Chonnam Med J.  2016 Jan;52(1):18-37. 10.4068/cmj.2016.52.1.18.

The Neurobiology of Bipolar Disorder: An Integrated Approach

Affiliations
  • 1Department of Psychiatry, Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan. muneerather2@gmail.com

Abstract

Bipolar disorder is a heterogeneous condition with myriad clinical manifestations and many comorbidities leading to severe disabilities in the biopsychosocial realm. The objective of this review article was to underline recent advances in knowledge regarding the neurobiology of bipolar disorder. A further aim was to draw attention to new therapeutic targets in the treatment of bipolar disorder. To accomplish these goals, an electronic search was undertaken of the PubMed database in August 2015 of literature published during the last 10 years on the pathophysiology of bipolar disorder. A wide-ranging evaluation of the existing work was done with search terms such as "mood disorders and biology," "bipolar disorder and HPA axis," "bipolar disorder and cytokines," "mood disorders and circadian rhythm," "bipolar disorder and oxidative stress," etc. This endeavor showed that bipolar disorder is a diverse condition sharing neurobiological mechanisms with major depressive disorder and psychotic spectrum disorders. There is convincing evidence of crosstalk between different biological systems that act in a deleterious manner causing expression of the disease in genetically predisposed individuals. Inflammatory mediators act in concert with oxidative stress to dysregulate hormonal, metabolic, and circadian homeostasis in precipitating and perpetuating the illness. Stress, whether biologically or psychologically mediated, is responsible for the initiation and progression of the diathesis. Bipolar spectrum disorders have a strong genetic component; severe life stresses acting through various paths cause the illness phenotype.

Keyword

Bipolar disorder; Circadian rhythm; Oxidative stress

MeSH Terms

Bipolar Disorder*
Circadian Rhythm
Comorbidity
Depressive Disorder, Major
Disease Susceptibility
Homeostasis
Neurobiology*
Oxidative Stress
Phenotype
Stress, Psychological

Figure

  • FIG. 1 The central role of glucocorticoid receptor in the biological functions of cortisol. Cortisol (CORT) enters the cytosol by passive diffusion and binds to the glucocorticoid receptor (GR) which is a dynamic multiprotein complex composed of an array of chaperones. These have inhibitory as well as facilitatory actions and induce conformational change, homodimerization and translocation of the glucocorticoid receptor. The GR homodimer shuttles to the nucleus where it binds to glucocorticoid response element (GRE) on the promoter region of the DNA resulting in gene expression. This attachment to the GRE is facilitated by steroid receptor coactivator-1 (SRC-1); the subsequent gene transcription plays diverse roles in physiological functioning. FKBP: FK506 binding protein, BAG 1: Bcl-2-associated gene product-1, PPID: petidylprolyl isomerase D.

  • FIG. 2 Pro and anti-inflammatory activities of IL-6. Anti-inflammatory activities of IL-6 include STAT3 dependent regeneration of cells and the induction of the hepatic acute phase response, mediated by membrane bound IL-6R (MB IL-6R). Pro-inflammatory activities of IL-6 via soluble IL-6R (sIL-6R) include recruitment of inflammatory cells and inhibition of regulatory T-cell differentiation. ADAM17 plays the key balancing role in determining the direction of IL-6 biological actions. ADAM17: a disintegrin and metalloproteinase 17, MNC: mononuclear cells, STAT3: signal transducer and activator of transcription 3.

  • FIG. 3 Development of psychiatric disorders as a consequence of prolonged SLS. When subjected to long lasting severe life stress (SLS) pathophysiological changes take place in the brain. NADPH oxidase (NOX) enzymes are induced, particularly NOX2 with ensuing increased generation of reactive oxygen species (ROS). The later enhance the release of glutamate (GLU) from neurons; prolonged glutamatergic discharge has such resultant effects as N-methyl D-aspartate (NMDA) receptor down regulation, loss of phenotype of inhibitory parvalbumin (PV) interneurons and apoptotic changes in the hippocampus. These alterations are similar to those seen in psychosis; the putative model of major psychiatric disorders described here is extrapolated from animal experiments carried out in rodents and primates.

