, M.M., Bigelow, L.B., Weinberger, D.R. & Kleinman, J.E. (2011) Expression of GABA signaling molecules KCC2, NKCC1, and GAD1 in cortical development and schizophrenia. J Neurosci, 31, 11088-11095.
    Ibla, J.C., Khoury, J., Kong, T., Robinson, A. & Colgan, S.P. (2006) Transcriptional repression of Na-K-2Cl cotransporter NKCC1 by hypoxia-inducible factor-1. Am J Physiol Cell Physiol, 291, C282-289.
    Inda, M.C., Delgado-Garcia, J.M. & Carrion, A.M. (2005) Acquisition, consolidation, reconsolidation, and extinction of eyelid conditioning responses require de novo protein synthesis. J Neurosci, 25, 2070-2080.
    Jahng, J.W. (2011) An animal model of eating disorders associated with stressful experience in early life. Horm Behav, 59, 213-220.
    Jeanneteau, F., Garabedian, M.J. & Chao, M.V. (2008) Activation of Trk neurotrophin receptors by glucocorticoids provides a neuroprotective effect. Proc Natl Acad Sci U S A, 105, 4862-4867.
    Jeanneteau, F.D., Lambert, W.M., Ismaili, N., Bath, K.G., Lee, F.S., Garabedian, M.J. & Chao, M.V. (2012) BDNF and glucocorticoids regulate corticotrophin-releasing hormone (CRH) homeostasis in the hypothalamus. Proc Natl Acad Sci U S A, 109, 1305-1310.
    Jiang, S., Li, X., Jin, W., Duan, X., Bo, L., Wu, J., Zhang, R., Wang, Y., Kang, R. & Huang, L. (2018) Ketamine-induced neurotoxicity blocked by N-Methyl-d-aspartate is mediated through activation of PKC/ERK pathway in developing hippocampal neurons. Neurosci Lett, 673, 122-131.
    Jin, S.X. & Feig, L.A. (2010) Long-term potentiation in the CA1 hippocampus induced by NR2A subunit-containing NMDA glutamate receptors is mediated by Ras-GRF2/Erk map kinase signaling. PLoS One, 5, e11732.
    Kahle, K.T. & Staley, K.J. (2008) The bumetanide-sensitive Na-K-2Cl cotransporter NKCC1 as a potential target of a novel mechanism-based treatment strategy for neonatal seizures. Neurosurg Focus, 25, E22.
    Keding, T.J. & Herringa, R.J. (2015) Abnormal structure of fear circuitry in pediatric post-traumatic stress disorder. Neuropsychopharmacology, 40, 537-545.
    Kendler, K.S., Thornton, L.M. & Gardner, C.O. (2000) Stressful life events and previous episodes in the etiology of major depression in women: an evaluation of the "kindling" hypothesis. Am J Psychiatry, 157, 1243-1251.
    Keshavarzy, F., Bonnet, C., Behzadi, G. & Cespuglio, R. (2015) Expression patterns of c-Fos early gene and phosphorylated ERK in the rat brain following 1-h immobilization stress: concomitant changes induced in association with stress-related sleep rebound. Brain Struct Funct, 220, 1793-1804.
    Kim, M.J., Dunah, A.W., Wang, Y.T. & Sheng, M. (2005) Differential roles of NR2A- and NR2B-containing NMDA receptors in Ras-ERK signaling and AMPA receptor trafficking. Neuron, 46, 745-760.
    Klengel, T., Mehta, D., Anacker, C., Rex-Haffner, M., Pruessner, J.C., Pariante, C.M., Pace, T.W., Mercer, K.B., Mayberg, H.S., Bradley, B., Nemeroff, C.B., Holsboer, F., Heim, C.M., Ressler, K.J., Rein, T. & Binder, E.B. (2013) Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nat Neurosci, 16, 33-41.
    Ko, M.C., Lee, M.C., Amstislavskaya, T.G., Tikhonova, M.A., Yang, Y.L. & Lu, K.T. (2014) Inhibition of NKCC1 attenuated hippocampal LTP formation and inhibitory avoidance in rat. PLoS One, 9, e106692.
    Ko, M.C., Lee, M.C., Tang, T.H., Amstislavskaya, T.G., Tikhonova, M.A., Yang, Y.L. & Lu, K.T. (2018) Bumetanide blocks the acquisition of conditioned fear in adult rats. Br J Pharmacol, 175, 1580-1589.
