| 研究生: |
林芷瑩 Lin, Chih-Ying |
|---|---|
| 論文名稱: |
探討丙戊酸在社交挫敗壓力之小鼠模式中對腸道菌相和多巴胺轉運子的影響 Exploration of the effects of valproic acid on gut microbiome and dopamine transporter in social defeat stress mouse model |
| 指導教授: |
張惠華
Chang, Hui Hua |
| 學位類別: |
碩士 Master |
| 系所名稱: |
醫學院 - 臨床藥學與藥物科技研究所 Institute of Clinical Pharmacy and Pharmaceutical sciences |
| 論文出版年: | 2019 |
| 畢業學年度: | 107 |
| 語文別: | 英文 |
| 論文頁數: | 311 |
| 中文關鍵詞: | 雙極症 、社交挫敗壓力 、丙戊酸 、腸道菌相 、多巴胺轉運子 |
| 外文關鍵詞: | bipolar disorder, social defeat, valproate, gut microbiota, dopamine transporter |
| 相關次數: | 點閱:83 下載:0 |
| 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 |
雙極症為一種嚴重且功能失調的情緒疾患。流行病學指出,使用丙戊酸治療的雙極症患者會增加其代謝異常的風險。雖然雙極症的病理生理學尚未被完全了解,據近年研究發現腸道菌相和多巴胺轉運子可能會影響大腦功能和周邊代謝。然而,目前研究針對微生物-腸-腦軸線和多巴胺轉運子,與丙戊酸的治療成效之關聯性,尚未定論。因此,本研究假設腸道菌相與多巴胺轉運子為丙戊酸治療中,作為與腦部治療成效及周邊代謝系統的共同調節因子。本研究使用7週齡雄性C57BL / 6N小鼠,為了誘導小鼠出現雙極症類似行為,施予社交挫敗壓力10天,而後於老鼠腹腔注射為期三週的350mg / kg 丙戊酸或10ml / kg生理食鹽水。本研究透過強迫游泳測試及社會偏好測試,檢測小鼠是否具憂鬱及社交異常行為。收集血液用於三酸甘油脂檢測和葡萄糖耐受性試驗。在丙戊酸治療前後分別收集新鮮糞便檢體,以Illumina次世代定序分析菌相16S rRNA的V3-V4區域,進一步透過線性判別分析(LEfSe),QIIME v1.80和mothur v.1.39.5整合生物資訊。我們利用西方墨點法測試小鼠紋狀體中多巴胺轉運子蛋白表現。統計顯著設於p <0.05。本研究結果顯示社交挫敗壓力誘導小鼠出現雙極症類似行為,包含社會迴避和類憂鬱行為。使用丙戊酸造成對照小鼠和社交挫敗小鼠其腸道微生物分布顯著不同。此外,Escherichia-Shigella屬在社交挫敗小鼠中含量顯著降低,而Enterobacter屬則顯著增加。藉由抗生素操弄腸道菌相可逆轉由壓力所誘導的憂鬱及社交異常行為,以及減緩在社交挫敗小鼠中由丙戊酸引起的高三酸甘油酯。除此之外,本研究亦發現社交挫敗小鼠腦部紋狀體的多巴胺轉運子表現量增加。使用丙戊酸處理後,社交挫敗小鼠的三酸甘油脂升高伴隨著多巴胺轉運子表現量下降,且三酸甘油脂與多巴胺轉運子的表現量具有顯著的負相關關係。根據以上結果,本研究推論腸道菌相和多巴胺轉運子在壓力狀態下扮演重要的角色,並提供雙極症疾患一個有潛力的新治療標的。
Bipolar disorder (BD) is a severe and dysfunctional mood disorder. Epidemiologic evidences have indicated increased risk of the metabolic abnormalities in BD patients treated with valproic acid (VPA). Although the pathophysiology of BD is not fully understood, recent reports suggested that gut microbiota and dopamine transporter (DAT) activity could affect brain function and metabolic status. Moreover, literatures about the potential role of the microbiota-gut-brain axis and DAT in the therapeutic effect of VPA is unknown. Thus, we hypothesized that gut microbiota and DAT could be common mediators to regulate therapeutic effects in the brain and influence metabolism in the peripheral tissue after VPA treatment. We used seven-week-old male C57BL/6N mice. To mimic bipolar disorder-like behaviours, the mice were under social defeat stress (SD) for 10 days, and then they were injected 350mg/kg VPA or 10ml/kg saline intraperitoneally for 3 weeks. There were two domains to confirm BD-like behaviour, including depression-like (forced swim test) and social behaviour (social preference test). The blood was collected for triglyceride detection and glucose tolerance test. Fresh fecal samples were individually collected at baseline and after VPA treatment. The microbial profiles of the samples were analyzed the V3-V4 regions of the bacterial 16S rRNA gene with next generation sequencing by Illumina. Bioinformatics analyses were performed using linear discriminant analysis effect size (LEfSe), QIIME v1.80 and mothur v.1.39.5. We applied western blotting to verify DAT protein expression on mice striatum. The significance was determined at p<0.05. In the current study, the results showed that social defeat stress developed BD-like behaviors mice, which exhibited social avoidance and depression-like behavior. The composition of gut microbiota was significantly different between saline-treated and VPA-treated control mice and the results were consistent in SD mice. We found that the genus Escherichia-Shigella were significantly less abundant in VPA-treated SD mice, together with the significant increase of Enterobacter when compared with vehicle-treated SD mice. Manipulation of composition of gut microbiota via antibiotic intervention reversed stress-induced depression-like and impaired social behavior, as well as alleviated triglyceride level caused by VPA in SD mice. In addition, SD mice were observed increased DAT expression in striatum. After treated with VPA, SD mice had elevated triglyceride levels along with decreased DAT expression. There was a significant negative correlation between triglyceride level and DAT expression. Taken together, the current study provided evidance of important roles of gut microbiota and DAT under stressful condition, and provided possible new therapeutic strategy in BD.