  • FIG. 4 The kynurenine pathway of tryptophan metabolism. Tryptophan is the biochemical precursor for the production of serotonin. Activation of the enzyme indoleamine 2, 3 dioxygenase (IDO) by pro-inflammatory cytokines (PIC) and reactive oxidative species (ROS) metabolizes tryptophan into kynurenine (KYN) which can then be metabolized by kynurenine aminotransferase (KAT) into the neuroprotective kynurenic acid or by kynurenine mono-oxygenase (KMO) into the potentially neurotoxic 3-hydroxykynurenine and subsequently to quinolinic acid (QA).

  • FIG. 5 The relationship between mood regulation and the circadian system. The circadian clock affects several systems and pathways which are supposedly the cause of mood disorders. In the majority of patients there are shared aberrant connections which lead to dysregulated daily oscillations. Circadian gene mutations possibly make a person more susceptible to affective disturbances and these are worsened by environmental variations in the daily timetable. 5-HT: 5-hydroxytryptamine, DA: dopamine, HPA-axis: hypothalamic pituitary axis, NA: norepinephrine, SCN: supraschiasmatic nucleus.

  • FIG. 6 The integrative model of bipolar disorder pathophysiology. Dysfunctions in crucial bodily homeostatic systems acting in an orchestrated manner feed into one another leading to a progressively worsening course of bipolar disorder. The result is a persistent symptomatic state, treatment resistance, psychosocial functional deterioration and numerous physical complications. BD: bipolar disorder, HPA axis: hypothalamic-pituitary-adrenal axis, ROS: reactive oxygen species.