    Kolata, S.M., Nakao, K., Jeevakumar, V., Farmer-Alroth, E.L., Fujita, Y., Bartley, A.F., Jiang, S.Z., Rompala, G.R., Sorge, R.E., Jimenez, D.V., Martinowich, K., Mateo, Y., Hashimoto, K., Dobrunz, L.E. & Nakazawa, K. (2018) Neuropsychiatric Phenotypes Produced by GABA Reduction in Mouse Cortex and Hippocampus. Neuropsychopharmacology, 43, 1445-1456.
    Koss, K.J. & Gunnar, M.R. (2018) Annual Research Review: Early adversity, the hypothalamic-pituitary-adrenocortical axis, and child psychopathology. J Child Psychol Psychiatry, 59, 327-346.
    Kosten, T.A., Kim, J.J. & Lee, H.J. (2012) Early life manipulations alter learning and memory in rats. Neurosci Biobehav Rev, 36, 1985-2006.
    Kotak, V.C., Mirallave, A., Mowery, T.M. & Sanes, D.H. (2017) GABAergic inhibition gates excitatory LTP in perirhinal cortex. Hippocampus, 27, 1217-1223.
    Krania, P., Dimou, E., Bantouna, M., Kouvaros, S., Tsiamaki, E., Papatheodoropoulos, C., Sarantis, K. & Angelatou, F. (2018) Adenosine A2A receptors are required for glutamate mGluR5- and dopamine D1 receptor-evoked ERK1/2 phosphorylation in rat hippocampus: involvement of NMDA receptor. J Neurochem, 145, 217-231.
    Krystal, A.D., Sutherland, J. & Hochman, D.W. (2012) Loop diuretics have anxiolytic effects in rat models of conditioned anxiety. PLoS One, 7, e35417.
    Kumar, A. & Foster, T.C. (2019) Alteration in NMDA Receptor Mediated Glutamatergic Neurotransmission in the Hippocampus During Senescence. Neurochem Res, 44, 38-48.
    Lang, U.E. & Borgwardt, S. (2013) Molecular mechanisms of depression: perspectives on new treatment strategies. Cell Physiol Biochem, 31, 761-777.
    Lee, H.H., Deeb, T.Z., Walker, J.A., Davies, P.A. & Moss, S.J. (2011) NMDA receptor activity downregulates KCC2 resulting in depolarizing GABAA receptor-mediated currents. Nat Neurosci, 14, 736-743.
    Lee, H.J., Berger, S.Y., Stiedl, O., Spiess, J. & Kim, J.J. (2001) Post-training injections of catecholaminergic drugs do not modulate fear conditioning in rats and mice. Neurosci Lett, 303, 123-126.
    Lee, R.S. (2016) Epigenetics and neuroendocrinology : clinical focus on psychiatry. Springer Berlin Heidelberg, New York, NY.
    Lemonnier, E., Degrez, C., Phelep, M., Tyzio, R., Josse, F., Grandgeorge, M., Hadjikhani, N. & Ben-Ari, Y. (2012) A randomised controlled trial of bumetanide in the treatment of autism in children. Transl Psychiatry, 2, e202.
    Lemonnier, E., Lazartigues, A. & Ben-Ari, Y. (2016) Treating Schizophrenia With the Diuretic Bumetanide: A Case Report. Clin Neuropharmacol, 39, 115-117.
    Li, J.T., Xie, X.M., Yu, J.Y., Sun, Y.X., Liao, X.M., Wang, X.X., Su, Y.A., Liu, Y.J., Schmidt, M.V., Wang, X.D. & Si, T.M. (2017) Suppressed Calbindin Levels in Hippocampal Excitatory Neurons Mediate Stress-Induced Memory Loss. Cell Rep, 21, 891-900.
    Liang, B. & Huang, J.H. (2017) Elevated NKCC1 transporter expression facilitates early post-traumatic brain injury seizures. Neural Regen Res, 12, 401-402.
    Lisman, J. (2017) Glutamatergic synapses are structurally and biochemically complex because of multiple plasticity processes: long-term potentiation, long-term depression, short-term potentiation and scaling. Philos Trans R Soc Lond B Biol Sci, 372.
    Lisman, J., Yasuda, R. & Raghavachari, S. (2012) Mechanisms of CaMKII action in long-term potentiation. Nat Rev Neurosci, 13, 169-182.
