Chonnam Med J. 2016 Jan;52(1):18-37. English.
Published online Jan 19, 2016.
© Chonnam Medical Journal, 2016
Review

The Neurobiology of Bipolar Disorder: An Integrated Approach

Ather Muneer
    • Department of Psychiatry, Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan.

Received November 09, 2015; Revised November 26, 2015; Accepted November 30, 2015.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

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.

Keywords
Bipolar disorder; Circadian rhythm; Oxidative stress

INTRODUCTION

Bipolar disorder (BD) is unique among psychiatric conditions in that its symptoms swing between two opposite mood states: mania and depression.1 Almost 70 years ago John Cade found out that lithium had beneficial effects in his patients suffering from "mania, dementia praecox and melancholia."2 The favorable outcome with lithium treatment in patients with major mental illnesses was a breakthrough in that era. Since that discovery, however, no pharmacological agent for the specific treatment of BD has been ascertained, and for all intents and purposes the management of this condition is with psychotherapeutic drugs developed for other indications.3 In the past few decades, more and more refined investigational techniques have been employed to uncover the pathophysiology of BD. As a result, several important discoveries have been made; the translational nature of the research gives rise to the expectation that new insights will lead to more effective treatments for patients with BD and that the fruits of scientific knowhow will be passed on from the bench to the bedside.

BD is a complex medical condition whose etiology involves genetic and epigenetic factors acting alongside environmental stresses in causing expression of the disease.4 This diathesis is currently viewed as a multisystem ailment that not only affects brain functioning but also results in physical comorbidities like cardiovascular disease, diabetes mellitus, disorders of immunity, and endocrine dysfunction. Genome-wide studies have failed to detect any single gene to account for the incidence of BD, fostering the prevailing assumption that it is a polygenic condition.5 The involved genes interact with life stresses to cause disruption in biological and homeostatic mechanisms. Research efforts in recent years have disclosed that dysregula tion of vital biochemical pathways acts in an orchestrated manner in the pathogenesis of BD. There is disruption of glucocorticoid signaling, immune-inflammatory imbalance, increased oxidative stress, abnormalities of tryptophan metabolism, and derangement of phospholipid turnover with shifts in the 1-carbon cycle of methionine and homocysteine. 6 These identified mechanistic pathways offer opportunities for the development of novel and state-of-the-art therapeutic agents, which hold the promise of opening fresh avenues in the treatment of BD.

Life stresses acting on predisposed individuals have an enduring effect on the neural substrate, causing rewiring of the nervous system with increased sensitization and proneness to recurrent affective episodes.7 There is mounting evidence that manic and major depressive exacerbations have a neurotoxic effect, damaging the neurons as well as the glial elements in the brain.8 The results of preclinical and human studies consistently show accumulating organ damage both in the center and in the periphery with illness progression. The neuroprogressive nature of BD is clinically manifested as increased frequency and severity of episodes, greater suicidal risk, and cognitive and functional impairment.9 In the final stages of the disorder there is no illness remission, persistence of inter-episode subthreshold affective symptoms, and eventual loss of autonomy.10 The course of BD is malignant in many cases; currently available medications fail to control the disease manifestations, with very high rates of polypharmacy, soaring frequency of treatment-emergent adverse effects, and meager compliance from the patients.11 Considering that BD is a prevalent condition, the application of less than optimal treatment strategies and consequent illness progression place a huge burden on the individual patient, his or her family, and society as a whole. In view of these concerns, it is vitally important to cultivate better understanding of the disease mechanisms so that adequate cures are made available to the numerous people afflicted by this intractable illness. The purpose of this was review to clarify the key interacting pathophysiological mechanisms that drive the disease process in bipolar spectrum disorders and to uncover potential new treatment targets for these and related mood disorders.

SEARCH STRATEGY

In August 2015, the PubMed electronic database was searched for literature on the neurobiology of mood disorders, with particular reference to BD. The investigative approach was broadly based to include articles on animal models with extrapolative value for human disease. During the appraisal of the literature, special emphasis was placed on the translational significance of the research work. This policy was employed with the specific intent to uncover the pathophysiology of bipolar spectrum disorders while at same time identifying novel therapeutic targets for the treatment of these conditions. The aim of conducting a wide-ranging evaluation of the extant body of work was driven by the understanding that the management of BD is particularly challenging considering the heterogeneity, developmental trajectory, and neuroprogressive nature of the disease. An attempt was made to highlight original concepts in the neurobiology of BD that were both innovative and had heuristic value. During the preparation of the article, only studies conducted in the last 10 years were included. Several combinations of search terms were used; some examples are "mood disorders and biology," "bipolar disorder and HPA axis," "bipolar disorder and cytokines," "bipolar disorder and circadian," "mood disorders and circadian rhythm," "mood disorders and inflammation," "bipolar disorder and oxidative stress," and "bipolar disorder and neurobiology." Investigative work on animals and humans as well as review articles were assessed; articles that were considered to be especially pertinent were read in full and their reference lists were also consulted. Finally, the data were integrated in a narrative review in a concise and coherent style.

