Next Article in Journal
Lutathera®: The First FDA- and EMA-Approved Radiopharmaceutical for Peptide Receptor Radionuclide Therapy
Next Article in Special Issue
The Impact of Childhood Maltreatment on Intravenous Ketamine Outcomes for Adult Patients with Treatment-Resistant Depression
Previous Article in Journal
Larvicidal and Enzymatic Inhibition Effects of Annona Muricata Seed Extract and Main Constituent Annonacin against Aedes Aegypti and Aedes Albopictus (Diptera: Culicidae)
Previous Article in Special Issue
VitalSign6: A Primary Care First (PCP-First) Model for Universal Screening and Measurement-Based Care for Depression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Treatment Targets Based on Insights in the Etiology of Depression: Role of IL-6 Trans-Signaling and Stress-Induced Elevation of Glutamate and ATP

Retired pharmacologist, Gänsbühlgartenweg 7, CH4132 Muttenz, Switzerland
Pharmaceuticals 2019, 12(3), 113; https://0-doi-org.brum.beds.ac.uk/10.3390/ph12030113
Submission received: 25 June 2019 / Revised: 10 July 2019 / Accepted: 26 July 2019 / Published: 29 July 2019
(This article belongs to the Special Issue Antidepressants: Mechanistic Insights and Future Directions)

Abstract

:
Inflammation and psychological stress are risk factors for major depression and suicide. Both increase central glutamate levels and activate the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system. Both factors also affect the function of the chloride transporters, Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2), and provoke interleukin-6 (IL-6) trans-signaling. This leads to measurable increases in circulating corticosteroids, catecholamines, anxiety, somatic and psychological symptoms, and a decline in cognitive functions. Recognition of the sequence of pathological events allows the prediction of novel targets for therapeutic intervention. Amongst others, these include blockade of the big-K potassium channel, blockade of the P2X4 channel, TYK2-kinase inhibition, noradrenaline α2B-receptor antagonism, nicotinic α7-receptor stimulation, and the Sgp130Fc antibody. A better understanding of downstream processes evoked by inflammation and stress also allows suggestions for tentatively better biomarkers (e.g., SERPINA3N, MARCKS, or 13C-tryptophan metabolism).

1. Introduction

Major depressive disorder (MDD) is worldwide a leading cause of years lived with disability [1,2]. Unfortunately, drug development in psychiatry has stagnated during the last decades [3,4]. However, continuing research has led to the recognition of the importance of the immune system in psychiatric disorders [5,6], including depression [7,8]. Based on insights provided by ongoing academic research efforts, it is possible to reinterpret the wealth of information on the pathophysiological processes in major depressive disorder. This better understanding enables proposals for novel pharmacological treatment targets. The current review starts off with a reinterpretation of existing disease knowledge. This is subsequently used to list hitherto underexplored pharmacological treatment options. Finally, it provides proposals for novel biomarkers for endophenotypic depression symptoms, as well as suicide risk.

2. Stress as a Risk Factor for Depression and Suicide

Stress, including early life stress, is an important risk factor for major depression [9,10,11,12,13,14,15]. Early life stress (childhood neglect, physical or sexual abuse, or early parental loss) not only constitutes a major risk factor for depression, but also significantly increases suicide risk [16,17]. Stressors like family or romantic conflicts and legal or disciplinary problems can trigger suicidal behaviors [17] and manic episodes [18,19]. Stress-induced depression is characterized by deep mental and physical fatigue, disturbed non-restorative sleep, irritability, emotionality, concentration problems and memory disturbances [9]. Psychological stress can be investigated under laboratory conditions by a tool called the Trier social stress test (TSST) [20]. Application of the TSST frequently evokes marked increases in the activity of the sympathetic nervous system and the hypothalamic-pituitary-adrenal (HPA) axis (seen as increases in circulating levels of catecholamines, adrenocorticotrophic hormone (ACTH), and cortisol, and tachycardia) [20,21]. Interestingly, the circulating levels of interleukin-6 (IL-6) are also increased, as is the activity of the pro-inflammatory nuclear-factor kappa-B (NFκB)-pathway in monocytes [21,22,23,24]. Notably, TSST-induced pro-inflammatory activity is particularly strong in individuals with a history of childhood maltreatment [22]. Both the corticosteroid “stress”-hormones and the sympathetic nervous system represent important effector systems. Chronic social stress in laboratory animals and in humans has been shown to stimulate gene transcription of pro-inflammatory proteins in monocytes via an activation of β-adrenoceptors [25,26] and α1B receptors [27]. Repeated social defeat in mice has been observed to increase the number of monocytes trafficking to the brain, while plasma levels of IL-6 and the weight of the spleen have been seen to increase [28]. Blockade of β-adrenoceptors during the defeat period has been shown to prevent these changes [28]. Notably, the observation that stress leads to activation of the immune system has been conceptualized as a preparation for subsequent wounding and possible infection [14,15]. This interpretation is supported by the observation that defeat-stress in mice improves bacterial clearance by phagocytes [29]. As in peripheral monocytes, in the central nervous system stress can result in activation of microglial cells [30,31,32] (reviewed by [5,33,34]). Stress-induced microglia activation is microscopically visible in terms of an alteration of the microglia phenotype [35,36].

2.1. Stress Modifies KCC2 and NKCC1 Activity

The direction of anion-flux through the gamma-aminobutyric acid-A (GABAA) channel depends on the electrochemical gradient for the chloride ion and is determined by the expression and activity of two trans-membrane chloride-transporters, K-Cl-cotransporter-2 (KCC2) and Na-K-Cl-cotransporter-1 (NKCC1) (reviewed by [37]). KCC2 extrudes chloride, while NKCC1 performs chloride-influx. A reduction in the expression of KCC2 or the inhibition of the activity impairs chloride-extrusion, which leads to higher intracellular chloride levels [37]. Under such conditions, a GABA-induced opening of the GABAA-channel causes a chloride efflux and thus a depolarization.
Corticotrophin-releasing factor (CRF)-producing cells within the paraventricular nucleus (PVN) are activated by glutamate-neurons and noradrenergic neurons [9,38,39] but under inhibitory control by GABA-neurons [38,40,41]. An altered direction of the chloride flux through GABA-channels thus has consequences for the activity of the HPA axis.
A variety of experiments in rodents show that stress affects the function of chloride transporters. For instance, acute restraint stress in rats has been shown to activate α1-adrenoceptors in the PVN [40], which suppressed the activity of the KCC2. Consequently, restraint stress was observed to lead to a depolarization shift, rendering GABA-agonists activating rather than inhibitory [40]. In mice, acute restraint stress has been seen to reduce the cell-surface expression of KCC2 [41]. The modified chloride-gradient has been observed to cause an increase in excitatory GABA transmission. Interestingly, under this particular condition, GABAA-mimetic neurosteroids were found to raise the firing frequency of CRF neurons and caused significant increases in circulating corticosterone-levels [41]. Moreover, the anxiolytic activity of neurosteroids has been seen to be altered to an anxiogenic effect [41]. In a model of chronic stress, Gao and colleagues [42] found that repeated exposure to unpredictable mild stress increased the protein levels of NKCC1 in CRF neurons of rat PVN. The NKCC1 levels remained elevated for at least 10 days [42]. Moreover, the authors confirmed that acute restraint stress decreased KCC2 levels in the PVN; however, this effect was short-lasting and returned to baseline within 5 days post stress. These data indicate that acute stress impairs GABA control over CRF neurons via a decrease in KCC2 function, whereas chronic stress-induced impairment is mediated via an increase in NKCC1 expression. The depolarizing activity of GABA is not restricted to the PVN and similar effects have been noticed in the spinal cord and are thought to play a role in neuropathic pain and hyperalgesia [43,44]. The effect of stress on chloride transporters and consequently on GABA-function may provide a mechanistic explanation for “the GABAergic deficit hypothesis of major depressive disorder” [14,45]. A dysfunctional GABA-inhibition would lead to a diminished suppression of glutamate neurons and to an increased activity of the sympathetic nervous system and HPA axis. The reader may notice that this process represents a vicious circle. A further vicious circle may be formed when high levels of cortisol activate the mineralo-corticoid receptor (MR) because MR-activation is known to increase the metabolic stability of NKCC1 [46].

2.2. Hyperactivation of the HPA Axis

A prolonged activation of the HPA axis may result in an exhaustion (atrophy) of cortisol production by the adrenal. Low circulating levels of cortisol are observed in a number of chronic stress disorders in humans. This includes atypical depression [47], posttraumatic stress disorder [48], and suicide attempts [49]. Since activation of the HPA axis inhibits immune-cell function [15,50,51], an exhausted cortisol production might lead to a hyper-immune state with increased levels of IL-6 [47,52].

2.3. Stress Induces Increased Central Glutamate Signaling

Immobilization stress in rats has been found to significantly increase extracellular glutamate levels in the hippocampus and frontal cortex [53,54]. A role of corticosterone in this response is indicated by the observation that adrenalectomy markedly attenuates the stress-induced increase in glutamate [53,54]. Additionally, chronic unpredictable mild stress has been observed to cause an increase in extracellular glutamate levels in these two brain areas, acting on N-methyl-D-aspartate (NMDA) receptors that couple to the NFκB signaling pathway [55]. Furthermore, foot-shock stress, as applied in the learned helplessness model, has been shown to cause a marked increase in neuronal glutamate release in the rat frontal cortex, which again is dependent on glucocorticoid receptor (GR) activation [56]. Finally, chronic mild stress has been found to increase the expression of NMDA-NR1 receptors on CRF neurons in the rat PVN, which in turn leads to an increased excitation of the HPA axis [39]. These data indicate that stress causes a corticosterone-driven increase in neuronal glutamate release [57]. Neuron-derived glutamate provokes adenosyl-triphosphate (ATP)-release by astrocytes, which represents a danger-signal for microglial cells [58]. Chronic psychosocial stress decreases astroglial plasticity in the rodent hippocampus and frontal cortex [59,60]. In another experiment, four sessions of restraint stress elevated corticosterone levels in mice in vivo. Blockade of corticosterone synthesis, blockade of the glucocorticoid receptor by RU486, or blockade of NMDA-receptors by MK801 prevented stress-induced microglia proliferation [50]. MK801 also prevented microglia-proliferation following exogenous corticosterone administration to non-stressed mice [50]. These results suggest that stress activates the GR receptor on neurons, which leads to increased extracellular levels of glutamate. This glutamate may subsequently activate NMDA receptors on astrocytes, and these then produce ATP (see Figure 1). Data by Ferrini and De Koninck indicate that ATP activates P2X4 receptors on microglia, and this provokes the release of brain-derived neurotrophic factor (BDNF) [44]. BDNF will activate tropomyosin receptor kinase-B (TrkB) receptors on neurons and reduce the activity of KCC2 (promoting GABA-induced depolarization) [44]. This summary indicates that stress in the CNS triggers a sequence of events that ultimately leads to an increase in neuronal network excitability [44].
ATP also stimulates microglia to assemble the components of the ‘NOD-, LRR- and pyrin-domain-containing protein 3′ (NLRP3) inflammasome [58]. The NLRP3 inflammasome mediates the cleavage of pro-interleukin-1β (pro-IL1β) to mature IL1β [34] and sensitizes microglia to generate a stronger pro-inflammatory response [61]. Frank et al. have exposed rats to inescapable tail shocks. They noted that the responsiveness of hippocampal microglia to lipopolysaccharide (LPS) was raised by the tail-shock procedure, but, notably, this sensitization was absent if the rats had been adrenalectomized or treated with the GR-antagonist RU486 prior to the tail shocks [61]. Chronic stress reduced the dendritic connectivity and size of the hippocampus and prefrontal cortex (PFC) [62,63,64], while inhibition of NMDA receptors or inhibition of glutamate-release blocked the effect of chronic stress on dendritic atrophy (reviewed by [57]). Stress-induced reduction in neuropil is likely to contribute to impairment in cognitive function [65,66].

2.4. Stress Increases Levels of Interleukin-6

In human subjects, physical and psychosocial stress can cause central and peripheral IL-6 release [15,67,68,69]. Daily life stressors evoked a particularly large increase in IL-6 in individuals who had suffered from childhood trauma [22,70]. In addition, rodent studies provide ample evidence for stress-induced IL-6 release. For instance, chronic unpredictable mild stress has been shown to elevate the levels of the IL-6 protein in the hippocampus and alter animal behavior [71]. Treatment with the NMDA-blocker ketamine has been found to provoke a rapid reduction in circulating IL-6 levels and normalized ‘depressed’ behavior [71]. Chronic mild stress, similar to other forms of stress such as acute foot shock or chronic intermittent cold exposure, has been seen to increase IL-6 levels in the hypothalamus of rats and activate the IL-6 – gp130 – STAT3 (signal transducer and activator-3) signaling pathway [72]. In apparent contradiction to these results is the observation that rats which are resilient to learned helplessness (a further rodent model of depression) have lower brain levels of IL-6 than their helpless counterparts [73]. The solution to this apparent conundrum is that IL-6 signals in two fundamentally different ways. When IL-6 acts via the membrane-bound IL-6 receptor (called “classical signaling”) it mainly causes neuron-protective effects [74,75,76]. However, when IL-6 acts in conjunction with the membrane-shed moiety of the IL-6 receptor (soluble IL6R, or sIL6R), a process called “trans-signaling”, it provokes mainly pathologic effects [75,77,78,79]. A detailed discussion about factors that influence the use of the two signaling modes is provided in Section 3.1 below.