    Liu, L., Wong, T.P., Pozza, M.F., Lingenhoehl, K., Wang, Y., Sheng, M., Auberson, Y.P. & Wang, Y.T. (2004) Role of NMDA receptor subtypes in governing the direction of hippocampal synaptic plasticity. Science, 304, 1021-1024.
    Liu, X.H., Zhu, R.T., Hao, B., Shi, Y.W., Wang, X.G., Xue, L. & Zhao, H. (2019) Norepinephrine Induces PTSD-Like Memory Impairments via Regulation of the beta-Adrenoceptor-cAMP/PKA and CaMK II/PKC Systems in the Basolateral Amygdala. Front Behav Neurosci, 13, 43.
    Lodge, D., Watkins, J.C., Bortolotto, Z.A., Jane, D.E. & Volianskis, A. (2019) The 1980s: D-AP5, LTP and a Decade of NMDA Receptor Discoveries. Neurochem Res, 44, 516-530.
    Loi, M., Mossink, J.C., Meerhoff, G.F., Den Blaauwen, J.L., Lucassen, P.J. & Joels, M. (2017) Effects of early-life stress on cognitive function and hippocampal structure in female rodents. Neuroscience, 342, 101-119.
    Lu, B., Nagappan, G. & Lu, Y. (2014) BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol, 220, 223-250.
    Lu, K.T., Huang, T.C., Tsai, Y.H. & Yang, Y.L. (2017) Transient receptor potential vanilloid type 4 channels mediate Na-K-Cl-co-transporter-induced brain edema after traumatic brain injury. J Neurochem, 140, 718-727.
    Lu, K.T., Huang, T.C., Wang, J.Y., You, Y.S., Chou, J.L., Chan, M.W., Wo, P.Y., Amstislavskaya, T.G., Tikhonova, M.A. & Yang, Y.L. (2015) NKCC1 mediates traumatic brain injury-induced hippocampal neurogenesis through CREB phosphorylation and HIF-1alpha expression. Pflugers Arch, 467, 1651-1661.
    Lucassen, P.J., Pruessner, J., Sousa, N., Almeida, O.F., Van Dam, A.M., Rajkowska, G., Swaab, D.F. & Czeh, B. (2014) Neuropathology of stress. Acta Neuropathol, 127, 109-135.
    MacKenzie, G. & Maguire, J. (2015) Chronic stress shifts the GABA reversal potential in the hippocampus and increases seizure susceptibility. Epilepsy Res, 109, 13-27.
    Malenka, R.C. & Nicoll, R.A. (1999) Long-term potentiation--a decade of progress? Science, 285, 1870-1874.
    Maren, S. (2001) Neurobiology of Pavlovian fear conditioning. Annu Rev Neurosci, 24, 897-931.
    Martin-Aragon Baudel, M.A., Poole, A.V. & Darlison, M.G. (2017) Chloride co-transporters as possible therapeutic targets for stroke. J Neurochem, 140, 195-209.
    Martinez-Canabal, A., Lopez-Oropeza, G., Gaona-Gamboa, A., Ballesteros-Zebadua, P., de la Cruz, O.G., Moreno-Jimenez, S. & Sotres-Bayon, F. (2019) Hippocampal neurogenesis regulates recovery of defensive responses by recruiting threat- and extinction-signalling brain networks. Sci Rep, 9, 2939.
    Maruyama, J., Kobayashi, Y., Umeda, T., Vandewalle, A., Takeda, K., Ichijo, H. & Naguro, I. (2016) Osmotic stress induces the phosphorylation of WNK4 Ser575 via the p38MAPK-MK pathway. Sci Rep, 6, 18710.
    Matosin, N., Halldorsdottir, T. & Binder, E.B. (2018) Understanding the Molecular Mechanisms Underpinning Gene by Environment Interactions in Psychiatric Disorders: The FKBP5 Model. Biol Psychiatry, 83, 821-830.
    Matsuzaki, M., Honkura, N., Ellis-Davies, G.C. & Kasai, H. (2004) Structural basis of long-term potentiation in single dendritic spines. Nature, 429, 761-766.
    McGaugh, J.L. & Roozendaal, B. (2009) Drug enhancement of memory consolidation: historical perspective and neurobiological implications. Psychopharmacology (Berl), 202, 3-14.
    McLaughlin, K.A., Busso, D.S., Duys, A., Green, J.G., Alves, S., Way, M. & Sheridan, M.A. (2014) Amygdala response to negative stimuli predicts PTSD symptom onset following a terrorist attack. Depress Anxiety, 31, 834-842.