Reference

1. Hirschfeld RM. Differential diagnosis of bipolar disorder and major depressive disorder. J Affect Disord. 2014; 169:Suppl 1. S12–S16.
Article
2. Parker G. John Cade. Am J Psychiatry. 2012; 169:125–126.
Article
3. Muneer A. Pharmacotherapy of bipolar disorder with quetiapine: a recent literature review and an update. Clin Psychopharmacol Neurosci. 2015; 13:25–35.
Article
4. Pregelj P. Gene environment interactions in bipolar disorder. Psychiatr Danub. 2011; 23:Suppl 1. S91–S93.
5. Craddock N, Sklar P. Genetics of bipolar disorder. Lancet. 2013; 381:1654–1662.
Article
6. Assies J, Mocking RJ, Lok A, Ruhé HG, Pouwer F, Schene AH. Effects of oxidative stress on fatty acid- and one-carbon-metabolism in psychiatric and cardiovascular disease comorbidity. Acta Psychiatr Scand. 2014; 130:163–180.
Article
7. Post RM. Kindling and sensitization as models for affective episode recurrence, cyclicity, and tolerance phenomena. Neurosci Biobehav Rev. 2007; 31:858–873.
Article
8. Grande I, Magalhães PV, Kunz M, Vieta E, Kapczinski F. Mediators of allostasis and systemic toxicity in bipolar disorder. Physiol Behav. 2012; 106:46–50.
Article
9. Gama CS, Kunz M, Magalhães PV, Kapczinski F. Staging and neuroprogression in bipolar disorder: a systematic review of the literature. Rev Bras Psiquiatr. 2013; 35:70–74.
Article
10. Rosa AR, Magalhães PV, Czepielewski L, Sulzbach MV, Goi PD, Vieta E, et al. Clinical staging in bipolar disorder: focus on cognition and functioning. J Clin Psychiatry. 2014; 75:e450–e456.
11. Weinstock LM, Gaudiano BA, Epstein-Lubow G, Tezanos K, Celis-Dehoyos CE, Miller IW. Medication burden in bipolar disorder: a chart review of patients at psychiatric hospital admission. Psychiatry Res. 2014; 216:24–30.
Article
12. Fries GR, Pfaffenseller B, Stertz L, Paz AV, Dargél AA, Kunz M, et al. Staging and neuroprogression in bipolar disorder. Curr Psychiatry Rep. 2012; 14:667–675.
Article
13. Girshkin L, Matheson SL, Shepherd AM, Green MJ. Morning cortisol levels in schizophrenia and bipolar disorder: a metaanalysis. Psychoneuroendocrinology. 2014; 49:187–206.
Article
14. Watson S, Gallagher P, Ritchie JC, Ferrier IN, Young AH. Hypothalamic-pituitary-adrenal axis function in patients with bipolar disorder. Br J Psychiatry. 2004; 184:496–502.
Article
15. Ellenbogen MA, Santo JB, Linnen AM, Walker CD, Hodgins S. High cortisol levels in the offspring of parents with bipolar disorder during two weeks of daily sampling. Bipolar Disord. 2010; 12:77–86.
Article
16. Binder EB. The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology. 2009; 34:Suppl 1. S186–S195.
Article
17. Scammell JG, Denny WB, Valentine DL, Smith DF. Overexpression of the FK506-binding immunophilin FKBP51 is the common cause of glucocorticoid resistance in three New World primates. Gen Comp Endocrinol. 2001; 124:152–165.
Article
18. Vermeer H, Hendriks-Stegeman BI, van der Burg B, van Buul-Offers SC, Jansen M. Glucocorticoid-induced increase in lymphocytic FKBP51 messenger ribonucleic acid expression: a potential marker for glucocorticoid sensitivity, potency, and bioavailability. J Clin Endocrinol Metab. 2003; 88:277–284.
Article
19. Huzayyin AA, Andreazza AC, Turecki G, Cruceanu C, Rouleau GA, Alda M, et al. Decreased global methylation in patients with bipolar disorder who respond to lithium. Int J Neuropsychopharmacol. 2014; 17:561–569.
Article
20. Yang X, Ewald ER, Huo Y, Tamashiro KL, Salvatori R, Sawa A, et al. Glucocorticoid-induced loss of DNA methylation in non-neuronal cells and potential involvement of DNMT1 in epigenetic regulation of Fkbp5. Biochem Biophys Res Commun. 2012; 420:570–575.
Article
21. Fries GR, Vasconcelos-Moreno MP, Gubert C, dos Santos BT, Sartori J, Eisele B, et al. Hypothalamic-pituitary-adrenal axis dysfunction and illness progression in bipolar disorder. Int J Neuropsychopharmacol. 2014; 18:pii: pyu043.
Article
22. Uyanik V, Tuglu C, Gorgulu Y, Kunduracilar H, Uyanik MS. Assessment of cytokine levels and hs-CRP in bipolar I disorder before and after treatment. Psychiatry Res. 2015; 228:386–392.
Article
23. Bai YM, Su TP, Tsai SJ, Wen-Fei C, Li CT, Pei-Chi T, et al. Comparison of inflammatory cytokine levels among type I/type II and manic/hypomanic/euthymic/depressive states of bipolar disorder. J Affect Disord. 2014; 166:187–192.
Article
24. Leboyer M, Soreca I, Scott J, Frye M, Henry C, Tamouza R, et al. Can bipolar disorder be viewed as a multi-system inflammatory disease. J Affect Disord. 2012; 141:1–10.
Article
25. Miller GE, Chen E, Sze J, Marin T, Arevalo JM, Doll R, et al. A functional genomic fingerprint of chronic stress in humans: blunted glucocorticoid and increased NF-kappaB signaling. Biol Psychiatry. 2008; 64:266–272.
Article
26. Jones KA, Thomsen C. The role of the innate immune system in psychiatric disorders. Mol Cell Neurosci. 2013; 53:52–62.
Article
27. Barbosa IG, Bauer ME, Machado-Vieira R, Teixeira AL. Cytokines in bipolar disorder: paving the way for neuroprogression. Neural Plast. 2014; 2014:360481.
Article
28. Brambilla P, Bellani M, Isola M, Bergami A, Marinelli V, Dusi N, et al. Increased M1/decreased M2 signature and signs of Th1/Th2 shift in chronic patients with bipolar disorder, but not in those with schizophrenia. Transl Psychiatry. 2014; 4:e406.
Article
29. Fiedorowicz JG, Prossin AR, Johnson CP, Christensen GE, Magnotta VA, Wemmie JA. Peripheral inflammation during abnormal mood states in bipolar I disorder. J Affect Disord. 2015; 187:172–178.
Article
30. Li H, Hong W, Zhang C, Wu Z, Wang Z, Yuan C, et al. IL-23 and TGF-1 levels as potential predictive biomarkers in treatment of bipolar I disorder with acute manic episode. J Affect Disord. 2015; 174:361–366.
Article
31. Wieck A, Grassi-Oliveira R, do Prado CH, Rizzo LB, de Oliveira AS, Kommers-Molina J, et al. Pro-inflammatory cytokines and soluble receptors in response to acute psychosocial stress: differential reactivity in bipolar disorder. Neurosci Lett. 2014; 580:17–21.
Article
32. Bai YM, Su TP, Li CT, Tsai SJ, Chen MH, Tu PC, et al. Comparison of pro-inflammatory cytokines among patients with bipolar disorder and unipolar depression and normal controls. Bipolar Disord. 2015; 17:269–277.
Article
33. Barbosa IG, Morato IB, de Miranda AS, Bauer ME, Soares JC, Teixeira AL. A preliminary report of increased plasma levels of IL-33 in bipolar disorder: further evidence of pro-inflammatory status. J Affect Disord. 2014; 157:41–44.
Article
34. Doganavsargil-Baysal O, Cinemre B, Aksoy UM, Akbas H, Metin O, Fettahoglu C, et al. Levels of TNF-α, soluble TNF receptors (sTNFR1, sTNFR2), and cognition in bipolar disorder. Hum Psychopharmacol. 2013; 28:160–167.
Article
35. Agorastos A, Hauger RL, Barkauskas DA, Moeller-Bertram T, Clopton PL, Haji U, et al. Circadian rhythmicity, variability and correlation of interleukin-6 levels in plasma and cerebrospinal fluid of healthy men. Psychoneuroendocrinology. 2014; 44:71–82.
Article
36. Munkholm K, Weikop P, Kessing LV, Vinberg M. Elevated levels of IL-6 and IL-18 in manic and hypomanic states in rapid cycling bipolar disorder patients. Brain Behav Immun. 2015; 43:205–213.
Article
37. Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai Z, et al. Severe depression is associated with increased microglial quinolinic acid in subregions of the anteriorcingulate gyrus: evidence for an immune-modulated glutamatergic neurotransmission. J Neuroinflammation. 2011; 8:94.
Article
38. Mihara M, Hashizume M, Yoshida H, Suzuki M, Shiina M. IL-6/IL-6 receptor system and its role in physiological and pathological conditions. Clin Sci (Lond). 2012; 122:143–159.
Article
39. Düsterhöft S, Höbel K, Oldefest M, Lokau J, Waetzig GH, Chalaris A, et al. A disintegrin and metalloprotease 17 dynamic interaction sequence, the sweet tooth for the human interleukin 6 receptor. J Biol Chem. 2014; 289:16336–16348.
Article
40. Rose-John S. IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int J Biol Sci. 2012; 8:1237–1247.
Article
41. Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The proand anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011; 1813:878–888.
42. Chalaris A, Rabe B, Paliga K, Lange H, Laskay T, Fielding CA, et al. Apoptosis is a natural stimulus of IL6R shedding and contributes to the proinflammatory trans-signaling function of neutrophils. Blood. 2007; 110:1748–1755.
Article
43. Barkhausen T, Tschernig T, Rosenstiel P, van Griensven M, Vonberg RP, Dorsch M, et al. Selective blockade of interleukin-6 trans-signaling improves survival in a murine polymicrobial sepsis model. Crit Care Med. 2011; 39:1407–1413.
Article
44. Scheller J, Chalaris A, Garbers C, Rose-John S. ADAM17: a molecular switch to control inflammation and tissue regeneration. Trends Immunol. 2011; 32:380–387.
Article
45. Waetzig GH, Rose-John S. Hitting a complex target: an update on interleukin-6 trans-signalling. Expert Opin Ther Targets. 2012; 16:225–236.
Article
46. Maes M, Anderson G, Kubera M, Berk M. Targeting classical IL-6 signalling or IL-6 trans-signalling in depression. Expert Opin Ther Targets. 2014; 18:495–512.
47. Fonseka TM, McIntyre RS, Soczynska JK, Kennedy SH. Novel investigational drugs targeting IL-6 signaling for the treatment of depression. Expert Opin Investig Drugs. 2015; 24:459–475.
Article
48. Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact. 2014; 224C:164–175.
Article
49. Nayernia Z, Jaquet V, Krause KH. New insights on NOX enzymes in the central nervous system. Antioxid Redox Signal. 2014; 20:2815–2837.
50. Sorce S, Krause KH. NOX enzymes in the central nervous system: from signaling to disease. Antioxid Redox Signal. 2009; 11:2481–2504.
Article
51. Sorce S, Schiavone S, Tucci P, Colaianna M, Jaquet V, Cuomo V, et al. The NADPH oxidase NOX2 controls glutamate release: a novel mechanism involved in psychosis-like ketamine responses. J Neurosci. 2010; 30:11317–11325.
Article
52. Hu X, Zhou H, Zhang D, Yang S, Qian L, Wu HM, et al. Clozapine protects dopaminergic neurons from inflammation-induced damage by inhibiting microglial overactivation. J Neuroimmune Pharmacol. 2012; 7:187–201.
Article
53. Fatokun AA, Stone TW, Smith RA. Oxidative stress in neurodegeneration and available means of protection. Front Biosci. 2008; 13:3288–3311.
Article
54. Morris G, Berk M. The many roads to mitochondrial dysfunction in neuroimmune and neuropsychiatric disorders. BMC Med. 2015; 13:68.
Article
55. Nguyen D, Alavi MV, Kim KY, Kang T, Scott RT, Noh YH, et al. A new vicious cycle involving glutamate excitotoxicity, oxidative stress and mitochondrial dynamics. Cell Death Dis. 2011; 2:e240.
Article
56. Ventriglio A, Gentile A, Baldessarini RJ, Bellomo A. Early-life stress and psychiatric disorders: epidemiology, neurobiology and innovative pharmacological targets. Curr Pharm Des. 2015; 21:1379–1387.
Article
57. Gu Y, Dee CM, Shen J. Interaction of free radicals, matrix metalloproteinases and caveolin-1 impacts blood-brain barrier permeability. Front Biosci (Schol Ed). 2011; 3:1216–1231.
Article
58. Siwek M, Sowa-Kućma M, Dudek D, Styczeń K, Szewczyk B, Kotarska K, et al. Oxidative stress markers in affective disorders. Pharmacol Rep. 2013; 65:1558–1571.
Article
59. Bitanihirwe BK, Woo TU. Oxidative stress in schizophrenia: an integrated approach. Neurosci Biobehav Rev. 2011; 35:878–893.
Article
60. Ramalingam M, Kim SJ. Reactive oxygen/nitrogen species and their functional correlations in neurodegenerative diseases. J Neural Transm (Vienna). 2012; 119:891–910.
Article
61. Andreazza AC, Kauer-Sant . 'anna M, Frey BN, Bond DJ, Kapczinski F, Young LT, et al. Oxidative stress markers in bipolar disorder: a meta-analysis. J Affect Disord. 2008; 111:135–144.
62. Miller E, Morel A, Saso L, Saluk J. Isoprostanes and neuroprostanes as biomarkers of oxidative stress in neurodegenerative diseases. Oxid Med Cell Longev. 2014; 2014:572491.
Article
63. Black CN, Bot M, Scheffer PG, Cuijpers P, Penninx BW. Is depression associated with increased oxidative stress? A systematic review and meta-analysis. Psychoneuroendocrinology. 2015; 51:164–175.
Article
64. Lima IM, Barros A, Rosa DV, Albuquerque M, Malloy-Diniz L, Neves FS, et al. Analysis of telomere attrition in bipolar disorder. J Affect Disord. 2014; 172C:43–47.
Article
65. Brown NC, Andreazza AC, Young LT. An updated meta-analysis of oxidative stress markers in bipolar disorder. Psychiatry Res. 2014; 218:61–68.
Article
66. Joshi YB, Praticò D. Lipid peroxidation in psychiatric illness: overview of clinical evidence. Oxid Med Cell Longev. 2014; 2014:828702.
Article
67. Schiavone S, Jaquet V, Sorce S, Dubois-Dauphin M, Hultqvist M, Bäckdahl L, et al. NADPH oxidase elevations in pyramidal neurons drive psychosocial stress-induced neuropathology. Transl Psychiatry. 2012; 2:e111.
Article
68. Liang D, Li G, Liao X, Yu D, Wu J, Zhang M. Developmental loss of parvalbumin-positive cells in the prefrontal cortex and psychiatric anxiety after intermittent hypoxia exposures in neonatal rats might be mediated by NADPH oxidase-2. Behav Brain Res. 2016; 296:134–140.
Article
69. Schiavone S, Colaianna M, Curtis L. Impact of early life stress on the pathogenesis of mental disorders: relation to brain oxidative stress. Curr Pharm Des. 2015; 21:1404–1412.
Article
70. Schiavone S, Jaquet V, Trabace L, Krause KH. Severe life stress and oxidative stress in the brain: from animal models to human pathology. Antioxid Redox Signal. 2013; 18:1475–1490.
Article
71. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007; 87:245–313.
Article
72. Heisler JM, O'Connor JC. Indoleamine 2,3-dioxygenase-dependent neurotoxic kynurenine metabolism mediates inflammation-induced deficit in recognition memory. Brain Behav Immun. 2015; 50:115–124.
Article
73. Müller N, Schwarz MJ. Neuroimmune-endocrine crosstalk in schizophrenia and mood disorders. Expert Rev Neurother. 2006; 6:1017–1038.
Article
74. Modabbernia A, Taslimi S, Brietzke E, Ashrafi M. Cytokine alterations in bipolar disorder: a meta-analysis of 30 studies. Biol Psychiatry. 2013; 74:15–25.
Article
75. Musselman D, Royster EB, Wang M, Long Q, Trimble LM, Mann TK, et al. The impact of escitalopram on IL-2-induced neuroendocrine, immune, and behavioral changes in patients with malignant melanoma: preliminary findings. Neuropsychopharmacology. 2013; 38:1921–1928.
Article
76. Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry. 2013; 70:31–41.
Article
77. Myint AM. Kynurenines: from the perspective of major psychiatric disorders. FEBS J. 2012; 279:1375–1385.
Article
78. Campbell BM, Charych E, Lee AW, Möller T. Kynurenines in CNS disease: regulation by inflammatory cytokines. Front Neurosci. 2014; 8:12.
Article
79. Schwarcz R, Bruno JP, Muchowski PJ, Wu HQ. Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci. 2012; 13:465–477.
Article
80. Dantzer R, Walker AK. Is there a role for glutamate-mediated excitotoxicity in inflammation-induced depression? J Neural Transm (Vienna). 2014; 121:925–932.