Abercrombie, E. D., Keefe, K. A., DiFrischia, D. S., & Zigmond, M. J. (1989). Differential effect of stress on in vivo dopamine release in striatum, nucleus accumbens, and medial frontal cortex. J Neurochem, 52(5), 1655-1658. doi:10.1111/j.1471-4159.1989.tb09224.x
Ahn, T., Bae, C. S., & Yun, C.-H. (2016). Acute stress-induced changes in hormone and lipid levels in mouse plasma (Vol. 61).
Ait-Belgnaoui, A., Durand, H., Cartier, C., Chaumaz, G., Eutamene, H., Ferrier, L., . . . Theodorou, V. (2012). Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology, 37(11), 1885-1895. doi:10.1016/j.psyneuen.2012.03.024
Anand, A., Barkay, G., Dzemidzic, M., Albrecht, D., Karne, H., Zheng, Q. H., . . . Yoder, K. K. (2011). Striatal dopamine transporter availability in unmedicated bipolar disorder. Bipolar Disord, 13(4), 406-413. doi:10.1111/j.1399-5618.2011.00936.x
Angst, J., Gamma, A., Benazzi, F., Ajdacic, V., Eich, D., & Rossler, W. (2003). Toward a re-definition of subthreshold bipolarity: epidemiology and proposed criteria for bipolar-II, minor bipolar disorders and hypomania. J Affect Disord, 73(1-2), 133-146.
Armario, A., Montero, J. L., & Balasch, J. (1986). Sensitivity of corticosterone and some metabolic variables to graded levels of low intensity stresses in adult male rats. Physiology & Behavior, 37(4), 559-561. doi:https://doi.org/10.1016/0031-9384(86)90285-4
Atmaca, M. (2009). Valproate and neuroprotective effects for bipolar disorder. Int Rev Psychiatry, 21(4), 410-413. doi:10.1080/09540260902962206
Au - Tatem, K. S., Au - Quinn, J. L., Au - Phadke, A., Au - Yu, Q., Au - Gordish-Dressman, H., & Au - Nagaraju, K. (2014). Behavioral and Locomotor Measurements Using an Open Field Activity Monitoring System for Skeletal Muscle Diseases. JoVE(91), e51785. doi:doi:10.3791/51785
Bachmanov, A. A., Reed, D. R., Beauchamp, G. K., & Tordoff, M. G. (2002). Food intake, water intake, and drinking spout side preference of 28 mouse strains. Behav Genet, 32(6), 435-443.
Bai, X., Hong, W., Cai, P., Chen, Y., Xu, C., Cao, D., . . . Jin, J. (2017). Valproate induced hepatic steatosis by enhanced fatty acid uptake and triglyceride synthesis. Toxicol Appl Pharmacol, 324, 12-25. doi:10.1016/j.taap.2017.03.022
Bai, Y. M., Su, T. P., Li, C. T., Tsai, S. J., Chen, M. H., Tu, P. C., & Chiou, W. F. (2015). Comparison of pro-inflammatory cytokines among patients with bipolar disorder and unipolar depression and normal controls. Bipolar Disord, 17(3), 269-277. doi:10.1111/bdi.12259
Bedarf, J. R., Hildebrand, F., Goeser, F., Bork, P., & Wullner, U. (2019). [The gut microbiome in Parkinson's disease]. Nervenarzt, 90(2), 160-166. doi:10.1007/s00115-018-0601-6
Belcastro, V., D'Egidio, C., Striano, P., & Verrotti, A. (2013). Metabolic and endocrine effects of valproic acid chronic treatment. Epilepsy Res, 107(1-2), 1-8. doi:10.1016/j.eplepsyres.2013.08.016
Berger, M., Gray, J. A., & Roth, B. L. (2009). The Expanded Biology of Serotonin. Annual Review of Medicine, 60(1), 355-366. doi:10.1146/annurev.med.60.042307.110802
Blaxter, M., Mann, J., Chapman, T., Thomas, F., Whitton, C., Floyd, R., & Abebe, E. (2005). Defining operational taxonomic units using DNA barcode data. Philos Trans R Soc Lond B Biol Sci, 360(1462), 1935-1943. doi:10.1098/rstb.2005.1725
Burokas, A., Arboleya, S., Moloney, R. D., Peterson, V. L., Murphy, K., Clarke, G., . . . Cryan, J. F. (2017). Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol Psychiatry, 82(7), 472-487. doi:10.1016/j.biopsych.2016.12.031
Can, A., Dao, D. T., Arad, M., Terrillion, C. E., Piantadosi, S. C., & Gould, T. D. (2012). The mouse forced swim test. J Vis Exp(59), e3638-e3638. doi:10.3791/3638
Caporaso, J. G., Kuczynski, J., Stombaugh, J., Bittinger, K., Bushman, F. D., Costello, E. K., . . . Knight, R. (2010). QIIME allows analysis of high-throughput community sequencing data. Nat Methods, 7(5), 335-336. doi:10.1038/nmeth.f.303
Carpenter, A. C., Saborido, T. P., & Stanwood, G. D. (2012). Development of Hyperactivity and Anxiety Responses in Dopamine Transporter-Deficient Mice. Developmental Neuroscience, 34(2-3), 250-257. doi:10.1159/000336824
Chambers, E. S., Viardot, A., Psichas, A., Morrison, D. J., Murphy, K. G., Zac-Varghese, S. E. K., . . . Frost, G. (2015). Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut, 64(11), 1744. doi:10.1136/gutjnl-2014-307913
Chang, C. M., Wu, C. S., Huang, Y. W., Chau, Y. L., & Tsai, H. J. (2016). Utilization of Psychopharmacological Treatment Among Patients With Newly Diagnosed Bipolar Disorder From 2001 to 2010. J Clin Psychopharmacol, 36(1), 32-44. doi:10.1097/jcp.0000000000000440
Chang, H. H., Yang, Y. K., Gean, P. W., Huang, H. C., Chen, P. S., & Lu, R. B. (2010). The role of valproate in metabolic disturbances in bipolar disorder patients. J Affect Disord, 124(3), 319-323. doi:10.1016/j.jad.2009.12.011
Chang, P. V., Hao, L., Offermanns, S., & Medzhitov, R. (2014). The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A, 111(6), 2247-2252. doi:10.1073/pnas.1322269111
Chang, T. T., Yeh, T. L., Chiu, N. T., Chen, P. S., Huang, H. Y., Yang, Y. K., . . . Lu, R. B. (2010). Higher striatal dopamine transporters in euthymic patients with bipolar disorder: a SPECT study with [99mTc] TRODAT-1. Bipolar Disord, 12(1), 102-106. doi:10.1111/j.1399-5618.2009.00771.x
Chang, T. T., Yeh, T. L., Chiu, N. T., Chen, P. S., Huang, H. Y., Yang, Y. K., . . . Lu, R. B. (2010). Higher striatal dopamine transporters in euthymic patients with bipolar disorder: a SPECT study with [Tc] TRODAT-1. Bipolar Disord, 12(1), 102-106. doi:10.1111/j.1399-5618.2009.00771.x
Chao, A. (1987). Estimating the population size for capture-recapture data with unequal catchability. Biometrics, 783-791.