    McLaughlin, N.C., Strong, D., Abrantes, A., Garnaat, S., Cerny, A., O'Connell, C., Fadok, R., Spofford, C., Rasmussen, S.A., Milad, M.R. & Greenberg, B.D. (2015) Extinction retention and fear renewal in a lifetime obsessive-compulsive disorder sample. Behav Brain Res, 280, 72-77.
    Medina, J.H. & Viola, H. (2018) ERK1/2: A Key Cellular Component for the Formation, Retrieval, Reconsolidation and Persistence of Memory. Front Mol Neurosci, 11, 361.
    Mejia-Gervacio, S., Murray, K. & Lledo, P.M. (2011) NKCC1 controls GABAergic signaling and neuroblast migration in the postnatal forebrain. Neural Dev, 6, 4.
    Mello-Carpes, P.B., da Silva de Vargas, L., Gayer, M.C., Roehrs, R. & Izquierdo, I. (2016) Hippocampal noradrenergic activation is necessary for object recognition memory consolidation and can promote BDNF increase and memory persistence. Neurobiol Learn Mem, 127, 84-92.
    Miller, G.E., Chen, E. & Parker, K.J. (2011) Psychological stress in childhood and susceptibility to the chronic diseases of aging: moving toward a model of behavioral and biological mechanisms. Psychol Bull, 137, 959-997.
    Miranda, M., Morici, J.F., Zanoni, M.B. & Bekinschtein, P. (2019) Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front Cell Neurosci, 13, 363.
    Monteggia, L.M., Luikart, B., Barrot, M., Theobold, D., Malkovska, I., Nef, S., Parada, L.F. & Nestler, E.J. (2007) Brain-derived neurotrophic factor conditional knockouts show gender differences in depression-related behaviors. Biol Psychiatry, 61, 187-197.
    Monyer, H., Burnashev, N., Laurie, D.J., Sakmann, B. & Seeburg, P.H. (1994) Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron, 12, 529-540.
    Morgan, C.A., 3rd, Doran, A., Steffian, G., Hazlett, G. & Southwick, S.M. (2006) Stress-induced deficits in working memory and visuo-constructive abilities in Special Operations soldiers. Biol Psychiatry, 60, 722-729.
    Morris, R.G. (1989) Synaptic plasticity and learning: selective impairment of learning rats and blockade of long-term potentiation in vivo by the N-methyl-D-aspartate receptor antagonist AP5. J Neurosci, 9, 3040-3057.
    Mount, D.B., Delpire, E., Gamba, G., Hall, A.E., Poch, E., Hoover, R.S. & Hebert, S.C. (1998) The electroneutral cation-chloride cotransporters. J Exp Biol, 201, 2091-2102.
    Myhrer, T. (2003) Neurotransmitter systems involved in learning and memory in the rat: a meta-analysis based on studies of four behavioral tasks. Brain Res Brain Res Rev, 41, 268-287.
    Navarro-Frances, C.I. & Arenas, M.C. (2014) Influence of trait anxiety on the effects of acute stress on learning and retention of the passive avoidance task in male and female mice. Behav Processes, 105, 6-14.
    Nelson, C.A., 3rd, Zeanah, C.H., Fox, N.A., Marshall, P.J., Smyke, A.T. & Guthrie, D. (2007) Cognitive recovery in socially deprived young children: the Bucharest Early Intervention Project. Science, 318, 1937-1940.
    Nishi, M., Horii-Hayashi, N. & Sasagawa, T. (2014) Effects of early life adverse experiences on the brain: implications from maternal separation models in rodents. Front Neurosci, 8, 166.
    Olsen, R.K., Moses, S.N., Riggs, L. & Ryan, J.D. (2012) The hippocampus supports multiple cognitive processes through relational binding and comparison. Front Hum Neurosci, 6, 146.
    Olsen, R.W. & Sieghart, W. (2009) GABA A receptors: subtypes provide diversity of function and pharmacology. Neuropharmacology, 56, 141-148.
    Olver, J.S., Pinney, M., Maruff, P. & Norman, T.R. (2015) Impairments of spatial working memory and attention following acute psychosocial stress. Stress Health, 31, 115-123.
    Orellana, J.A., Moraga-Amaro, R., Diaz-Galarce, R., Rojas, S., Maturana, C.J., Stehberg, J. & Saez, J.C. (2015) Restraint stress increases hemichannel activity in hippocampal glial cells and neurons. Front Cell Neurosci, 9, 102.