HYPOTHALAMIC-PITUITARY-ADRENAL AXIS DYSFUNCTION

Early in the trajectory of BD, episodes occur secondary to stress but there is blighted psychobiological resilience and defective coping that increase vulnerability to recurrent affective exacerbations with illness advancement. 12 This impairment is principally provoked by the hypothalamic-pituitary-adrenal (HPA) axis, which does not function properly in patients with BD.13 Patients with BD have a hyperactive HPA axis, high levels of systemic cortisol, and nonsuppression of its circulating levels in the dexamethasone suppression test or the dexamethasone/corticotrophin-releasing hormone (DEX/CRF) test.14 Furthermore, subjects with increased susceptibility, such as first-degree relatives, have also been shown to have increased baseline cortisol levels and aberrant responses to the DEX/CRF test.15 From this perspective, HPA axis irregularities seem to be a genetic attribute endowing vulnerability to mood disorders. The glucocorticoid receptor (GR) is the most important factor in the formulation of the cortisol response; it binds to the hormone in the cytosol and shuttles it to the nucleus where it functions as a transcription factor. The action of the GR is dependent on a hefty molecular complex consisting of several chaperone proteins and cofactors, including FK506 binding protein 51 (FKBP51).16 In vitro experiments in humans reveal that overexpression of FKBP51 reduces hormone binding affinity and nuclear translocation of GR, whereas high levels of this chaperone cause GR insensitivity and raised peripheral levels of cortisol in nonhuman primates.17 Intriguingly, this hormone acting via an intracellular ultra-short negative feedback loop for GR activity can stimulate the expression of FKBP51.18

There is a known familial contribution to the neurobiology of BD, and it is probable that most of the cortisol-GR-related mechanisms alluded to above are a sign of the putative genetic underpinning. In this regard, research has shown that the hereditary component in BD largely acts through gene-environment associations. In essence, biological and psychosocial stressors reprogram gene activity by altering epigenetic modifications, thus escalating the risk for the disease in predisposed people, as well as meddling with the illness trajectory in those with the expressed phenotype. Among the purported epigenetic modifications, alterations in DNA methylation have been repeatedly shown in bipolar patients.19 Significantly, in murine models, prolonged exposure to glucocorticoids is known to bring about changes in DNA methylation at the FKBP5 gene. In human studies, such alterations have been noted in the Fkbp5 gene in patients with a stressor-related comorbid condition of BD, namely, post-traumatic stress disorder. As such, FKBP5 methylation may be one of paths through which the HPA system acting in response to stress malfunctions in BD pathophysiology.20 Given that the mechanisms of HPA axis dysregulation are incompletely known at present, as is its role in dictating the risk of the disease in vulnerable subjects, current work is beginning to unravel the molecular targets of illness development and progression in BD.21 The crucial function of GR in the actions of cortisol is depicted schematically in Fig. 1.

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.

IMMUNOLOGICAL FACTORS

1. Immune-inflammatory imbalance

In bipolar patients, major mood episodes of either polarity result in an inflammatory response that has been convincingly shown in several studies. This is evident as an increase in the levels of proinflammatory cytokines (PIC) and C-reactive protein in the peripheral blood.22 The PIC include most importantly interleukin 1β (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α). Treatment with mood stabilizers and resolution of acute affective exacerbations has been shown to normalize the levels of some PIC like IL-6 but not TNF-α, whereas a chronic, low-level inflammatory state persists even in euthymic patients.23 This points to the fact that immune-inflammatory dysregulation is fundamental to the pathophysiological processes in BD.24

It is suggested that unrelieved strain in BD activates the HPA axis with increased secretion of cortisol; in addition, there is stimulation of the sympatho-adrenal medullary axis with increased circulating levels of epinephrine and norepinephrine. These stress hormones acting by virtue of their membrane and cytosolic receptors affect the transcription of genes encoding cytokines via decreased inhibition of the canonical nuclear factor-kappa B (NF-κB) and the noncanonical inflammatory signal transduction pathways like activator-protein 1 (AP-1), Janus kinasesignal transducer and activator of transcription (JAK-STAT) factors, and mitogen-activated protein kinases (MAPK).25 The cells of the innate immune system (for example, monocytes, macrophages, and T-lymphocytes) secrete PIC, chemokines, and cell adhesion molecules that diffuse to the brain and trigger microglia, causing neuroinflammation and further activating the HPA axis.26 The unrelenting secretion of stress hormones leads to a constant low-grade inflammatory milieu in the body that is considered to be liable for the neuroprogression of the bipolar diathesis and also predisposes to cardiovascular and metabolic abnormalities often encountered in these patients. 27 Additionally, with repeated mood episodes and advancement of BD, there is a shift of the immune response from T-helper 2 (TH2) cells, which mainly secrete anti-inflammatory mediators like IL-10, to TH1 cells, which produce PIC (for example, TNF-α).28 Several case-control studies, mainly having cross-sectional designs, have revealed dysregulation of the immune-inflammatory response in BD. Table 1 summarizes recently published work in this area.