3. Inflammation as a Risk Factor for Depression and Suicide

Whereas the previous sections dealt with the effect of stress on depression-related parameters, the next sections deal with inflammation. It may be noted that central and peripheral inflammation cause effects that resemble those that are due to stress. Miller and Raison [80] have formulated the “pathogen defense hypothesis of depression”, which posits that risk alleles for depression are the ones that inhibit growth of pathogens by a pro-inflammatory activity, and therefore are conserved in the human genome (Table 1).
Depression is comorbid with numerous disorders involving inflammation (Table 1; for reviews see [6,14,15,95,96]). Conversely, inflammation markers such as IL-6 are often elevated in depression. For instance, depressed patients with metabolic syndrome [97] or patients suffering from atypical depression (e.g., hyperphagia, weight gain, hypersomnia) display high plasma levels of IL-6 [98]. Meta-analyses of cytokine levels show that plasma IL-6 levels [99,100,101,102], tumor necrosis factor-α (TNFα) [99,100], and circulating levels of C-reactive protein (CRP) [101,102] are elevated in patients with MDD. Moreover, IL-6 has been identified as an important susceptibility gene for major depression [103]. Serum IL-6 and sIL6R have also been found to be higher after delivery, especially in women with a history of depression [104]. Clinical studies measuring sIL6R in unipolar depression are still sparse, but two meta-analyses in bipolar depression have reported that circulating levels of IL-6 and sIL6R were higher in patients than in healthy controls [105,106]. Clearly elevated levels of sIL6R in serum and cerebrospinal fluid (CSF) have been observed in patients with neurological inflammatory diseases such as multiple sclerosis [107]. In a systematic literature search of studies concerning cytokine levels in patients with suicidal ideation, suicide attempts, or suicide completion, elevated IL-6 in CSF, blood, and postmortem brain tissue was found in 8 out of 14 studies [108]. In suicide attempters, plasma IL-6 levels and CSF IL-6 levels did not correlate, and, interestingly, IL-6 levels were higher in plasma than in CSF ([109,110] but see also [111]). High plasma levels of IL-6 were associated with increased suicidal ideation [112] and suicide attempts [113], and they were independent of depression severity [112,113,114]. Moreover, IL-6 levels in plasma are also associated with suicide endophenotypic behaviors, such as increased extraversion, impulsivity, and violent attempts [109,115,116]. Measurements of sIL6R levels in individuals showing suicidal behavior have, apparently, not yet been performed [117]. Apart from these clinical studies, rodent depression models such as chronic unpredictable mild stress, learned helplessness, maternal separation, forced swim test/tail suspension test, prenatal stress, and olfactory bulbectomy are also associated with significant increases in IL1β, TNFα and IL-6 in the brain and blood [68,118].

3.1. IL-6 Trans-Signaling in Depression

Although leukocytes, fibroblasts, adipocytes, keratinocytes, and endothelial cells all secrete IL-6 [119], about 30% of circulating IL-6 is derived from adipose tissue [13]. Homeostatic production of IL-6 leads to plasma levels of 1–10 pg/mL, but during infection, inflammation, or cancer, these levels are elevated to the lower ng/mL range. Signal transduction by IL-6 involves the formation of a hexamer built from IL-6, the membrane-bound IL-6-receptor, and the gp130 protein [74,120,121]. This form of signaling is occasionally called “cis”-signaling but is more often referred to as “classical”-signaling. Expression of the IL-6-receptor (IL6R) is confined to neutrophils, monocytes, CD4 T-cells (but not CD8 T-cells), memory T-cells, and hepatic and osteoblast cell lines [122,123]. In the brain, IL6R is strongly expressed by microglia, but very weakly (or not at all) by astrocytes, oligodendrocytes, and neurons [77,124,125]. Two metallo-proteinases, ‘a desintegrin and metalloproteinase-17′ (ADAM17) and ADAM10, are able to cleave the extracellular part (the ‘ectodomain’) of the IL-6-receptor [74,122,126,127]. The resulting moiety (soluble IL6R, or ‘sIL6R’) still binds IL-6, and the IL-6/sIL6R complex can activate cells that express gp130, notably with no need for membrane-bound IL6R [122,127]. This is called IL-6 “trans-signaling” ([122]; for a review see [76]). ADAM10 and ADAM17 activity is induced by phorbol-esters, by the cytokines IL1β and TNFα, and by apoptotic pathways (e.g., DNA-damage, UV radiation, and Fas ligation) [127]. Additionally, CRP provokes an increase in sIL6R production [123,128]. Hence, in principle, sIL6R can be produced by the sympathetic nervous system (via the α1 receptor-Gq-protein-kinase-C (PKC)-diacylglycerol (DAG) signaling pathway (mimicked by phorbolesters)) and during inflammation (when levels of CRP, TNFα, or IL1β are elevated). Hepatocytes, neutrophils, and CD4+ T-cells represent the major sources of circulating sIL6R [122,127,128]. In human serum sIL6R is always present at relatively high concentrations of 25–75 ng/mL, and these levels are 2–3 fold increased during inflammation [119]. Soluble-IL6R circulates at elevated levels in various diseases [128], including major depressive disorder [129] and bipolar disorder [105,106]. The ratio of IL-6 to sIL6R/IL-6 determines to which degree trans-signaling will occur [119,123]. Importantly, circulating sIL6R may cross the blood brain barrier and cause IL-6 trans-signaling in the CNS [130].

3.2. IL-6 Trans-Signaling in the Brain

Mice with a genetic overexpression of IL-6 by astrocytes have been found to respond to restraint stress with an exaggerated rise in plasma corticosterone [131]. This is consistent with data in humans that IL-6 activates the HPA axis [132]. Since this is a neuronal response to IL-6, it is likely that it involves trans-signaling via sIL6R. Inflammation in the CNS leads to production of reactive oxygen species, whereas oxidative stress is known to decrease in cell-surface expression due to a rapid decline in KCC2 tyrosine-phosphorylation [133]. In addition, in sensory nerves it has been shown that IL-6 signaling also alters phosphorylation of NKCC1, which in this case led to higher cell-surface expression and higher intracellular chloride levels [134]. It is conceivable that IL-6 might provoke similar effects in central neurons. As discussed in earlier sections, both mechanisms, a decline in KCC2 and an increase in NKCC1, would contribute a depolarizing activity of GABAA. IL-6 trans-signaling therefore increases the synaptic excitation/inhibition ratio [135,136]. It is likely that IL-6 will not only enhance the activity of the HPA axis but also the activity of the sympathetic nervous system (c.f. [137]). Peripheral and central inflammation in animals indeed increases the activity of the sympathetic nervous system [138,139,140].
Under physiological conditions, astrocytes assume numerous supportive functions, including structural support, neurovascular coupling, regulation of extracellular K+, uptake of neurotransmitters, and metabolic support of neurons [141]. Astrocytes are essential for regulation of glucose uptake and lactate release, uptake of glutamate and release of glutamine (required for glutamate as well as GABA neurotransmission), and uptake of glutathione precursors and the release of glutathione [141,142]. During brain inflammation, however, microglia is polarized to the activated M1-phenotype, and this, in turn, stimulates astrocytes to attain what is called a “neurotoxic reactive”, or “A1”-phenotype ([143]; see Figure 2). A1 astrocytes secrete an unknown factor that is highly toxic to a subset of neurons and to mature oligodendrocytes [143,144]. The sIL6R/IL-6 complex is a conceivable candidate for this toxic factor, but this has not been tested yet. Since corticosteroids inhibit the pro-inflammatory phenotype of microglial cells [145], it is likely that during brain inflammation corticosteroids are neuroprotective, which is in contrast to the stress-induced pathology described in Section 2.3. This makes the glucocorticoid receptor both a part of the problem and a part of the solution [146].
The data summarized in this section show that IL-6 in the pro-inflammatory trans-signaling mode provokes much of the downstream effects that are also observed after exposure to stress. It is evident that both stress and inflammation lead to activation of the sympathetic nervous system, the HPA axis, and an enhanced GABA-depolarization (presumably leading to anxiety, psychopathology, and cognitive decline), as well as somatic ‘sickness’ symptoms (see Figure 3). Although causing a similar spectrum of symptoms, it is likely that the two inputs, stress or inflammation, will differ in the intensity of their biological outputs [47,147]. It will be interesting to read future scientific literature to see if it fits the idea that melancholic and atypical depression are the respective outputs of the stress and the inflammation arms.

4. Potential for Therapeutic Intervention

4.1. Intervening in the Sequence of Events Provoked by Stress

Stress has a profoundly negative effect on the viability of neurons (Figure 1). The mechanism has been investigated in detail in in vitro experiments [135,148,149]. Corticosterone-induced neuron death is associated with a decrease in the activity of protein kinase-B (PKB or Akt) and an increase in glycogen-synthase kinase-3 (GSK3)-activity [135,148,149]. Factors that increase PKB-activity like leptin [135], insulin-like growth factor-1, [149], or inhibition of GSK3 with lithium, inhibit neuronal loss [148]. Zhang et al. ([150]) describe that dexamethason-induced apoptosis of neonatal hippocampal neurons involves the assembly of the NLRP1 inflammasome via an increased K-efflux through ‘big-K’ (BK) potassium channels. The effect of dexamethason was ascribed to an immediate effect on the electrophysiology of BK-channels but also to an increase in mRNA and protein levels of BK following a chronic (28 days) treatment of mice in vivo [150]. The GR-antagonist RU486 and the big-K inhibitor iberiotoxin blocked dexamethason-induced apoptosis [150]. This short summary shows that negative effects of stress can be diminished by multiple mechanisms, including GSK3-inhibition and blockade of the big-K potassium channel. These represent potential drug-development targets for treatment of depression.
The next logical target for interruption of the sequence of events shown in Figure 1 is NMDA blockade. The antidepressant activity of the NMDA-channel blocker ketamine is now well established but there are still doubts as to whether the antidepressant activity is mediated by NMDA-inhibition [57,151,152]. Consistent with data in Figure 1, the antidepressant effect of ketamine involves a rapid increase in the expression of BDNF and subsequent TrkB receptor activation [57,152,153]. However, another NMDA-channel blocking drug, memantine, has been observed to fail to produce antidepressant activity in humans (reviewed by [151]). Whilst BDNF expression and TrkB-activation is induced also by effective treatments like electroconvulsive shock therapy or tricyclic antidepressants [153], memantine has been shown to fail to induce BDNF transcription [151]. This result indicates that the degree of NMDA blockade by memantine might be too small. Another argument that casts doubt on the NMDA-mechanism of action is the observation that a metabolite of ketamine, 2R,6R-hydroxy-norketamine, lacks affinity for the ketamine-binding site but has still been found to generate a rapid antidepressant-like effect in animal studies [152]. This latter argument is not as strong, since it may well be that the metabolite simply acts via a different pharmacological activity (e.g., via activation of opiate μ-receptors; see [152]). BDNF decreases the function of the chloride extruder KCC2, thus promoting a polarizing activity of GABA [43,44,154,155]. It has been argued that a high intracellular chloride concentration is beneficial for the formation of novel dendrites and synapses [134,156].
The last evident target from the cascade pictured in Figure 1 is the P2X4 channel. Three subunits of P2X are required to form a functional channel and each has to be stimulated by ATP to open the channel [157]. Due to its effects on the NLRP3 inflammasome-formation in microglia and macrophages, the P2X4-related (P2X7) channel is a well-known target for depression, and several selective P2X7-inhibitors are currently under development [157]. The P2X4 channel is mostly known for its role in the development of neuropathic pain, and some lead compounds for drug development have been described [157,158,159]. Development hurdles are selectivity, bioavailability, brain penetration, and poor water solubility, as well as species differences in pharmacology [157,158,159]. Importantly, the blockade of P2X4 prevents BDNF-release and this ultimately results in a reduction in mRNA levels of KCC2 [159].

4.2. Intervening in the Sequence of Events Provoked by Inflammation

The sequence of events shown in Figure 2 is by no means as thoroughly validated as the one discussed in Section 4.1. Nevertheless, concerning depression, there can be no doubt that IL-6 trans-signaling has important pathophysiological consequences (see Section 3.1 and Section 3.2). In the IL-6-receptor family, the receptors for leukemia inhibitory factor, ciliary neurotrophic factor, and others associate with the Janus-kinases JAK1 and JAK2 [160], but gp130 exclusively associates with the tyrosine kinase-2 (TYK2) [161]. Since the IL-6 receptor is non-signaling, this implies that IL-6 in both classic and trans-signaling modes requires TYK2 for downstream signaling. Therefore, one way to block pathological IL-6 trans-signaling would be via TYK2 kinase-inhibition. Large pharmaceutical companies apparently have realized that selective TYK2 inhibition (vis-à-vis other kinases, in particular also to JAK1 and JAK2) may represent a worthwhile development target [161,162,163]. However, an obvious disadvantage of this approach is that it will also inhibit the desirable IL-6 classic signaling. Antibodies directed against IL-6 or IL6R suffer from the same disadvantage. Importantly though, this is not the case for a recombinant derivative of the soluble gp130 protein, “sgp130Fc” [76]. Sgp130Fc is sgp130 bound to the Fc portion of IgG, and acts as a specific inhibitor of IL-6 trans-signaling [127]. During sepsis, sgp130Fc has been shown to inhibited sIL6R signaling while the anti-inflammatory classic signaling remained unaffected and regenerative proliferation was retained [76,127]. Sgp130Fc (Olamkicept® Conaris/Ferring) is in phase II clinical trials. Sgp130fc has shown efficacy in numerous preclinical inflammation models, including CNS inflammation [76], but its effect in depression models remains to be tested. Apart from the need for a parenteral route of administration, a further potential issue could be an insufficient blood-brain passage for those cases where the depressogenic inflammation is within the central nervous system.

4.3. Common Pathways Activated by Stress and Inflammation

A further useful approach to inhibit inflammation is by stimulation of the vagus nerve [164] or mimicking the effects of the vagus by nicotinic α7-agonists [165,166,167]. Electrical stimulation of the efferent vagus nerves in rats prevented LPS-induced endotoxic shock [168] and reduced secretion of IL-6 and TNFα [169]. It has been suggested that exercise, controlled breathing, relaxation therapies and fish-oil increase the activity of the vagus nerve and decrease production of TNFα and IL-6 [164]. Electrical stimulation of the vagus nerve has been shown to improve major depression in treatment-resistant patients [164,170,171,172]. There is furthermore an extensive preclinical literature that stimulation of the nicotinic α7-receptor with agonists like acetylcholine, choline, carbachol, nicotine, or the relatively selective agonist GTS-21 causes a reduction in inflammation-induced cytokine-release from human and rodent microglia cells and macrophages [165,167,168,172,173,174,175,176]. Although shown thus far during brain maturation only, activation of nicotinic α7-receptors has the propensity to modulate chloride transporter levels [177,178]. This makes nicotinic α7 receptor activation an exciting target for drug development in depression, but up to now no development compound has been tested for depression and as of the year 2019, no compound has reached the market.
A more global overview of the down-stream consequences of stress and inflammation is provided in Figure 3. Cognitive function is worsened by acute uncontrollable stress exposure and involves an increase in noradrenaline/α1-adrenoceptor/PKC activation in the prefrontal cortex (reviewed by [62]). This has led to the use of the α1-adrenoceptor antagonist prazosin for treatment of stress-related disorders [62]. Centrally acting α2-adrenoceptor agonists like clonidine or guanfacine are alternatives for prazosin [62]. These compounds reduce the activity of the sympathetic nervous system [179] while stimulating the activity of the parasympathetic nervous system [180]. Unfortunately, their profound hypotensive effect [179] is a disadvantage for use in psychiatric indications. There are three subtypes of α2 receptors in human genome and of these it is the α2A subtype that seems responsible for cardiovascular activity [181]. Notably, the α2B receptor has received considerable attention, because a mutation in the α2B receptor gene was associated with higher performance in a cognitive task for emotional stimuli and a stronger emotional memory [182,183,184]. The mutant form of the receptor (a deletion of three amino acids) is relatively resistant to receptor-desensitization [185] and gives rise to an elevated sympathetic outflow [186]. These findings suggest that a selective α2B-receptor antagonist could be useful to dampen excessive responding to emotional stimuli, in particular in homozygous carriers of the mutant form of the α2B-receptor.
There are a number of ways in which high intracellular chloride levels can be reduced. The most obvious is the inhibition of the NKCC1 transporter. This can be done with registered drugs like the diuretic bumetanide (review by [187]) or, interestingly, by oxytocin-spray [187]. A further possibility is inhibition of kinases like lysine deficient kinase (WNK) and SPS1-related proline/alanine-rich serine-threonine kinase (SPAK), as they regulate the cell surface-persistence of NKCC1 and KCC2 [188]. However, seemingly, the discovery and development of such kinase-inhibitors is not far advanced.