Article
81. Erhardt S, Olsson SK, Engberg G. Pharmacological manipulation of kynurenic acid: potential in the treatment of psychiatric disorders. CNS Drugs. 2009; 23:91–101.
82. Takaki J, Fujimori K, Miura M, Suzuki T, Sekino Y, Sato K. L-glutamate released from activated microglia downregulates astrocytic L-glutamate transporter expression in neuroinflammation: the 'collusion' hypothesis for increased extracellular L-glutamate concentration in neuroinflammation. J Neuroinflammation. 2012; 9:275.
Article
83. Savitz J, Dantzer R, Wurfel BE, Victor TA, Ford BN, Bodurka J, et al. Neuroprotective kynurenine metabolite indices are abnormally reduced and positively associated with hippocampal and amygdalar volume in bipolar disorder. Psychoneuroendocrinology. 2015; 52:200–211.
Article
84. Leibowitz A, Boyko M, Shapira Y, Zlotnik A. Blood glutamate scavenging: insight into neuroprotection. Int J Mol Sci. 2012; 13:10041–10066.
Article
85. Myint AM, Kim YK. Network beyond IDO in psychiatric disorders: revisiting neurodegeneration hypothesis. Prog Neuropsychopharmacol Biol Psychiatry. 2014; 48:304–313.
Article
86. Milhiet V, Boudebesse C, Bellivier F, Drouot X, Henry C, Leboyer M, et al. Circadian abnormalities as markers of susceptibility in bipolar disorders. Front Biosci (Schol Ed). 2014; 6:120–137.
87. Boudebesse C, Geoffroy PA, Bellivier F, Henry C, Folkard S, Leboyer M, et al. Correlations between objective and subjective sleep and circadian markers in remitted patients with bipolar disorder. Chronobiol Int. 2014; 31:698–704.
Article
88. Gonzalez R. The relationship between bipolar disorder and biological rhythms. J Clin Psychiatry. 2014; 75:e323–e331.
Article
89. Abreu T, Bragança M. The bipolarity of light and dark: a review on Bipolar Disorder and circadian cycles. J Affect Disord. 2015; 185:219–229.
Article
90. Scott J. Clinical parameters of circadian rhythms in affective disorders. Eur Neuropsychopharmacol. 2011; 21:Suppl 4. S671–S675.
Article
91. Ramanathan C, Xu H, Khan SK, Shen Y, Gitis PJ, Welsh DK, et al. Cell type-specific functions of period genes revealed by novel adipocyte and hepatocyte circadian clock models. PLoS Genet. 2014; 10:e1004244.
Article
92. Gonzalez R, Tamminga CA, Tohen M, Suppes T. The relationship between affective state and the rhythmicity of activity in bipolar disorder. J Clin Psychiatry. 2014; 75:e317–e322.
Article
93. Menaker M, Murphy ZC, Sellix MT. Central control of peripheral circadian oscillators. Curr Opin Neurobiol. 2013; 23:741–746.
Article
94. Ko CH, Takahashi JS. Molecular components of the mammalian circadian clock. Hum Mol Genet. 2006 Spec No 2; 15:R271–R277.
Article
95. McClung CA. Circadian genes, rhythms and the biology of mood disorders. Pharmacol Ther. 2007; 114:222–232.
Article
96. Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, et al. The nuclear receptor REV-ERB mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci U S A. 2012; 109:582–587.
Article
97. Charmandari E, Chrousos GP, Lambrou GI, Pavlaki A, Koide H, Ng SS, et al. Peripheral CLOCK regulates target-tissue glucocorticoid receptor transcriptional activity in a circadian fashion in man. PLoS One. 2011; 6:e25612.
Article
98. Landgraf D, McCarthy MJ, Welsh DK. Circadian clock and stress interactions in the molecular biology of psychiatric disorders. Curr Psychiatry Rep. 2014; 16:483.
Article
99. Cagampang FR, Bruce KD. The role of the circadian clock system in nutrition and metabolism. Br J Nutr. 2012; 108:381–392.
Article
100. Kim TW, Jeong JH, Hong SC. The impact of sleep and circadian disturbance on hormones and metabolism. Int J Endocrinol. 2015; 2015:591729.
Article
101. Bellet MM, Orozco-Solis R, Sahar S, Eckel-Mahan K, Sassone-Corsi P. The time of metabolism: NAD+, SIRT1, and the circadian clock. Cold Spring Harb Symp Quant Biol. 2011; 76:31–38.
Article
102. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. Circadian control of the NAD+ salvage pathway by CLOCKSIRT1. Science. 2009; 324:654–657.
Article
103. Pinnock SB, Herbert J. Brain-derived neurotropic factor and neurogenesis in the adult rat dentate gyrus: interactions with corticosterone. Eur J Neurosci. 2008; 27:2493–2500.
Article
104. AlAhmed S, Herbert J. Effect of agomelatine and its interaction with the daily corticosterone rhythm on progenitor cell proliferation in the dentate gyrus of the adult rat. Neuropharmacology. 2010; 59:375–379.
Article
105. Hope S, Melle I, Aukrust P, Steen NE, Birkenaes AB, Lorentzen S, et al. Similar immune profile in bipolar disorder and schizophrenia: selective increase in soluble tumor necrosis factor receptor I and von Willebrand factor. Bipolar Disord. 2009; 11:726–734.
Article
106. Hope S, Ueland T, Steen NE, Dieset I, Lorentzen S, Berg AO, et al. Interleukin 1 receptor antagonist and soluble tumor necrosis factor receptor 1 are associated with general severity and psychotic symptoms in schizophrenia and bipolar disorder. Schizophr Res. 2013; 145:36–42.
Article
107. Sinclair D, Fillman SG, Webster MJ, Weickert CS. Dysregulation of glucocorticoid receptor co-factors FKBP5, BAG1 and PTGES3 in prefrontal cortex in psychotic illness. Sci Rep. 2013; 3:3539.
Article
108. Wang JF, Shao L, Sun X, Young LT. Increased oxidative stress in the anterior cingulate cortex of subjects with bipolar disorder and schizophrenia. Bipolar Disord. 2009; 11:523–529.
Article
109. Etain B, Milhiet V, Bellivier F, Leboyer M. Genetics of circadian rhythms and mood spectrum disorders. Eur Neuropsychopharmacol. 2011; 21:Suppl 4. S676–S682.
Article
110. De Berardis D, Fornaro M, Serroni N, Campanella D, Rapini G, Olivieri L, et al. Agomelatine beyond borders: current evidences of its efficacy in disorders other than major depression. Int J Mol Sci. 2015; 16:1111–1130.
Article
111. Kauer-Sant'Anna M, Kapczinski F, Andreazza AC, Bond DJ, Lam RW, Young LT, et al. Brain-derived neurotrophic factor and inflammatory markers in patients with early- vs. late-stage bipolar disorder. Int J Neuropsychopharmacol. 2009; 12:447–458.
112. Andreazza AC, Kapczinski F, Kauer-Sant'Anna M, Walz JC, Bond DJ, Gonçalves CA, et al. 3-Nitrotyrosine and glutathione antioxidant system in patients in the early and late stages of bipolar disorder. J Psychiatry Neurosci. 2009; 34:263–271.
113. Elvsåshagen T, Vera E, Bøen E, Bratlie J, Andreassen OA, Josefsen D, et al. The load of short telomeres is increased and associated with lifetime number of depressive episodes in bipolar II disorder. J Affect Disord. 2011; 135:43–50.
Article
114. Strakowski SM, DelBello MP, Zimmerman ME, Getz GE, Mills NP, Ret J, et al. Ventricular and periventricular structural volumes in first- versus multiple-episode bipolar disorder. Am J Psychiatry. 2002; 159:1841–1847.
Article
115. Kapczinski F, Vieta E, Andreazza AC, Frey BN, Gomes FA, Tramontina J, et al. Allostatic load in bipolar disorder: implications for pathophysiology and treatment. Neurosci Biobehav Rev. 2008; 32:675–692.
Article
116. Gaali S, Kirschner A, Cuboni S, Hartmann J, Kozany C, Balsevich G, et al. Selective inhibitors of the FK506-binding protein 51 by induced fit. Nat Chem Biol. 2015; 11:33–37.
Article
117. Dounay AB, Tuttle JB, Verhoest PR. Challenges and opportunities in the discovery of new therapeutics targeting the kynurenine pathway. J Med Chem. 2015; 58:8762–8782.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr