簡易檢索 / 詳目顯示

研究生: 邱佳慧
Chiu, Chia-Hui
論文名稱: 利用整合性基因體方法的富集分析和多基因風險評分於雙極症的臨床效用
Leverage the enrichment analysis and polygenic risk score of integrative genomic approaches in the clinical utility of bipolar disorder
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: 2022
畢業學年度: 110
語文別: 英文
論文頁數: 402
中文關鍵詞: 雙極症路徑富集分析多基因風險評分基因體藥物富集評分成纖維細胞生長因子-2澱粉樣蛋白-β
外文關鍵詞: bipolar disorder, pathway enrichment analysis,, polygenic risk score, pharmagenic enrichment score, fibroblast growth factor-2, amyloid-β
相關次數: 點閱:96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
  • 摘要 I Abstract III Abbreviation VI Contents 1 List of Tables 5 List of Figures 11 List of Supplementary Appendix Tables 15 List of Supplementary Appendix Figures 16 Chapter 1 Introduction 17 1.1 Bipolar disorder (BD) 17 1.1.1 Clinical feature and epidemiology of BD 17 1.1.2 Pharmacotherapy for BD 17 1.2 Challenge in BD and its treatment 19 1.2.1 Neurocognitive function impairment of BD 19 1.2.2 Metabolic disturbance of BD 19 1.2.3 Variation in pharmacotherapy of BD 20 1.3 Genome-wide association study (GWAS) of BD 21 1.3.1 Genetic attribution of BD 21 1.3.2 GWAS to reveal biological insights into complex disorder 21 1.3.3 GWAS and BD disease 21 1.4 Post GWAS analysis 23 1.4.1 Enrichment analysis 23 1.4.2 Mendelian randomization 23 1.5 Polygenic risk score (PRS) 24 1.5.1 Introduction of PRS 24 1.5.2 PRS and disease risk 24 1.5.3 PRS and environmental factors toward the disease risk 24 1.6 Pharmagenic enrichment scores (PES) 25 Chapter 2 Objective of current study 26 Chapter 3 Material and Methods 27 3.1 Study design 27 3.2 Taiwan Biobank (TWBB) 27 3.3 Genome-wide association study (GWAS) and enrichment analysis 29 3.3.1 Subjects selection 29 3.3.2 GWAS 29 3.3.3 SNPs annotation 30 3.3.4 MAGMA gene-based analysis 30 3.3.5 Post GWAS analysis 31 3.4 Clinical measurement collection from the NCKUH cohort 32 3.4.1 Subject recruitment from NCKUH 32 3.4.2 Mood status and disease severity assessment 33 3.4.3 Blood sample collection 33 3.4.4 DNA extraction and genotyping 33 3.4.5 Physical and hematological parameters 34 3.4.6 Metabolic syndrome assessment 36 3.4.7 Treatment response assessment 36 3.4.8 Neurocognitive function assessment 36 3.4.9 SPECT with [99mTc] TRODAT-1 37 3.4.10 Questionnaire 37 3.5 Genotype for apolipoprotein E (APOE) carrier 38 3.6 Statistics 39 3.7 Mendelian randomization 40 3.8 Polygenic risk scores (PRS) 42 3.8.1 The PRS analysis process 42 3.8.2 Software and cohort data for PRS calculation 42 3.8.3 Quality control of base and target data 42 3.8.4 Process of PRS calculation 43 3.9 Pharmagenic enrichment score (PES) 45 Chapter 4 Results 47 4.1 GWAS in BD from the TWBB cohort 48 4.1.1 Characteristic of BD patients and non-BD subjects in TWBB 48 4.1.2 Principal component analysis for comorbidities in TWBB 49 4.1.3 Model selection for GWAS 50 4.1.4 GWAS in TWBB 51 4.2 Post GWAS analysis 55 4.2.1 Enrichment analysis for SNP overlapped genes 55 4.2.2 MAGMA gene-based analysis 56 4.2.3 Enrichment analysis for MAGMA genes 57 4.3 Clinical characteristics of BD and controls from the NCKUH cohort 59 4.3.1 Study population of the NCKUH cohort 59 4.3.2 Clinical characteristic of the BD patients and controls 59 4.3.3 GWAS replication study of BD in the NCKUH cohort 62 4.4 Fibroblast growth factor-2 (FGF2) in BD 62 4.4.1 Characteristics of BD and controls with FGF2 measurements 62 4.4.2 Causal relationship between FGF2 and BD risk using MR 63 4.4.3 Correlation between FGF2 and clinical indices in BD and controls 63 4.4.4 Characteristics of BD VPA users, non-VPA users and controls with FGF2 measurements 64 4.4.5 Correlation between FGF2 and clinical indices in BD VPA users, non-VPA users and controls 65 4.5 Amyloid-β (Aβ) in BD 66 4.5.1 Characteristics of BD and controls with Aβ measurements 66 4.5.2 Causal relationship between Aβ and BD risk using MR 67 4.5.3 Correlation between Aβ and clinical indices 67 4.6 Polygenic risk score (PRS) 70 4.6.1 PRS model construction for BD risk estimation 70 4.6.2 Association of PRS with BD disease risk in NCKUH cohort 71 4.6.3 Association of environmental factors and PRS with BD risk 72 4.6.4 Correlation between PRS and clinical outcomes 74 4.6.5 Enrichment analysis for gene sets within PRS 75 4.7 Pharmacological enrichment and pharmagenic enrichment score 77 4.7.1 Pharmacological enrichment 77 4.7.2 Gene-drug interaction within the pharmacological enrichment 78 4.7.3 Pharmagenic enrichment score (PES) 79 Chapter 5 Discussion 274 5.1 Characteristics of BD in Taiwanese population 274 5.1.1 Characteristics of BD and controls in the TWBB cohort 274 5.1.2 Characteristics of BD and controls in the NCKUH cohort 276 5.2 GWAS in Taiwanese BD population 277 5.2.1 Significant BD-associated loci 277 5.2.2 Significant BD-associated loci within the gene-based analysis 279 5.3 Explore the functional and pathway enrichment in BD 279 5.3.1 Brain tissue expression and network interaction of gene products 280 5.3.2 The functional and pathway enrichment in BD-related gene sets 281 5.3.3 Inflammation, metabolism, and neurological disturbance of BD 284 5.3.4 Suggesting molecular targets of BD 285 5.4 Investigate the role of fibroblast growth factor-2 in BD illness 285 5.4.1 Decreased FGF2 in BD patients 285 5.4.2 Correlation between FGF2 and clinical indices 285 5.4.3 Increased FGF2 levels in BD VPA users 286 5.4.4 Clinical utility of FGF2 in BD patients 286 5.5 Investigate the role of Aβ in BD illness 287 5.5.1 Decreased Aβ in BD illness 287 5.5.2 Cognitive deficits appeared before plaque deposition 290 5.5.3 Aβ morphology and neurological profile 290 5.5.4 Causal relationship between Aβ and BD risk 291 5.5.5 Correlation between Aβ and clinical indices in BD 291 5.6 Implication of polygenic score in BD for precise medication strategy 294 5.6.1 Polygenic risk score (PRS) 294 5.6.2 Pharmacological enrichment and pharmagenic enrichment score (PES) 295 5.7 Clinical utility of the study 300 5.8 Limitation 301 Chapter 6 Conclusion 302 Chapter 7 Reference 303

    Agnew-Blais, J., & Danese, A. (2016). Childhood maltreatment and unfavourable clinical outcomes in bipolar disorder: a systematic review and meta-analysis. The Lancet Psychiatry, 3(4), 342-349. doi:https://doi.org/10.1016/S2215-0366(15)00544-1
    Ahn, S. W., Baek, J. H., et al. (2017). Long-term response to mood stabilizer treatment and its clinical correlates in patients with bipolar disorders: a retrospective observational study. Int J Bipolar Disord, 5(1), 24. doi:10.1186/s40345-017-0093-5
    Aken, B. L., Achuthan, P., et al. (2016). Ensembl 2017. Nucleic Acids Research, 45(D1), D635-D642. doi:10.1093/nar/gkw1104
    Akita, S., Akino, K., et al. (2013). Basic Fibroblast Growth Factor in Scarless Wound Healing. Adv Wound Care (New Rochelle), 2(2), 44-49. doi:10.1089/wound.2011.0324
    Alam, S., Suzuki, H., et al. (2014). Alternative splicing regulation of APP exon 7 by RBFox proteins. Neurochem Int, 78, 7-17. doi:10.1016/j.neuint.2014.08.001
    American Psychiatric Association. (2013). Diagnostic and Statistical Manual of Mental Disorders (Fifth ed.).
    Andlauer, T. F., & Nöthen, M. M. (2020). Polygenic scores for psychiatric disease: from research tool to clinical application. Medizinische Genetik, 32(1), 39-45.