5. Biomarkers

5.1. Biomarker for IL-6 Trans-Signaling

In contrast to the psychological symptoms of depression (mood symptoms, anxiety, irritability, and cognitive alterations), the inflammation-induced vegetative symptoms (flue-like symptoms, fatigue, or anorexia) respond poorly to treatment with antidepressants [15,189,190]. This implies that a full remission of depression is achieved only when the driving force behind the vegetative symptoms is eliminated. IL-6 in its trans-signaling mode is the prime suspect for this symptom cluster (see Figure 3). Indeed, successful antidepressant treatment is associated with a reduction in plasma IL-6 levels [100,129,191,192,193,194,195]. However, since IL-6 levels do not necessarily reflect the degree of trans-signaling, one would need a marker for trans-signaling. Cellular internalization of the complex IL-6/sIL6R is slower than that of IL-6 bound to membrane-localized IL6R. Therefore, IL-6 trans-signaling leads to stronger and longer lasting intracellular signaling than IL-6 classic signaling [76]. SERine proteinase inhibitor-A3N (SERPINA3N) is an example of a gene that is regulated by trans-signaling but not via classic signaling [77], and probably there are more examples to be found. These genes could serve as biomarkers for the sickness syndrome-related symptoms of depression and suicide risk [123].

5.2. Biomarkers for Stress-Induced Effects

As a biomarker for the stress pathway to depression, one could propose salivary α-amylase [196,197]. Whether salivary α-amylase (sAA) is also a useful marker for suicidality is questionable, since relatives of suicide completers unexpectedly have been found to display a blunted stress-induced sAA response [198]. Stress-induced activation of the sympathetic nervous system can lead to activation of PKC, and this may promote sIL6R shedding and cognitive decline (see above). One of the substrates of PKC is the protein myristoylated alanine-rich C-kinase substrate (MARCKS) [199,200]. Interestingly, the expression level of MARCKS has been shown to be consistently increased in circulating leukocytes in different cohorts of suicidal bipolar depression-patients [201]. As a read-out of peripheral activity of the sympathetic nervous system, it might therefore be worthwhile to study phosphorylation levels of MARCKS in white blood cells of depression patients.
The parasympathetic nervous system is the physiological opponent of the sympathetic nervous system. A low activity of the parasympathetic nervous system can be quantified by low heart rate variability (HRV). Low HRV has been detected in humans suffering from stress [202] or depression [203,204,205] as well as in suicidal individuals [206]. Depression severity has been observed as being negatively correlated with HRV [203,204]. It would be useful to test if improvements in HRV correlate with amelioration of stress-induced depression symptoms.
Hypercortisolemia and dysregulation of the HPA axis are often found in severe forms of depression [81,207,208,209]. In contrast, atypical depression (characterized by hyperphagia and hypersomnia) is associated with low cortisol levels [207]. Hypocortisolemia is also frequently observed in fibromyalgia, chronic fatigue syndrome, and post-traumatic stress disorder [207]. In patients with depression, high cortisol levels in blood or saliva are associated with future completed suicide [210,211,212], but, remarkably, abnormally low cortisol levels are also a risk factor for suicide [49,198,213]. Moreover, there is evidence that glucocorticoids may diminish suicide numbers [214]. This short overview shows that both high and low cortisol levels are associated with depression and suicide risk, and this of course jeopardizes its utility as a biomarker.
In principle, the same is also true for the biomarker glutamine-synthetase. The enzyme glutamine-synthetase (glutamate ammonium ligase (GLUL)) is exclusively expressed in astrocytes and promotes the synthesis of glutamine from glutamate (reviewed: [215]). Astrocytes play a central role in both stress-induced activation of microglia and inflammation-induced neuron loss (Figure 1 and Figure 2). LPS and inflammatory cytokines inhibit glutamine-synthetase activity and also, as a consequence, glutamate uptake, glutamine synthesis, and neuronal-protection are diminished [216]. Conversely, the neuroprotective effect of astrocytes is increased after forced expression of GLUL [216]. The activity of GLUL in astrocytes is increased by glucocorticoids and glutamate but reduced by glutamine [217]. Dexamethasone has been found to induce an increase in synthesis and activity of GLUL in astrocytes in culture, whereas noradrenaline, in itself ineffective, potentiated GLUL activity [217]. Since GLUL activity is also stimulated by glutamate, the data by Hansson ([217]) indicate that all three stress factors (noradrenaline, corticosterone, and glutamate) increase GLUL function. Thus, ‘stress’ (Figure 1) and ‘inflammation’ (Figure 2) display opposite effects on GLUL function in astrocytes. Lithium, via an increase in β-catenin, activates the transcription of the GLUL gene [218,219]. Notably, β-catenin levels are diminished in post mortem brains of suicide victims [220], whereas Li has an anti-suicidal activity [221,222]. The proposal that low GLUL levels/activity could be a biomarker for suicide [215] is therefore jeopardized, due to the finding that GLUL levels become elevated under stress conditions.

5.3. Biomarkers for Inflammation-Induced Effects

The value of cortisol or GLUL levels as biomarkers for depression and suicide could eventually be rescued if we were able to distinguish between stress-induced and inflammation-induced consequences (Figure 3). While stress is thought to lead to depression with melancholic symptoms, inflammation tends to result in atypical depression [147,223]. Roughly one third of MDD patients have clearly elevated cytokines [224,225,226,227]. Patients in this subgroup are frequently obese [225,227,228] and/or suffer from metabolic syndrome [98,229] and cardiovascular disease [96]. They often display atypical features such as increased appetite, hypersomnia, and fatigue [230]. Their depression symptoms may preferentially respond to dietary interventions such as dietary restriction or the fish-oil component eicosapentaenoic acid [227,231]. It may well be that patients with atypical depression are the ones that display a distinct suicide-endophenotype with increased extraversion, impulsivity, and violent attempts [109,115,116]. Infectious agents such as human immunodeficiency virus (HIV), neuro-borreliosis, and Toxoplasma gondii are associated with agitation, aggression, and violent suicide attempts [224,226]. Numerous inflammatory mediators [115,228], including IL-6 in its trans-signaling mode, increase the expression of the enzyme indoleamine 2,3-dioxygenase (IDO) [156]. IDO converts L-tryptophan to L-kynurenine, and this is further metabolized to quinolinic acid. In suicidal individuals increased levels of IL-6 [108,114], L-kynurenine [232] and quinolinic acid [233] have all been reported. An IDO-induced increase in the production of kynurenine, and quinolinic acid have negative consequences for the availability of tryptophan for serotonin and melatonin synthesis. Consequently, CSF, plasma, and urine levels of the serotonin metabolite 5-hydroxy-indolic acid (5HIAA) are often strongly diminished in suicidal individuals [234]. The same is true for melatonin [235,236]. The enzymatic activity of IDO can be measured by a relatively simple, non-invasive method. Teraishi et al. ([237]) orally administered C13-labeled tryptophan to MDD patients and respective controls. Exhaled 13C-CO2 was quantified over the next three hours. Compared to the controls, in the MDD patients the recovery rate and peak levels of 13C-CO2 were significantly larger. This method would also be suitable to study IDO-activity in patients at an increased risk of suicide.

6. General Remarks

From the above it is evident that the subgroup of patients with major depressive disorder that suffer from an inflammation-prominent form of the disorder can be easily identified by multiple biomarkers (cytokine levels, IL-6-trans-signaling, low 5HIAA, low GLUL, low(er) cortisol, high kynurenine and quinolinic acid, and high tryptophan metabolism). Putative novel treatments for these particular patients are sGP130fc (particularly in case of an inflammation outside the brain), or TYK2-inhibition. The remaining group of MDD patients is probably divided in a group defined by stress-induced depression, and a group with a mix of inflammation and stress. High cortisol levels, high α-amylase, PKC-activity, MARCKS-phosphorylation, and high GLUL are tentatively biomarkers for this group of patients with stress-induced depression. Novel treatments for this group could comprise low-dose α2-adrenoceptor agonists, β-blockers, and treatments that reduce high intracellular chloride levels (shifting the effect of GABA from depolarizing to inhibitory). Other treatment targets for this group of patients could be NMDA-blockade, P2X4-blockade, inhibition of the BK-potassium channel, and/or GSK3-inhibition. Vagus nerve stimulation and nicotinic α7 receptor agonists are particularly interesting targets because these interventions might improve both stress- and inflammation-induced symptoms and would be particularly suited to patients in whom depression is driven by a mix of stress and inflammation factors. The information collected in the current review could be used to generate and improve sequential treatment optimization paradigms, such as for instance that described by Kraus et al. [238].

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interests.