TABLE 1
Circulating cytokine abnormalities in bipolar disorder

2. IL-6 trans-signaling in mood disorders

IL-6 is a ubiquitous inflammatory cytokine performing diverse biological actions that vary from regeneration and repair of cellular elements to augmenting the response to injury in various types of tissue damage.35 Several studies have documented an increase in peripheral circulating levels of IL-6 during acute mood episodes of either polarity.36 Because the brain is no longer considered privileged from the effects of peripheral inflammatory mediators, IL-6 gains access to this organ from the circulation. Upon stimulation, microglia and astrocytes also secrete IL-6 locally.37 However, the receptor IL-6R, which binds to this cytokine in nanomolar concentrations, is expressed by a few cell types only (including some leukocytes and hepatocytes). The complex of IL-6 and IL-6R binds with a second glycoprotein, gp 130, that subsequently dimerizes and kicks off intracellular signaling via the JAK/STAT pathways.38 Unlike IL-6R, gp 130 is expressed on all cells; however, the latter alone has no affinity for the cytokine and cells lacking IL-6R cannot respond to this mediator. Of note, a soluble form of IL-6R (sIL-6R) consisting of the extracellular portion of the receptor was detected in humans in plasma and other body fluids. sIL-6R is generated by partial proteolysis of the membrane-bound receptor by "a disintegrin and metalloprotease" (ADAM17).39 The sIL-6R interacts with IL-6 with similar binding properties as the membrane-bound IL-6R. IL-6 trans-signaling is the path by which gp 130–expressing cells, even in the absence of membrane-bound IL-6R, can be stimulated by the complex of IL-6 and sIL-6R.40 Because of this phenomenon, the inflammatory process can affect every organ system in the body, but under the steady state condition, uncontrolled inflammation is kept in check and physiological homeostasis is maintained.41

In the course of inflammation, proteolysis of the IL-6R from neutrophils by virtue of ADAM17 leads to the activation of endothelial cells which do not express IL-6R on their membranes and are therefore insensitive to the cytokine. Priming of the endothelial cells by the IL-6/sIL-6R complex leads to the secretion of the mononuclear cell attracting cytokine MCP-1. Thereby, the shedding of the IL-6R acts as a measure of preliminary injury as shown by the number of neutrophils involved, since these cells are the first to arrive at the site of damage.42 Furthermore, experimental models in mice have determined that classic IL-6 signaling has a regenerative and anti-apoptotic role during inflammation, for example, in the cecal puncture and ligation sepsis paradigm during which the animals are subjected to extreme stress. In contrast, IL-6 trans-signaling reflects the proinflammatory arm of the cytokine's biological activities.43 Since the proteolysis of IL-6R is mainly governed by ADAM17, it is highly likely that ADAM17 has a key function in inflammation-related phenomena (Fig. 2).44

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.

There are therapeutic implications in this pattern, as specific agents can be developed that block the proinflammatory properties of IL-6 without stalling its anti-inflammatory actions.45 Many if not all neural cells are the target of IL-6 trans-signaling, and inhibition of this activity can be expected to have important salutary affects in neuropsychiatric disorders.46 A soluble fusion glycoprotein sgp 130Fc has been engineered from the extracellular portion of gp 130 that exclusively restrains IL-6 trans-signaling. Administration of this engineered protein is a viable treatment approach in major psychiatric conditions, including BD.47 Thus, current evidence provides a rationale for undertaking preclinical studies in mood disorders to determine the therapeutic prospects of IL-6 blocking strategies.

OXIDATIVE STRESS

1. Physiological role of free radicals

In biological terms, oxidative stress can be considered as a continuing discrepant interaction between antioxidants and prooxidants with a tilt toward the latter.48 The result of this fact is the disproportionate formation of free radicals, the reactive oxygen species (ROS). At low physiological concentrations, ROS perform important functions in the central nervous system (CNS).49 These include regulation of the destiny of neurons either through growth or programmed cell death via stimulation of the AP-1 transcription factor and the nerve growth factor pathways.50 ROS take part in critical signaling cascades such as the regulation of the membrane potential and cellular H+ fluxes, the execution of cardiovascular homeostasis and management of blood pressure through the angiotensin II receptor, and the control of the glutamatergic neurotransmission via the N-methyl-D-aspartate (NMDA) receptor.51 In addition, ROS are engaged in the neuroinflammatory response by means of priming of the microglia.52 Free radical actions may have a key function in fine-tuning the responses of neuronal cells to adverse events, either by promoting resiliency through stress-induced molecular cascades such as MAPK or by getting rid of the severely impaired cells by apoptosis.53

2. Pathophysiological mechanisms

On the flip side, the buildup of ROS is found to enhance the vulnerability of brain tissue to damage and has an important part in the pathophysiology of a number of neuropsychiatric conditions. Unequivocal proof of increased brain oxidative damage has been revealed for neurodegenerative conditions like Alzheimer's and Parkinson's diseases, cerebrovascular disorders, demyelinating diseases, and severe psychiatric ailments such as schizophrenia, major depressive disorder, and BD.54 ROS cause glutamate excitotoxicity and alter mitochondrial activity. Mitochondrial dysfunction in turn causes NMDA receptor up-regulation and further increases oxidative stress, resulting in a detrimental self-sustaining and aggravating cellular process.55 Oxidative stress products such as superoxide and hydroxyl radicals have been shown to induce cortisol resistance by impairing the GR movement from the cytosol to the nucleus. HPA axis dysregulation by ROS in turn causes a proinflammatory response with increased circulating levels of PIC.56 Unrestricted ROS activity is also responsible for increased blood-brain barrier permeability through launching of matrix metalloproteinases and subsequent degradation of tight junctions, unrepressed neuroinflammation, and enhanced apoptosis of neurons.57