    Atanasova, B., Graux, J., et al. (2008). Olfaction: a potential cognitive marker of psychiatric disorders. Neurosci Biobehav Rev, 32(7), 1315-1325. doi:10.1016/j.neubiorev.2008.05.003
    Avram, S., Bologa, C. G., et al. (2020). DrugCentral 2021 supports drug discovery and repositioning. Nucleic Acids Research, 49(D1), D1160-D1169. doi:10.1093/nar/gkaa997
    Bai, H., & Zhang, Q. (2021). Activation of NLRP3 Inflammasome and Onset of Alzheimer's Disease. Front Immunol, 12, 701282. doi:10.3389/fimmu.2021.701282
    Bai, Y.-M., Li, C.-T., et al. (2016). Metabolic syndrome and adverse clinical outcomes in patients with bipolar disorder. BMC Psychiatry, 16(1), 448. doi:10.1186/s12888-016-1143-8
    Bai, Y. M., Chang, C. J., et al. (2013). Taiwan consensus of pharmacological treatment for bipolar disorder. J Chin Med Assoc, 76(10), 547-556. doi:10.1016/j.jcma.2013.06.013
    Bai, Y. M., Li, C. T., et al. (2016). Metabolic syndrome and adverse clinical outcomes in patients with bipolar disorder. BMC Psychiatry, 16(1), 448. doi:10.1186/s12888-016-1143-8
    Baird, A., & Klagsbrun, M. (1991). The Fibroblast Growth Factor Family An Overview. Annals of the New York Academy of Sciences, 638(1), xi-xii. doi:https://doi.org/10.1111/j.1749-6632.1991.tb49010.x
    Baptista, T., Rangel, N., et al. (2007). Metformin as an adjunctive treatment to control body weight and metabolic dysfunction during olanzapine administration: A multicentric, double-blind, placebo-controlled trial. Schizophrenia Research, 93(1), 99-108. doi:https://doi.org/10.1016/j.schres.2007.03.029
    Barbosa, I. G., Ferreira, G. C., et al. (2020). The Renin Angiotensin System and Bipolar Disorder: A Systematic Review. Protein Pept Lett, 27(6), 520-528. doi:10.2174/0929866527666200127115059
    Barbosa, I. G., Rocha, N. P., et al. (2012). Increased levels of adipokines in bipolar disorder. J Psychiatr Res, 46(3), 389-393. doi:10.1016/j.jpsychires.2011.11.010
    Barbosa, I. G., Rocha, N. P., et al. (2013). Increased BDNF levels in long-term bipolar disorder patients. Braz J Psychiatry, 35(1), 67-69. doi:10.1016/j.rbp.2012.05.011
    Bautista, L. E., Smeeth, L., et al. (2006). Estimation of bias in nongenetic observational studies using "mendelian triangulation". Annals of Epidemiology, 16(9), 675-680. doi:10.1016/j.annepidem.2006.02.001
    Bellenguez, C., Küçükali, F., et al. (2022). New insights into the genetic etiology of Alzheimer's disease and related dementias. Nat Genet, 54(4), 412-436. doi:10.1038/s41588-022-01024-z
    Benarroch, E. E. (2017). Anoctamins (TMEM16 proteins). Neurology, 89(7), 722. doi:10.1212/WNL.0000000000004246
    Benzel, I., Bansal, A., et al. (2007). Interactions among genes in the ErbB-Neuregulin signalling network are associated with increased susceptibility to schizophrenia. Behavioral and Brain Functions, 3(1), 31. doi:10.1186/1744-9081-3-31
    Bi, B. T., Lin, H. B., et al. (2012). Promotion of β-amyloid production by C-reactive protein and its implications in the early pathogenesis of Alzheimer's disease. Neurochem Int, 60(3), 257-266. doi:10.1016/j.neuint.2011.12.007
    Bienvenu, T., & Chelly, J. (2006). Molecular genetics of Rett syndrome: when DNA methylation goes unrecognized. Nat Rev Genet, 7(6), 415-426. doi:10.1038/nrg1878
    Billings, L. M., Oddo, S., et al. (2005). Intraneuronal Aβ Causes the Onset of Early Alzheimer’s Disease-Related Cognitive Deficits in Transgenic Mice. Neuron, 45(5), 675-688. doi:https://doi.org/10.1016/j.neuron.2005.01.040
    Boal, A. H., Smith, D. J., et al. (2016). Monotherapy With Major Antihypertensive Drug Classes and Risk of Hospital Admissions for Mood Disorders. Hypertension, 68(5), 1132-1138. doi:10.1161/HYPERTENSIONAHA.116.08188
    Bocchetta, A., Traccis, F., et al. (2016). Bipolar disorder and antithyroid antibodies: review and case series. Int J Bipolar Disord, 4(1), 5. doi:10.1186/s40345-016-0046-4
    Bourin, M., Chenu, F., et al. (2009). The role of sodium channels in the mechanism of action of antidepressants and mood stabilizers. Curr Drug Targets, 10(11), 1052-1060. doi:10.2174/138945009789735138
    Bourne, C., Aydemir, Ö., et al. (2013). Neuropsychological testing of cognitive impairment in euthymic bipolar disorder: an individual patient data meta-analysis. Acta Psychiatr Scand, 128(3), 149-162. doi:10.1111/acps.12133
    Brembeck, F. H., Rosário, M., et al. (2006). Balancing cell adhesion and Wnt signaling, the key role of beta-catenin. Curr Opin Genet Dev, 16(1), 51-59. doi:10.1016/j.gde.2005.12.007
    Browning, B. L., & Browning, S. R. (2016). Genotype Imputation with Millions of Reference Samples. Am J Hum Genet, 98(1), 116-126. doi:10.1016/j.ajhg.2015.11.020
    Bulik-Sullivan, B. K., Loh, P.-R., et al. (2015). LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nature Genetics, 47(3), 291-295. doi:10.1038/ng.3211
    Burgess, S., & Bowden, J. (2015). Integrating summarized data from multiple genetic variants in Mendelian randomization: bias and coverage properties of inverse-variance weighted methods. arXiv preprint arXiv:1512.04486.
    Burton, P. R., Clayton, D. G., et al. (2007). Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature, 447(7145), 661-678. doi:10.1038/nature05911
    Buysse, D. J., Reynolds, C. F., 3rd, et al. (1989). The Pittsburgh Sleep Quality Index: a new instrument for psychiatric practice and research. Psychiatry Res, 28(2), 193-213. doi:10.1016/0165-1781(89)90047-4
    Calkin, C. V., Chengappa, K. N. R., et al. (2022). Treating Insulin Resistance With Metformin as a Strategy to Improve Clinical Outcomes in Treatment-Resistant Bipolar Depression (the TRIO-BD Study): A Randomized, Quadruple-Masked, Placebo-Controlled Clinical Trial. J Clin Psychiatry, 83(2). doi:10.4088/JCP.21m14022
    Canevari, L., Abramov, A. Y., et al. (2004). Toxicity of amyloid beta peptide: tales of calcium, mitochondria, and oxidative stress. Neurochem Res, 29(3), 637-650. doi:10.1023/b:nere.0000014834.06405.af
    Cardoso, T., Bauer, I. E., et al. (2015). Neuroprogression and Cognitive Functioning in Bipolar Disorder: A Systematic Review. Curr Psychiatry Rep, 17(9), 75. doi:10.1007/s11920-015-0605-x
    Carlsen, J., de Olmos, J., et al. (1982). Tracing of two-neuron pathways in the olfactory system by the aid of transneuronal degeneration: Projections to the amygdaloid body and hippocampal formation. Journal of Comparative Neurology, 208(2), 196-208. doi:https://doi.org/10.1002/cne.902080208
    Cearns, M., Amare, A. T., et al. (2022). Using polygenic scores and clinical data for bipolar disorder patient stratification and lithium response prediction: machine learning approach. Br J Psychiatry, 1-10. doi:10.1192/bjp.2022.28
    Ceraso, A., Lin, J. J., et al. (2022). Maintenance Treatment With Antipsychotic Drugs in Schizophrenia: A Cochrane Systematic Review and Meta-analysis. Schizophr Bull, 48(4), 738-740. doi:10.1093/schbul/sbac041
    Cernea, S., Dima, L., et al. (2020). Pharmacological Management of Glucose Dysregulation in Patients Treated with Second-Generation Antipsychotics. Drugs, 80(17), 1763-1781. doi:10.1007/s40265-020-01393-x
    Chadwick, D. W. (1985). Concentration-effect relationships of valproic acid. Clin Pharmacokinet, 10(2), 155-163. doi:10.2165/00003088-198510020-00003
    Chahrour, M., Jung, S. Y., et al. (2008). MeCP2, a key contributor to neurological disease, activates and represses transcription. Science, 320(5880), 1224-1229. doi:10.1126/science.1153252
    Chakrabarty, P., Li, A., et al. (2015). IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron, 85(3), 519-533. doi:10.1016/j.neuron.2014.11.020
    Chang, H. H., Chen, P. S., et al. (2017). Effect of memantine on C-reactive protein and lipid profiles in bipolar disorder. J Affect Disord, 221, 151-157. doi:10.1016/j.jad.2017.05.052
    Chang, H. H., Tseng, H.-H., et al. (2021). Peripheral insulin sensitivity predicting cognitive function in euthymic bipolar disorder patients. CNS Spectr, 1-6. doi:10.1017/S1092852921000158
    Chang, H. H., Yang, Y. K., et al. (2010). The role of valproate in metabolic disturbances in bipolar disorder patients. J Affect Disord, 124(3), 319-323. doi:https://doi.org/10.1016/j.jad.2009.12.011
    Chatterjee, N., Wheeler, B., et al. (2013). Projecting the performance of risk prediction based on polygenic analyses of genome-wide association studies. Nat Genet, 45(4), 400-405, 405e401-403. doi:10.1038/ng.2579
    Chen, G. F., Xu, T. H., et al. (2017). Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol Sin, 38(9), 1205-1235. doi:10.1038/aps.2017.28
    Chen, P.-L., Avramopoulos, D., et al. (2009). Fine Mapping on Chromosome 10q22-q23 Implicates Neuregulin 3 in Schizophrenia. The American Journal of Human Genetics, 84(1), 21-34. doi:https://doi.org/10.1016/j.ajhg.2008.12.005
    Chen, P. S., Peng, G. S., et al. (2006). Valproate protects dopaminergic neurons in midbrain neuron/glia cultures by stimulating the release of neurotrophic factors from astrocytes. Molecular Psychiatry, 11(12), 1116-1125. doi:10.1038/sj.mp.4001893
    Chen, P. S., Tang, L. Y., et al. (2022). Roles of C-reactive protein polymorphisms and life event changes on cognitive function in bipolar patients receiving valproate. Int J Immunopathol Pharmacol, 36, 3946320221084835. doi:10.1177/03946320221084835
    Chen, Q., Zhang, F., et al. (2021). SIDT1-dependent absorption in the stomach mediates host uptake of dietary and orally administered microRNAs. Cell Research, 31(3), 247-258. doi:10.1038/s41422-020-0389-3
    Chen, S., Ye, J., et al. (2018). Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-κB pathway dependent of HDAC3. Journal of Neuroinflammation, 15(1), 150. doi:10.1186/s12974-018-1193-6
    Chen, Y., Xu, J., et al. (2020). Coronin 2B regulates dendrite outgrowth by modulating actin dynamics. FEBS Lett, 594(18), 2975-2987. doi:10.1002/1873-3468.13886
    Chen, Y. R., Liang, C. S., et al. (2021). Diagnostic accuracy of blood biomarkers for Alzheimer's disease and amnestic mild cognitive impairment: A meta-analysis. Ageing Res Rev, 71, 101446. doi:10.1016/j.arr.2021.101446
    Cheng, T. L., Chen, J., et al. (2017). Regulation of mRNA splicing by MeCP2 via epigenetic modifications in the brain. Sci Rep, 7, 42790. doi:10.1038/srep42790
    Cheng, Y.-C., Chen, C.-H., et al. (2018). Reliability and factor structure of the Chinese version of childhood trauma questionnaire-short form in patients with substance use disorder. Taiwanese J Psychiatry, 32(1), 52-62.
    Chiarella, E., Aloisio, A., et al. (2018). ZNF521 Has an Inhibitory Effect on the Adipogenic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. Stem Cell Rev Rep, 14(6), 901-914. doi:10.1007/s12015-018-9830-0
    Choi, S. W., Mak, T. S., et al. (2020). Tutorial: a guide to performing polygenic risk score analyses. Nat Protoc, 15(9), 2759-2772. doi:10.1038/s41596-020-0353-1
    Choi, S. W., & O'Reilly, P. F. (2019). PRSice-2: Polygenic Risk Score software for biobank-scale data. Gigascience, 8(7). doi:10.1093/gigascience/giz082
    Chouraki, V., Beiser, A., et al. (2015). Plasma amyloid-β and risk of Alzheimer's disease in the Framingham Heart Study. Alzheimers Dement, 11(3), 249-257.e241. doi:10.1016/j.jalz.2014.07.001
    Cipriani, A., Reid, K., et al. (2013). Valproic acid, valproate and divalproex in the maintenance treatment of bipolar disorder. Cochrane Database Syst Rev, 2013(10), Cd003196. doi:10.1002/14651858.CD003196.pub2
    Cirulli, V., & Yebra, M. (2007). Netrins: beyond the brain. Nature Reviews Molecular Cell Biology, 8(4), 296-306. doi:10.1038/nrm2142
    Cline, E. N., Bicca, M. A., et al. (2018). The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis, 64(s1), S567-s610. doi:10.3233/jad-179941
    Cooper, G. M., Coe, B. P., et al. (2011). A copy number variation morbidity map of developmental delay. Nat Genet, 43(9), 838-846. doi:10.1038/ng.909
    Cusi, A. M., Macqueen, G. M., et al. (2012). Patients with bipolar disorder show impaired performance on complex tests of social cognition. Psychiatry Res, 200(2-3), 258-264. doi:10.1016/j.psychres.2012.06.021
    D'Anna, L., Abu-Rumeileh, S., et al. (2017). Serum Interleukin-10 Levels Correlate with Cerebrospinal Fluid Amyloid Beta Deposition in Alzheimer Disease Patients. Neurodegener Dis, 17(4-5), 227-234. doi:10.1159/000474940
    Dandekar, M. P., Valvassori, S. S., et al. (2018). Glycogen Synthase Kinase-3β as a Putative Therapeutic Target for Bipolar Disorder. Curr Drug Metab, 19(8), 663-673. doi:10.2174/1389200219666171227203737
    Dauphinais, D., Knable, M., et al. (2011). Zonisamide for bipolar disorder, mania or mixed states: a randomized, double blind, placebo-controlled adjunctive trial. Psychopharmacol Bull, 44(1), 5-17.
    Davies, N. M., Holmes, M. V., et al. (2018). Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. Bmj, 362, k601. doi:10.1136/bmj.k601
    de Leeuw, C. A., Mooij, J. M., et al. (2015). MAGMA: Generalized Gene-Set Analysis of GWAS Data. PLOS Computational Biology, 11(4), e1004219. doi:10.1371/journal.pcbi.1004219
    de Rojas, I., Moreno-Grau, S., et al. (2021). Common variants in Alzheimer's disease and risk stratification by polygenic risk scores. Nat Commun, 12(1), 3417. doi:10.1038/s41467-021-22491-8
    Delgado, F. G., Cárdenas, P., et al. (2018). Valproic Acid Downregulates Cytokine Expression in Human Macrophages Infected with Dengue Virus. Diseases, 6(3). doi:10.3390/diseases6030059
    Dickerson, F., Stallings, C., et al. (2013). Elevated C-reactive protein and cognitive deficits in individuals with bipolar disorder. J Affect Disord, 150(2), 456-459. doi:10.1016/j.jad.2013.04.039
    do Prado, C. H., Rizzo, L. B., et al. (2013). Reduced regulatory T cells are associated with higher levels of Th1/TH17 cytokines and activated MAPK in type 1 bipolar disorder. Psychoneuroendocrinology, 38(5), 667-676. doi:10.1016/j.psyneuen.2012.08.005
    Dodart, J.-C., Bales, K. R., et al. (2002). Immunization reverses memory deficits without reducing brain Aβ burden in Alzheimer's disease model. Nature Neuroscience, 5(5), 452-457. doi:10.1038/nn842
    Doecke, J. D., Pérez-Grijalba, V., et al. (2020). Total Aβ(42)/Aβ(40) ratio in plasma predicts amyloid-PET status, independent of clinical AD diagnosis. Neurology, 94(15), e1580-e1591. doi:10.1212/wnl.0000000000009240
    Doganavsargil-Baysal, O., Cinemre, B., et al. (2013). Levels of TNF-α, soluble TNF receptors (sTNFR1, sTNFR2), and cognition in bipolar disorder. Hum Psychopharmacol, 28(2), 160-167. doi:10.1002/hup.2301
    Duan, Y., Liao, A. P., et al. (2007). beta-Catenin activity negatively regulates bacteria-induced inflammation. Lab Invest, 87(6), 613-624. doi:10.1038/labinvest.3700545
    Dudbridge, F. (2013). Power and predictive accuracy of polygenic risk scores. PLoS Genet, 9(3), e1003348. doi:10.1371/journal.pgen.1003348
    Dunn, O. J. (1961). Multiple Comparisons among Means. Journal of the American Statistical Association, 56(293), 52-64. doi:10.1080/01621459.1961.10482090
    Eggert, S., Thomas, C., et al. (2018). Trafficking in Alzheimer’s Disease: Modulation of APP Transport and Processing by the Transmembrane Proteins LRP1, SorLA, SorCS1c, Sortilin, and Calsyntenin. Molecular Neurobiology, 55(7), 5809-5829. doi:10.1007/s12035-017-0806-x
    Elsayed, M., Banasr, M., et al. (2012). Antidepressant effects of fibroblast growth factor-2 in behavioral and cellular models of depression. Biol Psychiatry, 72(4), 258-265. doi:10.1016/j.biopsych.2012.03.003
    Euesden, J., Lewis, C. M., et al. (2015). PRSice: Polygenic Risk Score software. Bioinformatics, 31(9), 1466-1468. doi:10.1093/bioinformatics/btu848
    Fagiolini, A., Frank, E., et al. (2005). Metabolic syndrome in bipolar disorder: findings from the Bipolar Disorder Center for Pennsylvanians. Bipolar Disord, 7(5), 424-430. doi:10.1111/j.1399-5618.2005.00234.x
    Ferreira, M. A., O'Donovan, M. C., et al. (2008). Collaborative genome-wide association analysis supports a role for ANK3 and CACNA1C in bipolar disorder. Nat Genet, 40(9), 1056-1058. doi:10.1038/ng.209
    Flores, C., Manitt, C., et al. (2005). Netrin receptor deficient mice exhibit functional reorganization of dopaminergic systems and do not sensitize to amphetamine. Mol Psychiatry, 10(6), 606-612. doi:10.1038/sj.mp.4001607
    Forlenza, O. V., Aprahamian, I., et al. (2016). Cognitive impairment in late-life bipolar disorder is not associated with Alzheimer's disease pathological signature in the cerebrospinal fluid. Bipolar Disord, 18(1), 63-70. doi:10.1111/bdi.12360
    Frappart, P. O., Lee, Y., et al. (2007). BRCA2 is required for neurogenesis and suppression of medulloblastoma. Embo j, 26(11), 2732-2742. doi:10.1038/sj.emboj.7601703
    Freshour, S. L., Kiwala, S., et al. (2020). Integration of the Drug–Gene Interaction Database (DGIdb 4.0) with open crowdsource efforts. Nucleic Acids Research, 49(D1), D1144-D1151. doi:10.1093/nar/gkaa1084
    Friedewald, W. T., Levy, R. I., et al. (1972). Estimation of the Concentration of Low-Density Lipoprotein Cholesterol in Plasma, Without Use of the Preparative Ultracentrifuge. Clinical Chemistry, 18(6), 499-502. doi:10.1093/clinchem/18.6.499
    Fuller, B. E., Rodriguez, V. L., et al. (2011). Prevalence of liver disease in veterans with bipolar disorder or schizophrenia. Gen Hosp Psychiatry, 33(3), 232-237. doi:10.1016/j.genhosppsych.2011.03.006
    Fullerton, J. M., & Nurnberger, J. I. (2019). Polygenic risk scores in psychiatry: Will they be useful for clinicians? F1000Res, 8. doi:10.12688/f1000research.18491.1
    Fusar-Poli, L., Amerio, A., et al. (2020). Lipid and Glycemic Profiles in Patients with Bipolar Disorder: Cholesterol Levels Are Reduced in Mania. Medicina (Kaunas), 57(1). doi:10.3390/medicina57010028
    Gaspar, H. A., & Breen, G. (2017). Drug enrichment and discovery from schizophrenia genome-wide association results: an analysis and visualisation approach. Sci Rep, 7(1), 12460. doi:10.1038/s41598-017-12325-3
    Ge, S. X., Jung, D., et al. (2020). ShinyGO: a graphical gene-set enrichment tool for animals and plants. Bioinformatics, 36(8), 2628-2629. doi:10.1093/bioinformatics/btz931
    Gehman, L. T., Stoilov, P., et al. (2011). The splicing regulator Rbfox1 (A2BP1) controls neuronal excitation in the mammalian brain. Nat Genet, 43(7), 706-711. doi:10.1038/ng.841
    Ghaemi, S. N., Gilmer, W. S., et al. (2009). A Double-Blind, Placebo-Controlled Pilot Study of Galantamine to Improve Cognitive Dysfunction in Minimally Symptomatic Bipolar Disorder. Journal of Clinical Psychopharmacology, 29(3).