    Osborne, D.M., Pearson-Leary, J. & McNay, E.C. (2015) The neuroenergetics of stress hormones in the hippocampus and implications for memory. Front Neurosci, 9, 164.
    Panet, R., Eliash, M., Pick, M. & Atlan, H. (2002) Na(+)/K(+)/Cl(-) cotransporter activates mitogen-activated protein kinase in fibroblasts and lymphocytes. J Cell Physiol, 190, 227-237.
    Paoletti, P., Bellone, C. & Zhou, Q. (2013) NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci, 14, 383-400.
    Patel, B., Mortensen, M. & Smart, T.G. (2014) Stoichiometry of delta subunit containing GABA(A) receptors. Br J Pharmacol, 171, 985-994.
    Pavlowsky, A., Wallace, E., Fenton, A.A. & Alarcon, J.M. (2017) Persistent modifications of hippocampal synaptic function during remote spatial memory. Neurobiol Learn Mem, 138, 182-197.
    Penza, K.M., Heim, C. & Nemeroff, C.B. (2003) Neurobiological effects of childhood abuse: implications for the pathophysiology of depression and anxiety. Arch Womens Ment Health, 6, 15-22.
    Peterlin, B.L., Katsnelson, M.J. & Calhoun, A.H. (2009) The associations between migraine, unipolar psychiatric comorbidities, and stress-related disorders and the role of estrogen. Curr Pain Headache Rep, 13, 404-412.
    Petrovic, M.M., Viana da Silva, S., Clement, J.P., Vyklicky, L., Mulle, C., Gonzalez-Gonzalez, I.M. & Henley, J.M. (2017) Metabotropic action of postsynaptic kainate receptors triggers hippocampal long-term potentiation. Nat Neurosci, 20, 529-539.
    Pfeffer, C.K., Stein, V., Keating, D.J., Maier, H., Rinke, I., Rudhard, Y., Hentschke, M., Rune, G.M., Jentsch, T.J. & Hubner, C.A. (2009) NKCC1-dependent GABAergic excitation drives synaptic network maturation during early hippocampal development. J Neurosci, 29, 3419-3430.
    Piala, A.T., Moon, T.M., Akella, R., He, H., Cobb, M.H. & Goldsmith, E.J. (2014) Chloride sensing by WNK1 involves inhibition of autophosphorylation. Sci Signal, 7, ra41.
    Plotsky, P.M. & Meaney, M.J. (1993) Early, postnatal experience alters hypothalamic corticotropin-releasing factor (CRF) mRNA, median eminence CRF content and stress-induced release in adult rats. Brain Res Mol Brain Res, 18, 195-200.
    Popoli, M., Yan, Z., McEwen, B.S. & Sanacora, G. (2011) The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci, 13, 22-37.
    Puskarjov, M., Kahle, K.T., Ruusuvuori, E. & Kaila, K. (2014) Pharmacotherapeutic targeting of cation-chloride cotransporters in neonatal seizures. Epilepsia, 55, 806-818.
    Raabe, F.J. & Spengler, D. (2013) Epigenetic Risk Factors in PTSD and Depression. Front Psychiatry, 4, 80.
    Rahmanzadeh, R., Mehrabi, S., Barati, M., Ahmadi, M., Golab, F., Kazmi, S., Joghataei, M.T., Seifi, M. & Gholipourmalekabadi, M. (2018) Effect of Co-administration of Bumetanide and Phenobarbital on Seizure Attacks in Temporal Lobe Epilepsy. Basic Clin Neurosci, 9, 408-416.
    Reincke, S.A. & Hanganu-Opatz, I.L. (2017) Early-life stress impairs recognition memory and perturbs the functional maturation of prefrontal-hippocampal-perirhinal networks. Sci Rep, 7, 42042.
    Ren, W., Liu, X., Cheng, L., Wang, G., Liu, X., Peng, L. & Wang, Y. (2019) Embryonic Ketamine Produces a Downregulation of Prefrontal Cortex NMDA Receptors and Anxiety-Like Behavior in Adult Offspring. Neuroscience, 415, 18-30.
    Revest, J.M., Le Roux, A., Roullot-Lacarriere, V., Kaouane, N., Vallee, M., Kasanetz, F., Rouge-Pont, F., Tronche, F., Desmedt, A. & Piazza, P.V. (2014) BDNF-TrkB signaling through Erk1/2 MAPK phosphorylation mediates the enhancement of fear memory induced by glucocorticoids. Mol Psychiatry, 19, 1001-1009.
    Rivera, C., Voipio, J. & Kaila, K. (2005) Two developmental switches in GABAergic signalling: the K+-Cl- cotransporter KCC2 and carbonic anhydrase CAVII. J Physiol, 562, 27-36.
    Robel, S., Buckingham, S.C., Boni, J.L., Campbell, S.L., Danbolt, N.C., Riedemann, T., Sutor, B. & Sontheimer, H. (2015) Reactive astrogliosis causes the development of spontaneous seizures. J Neurosci, 35, 3330-3345.
    Romeo, R.D. (2017) The impact of stress on the structure of the adolescent brain: Implications for adolescent mental health. Brain Res, 1654, 185-191.
    Roozendaal, B., Hahn, E.L., Nathan, S.V., de Quervain, D.J. & McGaugh, J.L. (2004) Glucocorticoid effects on memory retrieval require concurrent noradrenergic activity in the hippocampus and basolateral amygdala. J Neurosci, 24, 8161-8169.
    Roozendaal, B. & McGaugh, J.L. (2011) Memory modulation. Behav Neurosci, 125, 797-824.
    Ross, D.A., Arbuckle, M.R., Travis, M.J., Dwyer, J.B., van Schalkwyk, G.I. & Ressler, K.J. (2017) An Integrated Neuroscience Perspective on Formulation and Treatment Planning for Posttraumatic Stress Disorder: An Educational Review. JAMA Psychiatry, 74, 407-415.
    Roth, T.L., Zoladz, P.R., Sweatt, J.D. & Diamond, D.M. (2011) Epigenetic modification of hippocampal Bdnf DNA in adult rats in an animal model of post-traumatic stress disorder. J Psychiatr Res, 45, 919-926.
    Ryan, T.J., Roy, D.S., Pignatelli, M., Arons, A. & Tonegawa, S. (2015) Memory. Engram cells retain memory under retrograde amnesia. Science, 348, 1007-1013.
    Safren, S.A., Gershuny, B.S., Marzol, P., Otto, M.W. & Pollack, M.H. (2002) History of childhood abuse in panic disorder, social phobia, and generalized anxiety disorder. J Nerv Ment Dis, 190, 453-456.
    Sanz-Garcia, A., Knafo, S., Pereda-Perez, I., Esteban, J.A., Venero, C. & Armario, A. (2016) Administration of the TrkB receptor agonist 7,8-dihydroxyflavone prevents traumatic stress-induced spatial memory deficits and changes in synaptic plasticity. Hippocampus, 26, 1179-1188.
    Sawamura, T., Klengel, T., Armario, A., Jovanovic, T., Norrholm, S.D., Ressler, K.J. & Andero, R. (2016) Dexamethasone Treatment Leads to Enhanced Fear Extinction and Dynamic Fkbp5 Regulation in Amygdala. Neuropsychopharmacology, 41, 832-846.
    Sawyer, S.M., Afifi, R.A., Bearinger, L.H., Blakemore, S.J., Dick, B., Ezeh, A.C. & Patton, G.C. (2012) Adolescence: a foundation for future health. Lancet, 379, 1630-1640.
    Scheuer, S., Ising, M., Uhr, M., Otto, Y., von Klitzing, K. & Klein, A.M. (2016) FKBP5 polymorphisms moderate the influence of adverse life events on the risk of anxiety and depressive disorders in preschool children. J Psychiatr Res, 72, 30-36.
    Sheng, M., Cummings, J., Roldan, L.A., Jan, Y.N. & Jan, L.Y. (1994) Changing subunit composition of heteromeric NMDA receptors during development of rat cortex. Nature, 368, 144-147.
    Sivakumaran, S. & Maguire, J. (2016) Bumetanide reduces seizure progression and the development of pharmacoresistant status epilepticus. Epilepsia, 57, 222-232.
    Smith, C. & Squire, L.R. (2005) Declarative memory, awareness, and transitive inference. J Neurosci, 25, 10138-10146.
    Steiger, F., Nees, F., Wicking, M., Lang, S. & Flor, H. (2015) Behavioral and central correlates of contextual fear learning and contextual modulation of cued fear in posttraumatic stress disorder. Int J Psychophysiol, 98, 584-593.
    Stein, D.J., Scott, K., Haro Abad, J.M., Aguilar-Gaxiola, S., Alonso, J., Angermeyer, M., Demytteneare, K., de Girolamo, G., Iwata, N., Posada-Villa, J., Kovess, V., Lara, C., Ormel, J., Kessler, R.C. & Von Korff, M. (2010) Early childhood adversity and later hypertension: data from the World Mental Health Survey. Ann Clin Psychiatry, 22, 19-28.
    Stein, M.B., Walker, J.R., Anderson, G., Hazen, A.L., Ross, C.A., Eldridge, G. & Forde, D.R. (1996) Childhood physical and sexual abuse in patients with anxiety disorders and in a community sample. Am J Psychiatry, 153, 275-277.
    Strohle, A., Gensichen, J. & Domschke, K. (2018) The Diagnosis and Treatment of Anxiety Disorders. Dtsch Arztebl Int, 155, 611-620.
    Taiwan health and welfare report- Ministry of Health and Welfare (2019) Number of Abused Child and Youth. Number of Abused Child and Youth. Retrieved from
    https://dep.mohw.gov.tw/DOS/cp-2985-14085-113.html.
    Taliaz, D., Loya, A., Gersner, R., Haramati, S., Chen, A. & Zangen, A. (2011) Resilience to chronic stress is mediated by hippocampal brain-derived neurotrophic factor. J Neurosci, 31, 4475-4483.
    Tapia-Arancibia, L., Rage, F., Givalois, L. & Arancibia, S. (2004) Physiology of BDNF: focus on hypothalamic function. Front Neuroendocrinol, 25, 77-107.
    Teicher, M.H. & Samson, J.A. (2016) Annual Research Review: Enduring neurobiological effects of childhood abuse and neglect. J Child Psychol Psychiatry, 57, 241-266.
    Thoenen, H. (1991) The changing scene of neurotrophic factors. Trends Neurosci, 14, 165-170.
    Timmermans, W., Xiong, H., Hoogenraad, C.C. & Krugers, H.J. (2013) Stress and excitatory synapses: from health to disease. Neuroscience, 248, 626-636.
    Tiwari, A. & Gonzalez, A. (2018) Biological alterations affecting risk of adult psychopathology following childhood trauma: A review of sex differences. Clin Psychol Rev, 66, 69-79.
    Tsukahara, T., Masuhara, M., Iwai, H., Sonomura, T. & Sato, T. (2015) Repeated stress-induced expression pattern alterations of the hippocampal chloride transporters KCC2 and NKCC1 associated with behavioral abnormalities in female mice. Biochem Biophys Res Commun, 465, 145-151.
    Tu, B.X., Wang, L.F., Zhong, X.L., Hu, Z.L., Cao, W.Y., Cui, Y.H., Li, S.J., Zou, G.J., Liu, Y., Zhou, S.F., Zhang, W.J., Su, J.Z., Yan, X.X., Li, F. & Li, C.Q. (2019) Acute restraint stress alters food-foraging behavior in rats: Taking the easier Way while suffered. Brain Res Bull, 149, 184-193.
    Turner, B.D., Kashima, D.T., Manz, K.M., Grueter, C.A. & Grueter, B.A. (2018) Synaptic Plasticity in the Nucleus Accumbens: Lessons Learned from Experience. ACS Chem Neurosci, 9, 2114-2126.
    Tzanoulinou, S., Garcia-Mompo, C., Castillo-Gomez, E., Veenit, V., Nacher, J. & Sandi, C. (2014) Long-term behavioral programming induced by peripuberty stress in rats is accompanied by GABAergic-related alterations in the Amygdala. PLoS One, 9, e94666.
    US Department of Health and Human Services, A.f.C.a.F. (2017) Administration on Children, Youth and Families, Children’s Bureau (2017) Administration for Children and Families, Administration on Children, Youth and Families, Children’s Bureau., Child Maltreatment 2015. Retrieved from http://www.acf.hhs.gov/programs/cb/research-data-technology/statistics-research/child-maltreatment.
    van Stegeren, A.H. (2008) The role of the noradrenergic system in emotional memory. Acta Psychol (Amst), 127, 532-541.
    Villain, H., Benkahoul, A., Drougard, A., Lafragette, M., Muzotte, E., Pech, S., Bui, E., Brunet, A., Birmes, P. & Roullet, P. (2016) Effects of Propranolol, a beta-noradrenergic Antagonist, on Memory Consolidation and Reconsolidation in Mice. Front Behav Neurosci, 10, 49.
    Vinay, L. & Jean-Xavier, C. (2008) Plasticity of spinal cord locomotor networks and contribution of cation-chloride cotransporters. Brain Res Rev, 57, 103-110.
    Vogel, S. & Schwabe, L. (2016) Learning and memory under stress: implications for the classroom. NPJ Sci Learn, 1, 16011.
    Wang, D., Cui, Z., Zeng, Q., Kuang, H., Wang, L.P., Tsien, J.Z. & Cao, X. (2009) Genetic enhancement of memory and long-term potentiation but not CA1 long-term depression in NR2B transgenic rats. PLoS One, 4, e7486.
    Wang, Y., Liu, Q., Xie, J., Feng, R., Ma, F., Wang, F., Shen, S. & Wen, T. (2019) Dcf1 Affects Memory and Anxiety by Regulating NMDA and AMPA Receptors. Neurochem Res, 44, 2499-2505.
    Warrington, E.K. & Baddeley, A.D. (1974) Amnesia and memory for visual location. Neuropsychologia, 12, 257-263.
    Watanabe, M. & Fukuda, A. (2015) Development and regulation of chloride homeostasis in the central nervous system. Front Cell Neurosci, 9, 371.
    Weems, C.F., Klabunde, M., Russell, J.D., Reiss, A.L. & Carrion, V.G. (2015) Post-traumatic stress and age variation in amygdala volumes among youth exposed to trauma. Soc Cogn Affect Neurosci, 10, 1661-1667.
    Wei, L., Hao, J., Lacher, R.K., Abbott, T., Chung, L., Colangelo, C.M. & Kaffman, A. (2015) Early-Life Stress Perturbs Key Cellular Programs in the Developing Mouse Hippocampus. Dev Neurosci, 37, 476-488.
    Whitehead, G., Jo, J., Hogg, E.L., Piers, T., Kim, D.H., Seaton, G., Seok, H., Bru-Mercier, G., Son, G.H., Regan, P., Hildebrandt, L., Waite, E., Kim, B.C., Kerrigan, T.L., Kim, K., Whitcomb, D.J., Collingridge, G.L., Lightman, S.L. & Cho, K. (2013) Acute stress causes rapid synaptic insertion of Ca2+ -permeable AMPA receptors to facilitate long-term potentiation in the hippocampus. Brain, 136, 3753-3765.
    Williams, A.J. & Umemori, H. (2014) The best-laid plans go oft awry: synaptogenic growth factor signaling in neuropsychiatric disease. Front Synaptic Neurosci, 6, 4.
    Wochnik, G.M., Ruegg, J., Abel, G.A., Schmidt, U., Holsboer, F. & Rein, T. (2005) FK506-binding proteins 51 and 52 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor in mammalian cells. J Biol Chem, 280, 4609-4616.
    Yang, F.C. & Liang, K.C. (2014) Interactions of the dorsal hippocampus, medial prefrontal cortex and nucleus accumbens in formation of fear memory: difference in inhibitory avoidance learning and contextual fear conditioning. Neurobiol Learn Mem, 112, 186-194.
    Yang, Q., Zhu, G., Liu, D., Ju, J.G., Liao, Z.H., Xiao, Y.X., Zhang, Y., Chao, N., Wang, J., Li, W., Luo, J.H. & Li, S.T. (2017) Extrasynaptic NMDA receptor dependent long-term potentiation of hippocampal CA1 pyramidal neurons. Sci Rep, 7, 3045.
    Yang, X.L., Zeng, M.L., Shao, L., Jiang, G.T., Cheng, J.J., Chen, T.X., Han, S., Yin, J., Liu, W.H., He, X.H. & Peng, B.W. (2019) NFAT5 and HIF-1alpha Coordinate to Regulate NKCC1 Expression in Hippocampal Neurons After Hypoxia-Ischemia. Front Cell Dev Biol, 7, 339.
    Yehezkel., B.-A. (2002.09) Nature Reviews Neuroscience. 3, 728-739
    Yonelinas, A.P. (2013) The hippocampus supports high-resolution binding in the service of perception, working memory and long-term memory. Behav Brain Res, 254, 34-44.
    Zovkic, I.B., Meadows, J.P., Kaas, G.A. & Sweatt, J.D. (2013) Interindividual Variability in Stress Susceptibility: A Role for Epigenetic Mechanisms in PTSD. Front Psychiatry, 4, 60.

    下載圖示
    QR CODE