Clarke, G., Grenham, S., Scully, P., Fitzgerald, P., Moloney, R. D., Shanahan, F., . . . Cryan, J. F. (2013). The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry, 18(6), 666-673. doi:10.1038/mp.2012.77
Clarke, K. R. (1993). Non-parametric multivariate analyses of changes in community structure. Australian Journal of Ecology, 18(1), 117-143. doi:10.1111/j.1442-9993.1993.tb00438.x
Cousins, D. A., Butts, K., & Young, A. H. (2009). The role of dopamine in bipolar disorder. Bipolar Disord, 11(8), 787-806. doi:10.1111/j.1399-5618.2009.00760.x
Crane, J. D., Palanivel, R., Mottillo, E. P., Bujak, A. L., Wang, H., Ford, R. J., . . . Steinberg, G. R. (2014). Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nature Medicine, 21, 166. doi:10.1038/nm.3766
https://www.nature.com/articles/nm.3766#supplementary-information
Cryan, J. F., & Dinan, T. G. (2012). Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci, 13(10), 701-712. doi:10.1038/nrn3346
Dinan, T. G., & Cryan, J. F. (2013). Melancholic microbes: a link between gut microbiota and depression? Neurogastroenterol Motil, 25(9), 713-719. doi:10.1111/nmo.12198
Egger, J., & Brett, E. M. (1981). Effects of sodium valproate in 100 children with special reference to weight. British medical journal (Clinical research ed.), 283(6291), 577-581. doi:10.1136/bmj.283.6291.577
Fiorucci, S., & Distrutti, E. (2015). Bile Acid-Activated Receptors, Intestinal Microbiota, and the Treatment of Metabolic Disorders. Trends Mol Med, 21(11), 702-714. doi:10.1016/j.molmed.2015.09.001
Foster, J. A., & McVey Neufeld, K. A. (2013). Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci, 36(5), 305-312. doi:10.1016/j.tins.2013.01.005
Foster, M. T., Solomon, M. B., Huhman, K. L., & Bartness, T. J. (2006). Social defeat increases food intake, body mass, and adiposity in Syrian hamsters. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 290(5), R1284-R1293. doi:10.1152/ajpregu.00437.2005
Fu, J., Bonder, M. J., Cenit, M. C., Tigchelaar, E. F., Maatman, A., Dekens, J. A., . . . Zhernakova, A. (2015). The Gut Microbiome Contributes to a Substantial Proportion of the Variation in Blood Lipids. Circ Res, 117(9), 817-824. doi:10.1161/circresaha.115.306807
Furusawa, Y., Obata, Y., Fukuda, S., Endo, T. A., Nakato, G., Takahashi, D., . . . Ohno, H. (2013). Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature, 504(7480), 446-450. doi:10.1038/nature12721
Gacias, M., Gaspari, S., Santos, P.-M. G., Tamburini, S., Andrade, M., Zhang, F., . . . Casaccia, P. (2016). Microbiota-driven transcriptional changes in prefrontal cortex override genetic differences in social behavior. eLife, 5, e13442. doi:10.7554/eLife.13442
Ge, X., Ding, C., Zhao, W., Xu, L., Tian, H., Gong, J., . . . Li, N. (2017). Antibiotics-induced depletion of mice microbiota induces changes in host serotonin biosynthesis and intestinal motility. Journal of translational medicine, 15(1), 13-13. doi:10.1186/s12967-016-1105-4
Gohir, W., Whelan, F. J., Surette, M. G., Moore, C., Schertzer, J. D., & Sloboda, D. M. (2015). Pregnancy-related changes in the maternal gut microbiota are dependent upon the mother's periconceptional diet. Gut Microbes, 6(5), 310-320. doi:10.1080/19490976.2015.1086056
Golden, S. A., Covington, H. E., 3rd, Berton, O., & Russo, S. J. (2011). A standardized protocol for repeated social defeat stress in mice. Nat Protoc, 6(8), 1183-1191. doi:10.1038/nprot.2011.361
Gorelova, N., Mulholland, P. J., Chandler, L. J., & Seamans, J. K. (2012). The glutamatergic component of the mesocortical pathway emanating from different subregions of the ventral midbrain. Cereb Cortex, 22(2), 327-336. doi:10.1093/cercor/bhr107
Goto, T., & Toyoda, A. (2015). A Mouse Model of Subchronic and Mild Social Defeat Stress for Understanding Stress-induced Behavioral and Physiological Deficits. J Vis Exp(105). doi:10.3791/52973
Gower, J. C. (1966). Some distance properties of latent root and vector methods used in multivariate analysis. Biometrika, 53(3-4), 325-338. doi:10.1093/biomet/53.3-4.325
Grande, I., Berk, M., Birmaher, B., & Vieta, E. (2016). Bipolar disorder. The Lancet, 387(10027), 1561-1572. doi:10.1016/S0140-6736(15)00241-X
Green, A. L., Zhan, L., Eid, A., Zarbl, H., Guo, G. L., & Richardson, J. R. (2017). Valproate increases dopamine transporter expression through histone acetylation and enhanced promoter binding of Nurr1. Neuropharmacology, 125, 189-196. doi:10.1016/j.neuropharm.2017.07.020
Grover, M., & Kashyap, P. C. (2014). Germ-free mice as a model to study effect of gut microbiota on host physiology. Neurogastroenterol Motil, 26(6), 745-748. doi:10.1111/nmo.12366
Guida, F., Turco, F., Iannotta, M., De Gregorio, D., Palumbo, I., Sarnelli, G., . . . Maione, S. (2018). Antibiotic-induced microbiota perturbation causes gut endocannabinoidome changes, hippocampal neuroglial reorganization and depression in mice. Brain Behav Immun, 67, 230-245. doi:10.1016/j.bbi.2017.09.001
Guize, L., Pannier, B., Thomas, F., Bean, K., Jego, B., & Benetos, A. (2008). Recent advances in metabolic syndrome and cardiovascular disease. Arch Cardiovasc Dis, 101(9), 577-583. doi:10.1016/j.acvd.2008.06.011
Hossain, P., Kawar, B., & El Nahas, M. (2007). Obesity and diabetes in the developing world--a growing challenge. N Engl J Med, 356(3), 213-215. doi:10.1056/NEJMp068177
Huang, X., & Yang, Z. (2016). Resistin's, obesity and insulin resistance: the continuing disconnect between rodents and humans. J Endocrinol Invest, 39(6), 607-615. doi:10.1007/s40618-015-0408-2
Huo, R., Zeng, B., Zeng, L., Cheng, K., Li, B., Luo, Y., . . . Xie, P. (2017). Microbiota Modulate Anxiety-Like Behavior and Endocrine Abnormalities in Hypothalamic-Pituitary-Adrenal Axis. Front Cell Infect Microbiol, 7, 489. doi:10.3389/fcimb.2017.00489
Hylemon, P. B., Zhou, H., Pandak, W. M., Ren, S., Gil, G., & Dent, P. (2009). Bile acids as regulatory molecules. J Lipid Res, 50(8), 1509-1520. doi:10.1194/jlr.R900007-JLR200
Ianiro, G., Tilg, H., & Gasbarrini, A. (2016). Antibiotics as deep modulators of gut microbiota: between good and evil. Gut, 65(11), 1906. doi:10.1136/gutjnl-2016-312297
Iniguez, S. D., Riggs, L. M., Nieto, S. J., Dayrit, G., Zamora, N. N., Shawhan, K. L., . . . Warren, B. L. (2014). Social defeat stress induces a depression-like phenotype in adolescent male c57BL/6 mice. Stress, 17(3), 247-255. doi:10.3109/10253890.2014.910650
Isovich, E., Engelmann, M., Landgraf, R., & Fuchs, E. (2001). Social isolation after a single defeat reduces striatal dopamine transporter binding in rats. Eur J Neurosci, 13(6), 1254-1256.
Jernberg, C., Lofmark, S., Edlund, C., & Jansson, J. K. (2010). Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology, 156(Pt 11), 3216-3223. doi:10.1099/mic.0.040618-0
Jiang, H., Ling, Z., Zhang, Y., Mao, H., Ma, Z., Yin, Y., . . . Ruan, B. (2015). Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun, 48, 186-194. doi:10.1016/j.bbi.2015.03.016
Johnson, P. M., & Kenny, P. J. (2010). Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat Neurosci, 13(5), 635-641. doi:10.1038/nn.2519
Johnston, D. (1984). Valproic Acid: Update on Its Mechanisms of Action. Epilepsia, 25(s1), S1-S4. doi:10.1111/j.1528-1157.1984.tb05630.x
Kanemura, H., Sano, F., Maeda, Y.-i., Sugita, K., & Aihara, M. (2012). Valproate sodium enhances body weight gain in patients with childhood epilepsy: A pathogenic mechanisms and open-label clinical trial of behavior therapy. Seizure - European Journal of Epilepsy, 21(7), 496-500. doi:10.1016/j.seizure.2012.05.001
Kemp, D. E. (2014). Managing the side effects associated with commonly used treatments for bipolar depression. J Affect Disord, 169 Suppl 1, S34-44. doi:10.1016/s0165-0327(14)70007-2
Kessing, L. V., Vradi, E., & Andersen, P. K. (2015). Life expectancy in bipolar disorder. Bipolar Disord, 17(5), 543-548. doi:10.1111/bdi.12296
Kilbourne, A. M., Rofey, D. L., McCarthy, J. F., Post, E. P., Welsh, D., & Blow, F. C. (2007). Nutrition and exercise behavior among patients with bipolar disorder. Bipolar Disord, 9(5), 443-452. doi:10.1111/j.1399-5618.2007.00386.x
Kirchgessner, A. L., & Gershon, M. D. (1990). Innervation of the pancreas by neurons in the gut. J Neurosci, 10(5), 1626-1642.
Krishnan, V., Han, M.-H., Graham, D. L., Berton, O., Renthal, W., Russo, S. J., . . . Nestler, E. J. (2007). Molecular Adaptations Underlying Susceptibility and Resistance to Social Defeat in Brain Reward Regions. Cell, 131(2), 391-404. doi:https://doi.org/10.1016/j.cell.2007.09.018
Kuno, T., Hirayama-Kurogi, M., Ito, S., & Ohtsuki, S. (2018). Reduction in hepatic secondary bile acids caused by short-term antibiotic-induced dysbiosis decreases mouse serum glucose and triglyceride levels. Scientific Reports, 8(1), 1253. doi:10.1038/s41598-018-19545-1
Li, Y., Ding, L., Hassan, W., Abdelkader, D., & Shang, J. (2013). Adipokines and hepatic insulin resistance. J Diabetes Res, 2013, 170532. doi:10.1155/2013/170532
Lozupone, C., & Knight, R. (2005). UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol, 71(12), 8228-8235. doi:10.1128/AEM.71.12.8228-8235.2005
Lozupone, C., Lladser, M. E., Knights, D., Stombaugh, J., & Knight, R. (2010). UniFrac: an effective distance metric for microbial community comparison. The Isme Journal, 5, 169. doi:10.1038/ismej.2010.133
https://www.nature.com/articles/ismej2010133#supplementary-information
Luo, Y., Zeng, B., Zeng, L., Du, X., Li, B., Huo, R., . . . Xie, P. (2018). Gut microbiota regulates mouse behaviors through glucocorticoid receptor pathway genes in the hippocampus. Translational Psychiatry, 8(1), 187. doi:10.1038/s41398-018-0240-5
Lurie, I., Yang, Y. X., Haynes, K., Mamtani, R., & Boursi, B. (2015). Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. J Clin Psychiatry, 76(11), 1522-1528. doi:10.4088/JCP.15m09961
Lyte, M. (2004). Microbial endocrinology and infectious disease in the 21st century. Trends Microbiol, 12(1), 14-20.
M, D. E. H., Correll, C. U., Bobes, J., Cetkovich-Bakmas, M., Cohen, D., Asai, I., . . . Leucht, S. (2011). Physical illness in patients with severe mental disorders. I. Prevalence, impact of medications and disparities in health care. World Psychiatry, 10(1), 52-77.
Macfarlane, G. T., & Macfarlane, S. (2012). Bacteria, colonic fermentation, and gastrointestinal health. J AOAC Int, 95(1), 50-60.
McElroy, S. L., & Keck, P. E., Jr. (2014). Metabolic syndrome in bipolar disorder: a review with a focus on bipolar depression. J Clin Psychiatry, 75(1), 46-61. doi:10.4088/JCP.13r08634
McGaughey, K. D., Yilmaz-Swenson, T., Elsayed, N. M., Cruz, D. A., Rodriguiz, R. M., Kritzer, M. D., . . . Williamson, D. E. (2019). Relative abundance of Akkermansia spp. and other bacterial phylotypes correlates with anxiety- and depressive-like behavior following social defeat in mice. Scientific Reports, 9(1), 3281. doi:10.1038/s41598-019-40140-5
McVey Neufeld, K. A., Kay, S., & Bienenstock, J. (2018). Mouse Strain Affects Behavioral and Neuroendocrine Stress Responses Following Administration of Probiotic Lactobacillus rhamnosus JB-1 or Traditional Antidepressant Fluoxetine. Front Neurosci, 12, 294. doi:10.3389/fnins.2018.00294
Mesdjian, E., Ciesielski, L., Valli, M., Bruguerolle, B., Jadot, G., Bouyard, P., & Mandel, P. (1982). Sodium valproate: kinetic profile and effects on GABA levels in various brain areas of the rat. Prog Neuropsychopharmacol Biol Psychiatry, 6(3), 223-233.
Niimi, K., & Takahashi, E. (2019). New system to examine the activity and water and food intake of germ-free mice in a sealed positive-pressure cage. Heliyon, 5(8), e02176. doi:https://doi.org/10.1016/j.heliyon.2019.e02176
Novick, A. M., Forster, G. L., Tejani-Butt, S. M., & Watt, M. J. (2011). Adolescent social defeat alters markers of adult dopaminergic function. Brain research bulletin, 86(1-2), 123-128. doi:10.1016/j.brainresbull.2011.06.009
Ondov, B. D., Bergman, N. H., & Phillippy, A. M. (2011). Interactive metagenomic visualization in a Web browser. BMC Bioinformatics, 12(1), 385. doi:10.1186/1471-2105-12-385
Ostadhadi, S., Imran Khan, M., Norouzi-Javidan, A., & Dehpour, A.-R. (2016). Antidepressant effect of pramipexole in mice forced swimming test: A cross talk between dopamine receptor and NMDA/nitric oxide/cGMP pathway. Biomedicine & Pharmacotherapy, 81, 295-304. doi:https://doi.org/10.1016/j.biopha.2016.04.026
Perry, W., Minassian, A., Paulus, M. P., Young, J. W., Kincaid, M. J., Ferguson, E. J., . . . Geyer, M. A. (2009). A reverse-translational study of dysfunctional exploration in psychiatric disorders: from mice to men. Arch Gen Psychiatry, 66(10), 1072-1080. doi:10.1001/archgenpsychiatry.2009.58
Quast, C., Pruesse, E., Yilmaz, P., Gerken, J., Schweer, T., Yarza, P., . . . Glöckner, F. O. (2013). The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic acids research, 41(Database issue), D590-D596. doi:10.1093/nar/gks1219
Reinholz, J., Skopp, O., Breitenstein, C., Bohr, I., Winterhoff, H., & Knecht, S. (2008). Compensatory weight gain due to dopaminergic hypofunction: new evidence and own incidental observations. Nutrition & metabolism, 5, 35-35. doi:10.1186/1743-7075-5-35
Remage-Healey, L., & Michael Romero, L. (2002). Corticosterone and insulin interact to regulate plasma glucose but not lipid concentrations in molting starlings. Gen Comp Endocrinol, 129(2), 88-94.
Ridlon, J. M., Kang, D. J., & Hylemon, P. B. (2006). Bile salt biotransformations by human intestinal bacteria. J Lipid Res, 47(2), 241-259. doi:10.1194/jlr.R500013-JLR200
Ridlon, J. M., Kang, D. J., Hylemon, P. B., & Bajaj, J. S. (2014). Bile acids and the gut microbiome. Curr Opin Gastroenterol, 30(3), 332-338. doi:10.1097/mog.0000000000000057
Rosenberg, G. (2007). The mechanisms of action of valproate in neuropsychiatric disorders: can we see the forest for the trees? Cellular and Molecular Life Sciences, 64(16), 2090-2103. doi:10.1007/s00018-007-7079-x
Rubi, B., Ljubicic, S., Pournourmohammadi, S., Carobbio, S., Armanet, M., Bartley, C., & Maechler, P. (2005). Dopamine D2-like receptors are expressed in pancreatic beta cells and mediate inhibition of insulin secretion. J Biol Chem, 280(44), 36824-36832. doi:10.1074/jbc.M505560200
Savignac, H. M., Finger, B. C., Pizzo, R. C., O'Leary, O. F., Dinan, T. G., & Cryan, J. F. (2011). Increased sensitivity to the effects of chronic social defeat stress in an innately anxious mouse strain. Neuroscience, 192, 524-536. doi:10.1016/j.neuroscience.2011.04.054
Segata, N., Izard, J., Waldron, L., Gevers, D., Miropolsky, L., Garrett, W. S., & Huttenhower, C. (2011). Metagenomic biomarker discovery and explanation. Genome Biol, 12(6), R60. doi:10.1186/gb-2011-12-6-r60
Sharma, R. P., Rosen, C., Kartan, S., Guidotti, A., Costa, E., Grayson, D. R., & Chase, K. (2006). Valproic acid and chromatin remodeling in schizophrenia and bipolar disorder: preliminary results from a clinical population. Schizophr Res, 88(1-3), 227-231. doi:10.1016/j.schres.2006.07.015
Sigitova, E., Fisar, Z., Hroudova, J., Cikankova, T., & Raboch, J. (2017). Biological hypotheses and biomarkers of bipolar disorder. Psychiatry Clin Neurosci, 71(2), 77-103. doi:10.1111/pcn.12476
Snider, S. R., & Kuchel, O. (1983). Dopamine: an important neurohormone of the sympathoadrenal system. Significance of increased peripheral dopamine release for the human stress response and hypertension. Endocr Rev, 4(3), 291-309. doi:10.1210/edrv-4-3-291
Spohn, S. N., & Mawe, G. M. (2017). Non-conventional features of peripheral serotonin signalling - the gut and beyond. Nat Rev Gastroenterol Hepatol, 14(7), 412-420. doi:10.1038/nrgastro.2017.51
Strandwitz, P. (2018). Neurotransmitter modulation by the gut microbiota. Brain Research, 1693, 128-133. doi:https://doi.org/10.1016/j.brainres.2018.03.015
Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X. N., . . . Koga, Y. (2004). Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol, 558(Pt 1), 263-275. doi:10.1113/jphysiol.2004.063388
Thaiss, C. A., Zeevi, D., Levy, M., Zilberman-Schapira, G., Suez, J., Tengeler, A. C., . . . Elinav, E. (2014). Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell, 159(3), 514-529. doi:10.1016/j.cell.2014.09.048
Tidey, J. W., & Miczek, K. A. (1996). Social defeat stress selectively alters mesocorticolimbic dopamine release: an in vivo microdialysis study. Brain Res, 721(1-2), 140-149. doi:10.1016/0006-8993(96)00159-x
Tong, S. T. Y. (1992). The use of non-metric multidimensional scaling as an ordination technique in resource survey and evaluation: a case study from southeast Spain. Applied Geography, 12(3), 243-260. doi:https://doi.org/10.1016/0143-6228(92)90042-L
Valentini, M., Piermattei, A., Di Sante, G., Migliara, G., Delogu, G., & Ria, F. (2014). Immunomodulation by Gut Microbiota: Role of Toll-Like Receptor Expressed by T Cells. Journal of Immunology Research, 2014, 8. doi:10.1155/2014/586939
Vancampfort, D., Vansteelandt, K., Correll, C. U., Mitchell, A. J., De Herdt, A., Sienaert, P., . . . De Hert, M. (2013). Metabolic syndrome and metabolic abnormalities in bipolar disorder: a meta-analysis of prevalence rates and moderators. Am J Psychiatry, 170(3), 265-274. doi:10.1176/appi.ajp.2012.12050620
VanDongen, A. M., VanErp, M. G., & Voskuyl, R. A. (1986). Valproate reduces excitability by blockage of sodium and potassium conductance. Epilepsia, 27(3), 177-182.
Verrotti, A., D'Egidio, C., Mohn, A., Coppola, G., & Chiarelli, F. (2011). Weight gain following treatment with valproic acid: pathogenetic mechanisms and clinical implications. Obes Rev, 12(5), e32-43. doi:10.1111/j.1467-789X.2010.00800.x
Verrotti, A., la Torre, R., Trotta, D., Mohn, A., & Chiarelli, F. (2009). Valproate-induced insulin resistance and obesity in children. Horm Res, 71(3), 125-131. doi:10.1159/000197868
Vestri, H. S., Maianu, L., Moellering, D. R., & Garvey, W. T. (2007). Atypical antipsychotic drugs directly impair insulin action in adipocytes: effects on glucose transport, lipogenesis, and antilipolysis. Neuropsychopharmacology, 32(4), 765-772. doi:10.1038/sj.npp.1301142
Walf, A. A., & Frye, C. A. (2007). The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat Protoc, 2(2), 322-328. doi:10.1038/nprot.2007.44
Wang, G. J., Volkow, N. D., Logan, J., Pappas, N. R., Wong, C. T., Zhu, W., . . . Fowler, J. S. (2001). Brain dopamine and obesity. Lancet, 357(9253), 354-357. doi:10.1016/s0140-6736(00)03643-6
Wang, J., Michelhaugh, S. K., & Bannon, M. J. (2007). Valproate robustly increases Sp transcription factor-mediated expression of the dopamine transporter gene within dopamine cells. Eur J Neurosci, 25(7), 1982-1986. doi:10.1111/j.1460-9568.2007.05460.x
Whittaker, R. H. (1972). Evolution and measurement of species diversity. Taxon, 213-251.
Wikoff, W. R., Anfora, A. T., Liu, J., Schultz, P. G., Lesley, S. A., Peters, E. C., & Siuzdak, G. (2009). Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proceedings of the National Academy of Sciences, 106(10), 3698. doi:10.1073/pnas.0812874106
Yamashita, H., Fujisawa, K., Ito, E., Idei, S., Kawaguchi, N., Kimoto, M., . . . Tsuji, H. (2007). Improvement of Obesity and Glucose Tolerance by Acetate in Type 2 Diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) Rats. Bioscience, Biotechnology, and Biochemistry, 71(5), 1236-1243. doi:10.1271/bbb.60668
Yildirim, E., Zhang, Z., Uz, T., Chen, C.-q., Manev, R., & Manev, H. (2003). Valproate administration to mice increases histone acetylation and 5-lipoxygenase content in the hippocampus. Neuroscience Letters, 345(2), 141-143. doi:https://doi.org/10.1016/S0304-3940(03)00490-7
Zarate, C. A., Jr., Payne, J. L., Singh, J., Quiroz, J. A., Luckenbaugh, D. A., Denicoff, K. D., . . . Manji, H. K. (2004). Pramipexole for bipolar II depression: a placebo-controlled proof of concept study. Biol Psychiatry, 56(1), 54-60. doi:10.1016/j.biopsych.2004.03.013