    Ghaemi, S. N., Shields, G. S., et al. (2000). Cholesterol levels in mood disorders: high or low? Bipolar Disord, 2(1), 60-64. doi:10.1034/j.1399-5618.2000.020109.x
    Gill, H., Gill, B., et al. (2020). Antidepressant Medications and Weight Change: A Narrative Review. Obesity (Silver Spring), 28(11), 2064-2072. doi:10.1002/oby.22969
    Gitlin, M. J. (2018). Antidepressants in bipolar depression: an enduring controversy. Int J Bipolar Disord, 6(1), 25. doi:10.1186/s40345-018-0133-9
    Giudici, K. V., de Souto Barreto, P., et al. (2020). Assessment of Plasma Amyloid-β42/40 and Cognitive Decline Among Community-Dwelling Older Adults. JAMA Netw Open, 3(12), e2028634. doi:10.1001/jamanetworkopen.2020.28634
    Giuffrida, M. L., Caraci, F., et al. (2009). Beta-amyloid monomers are neuroprotective. J Neurosci, 29(34), 10582-10587. doi:10.1523/jneurosci.1736-09.2009
    Gould, T. D., & Manji, H. K. (2002). The Wnt Signaling Pathway in Bipolar Disorder. The Neuroscientist, 8(5), 497-511. doi:10.1177/107385802237176
    Grande, I., Berk, M., et al. (2016). Bipolar disorder. Lancet, 387(10027), 1561-1572. doi:10.1016/s0140-6736(15)00241-x
    Green, K. N., Billings, L. M., et al. (2006). Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer's disease. J Neurosci, 26(35), 9047-9056. doi:10.1523/jneurosci.2797-06.2006
    Greenwood, T. A., Lazzeroni, L. C., et al. (2019). Genome-wide Association of Endophenotypes for Schizophrenia From the Consortium on the Genetics of Schizophrenia (COGS) Study. JAMA Psychiatry, 76(12), 1274-1284. doi:10.1001/jamapsychiatry.2019.2850
    Grof, P., Duffy, A., et al. (2002). Is response to prophylactic lithium a familial trait? J Clin Psychiatry, 63(10), 942-947.
    Grover, S., Nehra, R., et al. (2017). Bipolar affective disorder and its impact on various aspects of marital relationship. Ind Psychiatry J, 26(2), 114-120. doi:10.4103/ipj.ipj_15_16
    Grundy, S. M., Cleeman, J. I., et al. (2005). Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation, 112(17), 2735-2752. doi:10.1161/circulationaha.105.169404
    Gubern, C., Camós, S., et al. (2014). Characterization of Gcf2/Lrrfip1 in experimental cerebral ischemia and its role as a modulator of Akt, mTOR and β-catenin signaling pathways. Neuroscience, 268, 48-65. doi:https://doi.org/10.1016/j.neuroscience.2014.02.051
    Haghighi, M., Bajoghli, H., et al. (2013). Assessment of cognitive impairments and seizure characteristics in electroconvulsive therapy with and without sodium valproate in manic patients. Neuropsychobiology, 67(1), 14-24. doi:10.1159/000343490
    Hall, R. D., & Macrides, F. (1983). Olfactory bulbectomy impairs the rat's radial-maze behavior. Physiol Behav, 30(5), 797-803. doi:10.1016/0031-9384(83)90180-4
    Hampel, H., Hardy, J., et al. (2021). The Amyloid-β Pathway in Alzheimer’s Disease. Molecular Psychiatry, 26(10), 5481-5503. doi:10.1038/s41380-021-01249-0
    Harandi, T. F., Taghinasab, M. M., et al. (2017). The correlation of social support with mental health: A meta-analysis. Electron Physician, 9(9), 5212-5222. doi:10.19082/5212
    Harris, M. A., Clark, J., et al. (2004). The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res, 32(Database issue), D258-261. doi:10.1093/nar/gkh036
    He, H., Hu, C., et al. (2020). Trends in the incidence and DALYs of bipolar disorder at global, regional, and national levels: Results from the global burden of Disease Study 2017. J Psychiatr Res, 125, 96-105. doi:10.1016/j.jpsychires.2020.03.015
    Hidese, S., Hattori, K., et al. (2020). Cerebrospinal fluid neuroplasticity-associated protein levels in patients with psychiatric disorders: a multiplex immunoassay study. Transl Psychiatry, 10(1), 161. doi:10.1038/s41398-020-0843-5
    Hilal, S., Ikram, M. A., et al. (2018). C-Reactive Protein, Plasma Amyloid-β Levels, and Their Interaction With Magnetic Resonance Imaging Markers. Stroke, 49(11), 2692-2698. doi:10.1161/strokeaha.118.022317
    Ho, A. M., Coombes, B. J., et al. (2020). Mood-Stabilizing Antiepileptic Treatment Response in Bipolar Disorder: A Genome-Wide Association Study. Clin Pharmacol Ther, 108(6), 1233-1242. doi:10.1002/cpt.1982
    Hsueh, Y. S., Lin, C. Y., et al. (2021). Changes in striatal dopamine transporters in bipolar disorder and valproate treatment. Eur Psychiatry, 64(1), e9. doi:10.1192/j.eurpsy.2021.1
    Huang, M. S., Wang, T. K., et al. (2014). Roles of carbonic anhydrase 8 in neuronal cells and zebrafish. Biochim Biophys Acta, 1840(9), 2829-2842. doi:10.1016/j.bbagen.2014.04.017
    Hui, Q., Jin, Z., et al. (2018). FGF Family: From Drug Development to Clinical Application. Int J Mol Sci, 19(7). doi:10.3390/ijms19071875
    Iosifescu, D. V., Moore, C. M., et al. (2009). Galantamine-ER for cognitive dysfunction in bipolar disorder and correlation with hippocampal neuronal viability: a proof-of-concept study. CNS Neurosci Ther, 15(4), 309-319. doi:10.1111/j.1755-5949.2009.00090.x
    Ito, K., Tatebe, T., et al. (2017). Memantine reduces the production of amyloid-β peptides through modulation of amyloid precursor protein trafficking. Eur J Pharmacol, 798, 16-25. doi:10.1016/j.ejphar.2017.02.001
    Iwatsubo, T., Mann, D. M., et al. (1995). Amyloid beta protein (A beta) deposition: A beta 42(43) precedes A beta 40 in Down syndrome. Ann Neurol, 37(3), 294-299. doi:10.1002/ana.410370305
    Iwatsubo, T., Odaka, A., et al. (1994). Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: evidence that an initially deposited species is A beta 42(43). Neuron, 13(1), 45-53. doi:10.1016/0896-6273(94)90458-8
    Jackson, L., Eldahshan, W., et al. (2018). Within the Brain: The Renin Angiotensin System. Int J Mol Sci, 19(3). doi:10.3390/ijms19030876
    Jakobsson, J., Zetterberg, H., et al. (2013). Altered concentrations of amyloid precursor protein metabolites in the cerebrospinal fluid of patients with bipolar disorder. Neuropsychopharmacology, 38(4), 664-672. doi:10.1038/npp.2012.231
    Janelidze, S., Stomrud, E., et al. (2016). Plasma β-amyloid in Alzheimer's disease and vascular disease. Sci Rep, 6, 26801. doi:10.1038/srep26801
    Jassal, B., Matthews, L., et al. (2020). The reactome pathway knowledgebase. Nucleic Acids Res, 48(D1), D498-d503. doi:10.1093/nar/gkz1031
    Johannessen, C. U., & Johannessen, S. I. (2003). Valproate: past, present, and future. CNS Drug Rev, 9(2), 199-216. doi:10.1111/j.1527-3458.2003.tb00249.x
    Johansson, V., Kuja-Halkola, R., et al. (2019). A population-based heritability estimate of bipolar disorder - In a Swedish twin sample. Psychiatry Res, 278, 180-187. doi:10.1016/j.psychres.2019.06.010
    Jung, G. A., Yoon, J. Y., et al. (2008). Valproic acid induces differentiation and inhibition of proliferation in neural progenitor cells via the beta-catenin-Ras-ERK-p21Cip/WAF1 pathway. BMC Cell Biol, 9, 66. doi:10.1186/1471-2121-9-66
    Kakudo, N., Shimotsuma, A., et al. (2007). Fibroblast growth factor-2 stimulates adipogenic differentiation of human adipose-derived stem cells. Biochemical and Biophysical Research Communications, 359(2), 239-244. doi:https://doi.org/10.1016/j.bbrc.2007.05.070
    Kamath, V., Paksarian, D., et al. (2018). Olfactory processing in bipolar disorder, major depression, and anxiety. Bipolar Disord, 20(6), 547-555. doi:https://doi.org/10.1111/bdi.12625
    Kanehisa, M., & Goto, S. (2000). KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res, 28(1), 27-30. doi:10.1093/nar/28.1.27
    Kao, C. Y., Hsu, Y. C., et al. (2013). The mood stabilizer valproate activates human FGF1 gene promoter through inhibiting HDAC and GSK-3 activities. J Neurochem, 126(1), 4-18. doi:10.1111/jnc.12292
    Karling, P., Maripuu, M., et al. (2016). Association between gastrointestinal symptoms and affectivity in patients with bipolar disorder. World J Gastroenterol, 22(38), 8540-8548. doi:10.3748/wjg.v22.i38.8540
    Kawaguchi, N., Toriyama, K., et al. (1998). De novo adipogenesis in mice at the site of injection of basement membrane and basic fibroblast growth factor. Proceedings of the National Academy of Sciences, 95(3), 1062-1066. doi:10.1073/pnas.95.3.1062
    Kim, J.-M., Stewart, R., et al. (2014). Longitudinal associations between serum cholesterol levels and suicidal ideation in an older Korean population. J Affect Disord, 152-154, 517-521. doi:https://doi.org/10.1016/j.jad.2013.08.008
    Kim, K. K., Adelstein, R. S., et al. (2009). Identification of neuronal nuclei (NeuN) as Fox-3, a new member of the Fox-1 gene family of splicing factors. J Biol Chem, 284(45), 31052-31061. doi:10.1074/jbc.M109.052969
    Kircher, M., Witten, D. M., et al. (2014). A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet, 46(3), 310-315. doi:10.1038/ng.2892
    Knecht, A. K., & Bronner-Fraser, M. (2002). Induction of the neural crest: a multigene process. Nat Rev Genet, 3(6), 453-461. doi:10.1038/nrg819
    Knott, S., Forty, L., et al. (2015). Epilepsy and bipolar disorder. Epilepsy & Behavior, 52, 267-274. doi:https://doi.org/10.1016/j.yebeh.2015.07.003
    Ko, H. M., Jin, Y., et al. (2018). Dual mechanisms for the regulation of brain-derived neurotrophic factor by valproic acid in neural progenitor cells. Korean J Physiol Pharmacol, 22(6), 679-688. doi:10.4196/kjpp.2018.22.6.679
    Koleck, T. A., Bender, C. M., et al. (2014). Apolipoprotein E genotype and cognitive function in postmenopausal women with early-stage breast cancer. Oncol Nurs Forum, 41(6), E313-325. doi:10.1188/14.Onf.E313-e325
    Koukopoulos, A., Serra, G., et al. (2012). The sustained mood-stabilizing effect of memantine in the management of treatment resistant bipolar disorders: findings from a 12-month naturalistic trial. J Affect Disord, 136(1-2), 163-166. doi:10.1016/j.jad.2011.09.040
    Koyama, A., Okereke, O. I., et al. (2012). Plasma amyloid-β as a predictor of dementia and cognitive decline: a systematic review and meta-analysis. Arch Neurol, 69(7), 824-831. doi:10.1001/archneurol.2011.1841
    Kuhn, R. M., Haussler, D., et al. (2013). The UCSC genome browser and associated tools. Brief Bioinform, 14(2), 144-161. doi:10.1093/bib/bbs038
    Kupferberg, A., Bicks, L., et al. (2016). Social functioning in major depressive disorder. Neuroscience & Biobehavioral Reviews, 69, 313-332. doi:https://doi.org/10.1016/j.neubiorev.2016.07.002
    Kusumastuti, K., & Jaeri, S. (2020). The effect of long-term valproic acid treatment in the level of total cholesterol among adult. Indian J Pharmacol, 52(2), 134-137. doi:10.4103/ijp.IJP_655_18
    Lövheim, H., Elgh, F., et al. (2017). Plasma concentrations of free amyloid β cannot predict the development of Alzheimer's disease. Alzheimers Dement, 13(7), 778-782. doi:10.1016/j.jalz.2016.12.004
    L. Pai, H.-C. W., R.-B. Lu, M.-L. Kuo. (1985). The questionnaire of life stress cognition of adults-revised (full text in Chinese). Chinese Journal of Mental Health (Taipei), 3, 195-205.