3. Redox balance in the brain

The assumed pathophysiological association between oxidative stress and mood disorders may be due to the fact that the nervous system is increasingly susceptible to oxidative damage for a number of reasons. First, of all the organs, the brain has the highest consumption rate of oxygen; it is roughly 2% of body weight but uses 20% of total inspired oxygen. This fact predisposes it to greater formation of ROS during the process of mitochondrial energy metabolism. Second, the brain's lipid content is very high, and lipids act as a substrate for the ROS. Third, there is the redox potential of several neurotransmitters, for instance, dopamine. Fourth, the defense systems against free radicals are relatively inefficient. Last, the brain has a high content of metal ions, for example, iron and copper, involved in redox reactions. 58 The protective arrangement against prooxidants is composed of an enzymatic and a nonenzymatic component. Glutathione peroxidase and glutathione reductase are recognized intracellular antioxidant enzymes. The former changes peroxides and hydroxyl radicals into benign moieties, with the oxidation of reduced glutathione (GSH) into the oxidized form glutathione disulfide (GSSG), and glutathione reductase regenerates GSH from GSSG. Additionally, other enzymes such as catalase (CAT) and superoxide dismutase (SOD) also take part in the cellular resistance against oxidative stress and act along with glutathione peroxidase, thus forming the principal enzymatic defense against accumulated free radicals. Further, glutathione-S-transferase and glucose-6-phosphate dehydrogenase are important in sustaining a stable provision of metabolic products like GSH and nicotinamide adenine dinucleotide-phosphate (NADPH), which are required for proper operation of the main antioxidant enzymes.59

4. Measurement of oxidative stress

The main ROS include the superoxide radical (O2˙-), hydrogen peroxide (H2O2), and the hydroxyl radical (OH˙). Interaction between ROS and nitric oxide results in the formation of highly reactive nitrogen species, such as the peroxynitrite ions (ONOO-), which are very damaging to the basic structural molecules of the cells.60 Because ROS and reactive nitrogen species possess very short half-lives, these are hard to quantify. Therefore, it is necessary to search for and determine indirect pointers of oxidative stress; these include lipid, protein, and DNA peroxidation indicators. Whereas measurements of key enzymes involved in the neutralization of free radicals also serve as putative markers.61 Biosignatures of oxidative stress in body fluids include oxidized proteins like protein carbonyls and oxidized lipids such as 4-hydroxynonenol, thiobarbituric acid–reactive substances, malondialdehyde, and isoprostanes, which are isomers resulting from the oxidation of arachidonic acid. In addition, neuroprostanes are emerging as definitive markers of oxidative stress emanating from the CNS.62 The oxidation of DNA at the guanine residue site produces 8-hydroxydeoxyguanosine, which is associated with telomere shortening detectable in peripheral blood cells or fibroblasts.63 Telomere shortening is an indicator of accelerated aging and premature mortality owing to the increased incidence of cardiovascular and other comorbidities in major psychiatric conditions.64 There are reliable data from work on circulating markers that the brain's main antioxidants, namely, GSH, CAT, SOD, and GSH peroxidase, have changed values in bipolar subjects. In this vein, recent meta-analyses have shown increased biomarkers of oxidative stress.65 Foremost discoveries are enhanced lipid peroxidation, DNA injury, and increased amino acid nitration in bipolar patients in contrast with healthy subjects, with lipid peroxidation being the predominant finding.66

5. NADPH oxidase enzymes as a potential therapeutic target

The sole purpose of NADPH oxidase (NOX) enzymes is the generation of ROS, and their induction is purportedly a significant cause of oxidative stress in major psychiatric disorders.67 These enzymes utilize cytoplasmic NADPH and catalyze the transfer of electrons to molecular O2 to produce ROS. Acting along the transmembrane electron transport chain, NOX enzymes generate ROS in the extracellular space or the lumen of intracellular organelles. Seven NOX genes have thus far been discovered, and the best described isoform is NOX2.68 These enzymes are found in several tissues of the body, and the presence of NOX transcripts has been confirmed in total brain samples as well as in neurons, microglia, and astrocytes. In the presence of severe life stress, these are induced, causing increased production of ROS in the brain and thereby playing a role in the expression of the main psychiatric disorders.69 Animal experiments in rodents and primates with paradigms such as social isolation and establishment of communal hierarchy models have been very helpful in revealing the contribution of these enzymes to prooxidant injury and the resulting expression of different types of abnormal behavior. In the case of persistent severe life stress, the neurophysiologic impact of excessive ROS generation gives rise to continued excitatory, glutamatergic transmission leading to changes suggestive of alterations seen in psychotic spectrum disorders. These include down-regulation of the NMDA receptors, failure of the inhibitory trait of GABAergic interneurons, and reduction of hippocampal volume.70 In view of these animal experiments, the cause of oxidative stress has been shown to be the increased expression of NOX enzymes. If these findings are confirmed in humans, these enzymes might emerge as potential therapeutic targets.71 Fig. 3 illustrates this supposed model of disease development in major psychiatric disorders.

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.

INFLAMMATION, KYNURENINES, AND MOOD DISORDERS

There is limited knowledge regarding the etiopathogenesis of affective disorders, which serves as a major obstacle in the discovery of effective preventive and curative therapies. Several converging sources of evidence point to a contribution of inflammation in the neurobiology of major depressive disorder and BD with the participation of several biological systems in the initiation and progression of these conditions.72 There is crosstalk among disturbed immune, hormonal, and metabolic systems causing aberration in the functioning of neurotransmitters and leading to the manifestations of affective disorders.73 With regard to BD, circulating levels of PIC have been shown to be increased during the course of acute episodes, returning to baseline with successful mood-stabilizing treatment.74 The strongest evidence for the contributory role of inflammation in the causation of major depressive disorder comes from the observation that cytokine immunotherapy based on chronic administration of interferon-alpha and/or IL-2 to patients with kidney cancer or melanoma with metastasis induces depressive episodes in a considerable number of patients.75 Furthermore, anti-TNFα treatment with etanercept and infliximab is emerging as a possible option in the management of resistant mood disorders, either as monotherapy or adjunctive therapy.76

PIC stimulate indoleamine 2,3-dioxygenase (IDO), which is a ubiquitous enzyme that metabolizes tryptophan along the kynurenine (KYN) pathway. The latter is an essential amino acid acquired from food; 5HT is produced from about 1% of the accessible tryptophan in the body, whereas the remaining 99% is metabolized in the hepatocytes by tryptophan 2,3-dioxygenase (TDO). TDO activity is chiefly governed by the amino acid's level itself, and therefore its biological action is usually steady and not influenced by inflammatory mediators.77 During conditions of inflammation and increased oxidative stress, IDO activation in the extrahepatic tissues shifts the metabolism of tryptophan away from the liver. KYN formed by the action of IDO is further metabolized to compounds that have neuroactive properties.78 Fig. 4 shows the main enzymes and products in the KYN pathway downstream of IDO.

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).

In circumstances of stress, extra KYN is formed in the brain in situ, whereas its central availability is further enhanced because it can pass through the blood-brain barrier. In fact, tryptophan and KYN can cross the blood-brain barrier via a sodium-independent large neutral amino acid transporter known as System L. By contrast, the remaining KYN metabolites are exclusively formed in the brain itself because these cannot cross from the periphery into the CNS.79 In summary, under the influence of biological stimuli triggered by stress, the KYN pathway is greatly stimulated in the brain. On the basis of the facts presented above, inflammation-induced mood disorders should be amenable to treatment by the following:

  • a) Targeting the development of inflammation, for example, with cytokine inhibitors or cytokine signaling pathway antagonists.

  • b) Preventing the formation of peripheral KYN with IDO competitive blocker, 1-methyl tryptophan. In the normal brain, up to 78% of KYN derives from the blood, while during systemic inflammation nearly all of the brain KYN originates from the periphery.

  • c) Stopping the transport of KYN into the brain by increasing the availability of large, branched-chain amino acids.

This approach would certainly work best when deployed in a preemptive manner before mood disorders occur.80 Once an affective episode has come about, the neuroactive KYN pathway metabolites are formed in situ in the brain and an alternate strategy aimed at repairing the inflamed brain must be put in place.

Among the products of the KYN pathway, kynurenic acid has a putative neuroprotective role by acting as an antagonist of NMDA receptor, while it also decreases glutamate levels via inhibition of α7-nicotinic receptors. 3-Hydroxykynurenine is a free radical generator, and quinolinic acid is an NMDA receptor agonist that also exerts neurotoxic effects via oxidative damage to lipids and interruption of the blood-brain barrier. The neurotoxic activity of quinolinic acid has been known for more than 30 years. This metabolite should a priori be a therapeutic target by blocking its formation or antagonizing its excitotoxic effect on NMDA glutamatergic receptors.81 However, in the context of an inflamed brain, the microglia are activated, which results in the release of large quantities of glutamate, a process facilitated by the uptake of extracellular glutamine that is converted to glutamate by the enzyme glutaminase. Moreover, oxidative stress generated by 3-hydroxykynurenine and 3-hydroxyanthranilic acid further contributes to microglia priming. In the normal brain, the clearance of glutamate is efficient because astrocytes take up this substance and convert it to glutamine via glutamine synthetase, which is recycled back to neurons for the continued formation of glutamate. However, in conditions of excitotoxicity, the extracellular concentration of glutamate can increase up to 100-fold, overwhelming this mechanism of glutamate reprocessing.82 Such pathologically increased levels of glutamate result in atrophic changes in key moodregulating areas like the hippocampus and amygdala.83

An important mechanism for the clearance of brain extracellular glutamate is represented by the extrusion of glutamate from the brain interstitial fluid into the circulation. The brain-to-blood movement of glutamate is limited owing to the relatively high levels of plasma glutamate, so that an efficient way to substantially enhance this process is to accelerate the degradation of glutamate in the blood by administering oxaloacetate, which activates glutamic oxaloacetate transaminase. This enzyme catalyzes the transformation of glutamate into 2-ketoglutarate. Blood glutamate scavenging can be enhanced further by adding recombinant glutamic oxaloacetate transaminase to oxaloacetate. The neuroprotective effect of blood glutamate scavenging has been confirmed in a number of experimental conditions associated with high brain concentrations of glutamate, such as traumatic brain injury, epilepsy, and ischemic stroke.84

Finally, to achieve personalized treatment it is necessaryto develop suitable biomarkers for identifying thosesubjects who would most likely gain by targeting the sequenceof actions leading from inflammation to major mooddisorders. The events to be marked in this chain are thetransport of KYN into the brain; the production and actionof KYN metabolites, particularly quinolinic acid; and possiblythe increased concentrations of glutamate in thebrain interstitial fluids.85

CIRCADIAN RHYTHM ABNORMALITIES IN MOOD DISORDERS

1. Circadian markers

Chronotropic representations have added to our knowledge of the neurobiology of mood disorders, and dysfunctions in the circadian system are linked with the bipolar phenotype. The evident abnormalities include hormonal dysregulations (melatonin and cortisol fluctuations), actigraphic patterns (sleep/wake cycles), and chronotypic activity preferences (dimensional variations), which serve as circadian markers.86 These aberrations are seen not only during acute affective exacerbations but also in the euthymic periods. Most of these indicators are also found in the healthy relatives of bipolar patients, thus signifying a robust familial predisposition. Therefore, these may be considered as genetic attributes of the disorder.87 A number of circadian genes are known to be correlated with BD: several studies have shown affirmative links for CLOCK, NPAS2, ARNTL1, NR1D1, PER3, RORB, and CSNK1-epsilon. Thus, disruption of the molecular circadian clock is supposedly involved in the genetic vulnerability to BD.88The circadian hypothesis has directed the use of treatment methods such as interpersonal and social rhythms therapy and chronotherapeutics (bright light therapy, total sleep deprivation). Furthermore, this hypothesis may elucidate how mood stabilizers, particularly lithium, which inhibits the enzyme GSK3β, resynchronize the molecular clock and bring about their therapeutic effects.89

2. Social zeitgeber theory

Over the years many theories have been expounded to clarify the etiology of mood disorders. The propositions include disturbance in monoamine neurotransmission, HPA axis malfunction, immune dysregulation, decreased neurogenesis, mitochondrial dysfunction, and altered neuropeptide signaling. Almost all patients with mood disorders have major interruptions in circadian rhythms and the sleep/wake cycle. No wonder that disruption in normal sleep pattern is one of the main diagnostic criteria for these conditions. Furthermore, environmental disturbances to circadian rhythms such as shift work, trans-meridian travel, and lopsided social timetables often induce or aggravate affective exacerbations.90 It has been shown in current research work that molecular clocks exist everywhere in the brain and body, where they partake in the working of nearly all physiological functions.91 The social zeitgeber premise of mood disorders assumes that stressful life events cause alterations in the sleep/wake pattern, which then affect molecular and cellular rhythms in susceptible people, inducing affective episodes. This postulate is supported by a number of clinical studies showing an unambiguous connection between the extent of rhythm disruptions and the severity of mood episodes; also, rhythms are re-established with successful treatment.92 In essence, all present therapies for mood disorders modify or even out circadian rhythms.

3. The molecular clock

The principal circadian synchronizer is placed in the suprachiasmatic nucleus of the hypothalamus where it gets light input by a direct connection, the retinohypothalamic tract. The suprachiasmatic nucleus subsequently conveys signals via direct and indirect projections all over the brain. It also manages the timing of the discharge of several peptides and hormones, including melatonin, the sleep-promoting agent. Of note, the suprachiasmatic nucleus appears to synchronize the timing of rhythms in the periphery through changes in body temperature, which serve as a common signal to harmonize multiple organ systems to the light/dark cycle.93 The molecular clock hub consists of a sequence of transcriptional and translational periodic loops that lead to the recurrent expression of clock genes on a time scale of just over 24 hours. In the primary feedback loop, circadian locomotor output cycles kaput (CLOCK) and brain and muscle Arnt-like protein 1 (BMAL1) heterodimerize and bind to E-box-containing sequences in a number of genes including the three period genes (Per1, Per2, and Per3) and two cryptochrome genes (Cry1 and Cry2). Next in the sequence, PER and CRY proteins dimerize and translocate to the nucleus, where CRY proteins can directly stall the action of CLOCK and BMAL1. Further to this mechanism, the CLOCK and BMAL1 proteins control the expression of Rev-erbα and Rora (retinoic acid–related orphan nuclear receptors), which subsequently inhibit or trigger Bmal1 transcription, respectively, by acting via the Rev-Erb/ROR response element in the promoter.

There are a number of important catalysts that adjust the timing of the molecular clock through the biochemical processes of phosphorylation, sumoylation, etc. Casein kinases phosphorylate the PER, CRY, and BMAL1 proteins, affecting their functioning and nuclear translocation. Another enzyme, glycogen synthase kinase 3 beta (GSK3β), also phosphorylates the PER2 protein, promoting its movement into the nucleus.94

4. Circadian mechanisms in mood disorders

The action of the antidepressant agomelatine, a melatonin receptor agonist and a weak 5-HT2C receptor antagonist, causes enhancement in monoaminergic neuronal activity, pointing to a regulatory function of melatonin in monoaminergic transmission and connecting the circadian and monoamine neurotransmitter systems.95 Circadian rhythm dysregulation results in an increase in PIC, and subsequently TNF-α, INF-γ, and IL-6 alter the sleep/wake cycle and circadian gene expression via NF-κB transduction. 96 Glucocorticoids show a robust diurnal variation in rhythm and crest in level just before wakening, when melatonin falls to undetectable levels in the peripheral blood. CRY proteins acting together with the GR in a ligand-dependent manner induce rhythmic suppression of its activity. Moreover, CLOCK directly acetylates GR, leading to indifference to cortisol in the morning and increased affinity at night when acetylation is annulled.97 The immediate control of GR function by circadian genes is allegedly of great importance in modulating the reaction to persistent stress.98

There is a very high incidence of metabolic diseases in mood disorders, whereas circadian oscillations in the liver, stomach, adipose tissue, and gut are very strong.99 The peptides that play a role in metabolism and feeding comprise ghrelin, orexin, leptin, and cholecystokinin, and these demonstrate a noteworthy circadian rhythm in activity. The association between corpulence, stress, and poor sleep signifies a vicious cycle, and mood disorders can be included in this sequence for a subgroup of patients.100

The nicotinamide cofactors NADPH/NADP+ and NADH/NAD+ have lately been recognized as indispensable associates of CLOCK, NPAS2, and SIRT1, establishing an immediate connection between the redox status of the cell and circadian rhythms.101 Besides, CLOCK/BMAL1 directly controls the expression of nicotinamide phosphoribosyltransferase in a circadian manner, determining the production of NAD+ in the cell over the daily cycle.102 Brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase B (TrkB) are known to be crucial in the effects of antidepressants and mood stabilizers, and both of these have a strong circadian rhythm in expression in the hippocampus. 103 BDNF is no longer effective as a neurotrophic agent in the nonexistence of a normal diurnal rhythm of glucocorticoids, demonstrating that the daily oscillations of cortisol control the salutary activity of psychotropic drugs on neuronal growth via BDNF/TrkB signaling in the hippocampus.104 Fig. 5 gives an overview of the links between mood disorders and the circadian system.