References

  1. Lim, S.S.; Vos, T.; Flaxman, A.D.; Danaei, G.; Shibuya, K.; Adair-Rohani, H.; Amann, M.; Anderson, H.R.; Andrews, K.G.; Aryee, M.; et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2224–2260. [Google Scholar] [CrossRef]
  2. Wittchen, H.U.; Jacobi, F.; Rehm, J.; Gustavsson, A.; Svensson, M.; Jonsson, B.; Olesen, J.; Allgulander, C.; Alonso, J.; Faravelli, C.; et al. The size and burden of mental disorders and other disorders of the brain in Europe 2010. Eur. Neuropsychopharmacol. 2011, 21, 655–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Monteggia, L.M.; Malenka, R.C.; Deisseroth, K. Depression: The best way forward. Nature 2014, 515, 200–201. [Google Scholar] [CrossRef] [PubMed]
  4. Morrow, J.K.; Tian, L.; Zhang, S. Molecular networks in drug discovery. Crit. Rev. Biomed. Eng. 2010, 38, 143–156. [Google Scholar] [CrossRef] [PubMed]
  5. Beumer, W.; Gibney, S.M.; Drexhage, R.C.; Pont-Lezica, L.; Doorduin, J.; Klein, H.C.; Steiner, J.; Connor, T.J.; Harkin, A.; Versnel, M.A.; et al. The immune theory of psychiatric diseases: A key role for activated microglia and circulating monocytes. J. Leukoc. Biol. 2012, 92, 959–975. [Google Scholar] [CrossRef] [PubMed]
  6. Najjar, S.; Pearlman, D.M.; Alper, K.; Najjar, A.; Devinsky, O. Neuroinflammation and psychiatric illness. J. Neuroinflamm. 2013, 10, 43. [Google Scholar] [CrossRef] [PubMed]
  7. Barnes, J.; Mondelli, V.; Pariante, C.M. Genetic Contributions of Inflammation to Depression. Neuropsychopharmacology 2017, 42, 81–98. [Google Scholar] [CrossRef] [PubMed]
  8. Setiawan, E.; Wilson, A.A.; Mizrahi, R.; Rusjan, P.M.; Miler, L.; Rajkowska, G.; Suridjan, I.; Kennedy, J.L.; Rekkas, P.V.; Houle, S.; et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry 2015, 72, 268–275. [Google Scholar] [CrossRef]
  9. Bartolomucci, A.; Leopardi, R. Stress and depression: Preclinical research and clinical implications. PLoS ONE 2009, 4, e4265. [Google Scholar] [CrossRef]
  10. Caspi, A.; Sugden, K.; Moffitt, T.E.; Taylor, A.; Craig, I.W.; Harrington, H.; McClay, J.; Mill, J.; Martin, J.; Braithwaite, A.; et al. Influence of life stress on depression: Moderation by a polymorphism in the 5-HTT gene. Science 2003, 301, 386–389. [Google Scholar] [CrossRef]
  11. Kendler, K.S.; Karkowski, L.M.; Prescott, C.A. Causal relationship between stressful life events and the onset of major depression. Am. J. Psychiatry 1999, 156, 837–841. [Google Scholar] [CrossRef] [PubMed]
  12. Kessler, R.C. The effects of stressful life events on depression. Annu. Rev. Psychol. 1997, 48, 191–214. [Google Scholar] [CrossRef] [PubMed]
  13. Littrell, J.L. Taking the Perspective that a Depressive State Reflects Inflammation: Implications for the Use of Antidepressants. Front. Psychol. 2012, 3, 297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sanacora, G.; Saricicek, A. GABAergic contributions to the pathophysiology of depression and the mechanism of antidepressant action. CNS Neurol. Disord. Drug. Targets 2007, 6, 127–140. [Google Scholar] [CrossRef] [PubMed]
  15. Slavich, G.M.; Irwin, M.R. From stress to inflammation and major depressive disorder: A social signal transduction theory of depression. Psychol. Bull. 2014, 140, 774–815. [Google Scholar] [CrossRef]
  16. Charney, D.S.; Manji, H.K. Life stress, genes, and depression: Multiple pathways lead to increased risk and new opportunities for intervention. Sci. STKE 2004, 2004, re5. [Google Scholar] [CrossRef] [PubMed]
  17. Nock, M.K.; Borges, G.; Bromet, E.J.; Cha, C.B.; Kessler, R.C.; Lee, S. Suicide and suicidal behavior. Epidemiol. Rev. 2008, 30, 133–154. [Google Scholar] [CrossRef]
  18. Ambelas, A. Psychologically stressful events in the precipitation of manic episodes. Br. J. Psychiatry 1979, 135, 15–21. [Google Scholar] [CrossRef]
  19. Malkoff-Schwartz, S.; Frank, E.; Anderson, B.P.; Hlastala, S.A.; Luther, J.F.; Sherrill, J.T.; Houck, P.R.; Kupfer, D.J. Social rhythm disruption and stressful life events in the onset of bipolar and unipolar episodes. Psychol. Med. 2000, 30, 1005–1016. [Google Scholar] [CrossRef]
  20. Kirschbaum, C.; Pirke, K.M.; Hellhammer, D.H. The Trier Social Stress Test—A tool for investigating psychobiological stress responses in a laboratory setting. Neuropsychobiology 1993, 28, 76–81. [Google Scholar] [CrossRef]
  21. Bierhaus, A.; Wolf, J.; Andrassy, M.; Rohleder, N.; Humpert, P.M.; Petrov, D.; Ferstl, R.; von Eynatten, M.; Wendt, T.; Rudofsky, G.; et al. A mechanism converting psychosocial stress into mononuclear cell activation. Proc. Natl. Acad. Sci. USA 2003, 100, 1920–1925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Carpenter, L.L.; Gawuga, C.E.; Tyrka, A.R.; Lee, J.K.; Anderson, G.M.; Price, L.H. Association between plasma IL-6 response to acute stress and early-life adversity in healthy adults. Neuropsychopharmacology 2010, 35, 2617–2623. [Google Scholar] [CrossRef] [PubMed]
  23. Dickerson, S.S.; Gable, S.L.; Irwin, M.R.; Aziz, N.; Kemeny, M.E. Social-evaluative threat and proinflammatory cytokine regulation: An experimental laboratory investigation. Psychol. Sci. 2009, 20, 1237–1244. [Google Scholar] [CrossRef]
  24. Pace, T.W.; Mletzko, T.C.; Alagbe, O.; Musselman, D.L.; Nemeroff, C.B.; Miller, A.H.; Heim, C.M. Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. Am. J. Psychiatry 2006, 163, 1630–1633. [Google Scholar] [CrossRef] [PubMed]
  25. Blandino, P., Jr.; Barnum, C.J.; Deak, T. The involvement of norepinephrine and microglia in hypothalamic and splenic IL-1beta responses to stress. J. Neuroimmunol. 2006, 173, 87–95. [Google Scholar] [CrossRef]
  26. Powell, N.D.; Sloan, E.K.; Bailey, M.T.; Arevalo, J.M.; Miller, G.E.; Chen, E.; Kobor, M.S.; Reader, B.F.; Sheridan, J.F.; Cole, S.W. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via beta-adrenergic induction of myelopoiesis. Proc. Natl. Acad. Sci. USA 2013, 110, 16574–16579. [Google Scholar] [CrossRef] [PubMed]
  27. Grisanti, L.A.; Woster, A.P.; Dahlman, J.; Sauter, E.R.; Combs, C.K.; Porter, J.E. Alpha1-adrenergic receptors positively regulate Toll-like receptor cytokine production from human monocytes and macrophages. J. Pharmacol. Exp. Ther. 2011, 338, 648–657. [Google Scholar] [CrossRef]
  28. Wohleb, E.S.; Hanke, M.L.; Corona, A.W.; Powell, N.D.; Stiner, L.M.; Bailey, M.T.; Nelson, R.J.; Godbout, J.P.; Sheridan, J.F. Beta-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J. Neurosci. 2011, 31, 6277–6288. [Google Scholar] [CrossRef]
  29. Bailey, M.T.; Engler, H.; Powell, N.D.; Padgett, D.A.; Sheridan, J.F. Repeated social defeat increases the bactericidal activity of splenic macrophages through a Toll-like receptor-dependent pathway. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 293, R1180–R1190. [Google Scholar] [CrossRef] [PubMed]
  30. Frank, M.G.; Baratta, M.V.; Sprunger, D.B.; Watkins, L.R.; Maier, S.F. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav. Immun. 2007, 21, 47–59. [Google Scholar] [CrossRef]
  31. Hinwood, M.; Morandini, J.; Day, T.A.; Walker, F.R. Evidence that microglia mediate the neurobiological effects of chronic psychological stress on the medial prefrontal cortex. Cereb. Cortex 2012, 22, 1442–1454. [Google Scholar] [CrossRef]
  32. Jankord, R.; Zhang, R.; Flak, J.N.; Solomon, M.B.; Albertz, J.; Herman, J.P. Stress activation of IL-6 neurons in the hypothalamus. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 299, R343–R351. [Google Scholar] [CrossRef] [Green Version]
  33. Audet, M.C.; Anisman, H. Interplay between pro-inflammatory cytokines and growth factors in depressive illnesses. Front. Cell Neurosci. 2013, 7, 68. [Google Scholar] [CrossRef] [Green Version]
  34. Kaufmann, F.N.; Costa, A.P.; Ghisleni, G.; Diaz, A.P.; Rodrigues, A.L.S.; Peluffo, H.; Kaster, M.P. NLRP3 inflammasome-driven pathways in depression: Clinical and preclinical findings. Brain Behav. Immun. 2017, 64, 367–383. [Google Scholar] [CrossRef]
  35. Tynan, R.J.; Naicker, S.; Hinwood, M.; Nalivaiko, E.; Buller, K.M.; Pow, D.V.; Day, T.A.; Walker, F.R. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain Behav. Immun. 2010, 24, 1058–1068. [Google Scholar] [CrossRef]
  36. Yirmiya, R.; Rimmerman, N.; Reshef, R. Depression as a microglial disease. Trends Neurosci. 2015, 38, 637–658. [Google Scholar] [CrossRef]
  37. Spitzer, N.C. How GABA generates depolarization. J. Physiol. 2010, 588, 757–758. [Google Scholar] [CrossRef]
  38. Levy, B.H.; Tasker, J.G. Synaptic regulation of the hypothalamic-pituitary-adrenal axis and its modulation by glucocorticoids and stress. Front. Cell Neurosci. 2012, 6, 24. [Google Scholar] [CrossRef]
  39. Zhou, J.J.; Gao, Y.; Zhang, X.; Kosten, T.A.; Li, D.P. Enhanced Hypothalamic NMDA Receptor Activity Contributes to Hyperactivity of HPA Axis in Chronic Stress in Male Rats. Endocrinology 2018, 159, 1537–1546. [Google Scholar] [CrossRef]
  40. Hewitt, S.A.; Wamsteeker, J.I.; Kurz, E.U.; Bains, J.S. Altered chloride homeostasis removes synaptic inhibitory constraint of the stress axis. Nat. Neurosci. 2009, 12, 438–443. [Google Scholar] [CrossRef]
  41. Sarkar, J.; Wakefield, S.; MacKenzie, G.; Moss, S.J.; Maguire, J. Neurosteroidogenesis is required for the physiological response to stress: Role of neurosteroid-sensitive GABAA receptors. J. Neurosci. 2011, 31, 18198–18210. [Google Scholar] [CrossRef]
  42. Gao, Y.; Zhou, J.J.; Zhu, Y.; Kosten, T.; Li, D.P. Chronic Unpredictable Mild Stress Induces Loss of GABA Inhibition in Corticotrophin-Releasing Hormone-Expressing Neurons through NKCC1 Upregulation. Neuroendocrinology 2017, 104, 194–208. [Google Scholar] [CrossRef]
  43. Dai, S.; Ma, Z. BDNF-trkB-KCC2-GABA pathway may be related to chronic stress-induced hyperalgesia at both the spinal and supraspinal level. Med. Hypotheses 2014, 83, 772–774. [Google Scholar] [CrossRef]
  44. Ferrini, F.; De Koninck, Y. Microglia control neuronal network excitability via BDNF signalling. Neural. Plast. 2013, 2013, 429815. [Google Scholar] [CrossRef]
  45. Luscher, B.; Shen, Q.; Sahir, N. The GABAergic deficit hypothesis of major depressive disorder. Mol. Psychiatry 2011, 16, 383–406. [Google Scholar] [CrossRef]
  46. Ding, B.; Frisina, R.D.; Zhu, X.; Sakai, Y.; Sokolowski, B.; Walton, J.P. Direct control of Na(+)-K(+)-2Cl(-)-cotransport protein (NKCC1) expression with aldosterone. Am. J. Physiol. Cell Physiol. 2014, 306, C66–C75. [Google Scholar] [CrossRef]
  47. Gold, P.W.; Chrousos, G.P. Organization of the stress system and its dysregulation in melancholic and atypical depression: High vs. low CRH/NE states. Mol. Psychiatry 2002, 7, 254–275. [Google Scholar] [CrossRef]
  48. Wahbeh, H.; Oken, B.S. Salivary cortisol lower in posttraumatic stress disorder. J. Trauma. Stress 2013, 26, 241–248. [Google Scholar] [CrossRef]
  49. Melhem, N.M.; Keilp, J.G.; Porta, G.; Oquendo, M.A.; Burke, A.; Stanley, B.; Cooper, T.B.; Mann, J.J.; Brent, D.A. Blunted HPA Axis Activity in Suicide Attempters Compared to those at High Risk for Suicidal Behavior. Neuropsychopharmacology 2016, 41, 1447–1456. [Google Scholar] [CrossRef]
  50. Nair, A.; Bonneau, R.H. Stress-induced elevation of glucocorticoids increases microglia proliferation through NMDA receptor activation. J. Neuroimmunol. 2006, 171, 72–85. [Google Scholar] [CrossRef]
  51. Nguyen, K.T.; Deak, T.; Owens, S.M.; Kohno, T.; Fleshner, M.; Watkins, L.R.; Maier, S.F. Exposure to acute stress induces brain interleukin-1beta protein in the rat. J. Neurosci. 1998, 18, 2239–2246. [Google Scholar] [CrossRef]
  52. Danese, A.; Moffitt, T.E.; Pariante, C.M.; Ambler, A.; Poulton, R.; Caspi, A. Elevated inflammation levels in depressed adults with a history of childhood maltreatment. Arch. Gen. Psychiatry 2008, 65, 409–415. [Google Scholar] [CrossRef]
  53. Lowy, M.T.; Gault, L.; Yamamoto, B.K. Adrenalectomy attenuates stress-induced elevations in extracellular glutamate concentrations in the hippocampus. J. Neurochem. 1993, 61, 1957–1960. [Google Scholar] [CrossRef]
  54. Moghaddam, B.; Bolinao, M.L.; Stein-Behrens, B.; Sapolsky, R. Glucocorticoids mediate the stress-induced extracellular accumulation of glutamate. Brain Res. 1994, 655, 251–254. [Google Scholar] [CrossRef]
  55. Munhoz, C.D.; Lepsch, L.B.; Kawamoto, E.M.; Malta, M.B.; Lima Lde, S.; Avellar, M.C.; Sapolsky, R.M.; Scavone, C. Chronic unpredictable stress exacerbates lipopolysaccharide-induced activation of nuclear factor-kappaB in the frontal cortex and hippocampus via glucocorticoid secretion. J. Neurosci. 2006, 26, 3813–3820. [Google Scholar] [CrossRef]
  56. Popoli, M.; Yan, Z.; McEwen, B.S.; Sanacora, G. The stressed synapse: The impact of stress and glucocorticoids on glutamate transmission. Nat. Rev. Neurosci. 2011, 13, 22–37. [Google Scholar] [CrossRef]
  57. Abdallah, C.G.; Sanacora, G.; Duman, R.S.; Krystal, J.H. The neurobiology of depression, ketamine and rapid-acting antidepressants: Is it glutamate inhibition or activation? Pharmacol. Ther. 2018, 190, 148–158. [Google Scholar] [CrossRef]
  58. Iwata, M.; Ota, K.T.; Li, X.Y.; Sakaue, F.; Li, N.; Dutheil, S.; Banasr, M.; Duric, V.; Yamanashi, T.; Kaneko, K.; et al. Psychological Stress Activates the Inflammasome via Release of Adenosine Triphosphate and Stimulation of the Purinergic Type 2X7 Receptor. Biol. Psychiatry 2016, 80, 12–22. [Google Scholar] [CrossRef]
  59. Banasr, M.; Duman, R.S. Glial loss in the prefrontal cortex is sufficient to induce depressive-like behaviors. Biol. Psychiatry 2008, 64, 863–870. [Google Scholar] [CrossRef]
  60. Czeh, B.; Simon, M.; Schmelting, B.; Hiemke, C.; Fuchs, E. Astroglial plasticity in the hippocampus is affected by chronic psychosocial stress and concomitant fluoxetine treatment. Neuropsychopharmacology 2006, 31, 1616–1626. [Google Scholar] [CrossRef]
  61. Frank, M.G.; Thompson, B.M.; Watkins, L.R.; Maier, S.F. Glucocorticoids mediate stress-induced priming of microglial pro-inflammatory responses. Brain Behav. Immun. 2012, 26, 337–345. [Google Scholar] [CrossRef] [PubMed]
  62. Arnsten, A.F. Stress weakens prefrontal networks: Molecular insults to higher cognition. Nat. Neurosci. 2015, 18, 1376–1385. [Google Scholar] [CrossRef] [PubMed]
  63. Liston, C.; Miller, M.M.; Goldwater, D.S.; Radley, J.J.; Rocher, A.B.; Hof, P.R.; Morrison, J.H.; McEwen, B.S. Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting. J. Neurosci. 2006, 26, 7870–7874. [Google Scholar] [CrossRef] [PubMed]
  64. Radley, J.J.; Rocher, A.B.; Miller, M.; Janssen, W.G.; Liston, C.; Hof, P.R.; McEwen, B.S.; Morrison, J.H. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cereb. Cortex 2006, 16, 313–320. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, G.; Henter, I.D.; Manji, H.K. A role for PKC in mediating stress-induced prefrontal cortical structural plasticity and cognitive function. Proc. Natl. Acad. Sci. USA 2009, 106, 17613–17614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hains, A.B.; Vu, M.A.; Maciejewski, P.K.; van Dyck, C.H.; Gottron, M.; Arnsten, A.F. Inhibition of protein kinase C signaling protects prefrontal cortex dendritic spines and cognition from the effects of chronic stress. Proc. Natl. Acad. Sci. USA 2009, 106, 17957–17962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Atzori, M.; Garcia-Oscos, F.; Mendez, J.A. Role of IL-6 in the etiology of hyperexcitable neuropsychiatric conditions: Experimental evidence and therapeutic implications. Future Med. Chem. 2012, 4, 2177–2192. [Google Scholar] [CrossRef] [PubMed]
  68. Berk, M.; Williams, L.J.; Jacka, F.N.; O’Neil, A.; Pasco, J.A.; Moylan, S.; Allen, N.B.; Stuart, A.L.; Hayley, A.C.; Byrne, M.L.; et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013, 11, 200. [Google Scholar] [CrossRef] [PubMed]
  69. Steptoe, A.; Hamer, M.; Chida, Y. The effects of acute psychological stress on circulating inflammatory factors in humans: A review and meta-analysis. Brain Behav. Immun. 2007, 21, 901–912. [Google Scholar] [CrossRef]
  70. Gouin, J.P.; Glaser, R.; Malarkey, W.B.; Beversdorf, D.; Kiecolt-Glaser, J.K. Childhood abuse and inflammatory responses to daily stressors. Ann. Behav. Med. 2012, 44, 287–292. [Google Scholar] [CrossRef]
  71. Wang, N.; Yu, H.Y.; Shen, X.F.; Gao, Z.Q.; Yang, C.; Yang, J.J.; Zhang, G.F. The rapid antidepressant effect of ketamine in rats is associated with down-regulation of pro-inflammatory cytokines in the hippocampus. Ups. J. Med. Sci. 2015, 120, 241–248. [Google Scholar] [CrossRef] [PubMed]
  72. Girotti, M.; Donegan, J.J.; Morilak, D.A. Influence of hypothalamic IL-6/gp130 receptor signaling on the HPA axis response to chronic stress. Psychoneuroendocrinology 2013, 38, 1158–1169. [Google Scholar] [CrossRef] [PubMed]
  73. Sukoff Rizzo, S.J.; Neal, S.J.; Hughes, Z.A.; Beyna, M.; Rosenzweig-Lipson, S.; Moss, S.J.; Brandon, N.J. Evidence for sustained elevation of IL-6 in the CNS as a key contributor of depressive-like phenotypes. Transl. Psychiatry 2012, 2, e199. [Google Scholar] [CrossRef] [PubMed]
  74. Erta, M.; Quintana, A.; Hidalgo, J. Interleukin-6, a major cytokine in the central nervous system. Int. J. Biol. Sci. 2012, 8, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
  75. Greenhill, C.J.; Rose-John, S.; Lissilaa, R.; Ferlin, W.; Ernst, M.; Hertzog, P.J.; Mansell, A.; Jenkins, B.J. IL-6 trans-signaling modulates TLR4-dependent inflammatory responses via STAT3. J. Immunol. 2011, 186, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  76. Rose-John, S. The Soluble Interleukin 6 Receptor: Advanced Therapeutic Options in Inflammation. Clin. Pharmacol. Ther. 2017, 102, 591–598. [Google Scholar] [CrossRef] [PubMed]
  77. Campbell, I.L.; Erta, M.; Lim, S.L.; Frausto, R.; May, U.; Rose-John, S.; Scheller, J.; Hidalgo, J. Trans-signaling is a dominant mechanism for the pathogenic actions of interleukin-6 in the brain. J. Neurosci. 2014, 34, 2503–2513. [Google Scholar] [CrossRef] [PubMed]
  78. Monje, M.L.; Toda, H.; Palmer, T.D. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003, 302, 1760–1765. [Google Scholar] [CrossRef]
  79. Wan, J.; Fu, A.K.; Ip, F.C.; Ng, H.K.; Hugon, J.; Page, G.; Wang, J.H.; Lai, K.O.; Wu, Z.; Ip, N.Y. Tyk2/STAT3 signaling mediates beta-amyloid-induced neuronal cell death: Implications in Alzheimer’s disease. J. Neurosci. 2010, 30, 6873–6881. [Google Scholar] [CrossRef]
  80. Miller, A.H.; Raison, C.L. The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2016, 16, 22–34. [Google Scholar] [CrossRef]
  81. Lee, S.; Jeong, J.; Kwak, Y.; Park, S.K. Depression research: Where are we now? Mol. Brain 2010, 3, 8. [Google Scholar] [CrossRef]
  82. Dumitrescu, A.L. Depression and Inflammatory Periodontal Disease Considerations-An Interdisciplinary Approach. Front. Psychol. 2016, 7, 347. [Google Scholar] [CrossRef]
  83. Jiang, M.; Qin, P.; Yang, X. Comorbidity between depression and asthma via immune-inflammatory pathways: A meta-analysis. J. Affect. Disord. 2014, 166, 22–29. [Google Scholar] [CrossRef]
  84. Siegert, R.J.; Abernethy, D.A. Depression in multiple sclerosis: A review. J. Neurol. Neurosurg. Psychiatry 2005, 76, 469–475. [Google Scholar] [CrossRef]
  85. Lu, M.C.; Guo, H.R.; Lin, M.C.; Livneh, H.; Lai, N.S.; Tsai, T.Y. Bidirectional associations between rheumatoid arthritis and depression: A nationwide longitudinal study. Sci Rep. 2016, 6, 20647. [Google Scholar] [CrossRef]
  86. Matcham, F.; Rayner, L.; Steer, S.; Hotopf, M. The prevalence of depression in rheumatoid arthritis: A systematic review and meta-analysis. Rheumatology 2013, 52, 2136–2148. [Google Scholar] [CrossRef]
  87. Nerurkar, L.; Siebert, S.; McInnes, I.B.; Cavanagh, J. Rheumatoid arthritis and depression: An inflammatory perspective. Lancet Psychiatry 2019, 6, 164–173. [Google Scholar] [CrossRef]
  88. Aarsland, D.; Pahlhagen, S.; Ballard, C.G.; Ehrt, U.; Svenningsson, P. Depression in Parkinson disease—Epidemiology, mechanisms and management. Nat. Rev. Neurol. 2011, 8, 35–47. [Google Scholar] [CrossRef]
  89. Jacob, E.L.; Gatto, N.M.; Thompson, A.; Bordelon, Y.; Ritz, B. Occurrence of depression and anxiety prior to Parkinson’s disease. Parkinsonism. Relat. Disord. 2010, 16, 576–581. [Google Scholar] [CrossRef]
  90. Benoit, M.; Berrut, G.; Doussaint, J.; Bakchine, S.; Bonin-Guillaume, S.; Fremont, P.; Gallarda, T.; Krolak-Salmon, P.; Marquet, T.; Mekies, C.; et al. Apathy and depression in mild Alzheimer’s disease: A cross-sectional study using diagnostic criteria. J. Alzheimers Dis. 2012, 31, 325–334. [Google Scholar] [CrossRef]
  91. Diniz, B.S.; Butters, M.A.; Albert, S.M.; Dew, M.A.; Reynolds, C.F., 3rd. Late-life depression and risk of vascular dementia and Alzheimer’s disease: Systematic review and meta-analysis of community-based cohort studies. Br. J. Psychiatry 2013, 202, 329–335. [Google Scholar] [CrossRef]
  92. Rutledge, T.; Reis, V.A.; Linke, S.E.; Greenberg, B.H.; Mills, P.J. Depression in heart failure a meta-analytic review of prevalence, intervention effects, and associations with clinical outcomes. J. Am. Coll. Cardiol. 2006, 48, 1527–1537. [Google Scholar] [CrossRef]
  93. Mezuk, B.; Eaton, W.W.; Albrecht, S.; Golden, S.H. Depression and type 2 diabetes over the lifespan: A meta-analysis. Diabetes Care 2008, 31, 2383–2390. [Google Scholar] [CrossRef]
  94. De Wit, L.; Luppino, F.; van Straten, A.; Penninx, B.; Zitman, F.; Cuijpers, P. Depression and obesity: A meta-analysis of community-based studies. Psychiatry Res. 2010, 178, 230–235. [Google Scholar] [CrossRef]
  95. Dunn, A.J.; Swiergiel, A.H.; de Beaurepaire, R. Cytokines as mediators of depression: What can we learn from animal studies? Neurosci. Biobehav. Rev. 2005, 29, 891–909. [Google Scholar] [CrossRef]
  96. Kopschina Feltes, P.; Doorduin, J.; Klein, H.C.; Juarez-Orozco, L.E.; Dierckx, R.A.; Moriguchi-Jeckel, C.M.; de Vries, E.F. Anti-inflammatory treatment for major depressive disorder: Implications for patients with an elevated immune profile and non-responders to standard antidepressant therapy. J. Psychopharmacol. 2017, 31, 1149–1165. [Google Scholar] [CrossRef]
  97. Silic, A.; Karlovic, D.; Serretti, A. Increased inflammation and lower platelet 5-HT in depression with metabolic syndrome. J. Affect. Disord. 2012, 141, 72–78. [Google Scholar] [CrossRef]
  98. Rudolf, S.; Greggersen, W.; Kahl, K.G.; Huppe, M.; Schweiger, U. Elevated IL-6 levels in patients with atypical depression but not in patients with typical depression. Psychiatry Res. 2014, 217, 34–38. [Google Scholar] [CrossRef]
  99. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctot, K.L. A meta-analysis of cytokines in major depression. Biol. Psychiatry 2010, 67, 446–457. [Google Scholar] [CrossRef]
  100. Goldsmith, D.R.; Rapaport, M.H.; Miller, B.J. A meta-analysis of blood cytokine network alterations in psychiatric patients: Comparisons between schizophrenia, bipolar disorder and depression. Mol. Psychiatry 2016, 21, 1696–1709. [Google Scholar] [CrossRef]
  101. Haapakoski, R.; Mathieu, J.; Ebmeier, K.P.; Alenius, H.; Kivimaki, M. Cumulative meta-analysis of interleukins 6 and 1beta, tumour necrosis factor alpha and C-reactive protein in patients with major depressive disorder. Brain Behav. Immun. 2015, 49, 206–215. [Google Scholar] [CrossRef]
  102. Howren, M.B.; Lamkin, D.M.; Suls, J. Associations of depression with C-reactive protein, IL-1, and IL-6: A meta-analysis. Psychosom. Med. 2009, 71, 171–186. [Google Scholar] [CrossRef]
  103. Zhang, C.; Wu, Z.; Zhao, G.; Wang, F.; Fang, Y. Identification of IL6 as a susceptibility gene for major depressive disorder. Sci. Rep. 2016, 6, 31264. [Google Scholar] [CrossRef] [Green Version]
  104. Maes, M.; Ombelet, W.; De Jongh, R.; Kenis, G.; Bosmans, E. The inflammatory response following delivery is amplified in women who previously suffered from major depression, suggesting that major depression is accompanied by a sensitization of the inflammatory response system. J. Affect. Disord. 2001, 63, 85–92. [Google Scholar] [CrossRef]
  105. 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. [Google Scholar] [CrossRef]
  106. Munkholm, K.; Brauner, J.V.; Kessing, L.V.; Vinberg, M. Cytokines in bipolar disorder vs. healthy control subjects: A systematic review and meta-analysis. J. Psychiatr. Res. 2013, 47, 1119–1133. [Google Scholar] [CrossRef]
  107. Michalopoulou, M.; Nikolaou, C.; Tavernarakis, A.; Alexandri, N.M.; Rentzos, M.; Chatzipanagiotou, S.; Cambouri, C.; Vassilopoulos, D. Soluble interleukin-6 receptor (sIL-6R) in cerebrospinal fluid of patients with inflammatory and non inflammatory neurological diseases. Immunol. Lett. 2004, 94, 183–189. [Google Scholar] [CrossRef]
  108. Gananca, L.; Oquendo, M.A.; Tyrka, A.R.; Cisneros-Trujillo, S.; Mann, J.J.; Sublette, M.E. The role of cytokines in the pathophysiology of suicidal behavior. Psychoneuroendocrinology 2016, 63, 296–310. [Google Scholar] [CrossRef]