    Lee, J., Baek, J. H., et al. (2020). Defining phenotypes of long-term lithium and valproate response, including combination therapy: a modified application of the Alda scale in patients with bipolar disorders. Int J Bipolar Disord, 8(1), 36. doi:10.1186/s40345-020-00199-w
    Lee, S. Y., Chen, S. L., et al. (2014). The effects of add-on low-dose memantine on cytokine levels in bipolar II depression: a 12-week double-blind, randomized controlled trial. J Clin Psychopharmacol, 34(3), 337-343. doi:10.1097/jcp.0000000000000109
    Li, H. J., Qu, N., et al. (2020). Further confirmation of netrin 1 receptor (DCC) as a depression risk gene via integrations of multi-omics data. Transl Psychiatry, 10(1), 98. doi:10.1038/s41398-020-0777-y
    Li, H. J., Zhang, C., et al. (2021). Novel Risk Loci Associated With Genetic Risk for Bipolar Disorder Among Han Chinese Individuals: A Genome-Wide Association Study and Meta-analysis. JAMA Psychiatry, 78(3), 320-330. doi:10.1001/jamapsychiatry.2020.3738
    Li, W., Ye, A., et al. (2020). Protective Mechanism and Treatment of Neurogenesis in Cerebral Ischemia. Neurochem Res, 45(10), 2258-2277. doi:10.1007/s11064-020-03092-1
    Li, W., Zhang, C. Y., et al. (2021). Identification of a Risk Locus at 7p22.3 for Schizophrenia and Bipolar Disorder in East Asian Populations. Front Genet, 12, 789512. doi:10.3389/fgene.2021.789512
    Li, X., Bijur, G. N., et al. (2002). Glycogen synthase kinase-3beta, mood stabilizers, and neuroprotection. Bipolar Disord, 4(2), 137-144. doi:10.1034/j.1399-5618.2002.40201.x
    Li, Z., Li, D., et al. (2022). Characterizing the polygenic overlaps of bipolar disorder subtypes with schizophrenia and major depressive disorder. J Affect Disord, 309, 242-251. doi:10.1016/j.jad.2022.04.097
    Lin, G. M., Chen, Y. J., et al. (2013). Cancer incidence in patients with schizophrenia or bipolar disorder: a nationwide population-based study in Taiwan, 1997-2009. Schizophr Bull, 39(2), 407-416. doi:10.1093/schbul/sbr162
    Lin, N., Ye, X., et al. (1999). Social support and depressed mood: a structural analysis. J Health Soc Behav, 40(4), 344-359.
    Lin, W. Y., Liu, Y. L., et al. (2020). Active Cigarette Smoking Is Associated With an Exacerbation of Genetic Susceptibility to Diabetes. Diabetes, 69(12), 2819-2829. doi:10.2337/db20-0156
    Lin, Y. S., Wang, H. Y., et al. (2016). Neuronal Splicing Regulator RBFOX3 (NeuN) Regulates Adult Hippocampal Neurogenesis and Synaptogenesis. PLoS One, 11(10), e0164164. doi:10.1371/journal.pone.0164164
    Liu, C.-H., Kang, E. Y.-C., et al. (2020). Association of ocular diseases with schizophrenia, bipolar disorder, and major depressive disorder: a retrospective case-control, population-based study. BMC Psychiatry, 20(1), 486. doi:10.1186/s12888-020-02881-w
    Lourida, I., Hannon, E., et al. (2019). Association of Lifestyle and Genetic Risk With Incidence of Dementia. JAMA, 322(5), 430-437. doi:10.1001/jama.2019.9879
    Lu, R. B., Wang, T. Y., et al. (2021). Add-on memantine may improve cognitive functions and attenuate inflammation in middle- to old-aged bipolar II disorder patients. J Affect Disord, 279, 229-238. doi:10.1016/j.jad.2020.10.003
    Lu, Y., Sun, X.-D., et al. (2014). Maintenance of GABAergic Activity by Neuregulin 1-ErbB4 in Amygdala for Fear Memory. Neuron, 84(4), 835-846. doi:https://doi.org/10.1016/j.neuron.2014.09.029
    Lumia, A. R., Teicher, M. H., et al. (1992). Olfactory bulbectomy as a model for agitated hyposerotonergic depression. Brain Res, 587(2), 181-185. doi:10.1016/0006-8993(92)90995-l
    Malhi, G. S., Adams, D., et al. (2009). The management of individuals with bipolar disorder: a review of the evidence and its integration into clinical practice. Drugs, 69(15), 2063-2101. doi:10.2165/11318850-000000000-00000
    Mallei, A., Shi, B., et al. (2002). Antidepressant treatments induce the expression of basic fibroblast growth factor in cortical and hippocampal neurons. Mol Pharmacol, 61(5), 1017-1024. doi:10.1124/mol.61.5.1017
    Mallolas, J., Hurtado, O., et al. (2006). A polymorphism in the EAAT2 promoter is associated with higher glutamate concentrations and higher frequency of progressing stroke. J Exp Med, 203(3), 711-717. doi:10.1084/jem.20051979
    Manchia, M., Adli, M., et al. (2013). Assessment of Response to Lithium Maintenance Treatment in Bipolar Disorder: A Consortium on Lithium Genetics (ConLiGen) Report. PLoS One, 8(6), e65636. doi:10.1371/journal.pone.0065636
    Mano, T., Sato, K., et al. (2021). Peripheral Blood BRCA1 Methylation Positively Correlates with Major Alzheimer's Disease Risk Factors. J Prev Alzheimers Dis, 8(4), 477-482. doi:10.14283/jpad.2021.31
    Martin, C. K., Han, H., et al. (2009). Effect of valproic acid on body weight, food intake, physical activity and hormones: results of a randomized controlled trial. J Psychopharmacol, 23(7), 814-825. doi:10.1177/0269881108091595
    Martinowich, K., Schloesser, R. J., et al. (2009). Bipolar disorder: from genes to behavior pathways. J Clin Invest, 119(4), 726-736. doi:10.1172/jci37703
    Marx, S., Dal Maso, T., et al. (2020). Transmembrane (TMEM) protein family members: Poorly characterized even if essential for the metastatic process. Semin Cancer Biol, 60, 96-106. doi:10.1016/j.semcancer.2019.08.018
    Mason, I. (2007). Initiation to end point: the multiple roles of fibroblast growth factors in neural development. Nat Rev Neurosci, 8(8), 583-596. doi:10.1038/nrn2189
    Matthews, D. R., Hosker, J. P., et al. (1985). Homeostasis model assessment: insulin resistance and β-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia, 28(7), 412-419. doi:10.1007/BF00280883
    McElroy, S. L., Suppes, T., et al. (2005). Open-label adjunctive zonisamide in the treatment of bipolar disorders: a prospective trial. J Clin Psychiatry, 66(5), 617-624. doi:10.4088/jcp.v66n0512
    McKnight, R. F., Adida, M., et al. (2012). Lithium toxicity profile: a systematic review and meta-analysis. Lancet, 379(9817), 721-728. doi:10.1016/s0140-6736(11)61516-x
    McLaren, W., Gil, L., et al. (2016). The Ensembl Variant Effect Predictor. Genome Biol, 17(1), 122. doi:10.1186/s13059-016-0974-4
    Medici, C. R., Videbech, P., et al. (2015). Mortality and secular trend in the incidence of bipolar disorder. J Affect Disord, 183, 39-44. doi:10.1016/j.jad.2015.04.032
    Mei, L., & Nave, K. A. (2014). Neuregulin-ERBB signaling in the nervous system and neuropsychiatric diseases. Neuron, 83(1), 27-49. doi:10.1016/j.neuron.2014.06.007
    Meng, Q., Wang, L., et al. (2020). Integrative analyses prioritize GNL3 as a risk gene for bipolar disorder. Mol Psychiatry, 25(11), 2672-2684. doi:10.1038/s41380-020-00866-5
    Metti, A. L., Cauley, J. A., et al. (2013). Plasma beta amyloid level and depression in older adults. J Gerontol A Biol Sci Med Sci, 68(1), 74-79. doi:10.1093/gerona/gls093
    Modabbernia, A., Taslimi, S., et al. (2013). Cytokine alterations in bipolar disorder: a meta-analysis of 30 studies. Biol Psychiatry, 74(1), 15-25. doi:10.1016/j.biopsych.2013.01.007
    Moechars, D., Dewachter, I., et al. (1999). Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain. J Biol Chem, 274(10), 6483-6492. doi:10.1074/jbc.274.10.6483
    Mula, M., Marotta, A. E., et al. (2010). Epilepsy and bipolar disorders. Expert Rev Neurother, 10(1), 13-23. doi:10.1586/ern.09.139
    Munkholm, K., Braüner, J. V., et al. (2013). Cytokines in bipolar disorder vs. healthy control subjects: a systematic review and meta-analysis. J Psychiatr Res, 47(9), 1119-1133. doi:10.1016/j.jpsychires.2013.05.018
    Musliner, K. L., Krebs, M. D., et al. (2020). Polygenic Risk and Progression to Bipolar or Psychotic Disorders Among Individuals Diagnosed With Unipolar Depression in Early Life. Am J Psychiatry, 177(10), 936-943. doi:10.1176/appi.ajp.2020.19111195
    Nascimento, K. K., Silva, K. P., et al. (2015). Plasma and cerebrospinal fluid amyloid-β levels in late-life depression: A systematic review and meta-analysis. J Psychiatr Res, 69, 35-41. doi:10.1016/j.jpsychires.2015.07.024
    National Center for Biotechnology Information. (2021). ADAMTS2 ADAM metallopeptidase with thrombospondin type 1 motif 2 [ Homo sapiens (human) ]. Retrieved from https://www.ncbi.nlm.nih.gov/gene/
    Nisha, Y., Bobby, Z., et al. (2018). Biochemical derangements related to metabolic syndrome in epileptic patients on treatment with valproic acid. Seizure, 60, 57-60. doi:10.1016/j.seizure.2018.06.003
    Nishi, A., Numata, S., et al. (2014). Meta-analyses of blood homocysteine levels for gender and genetic association studies of the MTHFR C677T polymorphism in schizophrenia. Schizophrenia Bulletin, 40(5), 1154-1163. doi:10.1093/schbul/sbt154
    Noble, J. M., Manly, J. J., et al. (2010). Association of C-reactive protein with cognitive impairment. Arch Neurol, 67(1), 87-92. doi:10.1001/archneurol.2009.308
    Nugent, M. A., & Iozzo, R. V. (2000). Fibroblast growth factor-2. Int J Biochem Cell Biol, 32(2), 115-120. doi:10.1016/s1357-2725(99)00123-5
    Nunes, Q. M., Li, Y., et al. (2016). Fibroblast growth factors as tissue repair and regeneration therapeutics. PeerJ, 4, e1535. doi:10.7717/peerj.1535
    Oprea, T. I., Bologa, C. G., et al. (2018). Unexplored therapeutic opportunities in the human genome. Nat Rev Drug Discov, 17(5), 317-332. doi:10.1038/nrd.2018.14
    Ostacher, M. J., Iosifescu, D. V., et al. (2014). Pilot investigation of isradipine in the treatment of bipolar depression motivated by genome-wide association. Bipolar Disord, 16(2), 199-203. doi:10.1111/bdi.12143
    Ouchi, N., Kihara, S., et al. (2000). Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation, 102(11), 1296-1301. doi:10.1161/01.cir.102.11.1296
    Ouchi, N., Parker, J. L., et al. (2011). Adipokines in inflammation and metabolic disease. Nat Rev Immunol, 11(2), 85-97. doi:10.1038/nri2921
    Ozaki, M. (2001). Neuregulins and the shaping of synapses. Neuroscientist, 7(2), 146-154. doi:10.1177/107385840100700209
    Pal Singh, G. (2008). A double-blind comparative study of clinical efficacy of verapamil versus lithium in acute mania. Int J Psychiatry Clin Pract, 12(4), 303-308. doi:10.1080/13651500802209670
    Pandey, G. N., Dwivedi, Y., et al. (2002). Protein Kinase C and Phospholipase C Activity and Expression of Their Specific Isozymes Is Decreased and Expression of MARCKS Is Increased in Platelets of Bipolar but Not in Unipolar Patients. Neuropsychopharmacology, 26(2), 216-228. doi:10.1016/S0893-133X(01)00327-X
    Parsons, C. G., Danysz, W., et al. (1999). Memantine is a clinically well tolerated N-methyl-D-aspartate (NMDA) receptor antagonist--a review of preclinical data. Neuropharmacology, 38(6), 735-767. doi:10.1016/s0028-3908(99)00019-2
    Patel, K., Scrimieri, F., et al. (2013). FAM190A Deficiency Creates a Cell Division Defect. The American Journal of Pathology, 183(1), 296-303. doi:https://doi.org/10.1016/j.ajpath.2013.03.020
    Paul, P., Nadella, R. K., et al. (2021). Association study of BDNF Val66Met gene polymorphism with bipolar disorder and lithium treatment response in Indian population. J Psychopharmacol, 2698811211032609. doi:10.1177/02698811211032609
    Peng, H., Wu, X., et al. (2021). Genetically predicted bipolar disorder is causally associated with an increased risk of breast cancer: a two-sample Mendelian randomization analysis. Ann Transl Med, 9(5), 401. doi:10.21037/atm-20-5372
    Peng, X., Xu, Z., et al. (2020). Association of plasma β-amyloid 40 and 42 concentration with type 2 diabetes among Chinese adults. Diabetologia, 63(5), 954-963. doi:10.1007/s00125-020-05102-x
    Pereira, A. C., Oliveira, J., et al. (2021). Inflammation in Bipolar Disorder (BD): Identification of new therapeutic targets. Pharmacol Res, 163, 105325. doi:10.1016/j.phrs.2020.105325
    Perez-Torrado, R., Yamada, D., et al. (2006). Born to bind: the BTB protein-protein interaction domain. Bioessays, 28(12), 1194-1202. doi:10.1002/bies.20500
    Piccinni, A., Origlia, N., et al. (2012). Plasma β-amyloid peptides levels: a pilot study in bipolar depressed patients. J Affect Disord, 138(1-2), 160-164. doi:10.1016/j.jad.2011.12.042
    Piccinni, A., Veltri, A., et al. (2013). Plasma amyloid-β levels in drug-resistant bipolar depressed patients receiving electroconvulsive therapy. Neuropsychobiology, 67(4), 185-191. doi:10.1159/000347082
    Piccolo, M., Belleau, E. L., et al. (2022). Alterations in resting-state functional activity and connectivity for major depressive disorder appetite and weight disturbance phenotypes. Psychol Med, 1-11. doi:10.1017/s0033291722001398
    Pickell, Z., Williams, A. M., et al. (2020). Histone Deacetylase Inhibitors: A Novel Strategy for Neuroprotection and Cardioprotection Following Ischemia/Reperfusion Injury. J Am Heart Assoc, 9(11), e016349. doi:10.1161/jaha.120.016349
    Pickup, J. C. (2004). Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care, 27(3), 813-823. doi:10.2337/diacare.27.3.813
    Pigott, K., Galizia, I., et al. (2016). Topiramate for acute affective episodes in bipolar disorder in adults. Cochrane Database Syst Rev, 9(9), Cd003384. doi:10.1002/14651858.CD003384.pub3
    Post, R. M., Altshuler, L. L., et al. (2010). Complexity of pharmacologic treatment required for sustained improvement in outpatients with bipolar disorder. J Clin Psychiatry, 71(9), 1176-1186; quiz 1252-1173. doi:10.4088/JCP.08m04811yel
    Post, R. M., Altshuler, L. L., et al. (2021). 25 Years of the International Bipolar Collaborative Network (BCN). Int J Bipolar Disord, 9(1), 13. doi:10.1186/s40345-020-00218-w
    Prashad, S., & Gopal, P. P. (2021). RNA-binding proteins in neurological development and disease. RNA Biol, 18(7), 972-987. doi:10.1080/15476286.2020.1809186
    Pruitt, K. D., Brown, G. R., et al. (2014). RefSeq: an update on mammalian reference sequences. Nucleic Acids Res, 42(Database issue), D756-763. doi:10.1093/nar/gkt1114
    Psychiatric Genomics Consortium. (2014). Biological insights from 108 schizophrenia-associated genetic loci. Nature, 511(7510), 421-427. doi:10.1038/nature13595
    Purcell, S. M., Wray, N. R., et al. (2009). Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature, 460(7256), 748-752. doi:10.1038/nature08185
    Qing, H., He, G., et al. (2008). Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer's disease mouse models. J Exp Med, 205(12), 2781-2789. doi:10.1084/jem.20081588
    Raghavan, N. S., Dumitrescu, L., et al. (2020). Association Between Common Variants in RBFOX1, an RNA-Binding Protein, and Brain Amyloidosis in Early and Preclinical Alzheimer Disease. JAMA Neurol, 77(10), 1288-1298. doi:10.1001/jamaneurol.2020.1760
    Rahe, R. H. (1975). Epidemiological studies of life change and illness. Int J Psychiatry Med, 6(1-2), 133-146. doi:10.2190/jgrj-kumg-gkka-hbge
    Rahman, N., Seal, S., et al. (2007). PALB2, which encodes a BRCA2-interacting protein, is a breast cancer susceptibility gene. Nature Genetics, 39(2), 165-167. doi:10.1038/ng1959
    Reay, W. R., Atkins, J. R., et al. (2020). Pharmacological enrichment of polygenic risk for precision medicine in complex disorders. Sci Rep, 10(1), 879. doi:10.1038/s41598-020-57795-0
    Reay, W. R., Atkins, J. R., et al. (2020). Pharmacological enrichment of polygenic risk for precision medicine in complex disorders. Sci Rep, 10(1), 879. doi:10.1038/s41598-020-57795-0
    Reay, W. R., & Cairns, M. J. (2021). Advancing the use of genome-wide association studies for drug repurposing. Nature Reviews Genetics, 22(10), 658-671. doi:10.1038/s41576-021-00387-z
    Reay, W. R., El Shair, S. I., et al. (2021). Genetic association and causal inference converge on hyperglycaemia as a modifiable factor to improve lung function. Elife, 10. doi:10.7554/eLife.63115
    Rees, E., Walters, J. T. R., et al. (2014). CNV analysis in a large schizophrenia sample implicates deletions at 16p12.1 and SLC1A1 and duplications at 1p36.33 and CGNL1. Human Molecular Genetics, 23(6), 1669-1676. doi:10.1093/hmg/ddt540
    Regier, D. A., Farmer, M. E., et al. (1990). Comorbidity of Mental Disorders With Alcohol and Other Drug Abuse: Results From the Epidemiologic Catchment Area (ECA) Study. JAMA, 264(19), 2511-2518. doi:10.1001/jama.1990.03450190043026
    Reimand, J., Isserlin, R., et al. (2019). Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat Protoc, 14(2), 482-517. doi:10.1038/s41596-018-0103-9
    Rice, H. C., de Malmazet, D., et al. (2019). Secreted amyloid-β precursor protein functions as a GABA(B)R1a ligand to modulate synaptic transmission. Science, 363(6423). doi:10.1126/science.aao4827
    Rosenblat, J. D., & McIntyre, R. S. (2017). Bipolar Disorder and Immune Dysfunction: Epidemiological Findings, Proposed Pathophysiology and Clinical Implications. Brain Sci, 7(11). doi:10.3390/brainsci7110144
    Rowland, T., Perry, B. I., et al. (2018). Neurotrophins, cytokines, oxidative stress mediators and mood state in bipolar disorder: systematic review and meta-analyses. Br J Psychiatry, 213(3), 514-525. doi:10.1192/bjp.2018.144
    Roy Chengappa, K. N., Schwarzman, L. K., et al. (2006). Adjunctive topiramate therapy in patients receiving a mood stabilizer for bipolar I disorder: a randomized, placebo-controlled trial. J Clin Psychiatry, 67(11), 1698-1706. doi:10.4088/jcp.v67n1105
    Ru, P., Hu, P., et al. (2016). Feedback Loop Regulation of SCAP/SREBP-1 by miR-29 Modulates EGFR Signaling-Driven Glioblastoma Growth. Cell Rep, 16(6), 1527-1535. doi:10.1016/j.celrep.2016.07.017
    Ruderfer, D. M., Ripke, S., et al. (2018). Genomic Dissection of Bipolar Disorder and Schizophrenia, Including 28 Subphenotypes. Cell, 173(7), 1705-1715.e1716. doi:https://doi.org/10.1016/j.cell.2018.05.046
    Russell, D. W. (1996). UCLA Loneliness Scale (Version 3): Reliability, Validity, and Factor Structure. Journal of Personality Assessment, 66(1), 20-40. doi:10.1207/s15327752jpa6601_2
    Sakai, Y., Hanafusa, H., et al. (2021). BRCA1-BARD1 Regulates Axon Regeneration in Concert with the Gqα-DAG Signaling Network. J Neurosci, 41(13), 2842-2853. doi:10.1523/jneurosci.1806-20.2021
    Salmaso, N., Stevens, H. E., et al. (2016). Fibroblast Growth Factor 2 Modulates Hypothalamic Pituitary Axis Activity and Anxiety Behavior Through Glucocorticoid Receptors. Biol Psychiatry, 80(6), 479-489. doi:10.1016/j.biopsych.2016.02.026
    Sanches, M., Colpo, G. D., et al. (2021). Decreased Plasma Levels of Angiotensin-Converting Enzyme Among Patients With Bipolar Disorder. Front Neurosci, 15, 617888. doi:10.3389/fnins.2021.617888
    Sanofi-aventis. (2020). Depakine® Chrono 500 mg [prescribing information]. Gentilly, France.
    Schulze, T. G., Detera-Wadleigh, S. D., et al. (2009). Two variants in Ankyrin 3 (ANK3) are independent genetic risk factors for bipolar disorder. Mol Psychiatry, 14(5), 487-491. doi:10.1038/mp.2008.134
    Seida, A., Wada, J., et al. (2003). Serum bFGF levels are reduced in Japanese overweight men and restored by a 6-month exercise education. International Journal of Obesity, 27(11), 1325-1331. doi:10.1038/sj.ijo.0802408
    Semenova, A. A., Kharitonova, E. V., et al. (2021). Valproic Acid Increases the Level of Expression of BDNF, GDNF and THEIR Receptors in the Embryonic Brain of Mice and Inducts the Stereotype Behavior. Cell and Tissue Biology, 15(6), 563-567. doi:10.1134/S1990519X21060092
    Shamsaei, F., Yadollahifar, S., et al. (2020). [Not Available]. Sleep Sci, 13(1), 65-69. doi:10.5935/1984-0063.20190135
    Sharma, K., Singh, J., et al. (2018). MeCP2 in central nervous system glial cells: current updates. Acta Neurobiol Exp (Wars), 78(1), 30-40.
    Shimizu, T., Pastuhov, S. I., et al. (2018). The C. elegans BRCA2-ALP/Enigma Complex Regulates Axon Regeneration via a Rho GTPase-ROCK-MLC Phosphorylation Pathway. Cell Rep, 24(7), 1880-1889. doi:10.1016/j.celrep.2018.07.049
    Sidor, M. M., & Macqueen, G. M. (2011). Antidepressants for the acute treatment of bipolar depression: a systematic review and meta-analysis. J Clin Psychiatry, 72(2), 156-167. doi:10.4088/JCP.09r05385gre
    Silverstone, P. H., & Birkett, L. (2000). Diltiazem as augmentation therapy in patients with treatment-resistant bipolar disorder: a retrospective study. J Psychiatry Neurosci, 25(3), 276-280.
    Simard, S., Shail, P., et al. (2018). Fibroblast growth factor 2 is necessary for the antidepressant effects of fluoxetine. PLoS One, 13(10), e0204980. doi:10.1371/journal.pone.0204980
    Simjanoski, M. (2021). Cognitive impairment in bipolar disorder in comparison to mild cognitive impairment and dementia: a systematic review. Trends Psychiatry Psychother. doi:10.47626/2237-6089-2021-0300
    Sjöholm, A., & Nyström, T. (2005). Endothelial inflammation in insulin resistance. Lancet, 365(9459), 610-612. doi:10.1016/s0140-6736(05)17912-4
    Smith, G. D., & Ebrahim, S. (2003). 'Mendelian randomization': can genetic epidemiology contribute to understanding environmental determinants of disease? Int J Epidemiol, 32(1), 1-22. doi:10.1093/ije/dyg070
    Smoller, J. W., & Finn, C. T. (2003). Family, twin, and adoption studies of bipolar disorder. Am J Med Genet C Semin Med Genet, 123c(1), 48-58. doi:10.1002/ajmg.c.20013
    Song, C., & Leonard, B. E. (2005). The olfactory bulbectomised rat as a model of depression. Neurosci Biobehav Rev, 29(4-5), 627-647. doi:10.1016/j.neubiorev.2005.03.010
    Stacey, D., Schubert, K. O., et al. (2018). A gene co-expression module implicating the mitochondrial electron transport chain is associated with long-term response to lithium treatment in bipolar affective disorder. Translational psychiatry, 8(1), 183-183. doi:10.1038/s41398-018-0237-0
    Stahl, E. A., Breen, G., et al. (2019). Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat Genet, 51(5), 793-803. doi:10.1038/s41588-019-0397-8
    Stahl, E. A., Breen, G., et al. (2019). Genome-wide association study identifies 30 loci associated with bipolar disorder. Nature Genetics, 51(5), 793-803. doi:10.1038/s41588-019-0397-8
    Stapp, E. K., Paksarian, D., et al. (2022). Mood and anxiety profiles differentially associate with physical conditions in US adolescents. J Affect Disord, 299, 22-30. doi:10.1016/j.jad.2021.11.056
    Steen, N. E., Aas, M., et al. (2016). Serum concentrations of mood stabilizers are associated with memory, but not other cognitive domains in psychosis spectrum disorders; explorative analyses in a naturalistic setting. Int J Bipolar Disord, 4(1), 24. doi:10.1186/s40345-016-0067-z
    Stein, E., Zou, Y., et al. (2001). Binding of DCC by netrin-1 to mediate axon guidance independent of adenosine A2B receptor activation. Science, 291(5510), 1976-1982. doi:10.1126/science.1059391
    Stelzer, G., Rosen, N., et al. (2016). The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Current Protocols in Bioinformatics, 54(1), 1.30.31-31.30.33. doi:https://doi.org/10.1002/cpbi.5
    Stentzel, U., van den Berg, N., et al. (2021). Telemedical care and quality of life in patients with schizophrenia and bipolar disorder: results of a randomized controlled trial. BMC Psychiatry, 21(1), 318. doi:10.1186/s12888-021-03318-8
    Sturchio, A., Dwivedi, A. K., et al. (2021). High cerebrospinal amyloid-β 42 is associated with normal cognition in individuals with brain amyloidosis. EClinicalMedicine, 38, 100988. doi:10.1016/j.eclinm.2021.100988
    Stygelbout, V., Leroy, K., et al. (2014). Inositol trisphosphate 3-kinase B is increased in human Alzheimer brain and exacerbates mouse Alzheimer pathology. Brain, 137(Pt 2), 537-552. doi:10.1093/brain/awt344
    Sucksdorff, D., Brown, A. S., et al. (2015). Parental and comorbid epilepsy in persons with bipolar disorder. J Affect Disord, 188, 107-111. doi:10.1016/j.jad.2015.08.051
    Sugaya, L., Hasin, D. S., et al. (2012). Child physical abuse and adult mental health: a national study. J Trauma Stress, 25(4), 384-392. doi:10.1002/jts.21719
    Sullivan, K. J., Blackshear, C., et al. (2021). Association of Midlife Plasma Amyloid-β Levels With Cognitive Impairment in Late Life: The ARIC Neurocognitive Study. Neurology. doi:10.1212/wnl.0000000000012482
    Sunderland, T., Linker, G., et al. (2003). Decreased β-Amyloid1-42 and Increased Tau Levels in Cerebrospinal Fluid of Patients With Alzheimer Disease. JAMA, 289(16), 2094-2103. doi:10.1001/jama.289.16.2094
    Swann, A. C. (2010). The strong relationship between bipolar and substance-use disorder. Annals of the New York Academy of Sciences, 1187(1), 276-293. doi:https://doi.org/10.1111/j.1749-6632.2009.05146.x
    Swanson, K. V., Deng, M., et al. (2019). The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol, 19(8), 477-489. doi:10.1038/s41577-019-0165-0
    Szklarczyk, D., Gable, A. L., et al. (2021). The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res, 49(D1), D605-d612. doi:10.1093/nar/gkaa1074
    Tapia-Rojas, C., Burgos, P. V., et al. (2016). Inhibition of Wnt signaling induces amyloidogenic processing of amyloid precursor protein and the production and aggregation of Amyloid-β (Aβ)(42) peptides. J Neurochem, 139(6), 1175-1191. doi:10.1111/jnc.13873
    Tegethoff, M., Belardi, A., et al. (2015). Association Between Mental Disorders and Physical Diseases in Adolescents From a Nationally Representative Cohort. Psychosomatic Medicine, 77(3).
    Tesli, M., Athanasiu, L., et al. (2010). Association analysis of PALB2 and BRCA2 in bipolar disorder and schizophrenia in a scandinavian case–control sample. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 153B(7), 1276-1282. doi:https://doi.org/10.1002/ajmg.b.31098
    Thomas, D. C., Lawlor, D. A., et al. (2007). Re: Estimation of bias in nongenetic observational studies using "Mendelian triangulation" by Bautista et al. Annals of Epidemiology, 17(7), 511-513. doi:10.1016/j.annepidem.2006.12.005
    Torkamani, A., Wineinger, N. E., et al. (2018). The personal and clinical utility of polygenic risk scores. Nat Rev Genet, 19(9), 581-590. doi:10.1038/s41576-018-0018-x
    Torres, I. J., Boudreau, V. G., et al. (2007). Neuropsychological functioning in euthymic bipolar disorder: a meta-analysis. Acta Psychiatr Scand Suppl(434), 17-26. doi:10.1111/j.1600-0447.2007.01055.x
    Tournikioti, K., Ferentinos, P., et al. (2017). Clinical and treatment-related predictors of cognition in bipolar disorder: focus on visual paired associative learning. Eur Arch Psychiatry Clin Neurosci, 267(7), 661-669. doi:10.1007/s00406-016-0743-0
    Tsitsipa, E., & Fountoulakis, K. N. (2015). The neurocognitive functioning in bipolar disorder: a systematic review of data. Ann Gen Psychiatry, 14, 42. doi:10.1186/s12991-015-0081-z
    Turner, C. A., Calvo, N., et al. (2008). The fibroblast growth factor system is downregulated following social defeat. Neurosci Lett, 430(2), 147-150. doi:10.1016/j.neulet.2007.10.041
    Turner, C. A., Clinton, S. M., et al. (2011). Fibroblast growth factor-2 (FGF2) augmentation early in life alters hippocampal development and rescues the anxiety phenotype in vulnerable animals. Proc Natl Acad Sci U S A, 108(19), 8021-8025. doi:10.1073/pnas.1103732108
    Turner, C. A., Watson, S. J., et al. (2012). The fibroblast growth factor family: neuromodulation of affective behavior. Neuron, 76(1), 160-174. doi:10.1016/j.neuron.2012.08.037
    Vagnoni, A., Perkinton, M. S., et al. (2012). Calsyntenin-1 mediates axonal transport of the amyloid precursor protein and regulates Aβ production. Hum Mol Genet, 21(13), 2845-2854. doi:10.1093/hmg/dds109
    Verma, S. S., de Andrade, M., et al. (2014). Imputation and quality control steps for combining multiple genome-wide datasets. Front Genet, 5, 370. doi:10.3389/fgene.2014.00370
    Verrotti, A., Trotta, D., et al. (2002). Valproate-induced hyperammonemic encephalopathy. Metab Brain Dis, 17(4), 367-373. doi:10.1023/a:1021918104127
    Vieta, E., & Rosa, A. R. (2007). Evolving trends in the long-term treatment of bipolar disorder. World J Biol Psychiatry, 8(1), 4-11. doi:10.1080/15622970601083280
    Visscher, P. M., Wray, N. R., et al. (2017). 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet, 101(1), 5-22. doi:10.1016/j.ajhg.2017.06.005
    Vosberg, D. E., Zhang, Y., et al. (2018). Mesocorticolimbic Connectivity and Volumetric Alterations in DCC Mutation Carriers. J Neurosci, 38(20), 4655-4665. doi:10.1523/jneurosci.3251-17.2018
    Vuong, C. K., Wei, W., et al. (2018). Rbfox1 Regulates Synaptic Transmission through the Inhibitory Neuron-Specific vSNARE Vamp1. Neuron, 98(1), 127-141.e127. doi:10.1016/j.neuron.2018.03.008
    Wainschtein, P., Jain, D., et al. (2021). Recovery of trait heritability from whole genome sequence data. bioRxiv, 588020. doi:10.1101/588020
    Wakabayashi, T., Yamaguchi, K., et al. (2019). Differential effects of diet- and genetically-induced brain insulin resistance on amyloid pathology in a mouse model of Alzheimer's disease. Mol Neurodegener, 14(1), 15. doi:10.1186/s13024-019-0315-7
    Walsh, D. M., Klyubin, I., et al. (2002). Amyloid-beta oligomers: their production, toxicity and therapeutic inhibition. Biochem Soc Trans, 30(4), 552-557. doi:10.1042/bst0300552
    Wang, W. Y., Tan, M. S., et al. (2015). Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease. Ann Transl Med, 3(10), 136. doi:10.3978/j.issn.2305-5839.2015.03.49
    Warde-Farley, D., Donaldson, S. L., et al. (2010). The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Research, 38(suppl_2), W214-W220. doi:10.1093/nar/gkq537
    Watanabe, K., Taskesen, E., et al. (2017). Functional mapping and annotation of genetic associations with FUMA. Nature Communications, 8(1), 1826. doi:10.1038/s41467-017-01261-5
    White, M. A., Whisenhunt, B. L., et al. (2002). Development and Validation of the Food-Craving Inventory. Obesity Research, 10(2), 107-114. doi:https://doi.org/10.1038/oby.2002.17
    Williams, L. J., Pasco, J. A., et al. (2016). Angiotensin Converting Enzyme Inhibitors and Risk of Mood Disorders. Psychother Psychosom, 85(4), 250-252. doi:10.1159/000444646
    Wu, C. K., Tseng, P. T., et al. (2016). Significantly higher peripheral fibroblast growth factor-2 levels in patients with major depressive disorder: A preliminary meta-analysis under MOOSE guidelines. Medicine (Baltimore), 95(33), e4563. doi:10.1097/md.0000000000004563
    Wu, L. S.-H., Huang, M.-C., et al. (2021). Genome-wide association study of early-onset bipolar I disorder in the Han Taiwanese population. Translational Psychiatry, 11(1), 301. doi:10.1038/s41398-021-01407-6
    Wu, X., Chen, P. S., et al. (2008). Histone deacetylase inhibitors up-regulate astrocyte GDNF and BDNF gene transcription and protect dopaminergic neurons. Int J Neuropsychopharmacol, 11(8), 1123-1134. doi:10.1017/s1461145708009024
    Yao, G., Chung, C. W., et al. (2002). Development and verification of validity and reliability of the WHOQOL-BREF Taiwan version. J Formos Med Assoc, 101(5), 342-351.
    Yarden, Y., & Sliwkowski, M. X. (2001). Untangling the ErbB signalling network. Nat Rev Mol Cell Biol, 2(2), 127-137. doi:10.1038/35052073
    Yates, A., Akanni, W., et al. (2015). Ensembl 2016. Nucleic Acids Research, 44(D1), D710-D716. doi:10.1093/nar/gkv1157
    Yatham, L. N., Kennedy, S. H., et al. (2018). Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) 2018 guidelines for the management of patients with bipolar disorder. Bipolar Disord, 20(2), 97-170. doi:10.1111/bdi.12609
    Yen, J. Y., Chang, S. J., et al. (2010). The high-sweet-fat food craving among women with premenstrual dysphoric disorder: emotional response, implicit attitude and rewards sensitivity. Psychoneuroendocrinology, 35(8), 1203-1212. doi:10.1016/j.psyneuen.2010.02.006
    Yokoyama, K., Tezuka, T., et al. (2011). NYAP: a phosphoprotein family that links PI3K to WAVE1 signalling in neurons. Embo j, 30(23), 4739-4754. doi:10.1038/emboj.2011.348
    Yuan, C., Jian, Z., et al. (2022). Type 2 diabetes mellitus increases risk of erectile dysfunction independent of obesity and dyslipidemia: A Mendelian randomization study. Andrology, 10(3), 518-524. doi:10.1111/andr.13132
    Zhan, T., Rindtorff, N., et al. (2017). Wnt signaling in cancer. Oncogene, 36(11), 1461-1473. doi:10.1038/onc.2016.304
    Zhang, D., Li, S., et al. (2021). Microglial activation contributes to cognitive impairments in rotenone-induced mouse Parkinson’s disease model. Journal of Neuroinflammation, 18(1), 4. doi:10.1186/s12974-020-02065-z
    Zhang, Y., Dong, Z., et al. (2020). NLRP3 inflammasome as a novel therapeutic target for Alzheimer’s disease. Signal Transduction and Targeted Therapy, 5(1), 37. doi:10.1038/s41392-020-0145-7
    Zhao, Y. Z., Lin, M., et al. (2016). Intranasal delivery of bFGF with nanoliposomes enhances in vivo neuroprotection and neural injury recovery in a rodent stroke model. J Control Release, 224, 165-175. doi:10.1016/j.jconrel.2016.01.017
    Zimbone, S., Monaco, I., et al. (2018). Amyloid Beta monomers regulate cyclic adenosine monophosphate response element binding protein functions by activating type-1 insulin-like growth factor receptors in neuronal cells. Aging Cell, 17(1), e12684. doi:https://doi.org/10.1111/acel.12684

    無法下載圖示 校內:2027-08-26公開
    校外:不公開
    電子論文尚未授權公開,紙本請查館藏目錄
    QR CODE