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.

CONTRIBUTORY NEUROBIOLOGICAL MECHANISMS IN MAJOR MENTAL DISORDERS

There are many commonalities in the psychopathology of major depressive disorder, BD, and schizophrenia. Schizoaffective disorder is an intermediary phenotype and the retention of the "not otherwise specified" category in current classificatory systems reflects that many patients with major mental disorders do not easily fall into clear-cut diagnoses. This is demonstrated in neurobiological groundwork, and recent studies bear out this actuality. Table 2 provides an outline of this area of research.

TABLE 2
Shared mechanisms in the pathophysiology of major psychiatric disorders

NEUROPROGRESSION IN BIPOLAR DISORDER

BD has its usual onset in adolescence or early adulthood following a stressful event. Its course is malignant in many cases, marked by repeated affective episodes of increasing severity, development of substance use disorders, and myriad physical diseases with increased morbidity and mortality. The allostatic load model has been proposed to explain the progressive nature of BD. Table 3 recapitulates representative studies delineating the pathophysiological foundations of this framework.

TABLE 3
Neurobiological correlates of illness progression in bipolar disorder

IDENTIFICATION OF NEW THERAPEUTIC TARGETS

Throughout this article, an effort has been made to allude to novel therapeutic measures for BD, which is in essence a recalcitrant condition. Refractoriness to conventional mood stabilizers is the rule rather than the exception and targeting the monoamine neurotransmitter systems results in less than satisfactory results for many patients. In this regard, Table 4 presents new therapeutic targets that may open fresh avenues in the treatment of BD.

TABLE 4
Potential new therapeutic targets with relevance to bipolar disorder

UNIFYING HYPOTHESIS OF THE NEUROBIOLOGY OF BIPOLAR DISORDER

Conceptualized as a polygenic condition, the expression of BD is secondary to a stressful event. There is evidence for HPA axis dysregulation, failure of mechanisms that mediate resiliency, and breakdown of homeostasis. Fig. 6 illustrates key biological abnormalities that act in concert and explains why so many body systems are adversely affected by the bipolar diathesis.

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.

LIMITATIONS

The following caveats should be kept in mind while deducing any inferences with respect to the neurobiology of BD:

  • 1) Not all predisposed individuals are afflicted by BD, which underscore the issues of genetic epistasis and hitherto little known mechanisms that mediate resiliency.

  • 2) There are large gaps in knowledge due to the absence of good animal models replicating BD.

  • 3) Technological advances are needed to reproduce findings from animal research in human samples.

FUTURE DIRECTIONS

Ongoing research on the neurobiology of BD is leading to a better understanding of the condition. Dysfunction of the molecular clockwork genes has repeatedly been shown and circadian disturbances are present not only during acute episodes but also in remission. Future research should clarify how a dysregulated internal clock precipitates affective episodes and suggest measures that could lead to the prevention and early treatment of mood disturbances, which come in innumerable flavors. A vicious cycle of immune-inflammatory imbalance, increased oxidative stress, and derangement of metabolic parameters accompanied by neurophysiological disturbances is at the foundation of this disorder. Further research should elaborate this mechanism, providing a composite view of the initiation and progression of BD. Finally, more effective and targeted therapies are likely to be discovered that are not palliative but curative in nature.

CONCLUSION

A broad assessment of the current literature was done to bring to light the underpinnings of mood disorders in general and BD in particular. These are stress-related conditions with overt expression in individuals with an underlying genetic vulnerability. Modern neuroscience is utilizing animal models and conducting human research with increasingly sophisticated methods to unravel their pathophysiology. Significant strides have been made in understanding the neurobiology of affective illnesses, and in this regard new targets and biomarkers have been identified. Diverse biological systems act in concert in perpetuating the disorders. While obstacles in research remain in the basic scientific and clinical domains, there is no doubt that a representation is emerging that is providing a consolidated view regarding the development of these intractable conditions. It is hoped that new knowledge will translate into novel therapeutic measures that have both preventive and curative value for patients with bipolar spectrum disorders.

Notes

CONFLICT OF INTEREST STATEMENT:None declared.

References

    1. Hirschfeld RM. Differential diagnosis of bipolar disorder and major depressive disorder. J Affect Disord 2014;169 Suppl 1:S12–S16.
    1. Parker G. John Cade. Am J Psychiatry 2012;169:125–126.
    1. Muneer A. Pharmacotherapy of bipolar disorder with quetiapine: a recent literature review and an update. Clin Psychopharmacol Neurosci 2015;13:25–35.
    1. Pregelj P. Gene environment interactions in bipolar disorder. Psychiatr Danub 2011;23 Suppl 1:S91–S93.
    1. Craddock N, Sklar P. Genetics of bipolar disorder. Lancet 2013;381:1654–1662.
    1. 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.
    1. Post RM. Kindling and sensitization as models for affective episode recurrence, cyclicity, and tolerance phenomena. Neurosci Biobehav Rev 2007;31:858–873.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Girshkin L, Matheson SL, Shepherd AM, Green MJ. Morning cortisol levels in schizophrenia and bipolar disorder: a metaanalysis. Psychoneuroendocrinology 2014;49:187–206.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Jones KA, Thomsen C. The role of the innate immune system in psychiatric disorders. Mol Cell Neurosci 2013;53:52–62.
    1. Barbosa IG, Bauer ME, Machado-Vieira R, Teixeira AL. Cytokines in bipolar disorder: paving the way for neuroprogression. Neural Plast 2014;2014:360481.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Waetzig GH, Rose-John S. Hitting a complex target: an update on interleukin-6 trans-signalling. Expert Opin Ther Targets 2012;16:225–236.
    1. 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.
    1. 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.
    1. Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact 2014;224C:164–175.
    1. Nayernia Z, Jaquet V, Krause KH. New insights on NOX enzymes in the central nervous system. Antioxid Redox Signal 2014;20:2815–2837.
    1. Sorce S, Krause KH. NOX enzymes in the central nervous system: from signaling to disease. Antioxid Redox Signal 2009;11:2481–2504.
    1. 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.
    1. 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.
    1. Fatokun AA, Stone TW, Smith RA. Oxidative stress in neurodegeneration and available means of protection. Front Biosci 2008;13:3288–3311.
    1. Morris G, Berk M. The many roads to mitochondrial dysfunction in neuroimmune and neuropsychiatric disorders. BMC Med 2015;13:68.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Bitanihirwe BK, Woo TU. Oxidative stress in schizophrenia: an integrated approach. Neurosci Biobehav Rev 2011;35:878–893.
    1. Ramalingam M, Kim SJ. Reactive oxygen/nitrogen species and their functional correlations in neurodegenerative diseases. J Neural Transm (Vienna) 2012;119:891–910.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Brown NC, Andreazza AC, Young LT. An updated meta-analysis of oxidative stress markers in bipolar disorder. Psychiatry Res 2014;218:61–68.
    1. Joshi YB, Praticò D. Lipid peroxidation in psychiatric illness: overview of clinical evidence. Oxid Med Cell Longev 2014;2014:828702.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 2007;87:245–313.
    1. 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.
    1. Müller N, Schwarz MJ. Neuroimmune-endocrine crosstalk in schizophrenia and mood disorders. Expert Rev Neurother 2006;6:1017–1038.
    1. 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.
    1. 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.
    1. 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.
    1. Myint AM. Kynurenines: from the perspective of major psychiatric disorders. FEBS J 2012;279:1375–1385.
    1. Campbell BM, Charych E, Lee AW, Möller T. Kynurenines in CNS disease: regulation by inflammatory cytokines. Front Neurosci 2014;8:12.
    1. Schwarcz R, Bruno JP, Muchowski PJ, Wu HQ. Kynurenines in the mammalian brain: when physiology meets pathology. Nat Rev Neurosci 2012;13:465–477.
    1. Dantzer R, Walker AK. Is there a role for glutamate-mediated excitotoxicity in inflammation-induced depression? J Neural Transm (Vienna) 2014;121:925–932.
    1. Erhardt S, Olsson SK, Engberg G. Pharmacological manipulation of kynurenic acid: potential in the treatment of psychiatric disorders. CNS Drugs 2009;23:91–101.
    1. 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.
    1. 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.
    1. Leibowitz A, Boyko M, Shapira Y, Zlotnik A. Blood glutamate scavenging: insight into neuroprotection. Int J Mol Sci 2012;13:10041–10066.
    1. Myint AM, Kim YK. Network beyond IDO in psychiatric disorders: revisiting neurodegeneration hypothesis. Prog Neuropsychopharmacol Biol Psychiatry 2014;48:304–313.
    1. 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.
    1. 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.
    1. Gonzalez R. The relationship between bipolar disorder and biological rhythms. J Clin Psychiatry 2014;75:e323–e331.
    1. 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.
    1. Scott J. Clinical parameters of circadian rhythms in affective disorders. Eur Neuropsychopharmacol 2011;21 Suppl 4:S671–S675.
    1. 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.
    1. 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.
    1. Menaker M, Murphy ZC, Sellix MT. Central control of peripheral circadian oscillators. Curr Opin Neurobiol 2013;23:741–746.
    1. Ko CH, Takahashi JS. Molecular components of the mammalian circadian clock. Hum Mol Genet 2006 Spec No 2;15:R271–R277.
    1. McClung CA. Circadian genes, rhythms and the biology of mood disorders. Pharmacol Ther 2007;114:222–232.
    1. 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.
    1. 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.
    1. Landgraf D, McCarthy MJ, Welsh DK. Circadian clock and stress interactions in the molecular biology of psychiatric disorders. Curr Psychiatry Rep 2014;16:483.
    1. Cagampang FR, Bruce KD. The role of the circadian clock system in nutrition and metabolism. Br J Nutr 2012;108:381–392.
    1. Kim TW, Jeong JH, Hong SC. The impact of sleep and circadian disturbance on hormones and metabolism. Int J Endocrinol 2015;2015:591729.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. Etain B, Milhiet V, Bellivier F, Leboyer M. Genetics of circadian rhythms and mood spectrum disorders. Eur Neuropsychopharmacol 2011;21 Suppl 4:S676–S682.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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.

Metrics
Share
Figures

1 / 6

Tables

1 / 4

PERMALINK