  109. Isung, J.; Aeinehband, S.; Mobarrez, F.; Nordstrom, P.; Runeson, B.; Asberg, M.; Piehl, F.; Jokinen, J. High interleukin-6 and impulsivity: Determining the role of endophenotypes in attempted suicide. Transl. Psychiatry 2014, 4, e470. [Google Scholar] [CrossRef]
  110. Lindqvist, D.; Janelidze, S.; Hagell, P.; Erhardt, S.; Samuelsson, M.; Minthon, L.; Hansson, O.; Bjorkqvist, M.; Traskman-Bendz, L.; Brundin, L. Interleukin-6 is elevated in the cerebrospinal fluid of suicide attempters and related to symptom severity. Biol. Psychiatry 2009, 66, 287–292. [Google Scholar] [CrossRef]
  111. Sasayama, D.; Hattori, K.; Wakabayashi, C.; Teraishi, T.; Hori, H.; Ota, M.; Yoshida, S.; Arima, K.; Higuchi, T.; Amano, N.; et al. Increased cerebrospinal fluid interleukin-6 levels in patients with schizophrenia and those with major depressive disorder. J. Psychiatr. Res. 2013, 47, 401–406. [Google Scholar] [CrossRef] [Green Version]
  112. O’Donovan, A.; Rush, G.; Hoatam, G.; Hughes, B.M.; McCrohan, A.; Kelleher, C.; O’Farrelly, C.; Malone, K.M. Suicidal ideation is associated with elevated inflammation in patients with major depressive disorder. Depress. Anxiety 2013, 30, 307–314. [Google Scholar] [CrossRef]
  113. Janelidze, S.; Mattei, D.; Westrin, A.; Traskman-Bendz, L.; Brundin, L. Cytokine levels in the blood may distinguish suicide attempters from depressed patients. Brain Behav. Immun. 2011, 25, 335–339. [Google Scholar] [CrossRef] [Green Version]
  114. Bergmans, R.S.; Kelly, K.M.; Mezuk, B. Inflammation as a unique marker of suicide ideation distinct from depression syndrome among U.S. adults. J. Affect. Disord 2019, 245, 1052–1060. [Google Scholar] [CrossRef]
  115. Brundin, L.; Erhardt, S.; Bryleva, E.Y.; Achtyes, E.D.; Postolache, T.T. The role of inflammation in suicidal behaviour. Acta Psychiatr. Scand. 2015, 132, 192–203. [Google Scholar] [CrossRef]
  116. Marsland, A.L.; Prather, A.A.; Petersen, K.L.; Cohen, S.; Manuck, S.B. Antagonistic characteristics are positively associated with inflammatory markers independently of trait negative emotionality. Brain Behav. Immun. 2008, 22, 753–761. [Google Scholar] [CrossRef] [Green Version]
  117. Marini, S.; Vellante, F.; Matarazzo, I.; De Berardis, D.; Serroni, N.; Gianfelice, D.; Olivieri, L.; Di Renzo, F.; Di Marco, A.; Fornaro, M.; et al. Inflammatory markers and suicidal attempts in depressed patients: A review. Int. J. Immunopathol. Pharmacol. 2016, 29, 583–594. [Google Scholar] [CrossRef] [Green Version]
  118. Kubera, M.; Obuchowicz, E.; Goehler, L.; Brzeszcz, J.; Maes, M. In animal models, psychosocial stress-induced (neuro)inflammation, apoptosis and reduced neurogenesis are associated to the onset of depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2011, 35, 744–759. [Google Scholar] [CrossRef]
  119. Baran, P.; Hansen, S.; Waetzig, G.H.; Akbarzadeh, M.; Lamertz, L.; Huber, H.J.; Ahmadian, M.R.; Moll, J.M.; Scheller, J. The balance of interleukin (IL)-6, IL-6.soluble IL-6 receptor (sIL-6R), and IL-6.sIL-6R.sgp130 complexes allows simultaneous classic and trans-signaling. J. Biol. Chem. 2018, 293, 6762–6775. [Google Scholar] [CrossRef]
  120. Boulanger, M.J.; Chow, D.C.; Brevnova, E.E.; Garcia, K.C. Hexameric structure and assembly of the interleukin-6/IL-6 alpha-receptor/gp130 complex. Science 2003, 300, 2101–2104. [Google Scholar] [CrossRef]
  121. Sohn, S.J.; Barrett, K.; Van Abbema, A.; Chang, C.; Kohli, P.B.; Kanda, H.; Smith, J.; Lai, Y.; Zhou, A.; Zhang, B.; et al. A restricted role for TYK2 catalytic activity in human cytokine responses revealed by novel TYK2-selective inhibitors. J. Immunol. 2013, 191, 2205–2216. [Google Scholar] [CrossRef]
  122. Briso, E.M.; Dienz, O.; Rincon, M. Cutting edge: Soluble IL-6R is produced by IL-6R ectodomain shedding in activated CD4 T cells. J. Immunol. 2008, 180, 7102–7106. [Google Scholar] [CrossRef]
  123. 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. [Google Scholar] [CrossRef]
  124. Burton, M.D.; Sparkman, N.L.; Johnson, R.W. Inhibition of interleukin-6 trans-signaling in the brain facilitates recovery from lipopolysaccharide-induced sickness behavior. J. Neuroinflamm. 2011, 8, 54. [Google Scholar] [CrossRef]
  125. Hsu, M.P.; Frausto, R.; Rose-John, S.; Campbell, I.L. Analysis of IL-6/gp130 family receptor expression reveals that in contrast to astroglia, microglia lack the oncostatin M receptor and functional responses to oncostatin M. Glia 2015, 63, 132–141. [Google Scholar] [CrossRef]
  126. Riethmueller, S.; Somasundaram, P.; Ehlers, J.C.; Hung, C.W.; Flynn, C.M.; Lokau, J.; Agthe, M.; Dusterhoft, S.; Zhu, Y.; Grotzinger, J.; et al. Proteolytic Origin of the Soluble Human IL-6R In Vivo and a Decisive Role of N-Glycosylation. PLoS Biol. 2017, 15, e2000080. [Google Scholar] [CrossRef]
  127. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 2011, 1813, 878–888. [Google Scholar] [CrossRef] [Green Version]
  128. Jones, S.A.; Novick, D.; Horiuchi, S.; Yamamoto, N.; Szalai, A.J.; Fuller, G.M. C-reactive protein: A physiological activator of interleukin 6 receptor shedding. J. Exp. Med. 1999, 189, 599–604. [Google Scholar] [CrossRef]
  129. Sluzewska, A.; Rybakowski, J.K.; Laciak, M.; Mackiewicz, A.; Sobieska, M.; Wiktorowicz, K. Interleukin-6 serum levels in depressed patients before and after treatment with fluoxetine. Ann. NY Acad. Sci. 1995, 762, 474–476. [Google Scholar] [CrossRef]
  130. Patel, A.; Zhu, Y.; Kuzhikandathil, E.V.; Banks, W.A.; Siegel, A.; Zalcman, S.S. Soluble interleukin-6 receptor induces motor stereotypies and co-localizes with gp130 in regions linked to cortico-striato-thalamo-cortical circuits. PLoS ONE 2012, 7, e41623. [Google Scholar] [CrossRef]
  131. Raber, J.; O’Shea, R.D.; Bloom, F.E.; Campbell, I.L. Modulation of hypothalamic-pituitary-adrenal function by transgenic expression of interleukin-6 in the CNS of mice. J. Neurosci. 1997, 17, 9473–9480. [Google Scholar] [CrossRef] [PubMed]
  132. Mastorakos, G.; Weber, J.S.; Magiakou, M.A.; Gunn, H.; Chrousos, G.P. Hypothalamic-pituitary-adrenal axis activation and stimulation of systemic vasopressin secretion by recombinant interleukin-6 in humans: Potential implications for the syndrome of inappropriate vasopressin secretion. J. Clin. Endocrinol. Metab. 1994, 79, 934–939. [Google Scholar] [CrossRef] [PubMed]
  133. Wake, H.; Watanabe, M.; Moorhouse, A.J.; Kanematsu, T.; Horibe, S.; Matsukawa, N.; Asai, K.; Ojika, K.; Hirata, M.; Nabekura, J. Early changes in KCC2 phosphorylation in response to neuronal stress result in functional downregulation. J. Neurosci. 2007, 27, 1642–1650. [Google Scholar] [CrossRef] [PubMed]
  134. Pieraut, S.; Lucas, O.; Sangari, S.; Sar, C.; Boudes, M.; Bouffi, C.; Noel, D.; Scamps, F. An autocrine neuronal interleukin-6 loop mediates chloride accumulation and NKCC1 phosphorylation in axotomized sensory neurons. J. Neurosci. 2011, 31, 13516–13526. [Google Scholar] [CrossRef] [PubMed]
  135. Garza, J.C.; Guo, M.; Zhang, W.; Lu, X.Y. Leptin restores adult hippocampal neurogenesis in a chronic unpredictable stress model of depression and reverses glucocorticoid-induced inhibition of GSK-3beta/beta-catenin signaling. Mol. Psychiatry 2012, 17, 790–808. [Google Scholar] [CrossRef] [PubMed]
  136. Hodes, G.E.; Menard, C.; Russo, S.J. Integrating Interleukin-6 into depression diagnosis and treatment. Neurobiol. Stress 2016, 4, 15–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Marz, P.; Cheng, J.G.; Gadient, R.A.; Patterson, P.H.; Stoyan, T.; Otten, U.; Rose-John, S. Sympathetic neurons can produce and respond to interleukin 6. Proc. Natl. Acad. Sci. USA 1998, 95, 3251–3256. [Google Scholar] [CrossRef] [Green Version]
  138. Helwig, B.G.; Craig, R.A.; Fels, R.J.; Blecha, F.; Kenney, M.J. Central nervous system administration of interleukin-6 produces splenic sympathoexcitation. Auton. Neurosci. 2008, 141, 104–111. [Google Scholar] [CrossRef] [Green Version]
  139. Palsson, J.; Ricksten, S.E.; Delle, M.; Lundin, S. Changes in renal sympathetic nerve activity during experimental septic and endotoxin shock in conscious rats. Circ. Shock 1988, 24, 133–141. [Google Scholar]
  140. Rudiger, A.; Jeger, V.; Arrigo, M.; Schaer, C.A.; Hildenbrand, F.F.; Arras, M.; Seifert, B.; Singer, M.; Schoedon, G.; Spahn, D.R.; et al. Heart rate elevations during early sepsis predict death in fluid-resuscitated rats with fecal peritonitis. Intensive Care Med. Exp. 2018, 6, 28. [Google Scholar] [CrossRef]
  141. Franke, H.; Verkhratsky, A.; Burnstock, G.; Illes, P. Pathophysiology of astroglial purinergic signalling. Purinergic Signal. 2012, 8, 629–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Jha, M.K.; Lee, W.H.; Suk, K. Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders. Biochem. Pharmacol. 2016, 103, 1–16. [Google Scholar] [CrossRef] [PubMed]
  143. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef] [PubMed]
  144. Hammond, T.R.; Gadea, A.; Dupree, J.; Kerninon, C.; Nait-Oumesmar, B.; Aguirre, A.; Gallo, V. Astrocyte-derived endothelin-1 inhibits remyelination through notch activation. Neuron 2014, 81, 588–602. [Google Scholar] [CrossRef] [PubMed]
  145. Boche, D.; Perry, V.H.; Nicoll, J.A. Review: Activation patterns of microglia and their identification in the human brain. Neuropathol. Appl. Neurobiol. 2013, 39, 3–18. [Google Scholar] [CrossRef] [PubMed]
  146. Pariante, C.M. The glucocorticoid receptor: Part of the solution or part of the problem? J. Psychopharmacol. 2006, 20, 79–84. [Google Scholar] [CrossRef]
  147. Lotrich, F.E. Inflammatory cytokine-associated depression. Brain Res. 2015, 1617, 113–125. [Google Scholar] [CrossRef]
  148. Boku, S.; Nakagawa, S.; Masuda, T.; Nishikawa, H.; Kato, A.; Kitaichi, Y.; Inoue, T.; Koyama, T. Glucocorticoids and lithium reciprocally regulate the proliferation of adult dentate gyrus-derived neural precursor cells through GSK-3beta and beta-catenin/TCF pathway. Neuropsychopharmacology 2009, 34, 805–815. [Google Scholar] [CrossRef]
  149. Nitta, A.; Zheng, W.H.; Quirion, R. Insulin-like growth factor 1 prevents neuronal cell death induced by corticosterone through activation of the PI3k/Akt pathway. J. Neurosci. Res. 2004, 76, 98–103. [Google Scholar] [CrossRef]
  150. Zhang, B.; Zhang, Y.; Wu, W.; Xu, T.; Yin, Y.; Zhang, J.; Huang, D.; Li, W. Chronic glucocorticoid exposure activates BK-NLRP1 signal involving in hippocampal neuron damage. J. Neuroinflamm. 2017, 14, 139. [Google Scholar] [CrossRef]
  151. Sanacora, G.; Schatzberg, A.F. Ketamine: Promising path or false prophecy in the development of novel therapeutics for mood disorders? Neuropsychopharmacology 2015, 40, 259–267. [Google Scholar] [CrossRef] [PubMed]
  152. Zanos, P.; Gould, T.D. Mechanisms of ketamine action as an antidepressant. Mol. Psychiatry 2018, 23, 801–811. [Google Scholar] [CrossRef] [PubMed]
  153. Bjorkholm, C.; Monteggia, L.M. BDNF—A key transducer of antidepressant effects. Neuropharmacology 2016, 102, 72–79. [Google Scholar] [CrossRef] [PubMed]
  154. Coull, J.A.; Beggs, S.; Boudreau, D.; Boivin, D.; Tsuda, M.; Inoue, K.; Gravel, C.; Salter, M.W.; De Koninck, Y. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature 2005, 438, 1017–1021. [Google Scholar] [CrossRef] [PubMed]
  155. Rivera, C.; Li, H.; Thomas-Crusells, J.; Lahtinen, H.; Viitanen, T.; Nanobashvili, A.; Kokaia, Z.; Airaksinen, M.S.; Voipio, J.; Kaila, K.; et al. BDNF-induced TrkB activation down-regulates the K+-Cl- cotransporter KCC2 and impairs neuronal Cl- extrusion. J. Cell Biol. 2002, 159, 747–752. [Google Scholar] [CrossRef] [PubMed]
  156. Kim, J.Y.; Liu, C.Y.; Zhang, F.; Duan, X.; Wen, Z.; Song, J.; Feighery, E.; Lu, B.; Rujescu, D.; St Clair, D.; et al. Interplay between DISC1 and GABA signaling regulates neurogenesis in mice and risk for schizophrenia. Cell 2012, 148, 1051–1064. [Google Scholar] [CrossRef]
  157. North, R.A.; Surprenant, A. Pharmacology of cloned P2X receptors. Annu. Rev. Pharmacol. Toxicol. 2000, 40, 563–580. [Google Scholar] [CrossRef]
  158. Ase, A.R.; Honson, N.S.; Zaghdane, H.; Pfeifer, T.A.; Seguela, P. Identification and characterization of a selective allosteric antagonist of human P2X4 receptor channels. Mol. Pharmacol. 2015, 87, 606–616. [Google Scholar] [CrossRef]
  159. Matsumura, Y.; Yamashita, T.; Sasaki, A.; Nakata, E.; Kohno, K.; Masuda, T.; Tozaki-Saitoh, H.; Imai, T.; Kuraishi, Y.; Tsuda, M.; et al. A novel P2X4 receptor-selective antagonist produces anti-allodynic effect in a mouse model of herpetic pain. Sci. Rep. 2016, 6, 32461. [Google Scholar] [CrossRef]
  160. Heinrich, P.C.; Behrmann, I.; Haan, S.; Hermanns, H.M.; Muller-Newen, G.; Schaper, F. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem. J. 2003, 374, 1–20. [Google Scholar] [CrossRef] [Green Version]
  161. Leitner, N.R.; Witalisz-Siepracka, A.; Strobl, B.; Muller, M. Tyrosine kinase 2—Surveillant of tumours and bona fide oncogene. Cytokine 2017, 89, 209–218. [Google Scholar] [CrossRef]
  162. He, X.; Chen, X.; Zhang, H.; Xie, T.; Ye, X.Y. Selective Tyk2 inhibitors as potential therapeutic agents: A patent review (2015–2018). Expert Opin. Ther. Pat. 2019, 29, 137–149. [Google Scholar] [CrossRef]
  163. Menet, C.J. Toward selective TYK2 inhibitors as therapeutic agents for the treatment of inflammatory diseases. Pharm. Pat. Anal. 2014, 3, 449–466. [Google Scholar] [CrossRef]
  164. Tracey, K.J. Reflex control of immunity. Nat. Rev. Immunol. 2009, 9, 418–428. [Google Scholar] [CrossRef] [Green Version]
  165. Bencherif, M.; Lippiello, P.M.; Lucas, R.; Marrero, M.B. Alpha7 nicotinic receptors as novel therapeutic targets for inflammation-based diseases. Cell Mol. Life Sci. 2011, 68, 931–949. [Google Scholar] [CrossRef]
  166. Egea, J.; Buendia, I.; Parada, E.; Navarro, E.; Leon, R.; Lopez, M.G. Anti-inflammatory role of microglial alpha7 nAChRs and its role in neuroprotection. Biochem. Pharmacol. 2015, 97, 463–472. [Google Scholar] [CrossRef]
  167. Wang, H.; Yu, M.; Ochani, M.; Amella, C.A.; Tanovic, M.; Susarla, S.; Li, J.H.; Wang, H.; Yang, H.; Ulloa, L.; et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 2003, 421, 384–388. [Google Scholar] [CrossRef]
  168. Borovikova, L.V.; Ivanova, S.; Zhang, M.; Yang, H.; Botchkina, G.I.; Watkins, L.R.; Wang, H.; Abumrad, N.; Eaton, J.W.; Tracey, K.J. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000, 405, 458–462. [Google Scholar] [CrossRef]
  169. De Jonge, W.J.; van der Zanden, E.P.; The, F.O.; Bijlsma, M.F.; van Westerloo, D.J.; Bennink, R.J.; Berthoud, H.R.; Uematsu, S.; Akira, S.; van den Wijngaard, R.M.; et al. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat. Immunol. 2005, 6, 844–851. [Google Scholar] [CrossRef]
  170. Bajbouj, M.; Merkl, A.; Schlaepfer, T.E.; Frick, C.; Zobel, A.; Maier, W.; O’Keane, V.; Corcoran, C.; Adolfsson, R.; Trimble, M.; et al. Two-year outcome of vagus nerve stimulation in treatment-resistant depression. J. Clin. Psychopharmacol. 2010, 30, 273–281. [Google Scholar] [CrossRef]
  171. Berry, S.M.; Broglio, K.; Bunker, M.; Jayewardene, A.; Olin, B.; Rush, A.J. A patient-level meta-analysis of studies evaluating vagus nerve stimulation therapy for treatment-resistant depression. Med. Devices 2013, 6, 17–35. [Google Scholar] [CrossRef] [Green Version]
  172. Kalkman, H.O.; Feuerbach, D. Modulatory effects of alpha7 nAChRs on the immune system and its relevance for CNS disorders. Cell Mol. Life Sci. 2016, 73, 2511–2530. [Google Scholar] [CrossRef]
  173. Cui, W.Y.; Zhao, S.; Polanowska-Grabowska, R.; Wang, J.; Wei, J.; Dash, B.; Chang, S.L.; Saucerman, J.J.; Gu, J.; Li, M.D. Identification and characterization of poly(I:C)-induced molecular responses attenuated by nicotine in mouse macrophages. Mol. Pharmacol. 2013, 83, 61–72. [Google Scholar] [CrossRef]
  174. Kalkman, H.O.; Feuerbach, D. Antidepressant therapies inhibit inflammation and microglial M1-polarization. Pharmacol. Ther. 2016, 163, 82–93. [Google Scholar] [CrossRef]
  175. Rosas-Ballina, M.; Goldstein, R.S.; Gallowitsch-Puerta, M.; Yang, L.; Valdes-Ferrer, S.I.; Patel, N.B.; Chavan, S.; Al-Abed, Y.; Yang, H.; Tracey, K.J. The selective alpha7 agonist GTS-21 attenuates cytokine production in human whole blood and human monocytes activated by ligands for TLR2, TLR3, TLR4, TLR9, and RAGE. Mol. Med. 2009, 15, 195–202. [Google Scholar] [CrossRef]
  176. Shytle, R.D.; Mori, T.; Townsend, K.; Vendrame, M.; Sun, N.; Zeng, J.; Ehrhart, J.; Silver, A.A.; Sanberg, P.R.; Tan, J. Cholinergic modulation of microglial activation by alpha 7 nicotinic receptors. J. Neurochem. 2004, 89, 337–343. [Google Scholar] [CrossRef]
  177. Liu, Z.; Neff, R.A.; Berg, D.K. Sequential interplay of nicotinic and GABAergic signaling guides neuronal development. Science 2006, 314, 1610–1613. [Google Scholar] [CrossRef]
  178. Sinkus, M.L.; Graw, S.; Freedman, R.; Ross, R.G.; Lester, H.A.; Leonard, S. The human CHRNA7 and CHRFAM7A genes: A review of the genetics, regulation, and function. Neuropharmacology 2015, 96, 274–288. [Google Scholar] [CrossRef] [Green Version]
  179. Van Zwieten, P.A.; Thoolen, M.J.; Timmermans, P.B. The hypotensive activity and side effects of methyldopa, clonidine, and guanfacine. Hypertension 1984, 6, II28–II33. [Google Scholar] [CrossRef]
  180. Chobanyan-Jurgens, K.; Jordan, J. Autonomic nervous system activity and inflammation: Good ideas, good treatments, or both? Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H1999–H2001. [Google Scholar] [CrossRef]
  181. Lomasney, J.W.; Lorenz, W.; Allen, L.F.; King, K.; Regan, J.W.; Yang-Feng, T.L.; Caron, M.G.; Lefkowitz, R.J. Expansion of the alpha 2-adrenergic receptor family: Cloning and characterization of a human alpha 2-adrenergic receptor subtype, the gene for which is located on chromosome 2. Proc. Natl. Acad. Sci. USA 1990, 87, 5094–5098. [Google Scholar] [CrossRef]
  182. De Quervain, D.J.; Kolassa, I.T.; Ertl, V.; Onyut, P.L.; Neuner, F.; Elbert, T.; Papassotiropoulos, A. A deletion variant of the alpha2b-adrenoceptor is related to emotional memory in Europeans and Africans. Nat. Neurosci. 2007, 10, 1137–1139. [Google Scholar] [CrossRef]
  183. Rasch, B.; Spalek, K.; Buholzer, S.; Luechinger, R.; Boesiger, P.; Papassotiropoulos, A.; de Quervain, D.J. A genetic variation of the noradrenergic system is related to differential amygdala activation during encoding of emotional memories. Proc. Natl. Acad. Sci. USA 2009, 106, 19191–19196. [Google Scholar] [CrossRef] [Green Version]
  184. Xie, W.; Cappiello, M.; Meng, M.; Rosenthal, R.; Zhang, W. ADRA2B deletion variant and enhanced cognitive processing of emotional information: A meta-analytical review. Neurosci. Biobehav. Rev. 2018, 92, 402–416. [Google Scholar] [CrossRef]
  185. Small, K.M.; Brown, K.M.; Forbes, S.L.; Liggett, S.B. Polymorphic deletion of three intracellular acidic residues of the alpha 2B-adrenergic receptor decreases G protein-coupled receptor kinase-mediated phosphorylation and desensitization. J. Biol. Chem. 2001, 276, 4917–4922. [Google Scholar] [CrossRef]
  186. Suzuki, N.; Matsunaga, T.; Nagasumi, K.; Yamamura, T.; Shihara, N.; Moritani, T.; Ue, H.; Fukushima, M.; Tamon, A.; Seino, Y.; et al. Alpha(2B)-adrenergic receptor deletion polymorphism associates with autonomic nervous system activity in young healthy Japanese. J. Clin. Endocrinol. Metab. 2003, 88, 1184–1187. [Google Scholar] [CrossRef]
  187. Ben-Ari, Y.; Damier, P.; Lemonnier, E. Failure of the Nemo Trial: Bumetanide Is a Promising Agent to Treat Many Brain Disorders but Not Newborn Seizures. Front. Cell Neurosci. 2016, 10, 90. [Google Scholar] [CrossRef]
  188. Shekarabi, M.; Zhang, J.; Khanna, A.R.; Ellison, D.H.; Delpire, E.; Kahle, K.T. WNK Kinase Signaling in Ion Homeostasis and Human Disease. Cell Metab. 2017, 25, 285–299. [Google Scholar] [CrossRef] [Green Version]
  189. Capuron, L.; Miller, A.H. Cytokines and psychopathology: Lessons from interferon-alpha. Biol. Psychiatry 2004, 56, 819–824. [Google Scholar] [CrossRef]
  190. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef]
  191. Cattaneo, A.; Gennarelli, M.; Uher, R.; Breen, G.; Farmer, A.; Aitchison, K.J.; Craig, I.W.; Anacker, C.; Zunsztain, P.A.; McGuffin, P.; et al. Candidate genes expression profile associated with antidepressants response in the GENDEP study: Differentiating between baseline ‘predictors’ and longitudinal ‘targets’. Neuropsychopharmacology 2013, 38, 377–385. [Google Scholar] [CrossRef]
  192. Hannestad, J.; DellaGioia, N.; Bloch, M. The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: A meta-analysis. Neuropsychopharmacology 2011, 36, 2452–2459. [Google Scholar] [CrossRef]
  193. Lanquillon, S.; Krieg, J.C.; Bening-Abu-Shach, U.; Vedder, H. Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology 2000, 22, 370–379. [Google Scholar] [CrossRef]
  194. O’Brien, S.M.; Scully, P.; Fitzgerald, P.; Scott, L.V.; Dinan, T.G. Plasma cytokine profiles in depressed patients who fail to respond to selective serotonin reuptake inhibitor therapy. J. Psychiatr. Res. 2007, 41, 326–331. [Google Scholar] [CrossRef]
  195. Yang, J.J.; Wang, N.; Yang, C.; Shi, J.Y.; Yu, H.Y.; Hashimoto, K. Serum interleukin-6 is a predictive biomarker for ketamine’s antidepressant effect in treatment-resistant patients with major depression. Biol. Psychiatry 2015, 77, e19–e20. [Google Scholar] [CrossRef]
  196. Maruyama, Y.; Kawano, A.; Okamoto, S.; Ando, T.; Ishitobi, Y.; Tanaka, Y.; Inoue, A.; Imanaga, J.; Kanehisa, M.; Higuma, H.; et al. Differences in salivary alpha-amylase and cortisol responsiveness following exposure to electrical stimulation versus the Trier Social Stress Tests. PLoS ONE 2012, 7, e39375. [Google Scholar] [CrossRef]
  197. Schumacher, S.; Kirschbaum, C.; Fydrich, T.; Strohle, A. Is salivary alpha-amylase an indicator of autonomic nervous system dysregulations in mental disorders?—A review of preliminary findings and the interactions with cortisol. Psychoneuroendocrinology 2013, 38, 729–743. [Google Scholar] [CrossRef]
  198. McGirr, A.; Diaconu, G.; Berlim, M.T.; Pruessner, J.C.; Sable, R.; Cabot, S.; Turecki, G. Dysregulation of the sympathetic nervous system, hypothalamic-pituitary-adrenal axis and executive function in individuals at risk for suicide. J. Psychiatry Neurosci. 2010, 35, 399–408. [Google Scholar] [CrossRef]
  199. Harwood, A.J. Lithium and bipolar mood disorder: The inositol-depletion hypothesis revisited. Mol. Psychiatry 2005, 10, 117–126. [Google Scholar] [CrossRef]
  200. Manji, H.K.; Bebchuk, J.M.; Moore, G.J.; Glitz, D.; Hasanat, K.A.; Chen, G. Modulation of CNS signal transduction pathways and gene expression by mood-stabilizing agents: Therapeutic implications. J. Clin. Psychiatry 1999, 60 (Suppl. 2), 27–39; 0–21, 113–116. [Google Scholar]
  201. Le-Niculescu, H.; Levey, D.F.; Ayalew, M.; Palmer, L.; Gavrin, L.M.; Jain, N.; Winiger, E.; Bhosrekar, S.; Shankar, G.; Radel, M.; et al. Discovery and validation of blood biomarkers for suicidality. Mol. Psychiatry 2013, 18, 1249–1264. [Google Scholar] [CrossRef]
  202. Kim, H.G.; Cheon, E.J.; Bai, D.S.; Lee, Y.H.; Koo, B.H. Stress and Heart Rate Variability: A Meta-Analysis and Review of the Literature. Psychiatry Investig. 2018, 15, 235–245. [Google Scholar] [CrossRef] [Green Version]
  203. Hartmann, R.; Schmidt, F.M.; Sander, C.; Hegerl, U. Heart Rate Variability as Indicator of Clinical State in Depression. Front. Psychiatry 2018, 9, 735. [Google Scholar] [CrossRef]
  204. Kemp, A.H.; Quintana, D.S.; Gray, M.A.; Felmingham, K.L.; Brown, K.; Gatt, J.M. Impact of depression and antidepressant treatment on heart rate variability: A review and meta-analysis. Biol. Psychiatry 2010, 67, 1067–1074. [Google Scholar] [CrossRef]
  205. Kemp, A.H.; Quintana, D.S.; Quinn, C.R.; Hopkinson, P.; Harris, A.W. Major depressive disorder with melancholia displays robust alterations in resting state heart rate and its variability: Implications for future morbidity and mortality. Front. Psychol. 2014, 5, 1387. [Google Scholar] [CrossRef]
  206. Wilson, S.T.; Chesin, M.; Fertuck, E.; Keilp, J.; Brodsky, B.; Mann, J.J.; Sonmez, C.C.; Benjamin-Phillips, C.; Stanley, B. Heart rate variability and suicidal behavior. Psychiatry Res. 2016, 240, 241–247. [Google Scholar] [CrossRef]
  207. Krishnan, V.; Nestler, E.J. The molecular neurobiology of depression. Nature 2008, 455, 894–902. [Google Scholar] [CrossRef]
  208. Pariante, C.M.; Lightman, S.L. The HPA axis in major depression: Classical theories and new developments. Trends Neurosci. 2008, 31, 464–468. [Google Scholar] [CrossRef]
  209. Thase, M.E. Using biomarkers to predict treatment response in major depressive disorder: Evidence from past and present studies. Dialogues Clin. Neurosci. 2014, 16, 539–544. [Google Scholar]
  210. Coryell, W.; Schlesser, M. The dexamethasone suppression test and suicide prediction. Am. J. Psychiatry 2001, 158, 748–753. [Google Scholar] [CrossRef]
  211. Jokinen, J.; Nordstrom, P. HPA axis hyperactivity as suicide predictor in elderly mood disorder inpatients. Psychoneuroendocrinology 2008, 33, 1387–1393. [Google Scholar] [CrossRef]
  212. Kamali, M.; Saunders, E.F.; Prossin, A.R.; Brucksch, C.B.; Harrington, G.J.; Langenecker, S.A.; McInnis, M.G. Associations between suicide attempts and elevated bedtime salivary cortisol levels in bipolar disorder. J. Affect. Disord 2012, 136, 350–358. [Google Scholar] [CrossRef] [Green Version]
  213. Pfennig, A.; Kunzel, H.E.; Kern, N.; Ising, M.; Majer, M.; Fuchs, B.; Ernst, G.; Holsboer, F.; Binder, E.B. Hypothalamus-pituitary-adrenal system regulation and suicidal behavior in depression. Biol. Psychiatry 2005, 57, 336–342. [Google Scholar] [CrossRef]
  214. Woo, J.M.; Gibbons, R.D.; Qin, P.; Komarow, H.; Kim, J.B.; Rogers, C.A.; Mann, J.J.; Postolache, T.T. Suicide and prescription rates of intranasal corticosteroids and nonsedating antihistamines for allergic rhinitis: An ecological study. J. Clin. Psychiatry 2011, 72, 1423–1428. [Google Scholar] [CrossRef]
  215. Kalkman, H.O. Circumstantial evidence for a role of glutamine-synthetase in suicide. Med. Hypotheses 2011, 76, 905–907. [Google Scholar] [CrossRef]
  216. Zou, J.; Wang, Y.X.; Dou, F.F.; Lu, H.Z.; Ma, Z.W.; Lu, P.H.; Xu, X.M. Glutamine synthetase down-regulation reduces astrocyte protection against glutamate excitotoxicity to neurons. Neurochem. Int. 2010, 56, 577–584. [Google Scholar] [CrossRef] [Green Version]
  217. Hansson, E. Regulation of glutamine synthetase synthesis and activity by glucocorticoids and adrenoceptor activation in astroglial cells. Neurochem. Res. 1989, 14, 585–587. [Google Scholar] [CrossRef]
  218. Cadoret, A.; Ovejero, C.; Terris, B.; Souil, E.; Levy, L.; Lamers, W.H.; Kitajewski, J.; Kahn, A.; Perret, C. New targets of beta-catenin signaling in the liver are involved in the glutamine metabolism. Oncogene 2002, 21, 8293–8301. [Google Scholar] [CrossRef]
  219. Marcus, S.R.; Nadiger, H.A.; Chandrakala, M.V.; Rao, T.I.; Sadasivudu, B. Acute and short-term effects of lithium on glutamate metabolism in rat brain. Biochem. Pharmacol. 1986, 35, 365–369. [Google Scholar] [CrossRef]
  220. Ren, X.; Rizavi, H.S.; Khan, M.A.; Dwivedi, Y.; Pandey, G.N. Altered Wnt signalling in the teenage suicide brain: Focus on glycogen synthase kinase-3beta and beta-catenin. Int. J. Neuropsychopharmacol. 2013, 16, 945–955. [Google Scholar] [CrossRef]
  221. Baldessarini, R.J.; Tondo, L.; Hennen, J. Lithium treatment and suicide risk in major affective disorders: Update and new findings. J. Clin. Psychiatry 2003, 64 (Suppl. 5), 44–52. [Google Scholar]
  222. Beurel, E.; Jope, R.S. Inflammation and lithium: Clues to mechanisms contributing to suicide-linked traits. Transl. Psychiatry 2014, 4, e488. [Google Scholar] [CrossRef]
  223. Lamers, F.; de Jonge, P.; Nolen, W.A.; Smit, J.H.; Zitman, F.G.; Beekman, A.T.; Penninx, B.W. Identifying depressive subtypes in a large cohort study: Results from the Netherlands Study of Depression and Anxiety (NESDA). J. Clin. Psychiatry 2010, 71, 1582–1589. [Google Scholar] [CrossRef]
  224. Arling, T.A.; Yolken, R.H.; Lapidus, M.; Langenberg, P.; Dickerson, F.B.; Zimmerman, S.A.; Balis, T.; Cabassa, J.A.; Scrandis, D.A.; Tonelli, L.H.; et al. Toxoplasma gondii antibody titers and history of suicide attempts in patients with recurrent mood disorders. J. Nerv. Ment. Dis. 2009, 197, 905–908. [Google Scholar] [CrossRef]
  225. Lamers, F.; Bot, M.; Jansen, R.; Chan, M.K.; Cooper, J.D.; Bahn, S.; Penninx, B.W. Serum proteomic profiles of depressive subtypes. Transl. Psychiatry 2016, 6, e851. [Google Scholar] [CrossRef]
  226. Okusaga, O.; Langenberg, P.; Sleemi, A.; Vaswani, D.; Giegling, I.; Hartmann, A.M.; Konte, B.; Friedl, M.; Groer, M.W.; Yolken, R.H.; et al. Toxoplasma gondii antibody titers and history of suicide attempts in patients with schizophrenia. Schizophr. Res. 2011, 133, 150–155. [Google Scholar] [CrossRef]
  227. Rapaport, M.H.; Nierenberg, A.A.; Schettler, P.J.; Kinkead, B.; Cardoos, A.; Walker, R.; Mischoulon, D. Inflammation as a predictive biomarker for response to omega-3 fatty acids in major depressive disorder: A proof-of-concept study. Mol. Psychiatry 2016, 21, 71–79. [Google Scholar] [CrossRef]
  228. MacKenzie, E.M.; Odontiadis, J.; Le Melledo, J.M.; Prior, T.I.; Baker, G.B. The relevance of neuroactive steroids in schizophrenia, depression, and anxiety disorders. Cell Mol. Neurobiol. 2007, 27, 541–574. [Google Scholar] [CrossRef]
  229. Rethorst, C.D.; Bernstein, I.; Trivedi, M.H. Inflammation, obesity, and metabolic syndrome in depression: Analysis of the 2009-2010 National Health and Nutrition Examination Survey (NHANES). J. Clin. Psychiatry 2014, 75, e1428–e1432. [Google Scholar] [CrossRef]
  230. Capuron, L.; Lasselin, J.; Castanon, N. Role of Adiposity-Driven Inflammation in Depressive Morbidity. Neuropsychopharmacology 2017, 42, 115–128. [Google Scholar] [CrossRef]
  231. Perez-Cornago, A.; de la Iglesia, R.; Lopez-Legarrea, P.; Abete, I.; Navas-Carretero, S.; Lacunza, C.I.; Lahortiga, F.; Martinez-Gonzalez, M.A.; Martinez, J.A.; Zulet, M.A. A decline in inflammation is associated with less depressive symptoms after a dietary intervention in metabolic syndrome patients: A longitudinal study. Nutr. J. 2014, 13, 36. [Google Scholar] [CrossRef]
  232. Sublette, M.E.; Galfalvy, H.C.; Fuchs, D.; Lapidus, M.; Grunebaum, M.F.; Oquendo, M.A.; Mann, J.J.; Postolache, T.T. Plasma kynurenine levels are elevated in suicide attempters with major depressive disorder. Brain Behav. Immun. 2011, 25, 1272–1278. [Google Scholar] [CrossRef] [Green Version]
  233. Erhardt, S.; Lim, C.K.; Linderholm, K.R.; Janelidze, S.; Lindqvist, D.; Samuelsson, M.; Lundberg, K.; Postolache, T.T.; Traskman-Bendz, L.; Guillemin, G.J.; et al. Connecting inflammation with glutamate agonism in suicidality. Neuropsychopharmacology 2013, 38, 743–752. [Google Scholar] [CrossRef]
  234. Sadkowski, M.; Dennis, B.; Clayden, R.C.; Elsheikh, W.; Rangarajan, S.; Dejesus, J.; Samaan, Z. The role of the serotonergic system in suicidal behavior. Neuropsychiatr. Dis. Treat. 2013, 9, 1699–1716. [Google Scholar] [CrossRef] [Green Version]
  235. Sandyk, R.; Awerbuch, G.I. Nocturnal melatonin secretion in multiple sclerosis patients with affective disorders. Int. J. Neurosci. 1993, 68, 227–240. [Google Scholar] [CrossRef]
  236. Stanley, M.; Brown, G.M. Melatonin levels are reduced in the pineal glands of suicide victims. Psychopharmacol. Bull. 1988, 24, 484–488. [Google Scholar]
  237. Teraishi, T.; Hori, H.; Sasayama, D.; Matsuo, J.; Ogawa, S.; Ota, M.; Hattori, K.; Kajiwara, M.; Higuchi, T.; Kunugi, H. (13)C-tryptophan breath test detects increased catabolic turnover of tryptophan along the kynurenine pathway in patients with major depressive disorder. Sci. Rep. 2015, 5, 15994. [Google Scholar] [CrossRef]
  238. Kraus, C.; Kadriu, B.; Lanzenberger, R.; Zarate, C.A., Jr.; Kasper, S. Prognosis and improved outcomes in major depression: A review. Transl. Psychiatry 2019, 9, 127. [Google Scholar] [CrossRef]
Figure 1. This scheme is based upon data published by [44,58]. Stress activates a sequence of events that involves neuron-derived glutamate, astrocyte-derived ATP, and microglia-derived brain-derived neurotrophic factor (BDNF), and ultimately results in higher network excitability. Since stress induces dendritic atrophy, the elevated network excitability might serve a repair function. Legend: GR, glucocorticoid receptor; NMDA, N-methyl-D-aspartate; TrkB, tropomyosin receptor kinase-B; KCC2, K-Cl-cotransporter-2.
Figure 1. This scheme is based upon data published by [44,58]. Stress activates a sequence of events that involves neuron-derived glutamate, astrocyte-derived ATP, and microglia-derived brain-derived neurotrophic factor (BDNF), and ultimately results in higher network excitability. Since stress induces dendritic atrophy, the elevated network excitability might serve a repair function. Legend: GR, glucocorticoid receptor; NMDA, N-methyl-D-aspartate; TrkB, tropomyosin receptor kinase-B; KCC2, K-Cl-cotransporter-2.
Pharmaceuticals 12 00113 g001
Figure 2. This scheme is based upon data published by [143]. It assumes that inflammation causes interleukin-6 (IL6)-release and shedding of sIL6R from microglia. Trans-activation of astrocytes would lead to the ‘neurotoxic-reactive’ phenotype that may cause damage to neurons. Activation of the glucocorticoid receptor would limit the inflammation-induced sequence of events. Legend: sIL6R, soluble IL-6-receptor.
Figure 2. This scheme is based upon data published by [143]. It assumes that inflammation causes interleukin-6 (IL6)-release and shedding of sIL6R from microglia. Trans-activation of astrocytes would lead to the ‘neurotoxic-reactive’ phenotype that may cause damage to neurons. Activation of the glucocorticoid receptor would limit the inflammation-induced sequence of events. Legend: sIL6R, soluble IL-6-receptor.
Pharmaceuticals 12 00113 g002
Figure 3. A graphical summary of the biological effects induced by stress and inflammation. As reviewed in Section 2 and Section 3 of this manuscript, inflammation and psychological stress activate the hypothalamic-pituitary-adrenal (HPA) axis, the sympathetic nervous system, and increase glutamate, while the inhibitory effect of gamma-aminobutyric acid (GABA) is diminished. This leads to measurable increases in circulating corticosteroids, catecholamines, anxiety, somatic and psychopathological symptoms, and a decline in cognitive functions. Both stress and inflammation affect the function of the chloride transporters, Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2), and provoke IL-6 trans-signaling. Although there is a large qualitative overlap, it may be that stress and inflammation lead to quantitative differences in the severity of the individual effects. Legend: BP = blood pressure, CS = corticosterone, HR = heart rate, NA = noradrenaline, ns = nervous system, PKC = protein kinase C, sIL6R = soluble IL6 receptor.
Figure 3. A graphical summary of the biological effects induced by stress and inflammation. As reviewed in Section 2 and Section 3 of this manuscript, inflammation and psychological stress activate the hypothalamic-pituitary-adrenal (HPA) axis, the sympathetic nervous system, and increase glutamate, while the inhibitory effect of gamma-aminobutyric acid (GABA) is diminished. This leads to measurable increases in circulating corticosteroids, catecholamines, anxiety, somatic and psychopathological symptoms, and a decline in cognitive functions. Both stress and inflammation affect the function of the chloride transporters, Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2), and provoke IL-6 trans-signaling. Although there is a large qualitative overlap, it may be that stress and inflammation lead to quantitative differences in the severity of the individual effects. Legend: BP = blood pressure, CS = corticosterone, HR = heart rate, NA = noradrenaline, ns = nervous system, PKC = protein kinase C, sIL6R = soluble IL6 receptor.
Pharmaceuticals 12 00113 g003
Table 1. Depression is comorbid with inflammatory disorders.
Table 1. Depression is comorbid with inflammatory disorders.
ComorbidityCitation
Viral infection (e.g., HIV*)[36,81]
Bacterial infection (e.g., periodontitis)[68,82]
Allergic inflammation (e.g., asthma)[83]
Autoimmune disease (MS)[84]
Autoimmune disease (RA)[85,86,87]
Neurological disorder (Parkinson’s)[88,89]
Neurological disorder (Alzheimer’s)[90,91]
Cardiovascular disease (heart failure)[92]
Diabetes[93]
Obesity[94]
*HIV, human immunodeficiency virus; MS, multiple sclerosis; RA, rheumatoid arthritis.

Share and Cite

MDPI and ACS Style

Kalkman, H.O. Novel Treatment Targets Based on Insights in the Etiology of Depression: Role of IL-6 Trans-Signaling and Stress-Induced Elevation of Glutamate and ATP. Pharmaceuticals 2019, 12, 113. https://0-doi-org.brum.beds.ac.uk/10.3390/ph12030113

AMA Style

Kalkman HO. Novel Treatment Targets Based on Insights in the Etiology of Depression: Role of IL-6 Trans-Signaling and Stress-Induced Elevation of Glutamate and ATP. Pharmaceuticals. 2019; 12(3):113. https://0-doi-org.brum.beds.ac.uk/10.3390/ph12030113

Chicago/Turabian Style

Kalkman, Hans O. 2019. "Novel Treatment Targets Based on Insights in the Etiology of Depression: Role of IL-6 Trans-Signaling and Stress-Induced Elevation of Glutamate and ATP" Pharmaceuticals 12, no. 3: 113. https://0-doi-org.brum.beds.ac.uk/10.3390/ph12030113

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop