Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2012 Jun;10(2):97–123. doi: 10.2174/157015912800604542

Immunomodulation Mechanism of Antidepressants: Interactions between Serotonin/Norepinephrine Balance and Th1/Th2 Balance

Matteo Martino 1,*, Giulio Rocchi 1, Andrea Escelsior 1, Michele Fornaro 1,2
PMCID: PMC3386509  PMID: 23204981

Abstract

Neurotransmitters and hormones regulate major immune functions, including the selection of T helper (Th)1 or Th2 cytokine responses, related to cell-mediated and humoral immunity, respectively. A role of imbalance and dynamic switching of Th1/Th2 system has been proposed, with relative displacement of the immune reserve in relation to complex interaction between Th1/Th2 and neuro-hormonal balance fluctuations, in the pathogenesis of various chronic human diseases, probably also including psychiatric disorders. Components of the stress system such as norepinephrine (NE) and glucocorticoids appear to mediate a Th2 shift, while serotonin (5-HT) and melatonin might mediate a Th1 shift. Some antidepressants would occur affecting these systems, acting on neurotransmitter balance (especially the 5-HT/NE balance) and expression levels of receptor subtypes, which in turn affect cytokine production and relative Th1/Th2 balance. It could be therefore hypothesized that the antidepressant-related increase in NE tone enhances the Th2 response, while the decrease in NE tone or the increase in 5-HT tone enhances the Th1 response. However, the neurotransmitter and Th1/Th2 balance modulation could be relative, aiming to restore physiological levels a previous imbalance in receptor sensitivity and cytokine production. The considerations on neuro-immunomodulation could represent an additional aid in the study of pathophysiology of psychiatric disorders and in the choice of specific antidepressants in specific clusters of symptoms, especially in comorbidity with internal pathologies. Furthermore limited data, reviewed here, have shown the effectiveness of some antidepressants as pure immunomodulators. However, these considerations are tentative and require experimental confirmation or refutation by future studies.

Keywords: Antidepressants, cytokines, neurotransmitters, receptors, Th1, Th2.

INTRODUCTION

In recent years, a growing number of studies are highlighting the links between the immune system and the neuroendocrine system. Neurotransmitters and hormones regulate major immune functions, such as antigen presentation, antibody production, lymphocyte activity, proliferation and traffic, and the secretion of cytokines including the selection of Th1 or Th2 cytokine responses [1]. For several years a role of altered Th1/Th2 cytokine balance in the pathogenesis of various human diseases [2], including psychiatric disorders has been proposed [3].

Recently, it was shown that some psychotropic drugs modify the serum concentrations of several cytokines, both in depressed patients and in healthy controls [4], making plausible an immunomodulatory action of these drugs. In fact, the modulation of neurotransmitter systems operated by these molecules seems to affect cytokine production and, consequently, Th1/Th2 balance.

Frequent comorbidities have been reported between psychiatric disorders, such as depression, and several internal pathologies, such as cardiovascular disease [5], cerebrovascular disease [6], inflammatory bowel diseases (depression and Crohn’s disease versus stress factors and ulcerative colitis) [7, 8], psoriasis [9], chronic obstructive pulmonary disease (COPD) [10], rheumatoid arthritis [11], multiple sclerosis [12], systemic lupus erythematosus [13], diabetes [14], thyroid disorders [15], neurodegenerative disorders [16], HCV [17], HIV [18] and cancer [19]. In this regard, data are emerging on both difficulties and potential uses of antidepressants in internistic comorbidities and also as pure immunomodulators.

A large amount of data has been published on cytokine balances in several human diseases, with still incomplete and sometimes contradictory results. The Th1/Th2 balance, although simplistic from an immunological point of view, represents the basis on which most of the data have been accumulated. Therefore, an overview on this topic is useful to define a reference system in order to frame the interactions between the neuroendocrine system, the immune system and drugs such as antidepressants, which represent the core of interest of this work.

The purpose of this study is to collect available data on the effects of neurotransmitters on cytokine production and compare them with those available on the effects of antidepressants on neurotransmitter and cytokine modulation, providing an overview on the role of Th1/Th2 balance in several chronic diseases, in order to analyze some aspects of the neuro-endocrine-immune (NEI) system that could find a clinical use.

MATERIALS AND METHODS

A MEDLINE search (1980 - April 2011) was conducted for English-language literature containing combinations of the following key words: “antidepressants”, “SSRI”, “venlafaxine”, “duloxetine”, “bupropion”, “mirtazapine”, “agomelatine”, “monoamine oxidase inhibitors”, “tricyclics”, “norepinephrine”, “serotonin”, “dopamine”, “acetylcholine”, “cortisol”, “melatonin”, “receptors”, “cytokines”, “Th1”, “Th2”, “asthma”, “allergic rhinitis”, “eczema”, “rheumatoid arthritis”, “multiple sclerosis”, “type 1 diabetes mellitus”, “immune thyroid disease”, “Behcet’s disease”, “IgA nephropathy”, “metabolic syndrome”, “essential hypertension”, “dyslipidemia”, “obesity”, “insulin resistance”, “type 2 diabetes mellitus”, “hypercoagulability”, “atherosclerosis”, “cardiovascular diseases”, “acute coronary syndromes”, “acute myocardial infarction”, “chronic heart failure”, “psoriasis”, “chronic obstructive bronchopneumopathy disease”, “Crohn’s disease”, “ulcerative colitis”, “osteoporosis”, “ageing”, “Alzheimer’s disease”, “Parkinson’s disease”, “HIV”, “HCV”, “herpes virus”, “leishmania”, “syphilis”, “tuberculosis”, “cancer”, “metastasis”, “psychiatric disorders”, “mood disorders”, “anxiety disorders”. The references of published articles identified in the initial search process were also examined for any additional studies appropriate for the review.

NEUROHORMONAL-CYTOKINES INTERACTIONS AND NEI STRESS-ADAPTATION SYSTEM

Cytokines and Th1/Th2 Balance

Since several years it was proposed that naïve T lymphocytes (Th0) differentiate into subsets of CD4+ T cells that produce distinct types of cytokines [20]. Interleukin (IL)-12, produced by antigen-presenting cells such as monocytes/macrophages and dendritic cells, in concert with IL-18 and Interferon (IFN)-γ promote the differentiation of Th0 cells towards the Th1 phenotype; Th1 cells primarily secrete IFN-γ, IL-2 and Tumor Necrosis Factor (TNF)-β; IL-1, IL-12, TNF-α and IFN-γ (in addition to downregulation of Tumor Growth Factor (TGF)-β) also stimulate the functional activity of cytotoxic cells, natural killer cells and activated macrophages, which are the major components of cell-mediated immunity [1]. Th2 cells produce IL-4, IL-5, IL-6, IL-10 and IL-13 that promote Th2 response stimulating the growth and activation of mast cells (resulting in degranulation and histamine release) and eosinophils (under action of IL-5), the differentiation of B cells into antibody-secreting B cells and the B cell immunoglobulin switching to IgE (under action of IL-4 and IL-13), which are the major components of humoral immunity [1]. These T cells subsets reciprocally regulate themselves since IL-12 and IFN-γ inhibit Th2, while IL-4 and IL-10 inhibit Th1 cell activities: so Th1 and Th2 responses are mutually inhibitory. In this regard, the Th1/Th2 balance hypothesis involves homeostasis between Th1 and Th2 activity that directs different immune response pathways: Th1 cells lead the cell-mediated immunity to fight intracellular pathogens such as viruses and eliminate cancerous cells, while Th2 cells lead the humoral activity to fight extracellular organisms [2].

Neurotransmitters and Cytokines (Table 1)

Table 1.

Neurotransmitters and Cytokines

Neurotransmitters Receptors Cytokines Th1/Th2 Balance Hypothetic Shifts References
Norepinephrine β2 adrenoreceptors ↓IL-12, IL-1, IFN-γ, TNF-α ↑IL-10, IL-4, IL6 Th2 shift Elenkov, 2008 [1]
Serotonin 5-HT1 receptors ↑IL-1β, IFN-γ, TNF-α Conflicting data Th1 shift Kubera et al., 2005 [32] Conflicting data
Dopamine D1-like receptors Th2 differentiation in CD4 naïve Conflicting data Th2 shift Nakano et al., 2009 [39] Conflicting data
Acetylcholine N ACh receptors Upregulation Th1 transcription factors Th1 shift Kikuchi et al., 2008 [42]
Histamine H1 and H2 receptors ↓IL-12, TNF-α, IFN-γ ↑IL-10, IL-6 Th2 shift Elenkov, 2008 [1]
Glucocorticoids Cytoplasmic/nuclear GR ↓IL-12, IL-1, IFN-γ, TNF-α ↑IL-10, IL-4, IL-13 Th2 shift Elenkov, 2008 [1]
Melatonin RZR/ROR MT receptors ↑IL-2, IFN-γ, IL-6 / ↓IL-10 Th1 shift Kuhlwein and Irwin, 2001 [43] Garcia-Maurino et al., 1997 [44]

The table summarizes cytokines modulation, receptors involved and hypothetical influence on Th1/Th2 balance for each neurotransmitter. (IL: Interleukin; IFN: Interferon; TNF: Tumor Necrosis Factor).

Evidence accumulated over the last 2-3 decades indicates that neurohormonal messages from the brain superimpose upon and interweave with cytokine balance and immune functions [1].

Cytokine-releasing cells express functional receptors for neurotransmitters, glucocorticoids and cytokines which include the G-linked NE and 5-HT receptors such as β2 [21], 5-HT1A, 5-HT2A, 5-HT1B, 5-HT3 [22-26] and 5-HT transporter [23], the glucocorticoid and mineral corticoid receptors [27], and the five families of cytokine receptors, which are constitutively expressed [28, 29]. NE and 5-HT receptors activate cAMP-dependent pathways which affect both the synthesis and release of cytokines and cellular proliferation [22], while, upon binding cortisol, cytoplasmic/nuclear glucocorticoid receptors undergo dimerization and translocation to the nucleus where they act to modulate lymphocyte proliferation and cytokine gene transcription [30].

Recent evidences indicate that both glucocorticoids and catecholamines (NE and epinephrine) systematically mediate a Th2 shift suppressing Th1-cytokine, such as IL-12, IFN-γ, IL-1, TNF-α, and up-regulating Th2-cytokine, such as IL-10, IL-4 and IL-6, production, through stimulation of classic cytoplasmic/nuclear glucocorticoid receptors and β2 adrenergic receptors, respectively [1, 31]. Specifically, since β2 adrenergic receptors are expressed on Th1 cells, but not on Th2 cells [21], NE does not affect directly the cytokine production by Th2 cells; however NE stimulation of β2 adrenergic receptors on Th1 cells suppresses the production of IL-12, the main inducer of Th1 response that stimulates the release of IFN-γ and inhibits that of IL-4; so the NE suppression of IL-12 on Th1 cells indirectly determines the reduction of IFN-γ and the increase of IL-4, thus shifting the Th1/Th2 balance towards Th2 polarization [1]. For instance, since catecholamines up-regulate IL-6 production, the chronic hyper-noradrenergic state may drive the increase in systemic IL-6 levels [1]. Histamine, through activation of H1 and H2 histamine receptors inhibits IL-12, TNF-α and INF-γ, but potentiates IL-10 and IL-6, driving to a Th2 shift [1].

On the other hand, 5-HT appears to mediate a Th1 shift. Previous studies have shown that physiological doses of 5-HT (0.15 a 1.5 µg/mL) induce the secretion of the pro-inflammatory cytokines IL-1β, IFN-γ and TNF-α, while at supraphysiological doses (15 µg/mL) the secretion of the same cytokines decreased [32]. Indeed, some evidence shows that 5-HT seems to increase IL-10 and IL-6 and decrease IL-12, by binding to 5-HT3, 5-HT4 and 5-HT7 receptors [33], while other studies demonstrate that 5-HT reduces the formation of antibodies, the humoral arm of the immune response mediated at least in part by the Th2 system, through the group of 5-TH1 receptors [34]. Overall, the increase in serotonergic tone would lead to a reduction in noradrenergic tone [35] and in dopaminergic tone [36], reduction in turn correlated with the increase of the cholinergic one [37, 38]. Some evidences show that dopamine (DA) dose dependently polarize the Th2 differentiation in naive CD4+ T cells via D1-like receptors [39], although other studies bring back conflicting data [40, 41]. Stimulation of nicotinic cholinergic receptors appears instead to shift T cells toward Th1 pattern through upregulation of Th1-specific transcription factors [42]. Finally it should be noted that 5-HT is also the precursor of melatonin, which enhances Th1 response increasing the production of IL-2 and INF-γ and decreasing the production of IL-10, via nuclear receptor-mediated transcriptional control like the melatonin receptor RZR / ROR [43, 44]; also, one study reported an increase in production of IL-6 by the same melatonin [44].

The complex mode of action of neurotransmitters on the Th1/Th2 balance should be analyzed taking into account the molecular context in which each neurotransmitter acts, impact of which also depends on T cells activation state, receptor subtypes expression, neurotransmitter dose and other co-released molecules [45].

The NEI Stress-adaptation System

From a physiological perspective, environmental stress activates noradrenergic system, hypothalamus-pituitary-adrenal (HPA) axis with cortisol release and Th2 shift, components of the NEI stress-adaptation system [46]. Such systems, in turn, induce a compensatory response with feedback mechanisms in serotonergic system and Th1 components [47]: for example, cortisol downregulates 5-HT1A somatodendritic presynaptic autoreceptors in the raphe nuclei (which play an inhibitory role on 5-HT release), thus enhancing serotonergic tone [47], and, in turn, influencing Th1 response [32]. In adaptive stress (mainly predictable and voluntary), these systems ideally compensate in order to achieve adaptation to higher levels of stress, through epigenetic mechanisms that converge on receptor sensitization/desensitization (such as 5-HT1A), regulation of neurotrophic factor expression (such as BDNF, VEGF and NGF), neurogenesis and synaptic enhancement (due to NMDA receptor modulation and LTP mechanism) on specific neural networks (such as hippocampus network), restoring neurotransmitter and cytokine levels on physiological values (similarly to neuromuscular system adaptation during repeated physical exercise) [47-49]. During inflammation, the activation of the stress system, through induction of a Th2 shift protects the organism from systemic "overshooting" with Th1/pro-inflammatory cytokines [1]. Under certain conditions, however, stress hormones and activation of the corticotropin-releasing hormone (CRH)/substance P - histamine axis may actually facilitate inflammation, through induction of IL-1, IL-6, IL-8, IL-18, TNF-α and CRP production [1]. On the other hand, pro-inflammatory cytokines, such as IL-1, IL-6 and TNF-α, released during immune response and inflammation activate the central components of the stress system, activate the hepatic synthesis of acute phase proteins and alter neurotransmitter networks activity, inducing the “sickness behavior” (fever, sleepiness, fatigue, loss of appetite and decreased libido) [1].

HUMAN CHRONIC DISEASES AND TH1/TH2 BALANCE

For several years a role of altered Th1/Th2 cytokine balance in the pathogenesis of various human diseases has been proposed [2]: a distinct Th1/Th2 divergence determine resistance versus susceptibility to several diseases [50].

Autoimmune and Allergic Diseases

Several autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, type 1 diabetes mellitus and immune thyroid disease, are characterized by dominant Th1 responses, with an excess of IL-12 and TNF-α production and a deficit of IL-10 production, critical factors in the proliferation and differentiation of Th1-related autoreactive cells [51]. On the other hand, allergic diseases, such as asthma, allergic rhinitis, eczema and IgE-mediated food allergy, are characterized by dominant Th2 responses, with overproduction of IL-4, IL-5, IL-9 and IL-13, resulting in activation of mast cells (and consequent histamine release), eosinophils and shifting to IgE production [52, 53].

Multiple sclerosis seems to be associated with a predominantly Th1 cytokine pattern, characterized by increased levels of IFN-γ, TNF-α, IL-12, IL-23 and IL-27 as well as IL-6 and decreased levels of IL-10 and TGF-β [54-56], although other cell subsets such as Th17 cells, regulatory T cells and B cells also appear to be involved [57]. However, this framework could represent the result of a multistep process related to genetic trait, virus infection, activation of the stress system and fluctuations in the Th1-Th2 balance. Stressful life events seem to reduce the clearance of virus infection or permit the episodic reactivation and dissemination of latent viruses (such as herpes virus), probably through overactivation of the HPA axis and the Th2 switch (current multiple sclerosis treatment includes agents for acute relapses, such as corticosteroids, and disease-modifying agents, such as the antiviral IFN-β, which has a beneficial effect on relapsing/remitting multiple sclerosis [58]); patients with multiple sclerosis generate autoreactive T cells that at some points differentiate into Th1 phenotype cells, which are the major players in the late disease maintaining a continuous destructive cell mediated immune response against brain and spinal cord antigens resulting in demyelination [59-61].

In rheumatoid arthritis, Th1 cytokines such as TNF-α, IFN-γ, IL-1, IL-12 and IL-18, as well as IL-23/IL-17 axis, contribute to joint inflammation, cell infiltration and destruction [62-68]; there also appears to be a compensatory anti-inflammatory response in synovial membrane, suggesting a protective role of Th2 cytokines [62]. However, transient Th2 phenotype, characterized by predominance of IL-4 and IL-13 and marked absence of IFN-γ, might be relevant in the early disease [69] and linked to B cell hyperactivity resulting in aberrant autoantibodies (e.g. rheumatoid factor and anti-cyclic-citrullinated antibodies) and immune complexes, that can also be found prior to the clinical onset and may be important in the initiation of the disease [66-68].

Studies on type 1 diabetes have shown that destructive insulitis is mediated through a Th1-dominated autoimmune response associated with an increased expression of IL-1, IL-2, IL-12, TNF-α and IFN-γ [70], as well with unregulated IL-23/IL-17 response [71], and cell mediated beta-cell destruction; this has been suggested by some to arise relatively late in the disease process after a latent period of non-destructive Th2 islet autoimmunity associated with an increased expression of IL-4, IL-10 and TGF-β and with the appearance of autoantibodies targeting, for example, insulin and glutamic acid decarboxylase [72].

About autoimmune thyroid diseases, some studies suggest that Hashimoto thyroiditis is characterized by Th1-dominant response associated with an increased expression of IFN-γ, TNF-α, IL-2, IL-12, IL-18, IL-1β, IL-8 and Th17 system and cell mediated thyroid infiltration and destruction [73-75]. On the other hand, the predominance of Th2 cytokines such as IL-4, IL-5, IL-6 and IL-10 and the related humoral pattern of immune reaction associated with B cell activation and production of autoantibodies against thyroid antigens, characterize Graves disease and early stages of Hashimoto thyroiditis: in Graves disease autoantibodies hyper-stimulate thyroid function, while in Hashimoto thyroiditis, on the contrary, the autoantibodies presence precedes the onset of subclinical and clinical hypothyroidism (thyroid peroxidase antibodies are quantitatively related with the IL-10 levels, which then gradually decrease as the disease progresses) [73-77].

Other diseases characterized by a Th1 polarized response are Behcet’s disease, in which there are increased levels of IFN-γ, IL-12 and IL-13 and reduced levels of IL-4 [78] as well as vitamin D [79], and IgA nephropathy, in which there is an increase in the ratio IFN/IL-10 and increased levels of IFN-γ, TNF, IL-1, IL-2 and IL-8 [80, 81].

Interestingly, some studies show that a hypoactive stress system may facilitate or sustain the Th1 shift in Th1-mediated diseases, such as rheumatoid arthritis [82-85].

On the other hand, systemic lupus erythematosus seems to be an autoimmune disease characterized by dominant Th2 response; however, during advanced stages of disease there is substantial ongoing Th1 cell-mediated inflammation. An important genetic load and environmental factors are involved in the pathophysiology of the disease, such as sunlight exposure and abnormal estrogen metabolism or environmental estrogen exposure (in postmenopause or in contraception use), whereas there is little evidence of infection; notably, estrogens, as well as sunlight, seem to shift the cytokine balance towards Th2 response, increasing IL-4, IL,10 and TGF-β and decreasing IFN-γ, TNF-α and IL-12, whereas androgens seem to enhance the Th1 response [86, 87]: the shifting to Th2 response, characterized by the increase of IL-10 and IL-6 and the decrease of IL-2 and IL-12, together with defective immune regulatory mechanisms such as the clearance of apoptotic cells and immune complexes, leads to B cell hyperactivity and production of pathogenic autoantibodies [86]. During advanced stages of disease there is substantial ongoing Th1 (and possibly Th17) cell-mediated inflammation, such as in lupus nephritis, characterized by increase in IFN-γ, TNF-α, IL-12, IL-17, IL-23 and IL-18 [88-93], suggesting diversified immuno-modulatory interventions depending on the stage of disease [88].

Regarding allergic diseases, although the onset of acute atopic dermatitis is strongly associated with the production of Th2 cytokines, notably IL-4, IL-5, IL-13 and IL-31, the expression of IFN-γ and IL-17 rather than IL-4 predominates in older lesions, indicating a cytokine switch from Th2 in the acute phase towards Th1 in the chronic phase [94].

Also, allergic asthma seems to be mediated by a Th2-dominated response, related to IL-4, IL-5, IL-9 and IL-13 increase [95-96]; however, after migration to asthmatic lung, cells switch on Th1 cytokines such as IFN-γ and TNF-α and, together with tissue-infiltrating Th17 cells, may modulate the local chronic inflammatory response [97-99]; moreover, Th2 cells are recruited efficiently to sites of tissue inflammation, creating a vicious circle in association with disease exacerbations related for example to bacterial or viral respiratory tract infections [100].

In summary, several immune-mediated diseases seem to be characterized by a multistep process resulting from complex interaction between predisposing genetic traits, infections, episodic and chronic activation of the neuroendocrine stress system and fluctuations in the Th1-Th2 system. The changes in cytokines expression show often a mixed pattern. Nevertheless, in the early stages Th2 activation is often detected, correlated with humoral response and allergic reactions or production of autoantibodies, which in some cases appear clinically silent, while in other determine clinically significant pathophysiological phenomena. In the late stages the immune response seems to show in several diseases a switch from Th2 to Th1 correlated to cell-mediated organ inflammation. Episodic or chronic activation of the neuroendocrine stress system due to several causes, including stressful events and intercurrent infections together with genetic polymorphisms and epigenetic factors, affects the Th2 response, while the Th2-to-Th1 switch has been in some cases linked to a hypoactive stress system.

Inflammatory, Metabolic and Cardiovascular Diseases

Chronic activation of the Th2-related stress system seems to lead to a Th1 switch with elevation of both Th1 and Th2 inflammatory cytokines that result in chronic systemic inflammation associated with a cluster of metabolic disturbances named metabolic syndrome, including arterial hypertension, dyslipidemia and obesity (specifically the visceral type), insulin resistance and/or diabetes type 2, in addition to endothelial inflammation and hypercoagulability of the blood [101]. These immune and metabolic changes increase all cause mortality, primarily cardiovascular due to atherosclerosis, and also cancer and infection-related [1, 102].

Essential hypertension seems to be associated with a cytokine pattern biased towards the Th2 system, with an increase of IL-4, IL-7, IL-13 [103], IL-6 [104] and TGF-β [105], as well as the pro-inflammatory cytokine TNF-α [104]. This pattern could be related to the hyper-reactivity of the stress system found in patients with essential hypertension. Mental stress induces phenylethanolamine N-methyltransferase, which may act as a DNA methylase, causing the NET gene silencing, which in turn, may exaggerate autonomic responsiveness [106]: this epigenetic modification has been hypothesized to underlie both essential hypertension and panic disorder, conditions frequently associated to each other [107]; moreover, cortisol responses to mental stress are greater and more persistent in people at high risk for hypertension relative to low-risk normotensives [108, 109].

Dyslipidemia is associated with an impaired Th1 and enhanced Th2 responses [110]. While an increase of IFN-γ was found in course of moderate hypercholesterolemia, severe hypercholesterolemia seems to be characterized by a Th2 shift, with an increase of IL-4 [111]: notably, cortisol and catecholamines, released during stress response, are involved in fat mobilization [112, 113].

Obesity, especially the visceral type, is associated with a predominantly Th1 cytokine pattern, with increased levels of IFN-γ, TNF-α, IL-1 and IL-8, as well as of the Th2 proinflammatory cytokine IL-6 [114-118]; furthermore leptin, an adipocyte-secreted hormone, modulates Th cells toward a Th1 phenotype [118]. However, it was shown in an animal model that stress exposure during the adolescent period decreases subcutaneous fat content, without change in visceral fat, and consequently increases the visceral fat / subcutaneous fat ratio in adulthood; treatment with SSRIs during stress exposure prevented later effects on body fat distribution [119]. It is possible that, while Th2-related acute stress reduces the subcutaneous fat to the mobilization of energy reserves, chronic stress, in association with a chronic activation of the HPA axis and a Th2-to-Th1 switch, facilitates Th1 inflammation and visceral fat distribution. HPA axis activation increases cortisol production; Cushing’s syndrome is the best evidence showing a link between chronic hypercortisolemia and accumulation of central fat; since fat cells catabolize glucocorticoids, hypertrophy of visceral fat could represent, among others, an adaptive response to chronic hyperactivity of the HPA axis, and so an adaptive role in response to stress; in fact abdominal obesity is associated with an increased cortisol clearance [120, 121].

Insulin resistance seems to be associated with a predominantly Th1 cytokine pattern: TNF-α (and to a lesser extent IL-1 and IL-6), produced by visceral fat among others, inhibits insulin signaling pathways, leading to insulin resistance [116, 122-124]. Type 2 diabetes is associated with an elevation of Th1 subset, with an increase of IFN-γ and IL-17 [125], while insulin induces a shift in T cell differentiation towards a Th2 response, decreasing IFN-γ and increasing IL-4 [126]. Resulting hyperglycemia determines, besides other effects, a decrease of LDL clearance and endothelial dysfunction, implicated in the pathogenesis of atherosclerosis [116, 122].

Atherosclerosis seems to be a chronic inflammatory disease of vascular endothelium related to a prevalent Th1 cytokine profile, in most cases initiated by chronic hypercholesterolemia, with an increase of Th1 cytokines with atherogenic activity such as IFN-γ, TNF-α, IL-1β, IL-8, IL-12 and IL-18 and a decrease of Th2 cytokines with anti-atherogenic properties such as IL-4, IL-5, IL-10 and IL-13 [127-130], while the increase of systemic mediators such as IL-6 appears to lead to the increased tendency of thrombosis [131]. The Th2-related dyslipidemia appears to be associated in chronic with the increase in oxidized LDL, especially in the presence of hyperglycemia and smoking (in addition probably to other factors such as infections with chlamydia pneumoniae and/or human cytomegalovirus [131]), resulting in the activation of macrophages and T cells that release predominantly Th1 cytokines, and so lead to chronic inflammation of the vessel wall and plaque formation [127-129].

In this regard it is notable that TNF augments inflammation, TNF and IFN-γ induce coagulation, IL-1β induces coagulation and fibrinolysis, IL-8 augments synergistic inflammation and coagulation, IL-6 augments coagulation and inhibits fibrinolysis, IL-10 inhibits inflammation and fibrinolysis and IL-4, IL-13 and TGF-β act for anticoagulation [132]: so hypercoagulability of the blood is associated with an inflammatory state [133] characterized by Th1 predominant response, not only present in atherosclerosis, but also in visceral obesity [134], disseminated intravascular coagulation [135] and cancer [136].

Atherosclerosis represents one of the key factors of cardiovascular and cerebrovascular diseases: the inflammation-related predominant Th1 response has also been implicated in the pathophysiology of chronic heart failure [137, 138] and heart failure after acute myocardial infarction [139]. The stress-related Th2 predominant response seems to be associated with acute coronary syndromes: acute exogenous or endogenous stress factors can trigger unstable angina, sudden cardiac death and acute myocardial infarction [140, 141], condition in which was found, in relation to sympathetic system activation and catecholamines release, an increase of IL-6 [142, 143] and IL-10 [144, 145]. Th2 polarized response also promotes fibrosis in general, and cardiac fibrosis in particular [146]. Regarding the cytokine imbalance in cardiovascular disease it should be noticed that atorvastatin can regulate the polarization of Th1/Th2 through the inhibition of Th1 cytokines [138, 139], while beta-blockers and angiotensin-converting enzyme inhibitors can decrease serum levels of IL-6, which correlates with NYHA class in chronic heart failure [147]. Levels of IL-6 in the serum of acute stroke patients have been reported to also correlate with stroke severity; serum concentrations of IL-6, IL-8 and IL-10 were higher in asphyxiated neonates; while cerebral ischemia initiates a Th1-prevalent inflammatory response associated with induction of a variety of cytokines, including TNF-α, IL-1β, IL-8, IL-18 and IL-6 [148-151].

Mixed Th1/Th2 pattern, related to chronic hyperactivation of proinflammatory cytokines mainly belonging to the Th1 group, seems to be associated to the pathophysiology and evolution of a number of other conditions. Several inflammatory diseases show a prevalence of Th1 response, such as psoriasis, in which there are high levels of IL-2 and IFN-γ and low levels of IL-4 and IL-10 [152, 153]. In COPD, the increase of the Th1 cytokines IFN-γ, TNF-α, IL-2 and IL-8 and the decrease of the Th2 cytokines IL-4 and TGF-β seem to be associated with progression to advanced stages of the disease [154, 155]. Furthermore, alterations of Th1/Th2 balance also appear to be relevant in the difference in clinical and pathophysiological presentation of diseases such as inflammatory bowel disease: the prevalence of a Th1 response involving overproduction of IL-12, IFN-γ and TNF-α manifests in the form of Crohn’s disease, while the prevalence of a Th2 response probably driven by overproduction of IL-13 manifests in the ulcerative colitis [156]. Osteoporosis is associated with low-grade inflammation both Th1 than Th2 type, since the bone turnover is regulated in a complex way: TNF-α, IL-1, IL-6, IL-11, IL-17 and glucocorticoids enhance bone resorption, while IFN-γ, IL-12, IL-18 and IL-4 inhibit bone resorption; osteoporosis is in fact associated both with diseases with a predominant Th1 cytokine pattern, such as atherosclerosis and rheumatoid arthritis, and conditions with a Th2-dominated cytokine pattern, such as ageing, menopause, pregnancy, HIV, steroid or cyclosporine therapy [157].

Other physiological and paraphysiological conditions are associated with changes in cytokine balance potentially favoring different types of diseases: ageing, pregnancy and menopause that have a Th2-dominated cytokine balance. During reproductive adulthood there is a relative bias towards Th1 immunity, which reverses to Th2 dominance, together with an increase of pro-inflammatory cytokines such as TNF-α, IL-1 and IL-6, during post-reproductive senescence, with impaired cellular response and increased susceptibility to viruses, intracellular pathogens and neoplastic diseases [158, 159]; during pregnancy a Th2 shift might be favored by the increase of cortisol and endorphins releases [160].

Neuroinflammation

Neuroinflammation appears to be central in degenerative diseases such as Parkinson disease and Alzheimer disease. Cytokines are expressed physiologically in CNS cells and are important for the development and function of the brain [58]. Furthermore, increasing evidence suggests that changes in cytokines levels outside the brain cause changes in cytokine expression and activity in the brain, and vice versa [161]. In the brain, activated glia, such as microglia and astrocytes, are the major cell types that participate in the inflammatory system both as sources and targets of cytokines [162]; the activation of these cellular systems seems to promote apoptotic cell death and subsequent phagocytosis of DA neurons in Parkinson disease or to be associated with amyloid plaques formation and neurodegeneration of hippocampus and other brain areas in Alzheimer disease [162-164]. In parkinsonian patients, increased levels of TNF-α, IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-10 and TGF concentration in serum, cerebrospinal fluid and nigrostriatal DA regions and decreased ones of neurotrophins were found [163-167]. Yet, Th1 cytokines such as IFN-γ seem to be implicate in apoptotic neuro-degeneration [168-170], while Th2 cytokines such as IL-10 seem to protect against inflammation-mediated degeneration of dopaminergic neurons in the substantia nigra [171, 172], possibly representing a compensatory increase. Furthermore, while damage of DA neurons in the nigrostriatal system seems to be associated with an exacerbated pro-inflammatory response [173], levodopa treatment correlates with reduction of IL-2 production [174]. In Alzheimer disease patients, increased levels of Th1 cytokines such as IFN-γ, and decreased levels of Th2 cytokines such as IL-4 have been found [175]. In a study on dementia-free subjects high levels of IL-1β and TNF-α were associated with an increased risk of developing Alzheimer’s disease [58], while the induction of Th2-biased responses seems to improve the clinical and pathophysiological condition in an Alzheimer’s mouse model, increasing IL-10 and reducing IFN-γ, TNF-α, IL-2 and IL-4 [176, 177]. Yet also Th2 cytokines such as IL-6 seem to have a pathogenic role in neurodegeneration [58, 178]; in this respect it is notable that brain NE levels, which potentially enhances Th2 response, may be elevated at least in some cases or in some phases of Alzheimer disease, considering a direct evidence of elevation in NE or in its metabolites in Alzheimer’s disease and the frequent comorbidity with bipolar disorder or hypertension, disorders in which elevated NE may be involved [179].

In summary, neuroinflammatory pathologies seem to be polarized to Th1 response, although Th2 component appears to play a compensatory role or to influence some stages of disease.

Chronic Infections and Cancer

The Th1-to-Th2 switch hypothesis has been proposed for the first time in chronic infections like HIV, in which it is highlighted that Th1 hyperactivation would result in a resistance or infection control, while the decline in Th1 cell activity and the increase in Th2 cell activity would result in a progression of the infection [180]. It was shown that environmental or internal stressors play a role in HIV disease progression, affecting immunity (CD4 cell counts) and viral set-point (plasma viral load) both in simian [181, 182] and in human [183, 184], and this could be correlated with a Th1-to-Th2 switch dependent on catecholaminergic and HPA axis activation [185-187]; moreover, the Th1-to-Th2 switch could be indicative of an allergic response to viral proteins [188] or other factors. Nevertheless, it was proposed that Th2 cytokine inhibitors and immune response modifiers could lead to a Th2-to-Th1 reversion, inducing a revival of the anti-viral Th1 response [188]. In other chronic systemic infections, a disease control due to Th1 hyperactivation and a disease progression due to Th2 switch were also observed [189], such as in HCV [190-192], herpes virus [193] (the use of topical immunomodulators which stimulate the release of Th1 type cytokines against HSV infected cells, as well as against human papillomavirus and cancerous skin lesions has been proposed [194, 195]), leishmania [196], syphilis [197] and tuberculosis [198-200] (in which HPA axis activation causes increased cortisol secretion possibly resulting in Th2 shift [201]), as well as in atopic diseases such as atopic eczema (in which a dominant Th2 response would replace the Th1/Th2 mixed pattern during the evolution of eczematous lesions) [1].

Regarding cancer, a Th1 phenotype appears to exert anticancer effects [202, 203], although the Th1 cytokine effects may be bimodal, since the inflammatory nature of the Th1 response may allow the development of DNA damage that facilitates transformation [204]. On the other hand, the progression from primary tumor to the metastatic phenotype could correlate to a Th2 switch [205], since it was found, with the passage of time, an increase of Th2 cytokines such as IL-4, IL-6, IL-10 and TGF-β and a decrease of Th1 cytokines such as TNF, IFN-γ and IL-1 in several metastatic cancers that became more evident during disease progression, such as hepatocellular carcinoma [206, 207], head and neck squamous cell carcinoma [208, 209], hypopharyngeal carcinoma [210], esophageal squamous cell carcinoma [211], pulmonary adenocarcinoma [212], breast cancer [213], melanoma [214], lymphoma [215, 216] and pancreatic carcinoma [217-219]. The Th1-to-Th2 switch mechanism could be influenced either by systemic factors such as environmental events that activate the stress system [220] or by local factors such as hypoxia [221]. Similarly to what was observed about the progression of HIV in stress conditions, it was demonstrated that the persistent activation of sympathetic and HPA systems in chronic stress response impaired the immune surveillance of tumors, resulting in the progression of some types of cancer, in association with a shift in the balance of Th1-Th2 immune response [220, 222], such as in breast cancer [223]. In this regard, the connection between the neurometabolic and immune systems appears to be relevant. It has been shown that the indoleamine 2,3-dioxygenase (IDO) enzyme, involved in the catabolic pathway of tryptophan (precursor of 5-HT), is overexpressed both in tumor cell and antigen-presenting cell in tumor draying lymph node, where it promotes the establishment of peripheral immune tolerance to tumor antigens [224]. Therefore, in course of chronic immune activation, decreased serum tryptophan, associated with 5-HT reduced synthesis [225, 226] and hypercortisolism [226], was found parallel to the course of malignant diseases [227]. These alterations appear to be associated, on one hand, with generation of immune tolerance in patients with advanced cancer (facilitating the survival and growth of tumor cell expressing unique antigens that would be recognized normally as foreign by T cells) [224, 225, 227], on the other, with irritability, lack of impulse control and mood disorders [225-227]. Some experimental approaches with IDO inhibitors are underway as a new immunomodulatory treatment [224, 228]. Interestingly, HIV infection is also associated with increased tryptophan catabolism by the IDO, contributing to perpetuation of HIV infection into its chronic phase by dampening efficient immune anti-viral responses [229]. Similarly, an influence of tryptophan degradation on different aspects of autoimmune disorders such as multiple sclerosis [230], rheumatoid arthritis [231] or systemic lupus erythematosus was detected [232]. A compensatory increase in IDO activity in models of rheumatoid arthritis was shown, indeed systemic inhibition of tryptophan catabolism correlated early with an increased production of IFN-γ and IL-17 and later with an increased infiltration of Th1 and Th17 cells in the inflamed joints, increasing disease severity [231]. Furthermore, it was noted that high IDO activity in winter predicted subsequent activation of systemic lupus erythematosus in spring and summer [232]. On the other hand there is evidence that low tyrosine and phenylalanine diet limit tumor growth in animal models, in relation to the reduction in blood levels of these amino acids involved in the catecholamine synthetic pathway; however, this diet has not found clinical applicability in the treatment of patients with advanced cancer for tolerability issues [233].

So, insufficient immunosurveillance is an important aspect in early tumorigenesis and in the pathogenesis of malignant disease; in the later course of cancer, the development of immunodeficiency is considered to be the major reason for disease progression and death [227], and mood symptoms could indicate an immunological failure associated with tumor progression [225-227].

In some cases specific therapeutic actions seem to reverse such changes to a Th1 response, increasing Th1 cytokines and decreasing Th2 cytokines, such as operations [211] and chemotherapy [234], but theoretically it is possible that immunomodulation actions on the stress system could enhance the Th1/Th2 balance restore.

In summary, the Th1-Th2 switch paradigm, in which perturbations of the system allow to switch from Th1 to Th2 response or vice versa, can influence the pathophysiology and evolution of several diseases [235]. Environmental or internal stress factors occurring on Th1 chronic activation could induce a Th2 switch which would lead to a progression of the underlying pathology for relative displacement of the immune reserve, in relation to interaction between fluctuations of Th1/Th2 balance and neuro-hormonal balance.

Psychiatric Diseases

In recent years, a growing body of data was collected on the presence of hormonal and cytokine changes also in various psychiatric disorders, particularly mood and anxiety disorders [3]. In Major Depressive Disorder (MDD), neuroendocrine-immune interactions, tightly regulated by HPA axis under homeostatic conditions [236], cause imbalances in levels of neurotransmitters such as 5-HT and NE, hormones such as cortisol, and also in cytokines such as TNFα and IL-6 (variations of which show the closest association with depression), and also IL-1β, IL-4, IL-2, IL-8, IL-10 and IFN-γ, that contribute to the behavioral and immune disturbances observed in these patients [237-245]. MDD seems to be associated to HPA axis hyperactivity [241], as confirmed by hypercortisolism, one of the most consistent clinical features of MDD [246, 247], and desensitization of glucocorticoid receptor, the most important regulator of HPA axis negative feedback system [248] (variations in cortisol levels are directly related to the activation index of HPA axis [249]). Previous studies reported that MDD patients without pharmacological treatment present polarization towards a Th2 type circulatory cytokine profile [250-252] and other alterations in the immune response such as changes in antibody levels and complement deficiencies [253], numeric alterations in several sub-sets of lymphocytes [254] and increase in the development of some infectious and tumorous conditions [255] in non-treated patients. This could be explained by the fact that hypercortisolism and NE systematically mediate a Th2 shift [1, 31, 250]. HPA axis hyperactivity could lead in some cases to deficiency in 5-HT compensatory tone. In fact, in animal models of depression cortisol has been shown to negatively regulate the 5-HT1A receptor in a dose-dependent manner [256], and other research has shown in lymphocytes from MDD patients a decrease in the density of the 5-HT transporter [23] and inhibition of 5-HT1A receptor function [22]. So, chronic excess in cortisol levels, index of HPA axis hyperactivity, downregulating 5-HT1A postsynaptic receptors in amygdala and cortex (which play an inhibitory role on glutamatergic tone), could lead to amygdala glutamatergic hyperactivity (and excitotoxicity) due to decrease in 5-HT activity, altering dendrites and spine number in such neurons network [47, 257]; 5-HT activity deficiency, in turn, might be related to lower levels of Th1 cytokine with Th2 shift and to the onset of anxiety and depressive symptoms [258].

Similar cytokine findings have been evidenced also in the course of anxiety disorders: in obsessive compulsive disorder higher levels of IL-6 and TNF-α were found, while in posttraumatic stress disorder, in addition to IL-6 and TNF-α increasing, also low cortisol and high DHEA levels were found [259-261]; furthermore, a systematic shift in cytokine balance towards a Th2 profile has been suggested in the Gulf War syndrome [262]. The alteration in cytokine patterns does not seem to be much different in anxious and depressed patients, possibly reflecting a substantial psychopathological overlap or the frequent comorbidity of anxiety and depressive symptoms in the same patient, making it difficult to distinguish homogeneous samples structured on the distinction of individual psychopathological dimensions [263]. In addition, there may be other confounding factors that make it difficult to relate specific sets of symptoms with specific patterns of cytokine alterations. For example, the frequent comorbidity of cigarette smoking with mood disorders [264] may contaminate the cytokine pattern related to HPA axis hyperactivity [1], often detected in the course of mood disorders [265], pursuant to a stimulation of Th1-type cytokines due to chronic activation of nicotinic receptors by nicotine in cigarettes [42]. Another confounding factor could be represented by body weight, since adipose tissue secretes TNF-α and IL-6 so that plasma levels of these cytokines are proportional to body mass index and are further elevated in patients with visceral obesity [266].

So, depressive and anxiety syndromes seem to be mainly characterized by chronic hyposerotonergic state, HPA axis hyperactivation and Th2 shift. Nevertheless, other studies suggest an imbalance Th1/Th2 shifted towards Th1 in depression [267, 268]. In this regard, a dimensional approach could lead to further insight of the issue. In melancholic depression, condition in which patients have anxiety, insomnia, anorexia and circadian variation with worsening in the morning, appears to be associated with significantly higher CSF NE and plasma cortisol levels that are increased around the clock, with inappropriately high plasma ACTH and CSF CRH levels considering the degree of their hypercortisolism. These data suggest a central hyper-noradrenergic state in association to hyperfunction of central CRH pathway [269]; furthermore, the chronic hyper-noradrenergic state may drive the increase in systemic IL-6 levels, since NE up-regulates IL-6 production, and, theoretically, to a Th2 shift [1]. On the other hand, atypical depression, condition in which patients have hypersomnia, hyperphagia and fatigue, appears to be associated with a central hypo-noradrenergic state in association to hypofunction of central CRH pathway [270, 271] and so, hypothetically, to a Th1 shift. It is noteworthy that atypical depression, ideally characterized by Th1 shift, seems to belong to bipolar depression [272]. On the other hand, during mania, IL-2, IL-4, IL-6 and IL-10 were found to be increased while IFN-γ decreased, ideally reflecting a Th2 shift [273-275]. Finally, both childhood and adult schizophrenia have been shown to be accompanied by elevated expression of TNF-α, IL-1 and IL-6 [276, 277].

In summary, unipolar depressive disorders and anxiety disorders might be intended as pathologies of NEI stress-adaptation system, mainly characterized by functional alterations and neuronal remodeling of anterior cingulate cortex, amygdala, hippocampus and HPA axis manifested in chronic hypernoradrenergic/hyposerotonergic state, HPA axis hyperactivation and Th2 shift or, in some case, Th1 switch [245, 250, 251, 257, 278]. On the other hand, bipolar depression might be included in primitive affective disorders of cortico-limbic systems (cingulate cortex, striatum, putamen, thalamus, prefrontal cortex and amygdala) [279-281], which in turn could secondarily involve the NEI stress system, since bipolar and anxiety disorders have both been associated with abnormality in the amygdala, one of the main nuclei involved in the regulation of HPA axis [282, 283]. It is possible that the dimensional approach in psychiatric diagnoses, together with attention to various confounding factors, could create homogeneous groups of patients that match the underlying neuroendocrine-immune alterations, to which the models of imbalance and dynamic switching of Th1/Th2 system, already used in other human diseases, could be applied.

ANTIDEPRESSANTS AND TH1/TH2 BALANCE

Antidepressants, Neurotransmission and Cytokines (Table 2)

Table 2.

Antidepressants, Neurotransmission and Cytokines

Antidepressants Pharmacodynamics Neuromodulation Cytokines Th1/Th2 Balance Hypothetic Shifts References
SSRIs: Paroxetine, Sertraline, Fluoxetine, Escitalopram SERT inhibition (+ M1 antagonism: paroxetine; 5HT2C antagonism: fluoxetine; DAT inhibition: sertraline) Desensitization of somatodendritic 5-HT1A autoreceptor (± 5-HT2(A/C) receptors) → 5-HT firing enhanced + NE firing reduced + cortisol decreased ↓IL-4, IL-10, IL-13, IL-6 / ↑IL-1β, IFN-γ, ↑↓IL-2 (Conflicting data) Th1 shift Hernandez et al., 2008 [252] (others)
SNRIs: Venlafaxine SERT inhibition ± NET inhibition (only at high dose) Desensitization of somatodendritic 5-HT1A autoreceptors (± 5-HT2(A/C) receptors) → 5-HT firing enhanced ± desensitization of α2 terminal receptors (± hypersensitization of α1 and desensitization of β1 receptor) → NE firing enhanced (i.e. in PFC and hippocampus) and DA release enhanced in PFC (only at high dose) ↓IL-12, TNF-α, IFN-γ / ↑IL-10, TGF-β1 / ↓IL-6 (low dose) ↑IL-6 (high dose) Th1 shift (low dose) Th2 shift (high dose) Kubera et al., 2001 [303] (others) Vollmar et al., 2008 [305] Kubera et al., 2004 [309]
SNRIs: Duloxetine SERT inhibition + NET inhibition Desensitization of somatodendritic 5-HT1A autoreceptors (± 5-HT2(A/C) receptors) → 5-HT firing enhanced + desensitization of α2 terminal receptors (± hypersensitization of α1 and desensitization of β1 receptor) → NE firing enhanced (i.e. in PFC and hippocampus) and DA release enhanced in PFC ↑IL-6 Th2 shift Fornaro et al., 2011 [318]
NRIs: Reboxetine NET inhibition Desensitization of α2 terminal auto- and heteroreceptor (± hypersensitization of α1 and desensitization of β1 receptor) → NE firing enhanced (i.e. in PFC and hippocampus), DA release enhanced in PFC and 5-HT firing enhanced + cortisol increased ↑IL-10 / ↓IL-1β, TNF-α, IL-6 Th2 shift McNamee et al., 2010 [319] O’Sullivan et al., 2009 [322] (others)
NDRIs: Bupropion NET inhibition + DAT inhibition (+ NACh receptors antagonism) Desensitization of α2 terminal auto- and heteroreceptor (± hypersensitization of α1 and desensitization of β1 receptor) → NE firing enhanced (i.e. in PFC and hippocampus) and DA release enhanced in PFC and in nucleus accumbens ↑IL-10 / ↓TNF-α, IFN-γ, IL-1β Th2 shift Brustolim et al., 2006 [336]
Atypical: Mirtazapine α2, 5HT2, 5HT3, H1 receptors antagonism Desensitization of α2 terminal heteroreceptor → 5-HT firing enhanced (75%) only at 5-HT1-mediated transmission + NE (30%) and DA release enhanced in PFC + melatonin increased and cortisol decreased ↑TNF-α ↓IL-6 / ↓IFN-γ ↑IL-4 Th1 shift Kraus et al., 2002 [342] Kubera et al., 2006 [343]

The table summarizes mechanisms of action, neuromodulatory effects, cytokines modulation and hypothetical influence on Th1/Th2 balance for each class of antidepressants. (SSRIs: Selective Serotonin Reuptake Inhibitors; SNRIs: Serotonin Norepinephrine Reuptake Inhibitors; NRIs: Norepinephrine Reuptake Inhibitors; NDRIs: Norepinephrine Dopamine Reuptake Inhibitors; 5HT: Serotonin; NE: Norepinephrine; DA: Dopamine; H: Histamine; ACh: Acetylcholine; SERT: Serotonin Transporter; DAT: Dopamine Transporter; NET; Norepinephrine Transporter; IL: Interleukin; IFN: Interferon; TNF: Tumor Necrosis Factor; TGF; Tumor Growth Factor; PFC: Prefrontal Cortex)

Some antidepressants would occur affecting NEI system, acting on neurotransmitter balance (especially on the 5-HT/NE balance) and levels of expression and sensitivity of individual receptor subtypes, which in turn affect the cytokine production and the relative Th1/Th2 balance.

Selective Serotonin Reuptake Inhibitors (SSRIs)

In rigorous and long term clinical study, SSRIs seem to increase Th1 cytokines, such as IL-1β, IL-2 and IFN-γ, and decrease Th2 cytokines, such as IL-4, IL-10 and IL-13, and cortisol levels after 52 weeks treatment in depressed patients [252], although other in vitro and short term ex-vivo studies reported conflicting results, showing decrease in IL-1β, IL-6, IL-10, IFN-γ and TNF-α after SSRI treatment in a dose dependent manner [284-288]. In that study, administration of SSRI in MDD patients, confirming baseline high levels of cortisol, IL-4, IL-13 and IL-10 (Th2) compared with healthy volunteers, induced clinical remission at week 20 of treatment, concomitantly with an increase in IL-2 and IL-1β levels (Th1) without changes in cortisol level. At week 52 of treatment, SSRI administration induced an increase in IL-1β and IFN-γ levels (Th1), together with a reduction in IL-4, IL-13 and IL-10 levels (Th2) and in cortisol levels (a 30% diminution compared to baseline) [252]. Variations in these parameters could be caused by SSRI effects both on 5-HT and glucocorticoid receptors, as a result of chronic intake of these drugs. SSRIs exert a relatively selective blockade of 5-HT transporter [289], progressively increasing 5-HT levels, also in the circulation [290, 291], and influencing the immune response in a dose-dependent manner [252]. As a consequence, long-term SSRI treatment desensitizes the inhibitory somatodendritic 5-HT1A autoreceptors in the dorsal and medial raphe, and 5-HT neurotransmission is enhanced [292-294]. Furthermore, a desensitization of 5-HT2A and 5-HT2C receptors occurs as a consequence of prolonged exposure to elevate levels of 5-HT [295, 296]. Finally, since 5-HT neurons exert a tonic inhibitory effect on locus coeruleus neurons, it appears that enhancing 5-HT neurotransmission by sustained SSRI administration leads to a reduction in the firing rate of noradrenergic neurons [35]. Thus, drug-mediated enhancement of 5-HT activity exerts immunostimulatory effects on Th1 cytokines [32], possibly acting on 5-HT1A receptors, and concomitant immunoinhibitory effects on Th2 cytokines. Furthermore, it has been proposed that long term SSRI treatment in depressed patients causes a decrease in circulating cortisol levels by reestablishing the down-regulated glucocorticoid receptor sensitivity [27], thus restoring negative feedback by cortisol on the HPA axis [297-299]. Finally it was shown that paroxetine attenuated cyclooxygenase (COX)-2 expression in human T cells [300], considering that COX inhibition due to NSAIDs results in augmentation of the Th1 response by limiting prostanoid synthesis [301].

Serotonin Norepinephrine Reuptake Inhibitors (SNRIs)

Venlafaxine, a SNRI, appears to have a more complex action on cytokine levels [302]. In several clinical and preclinical studies it was observed that venlafaxine reduces blood levels of IL-12, TNF-α, IFN-γ and increases those of IL-10 and TGF-β1 [303-306]. However, for discussion purpose, it is important to emphasize the dose-dependent effects of venlafaxine on cytokines such as IL-6, a molecule involved in the acute phase response and in the control of Th1/Th2 differentiation towards a Th2 polarization [307]: at low dose venlafaxine appears to reduce serum levels of IL-6 [305, 308], while at higher dose it seems to rather increase levels of IL-6 [309]. These data could be related to the peculiar pharmacodynamics of venlafaxine: the effects on neurotransmission and receptors expression do not seem to differ much from those of SSRIs, at least at low dose [310-312]; nevertheless at higher dose venlafaxine acts as a real SNRI: while at low dose the molecule mainly blocks the reuptake of 5-HT, at high dose the molecule blocks the reuptake of 5-HT and NE to the same extent [313]. Duloxetine, another SNRI, in contrast to venlafaxine has a greater affinity for the NE transporter, blocking to the same extent the reuptake of NE and 5-HT at standard dose [314]. Consequently, chronic administration of this molecule leads, in addition to the desensitization of dorsal raphe nucleus somatodendritic 5-HT1A autoreceptors with increased serotonergic firing [315, 316], also an increase in NE release in frontal cortex and hippocampus due to the desensitization of α2-adrenergic terminal autoreceptor [315, 317]. Our preliminary evidence has shown an increase in the serum levels of IL-6 associated with 6 weeks duloxetine treatment in depressed patients; if the increased levels of IL-6 by duloxetine were confirmed on larger samples, it could be assumed that at the basis of changes in IL-6 serum levels, considering them as a marker of Th1/Th2 balance, operates a 5-HT/NE balance [318].

Norepinephrine Reuptake Inhibitors (NRIs)

To date there are no clinical studies on the interaction between the NRIs reboxetine and atomoxetine and cytokine production. In preclinical studies, treatment with reboxetine seems to enhance IL-10 expression [319], as well as IL1β and its negative regulators, IL-1 receptor antagonist and IL-1 type II receptor, in rat cortex [320]. In a study it was observed that pretreatment with reboxetine inhibited the increase of IL-6 by exposure to IFN-γ in murine cells [321]. In another work, treatment with atomoxetine reduced rat cortical gene expression of IL-1β and TNF-α [322]. Overall, the limited available data could suggest that this class of molecules may shift the Th1/Th2 balance towards Th2 response. It is noteworthy that despite the NRIs selectively block NE reuptake, chronic administration of these molecules causes profound changes in neurotransmission of several systems. Inhibition of brain NE reuptake by acute reboxetine administration results in an inhibition of central noradrenergic activity via local increase in NE concentration at inhibitory α2 autoreceptors [323]; however, the sustained elevation of NE synaptic availability due to long term treatment results in desensitization and downregulation of α2 terminal receptor at least at level of prefrontal cortex and dorsal hippocampus, in turn strengthening NE transmission in these areas [324] and attenuating the inhibitory restraint on sympathetic outflow, obtaining an opposite result on noradrenergic tone in relation to time of action and receptor sensitivity [323]. In addition repeated administration of NRIs results in hypersensitization of α1 adrenergic receptor [325, 326] and desensitization of β1 adrenergic receptor [327, 328]. NRIs induce an increase in burst firing of DA cell in ventral tegmental area [329] and selectively increase DA availability in medial prefrontal cortex [329, 330], since in such area NE transporter even reuptakes DA. The desensitization of α2 adrenergic heteroreceptors would increase tonic activation of postsynaptic 5-HT1A receptors [331]. Finally, NRIs seem to have a complex action on acetylcholine (ACh), inhibiting the nicotinic receptors function [332] and also increasing cortical and hippocampal ACh [333], and appear to stimulate the secretion of cortisol (as well as adrenocorticotropin, growth hormone and prolactin) [334, 335].

Norepinephrine Dopamine Reuptake Inhibitors (NDRIs)

Some evidences showed that bupropion, a NDRI, reduces blood levels of TNF-α, IFN-γ, IL-1β and increases those of IL-10 (effect largely abrogated by β-adrenergic or D1 receptor antagonists and not by a D2 antagonist) [336] and IL-8 [337], shifting the Th1/Th2 balance towards Th2 polarization. Bupropion with its metabolites inhibits reuptake for human transporters for both DA and NE, with slightly greater functional potency at DA transporter than at NE transporter, increasing DA and NE concentrations in prefrontal cortex and DA concentrations in nucleus accumbens [338]. Inhibition of 5-HT reuptake via the 5-HT transporter was negligible even at the highest concentration tested [338]. Bupropion and its metabolites do not have appreciable affinity for postsynaptic receptors including histamine, α- or β-adrenergic, 5-HT or DA receptors [338, 339]. Most recent evidence showed that this molecule behaves as a noncompetitive antagonist of several nicotinic ACh receptors [340, 341].

Other Classes of Antidepressants

Other classes of antidepressants modulate neuro-transmission in more complex ways than selective reuptake inhibitors and their relatively homogeneous effects on specific neurotransmitters, making it even more difficult to examine the relationship between neurotransmitter modulation and Th1/Th2 balance.

Among these, mirtazapine seems to alter plasma concentrations of several cytokines, increasing levels of TNF-α [342] and reducing those of IL-6 [343] on one hand, inhibiting the production of IFN-γ and increasing the production of IL-4 [343] on the other. Mirtazapine blocks α2 adrenoceptors on noradrenergic neurons (autoreceptors) and on serotonergic neurons (heteroreceptors), 5-HT2 and 5-HT3 receptors and H1 receptor [344]. Long term administration of mirtazapine desensitizes terminal α2 adrenergic hetero-receptor, but not terminal α2 adrenergic autoreceptor [345], leading to an increase in the firing rate of 5-HT neurons (75%) and in the firing rate of NE neurons (30%) [346]. Since mirtazapine blocks 5-HT2 and 5-HT3 receptors, only 5-HT1-mediated transmission is enhanced [347] (the 5HT1A receptor stimulation seems to be directly related to the increase of TNF [348]) and, on the other hand, there is a reinforcement of DA and NE tone in prefrontal cortex [349]. It should also be noted that mirtazapine increases melatonin and decreases cortisol levels [350], effects that overall could account for a shift in balance towards the Th1 response.

Trazodone, a molecule that blocks 5HT2(A/C), H1 and α1 receptors and also 5-HT transporter, but only at high dose [351], seems to reduce levels of TNF-α, IL-1β [352] and IFN-γ as well [353].

Agomelatine, a novel melatonergic MT1 and MT2 receptors agonist and a 5-HT2C receptor antagonist [354], seems to stimulate IL-2 production, like melatonin, via MT1 receptor agonism in human lymphocytes [355]; chronic administration modulates chronobiological rhythms, including temporal organization of cortisol secretion [356] (MT and 5-HT2C receptors exhibit a circadian rhythms of expression), and desensitizes 5-HT2C receptor, which results in NE and DA enhancing only in frontal cortex [354], without affecting sensitization of 5-HT1A receptors [357] or that of suprachiasmatic nucleus neuronal responses [358].

Some studies on tricyclic drugs show that this class of molecules inhibits the secretion of IL-1β, IL-2, TNF-α, IFN-γ [284, 359] and stimulates the production of IL-10 [360]. Also, one clinical study reports an increase of IL-6 following tricyclic administration [309], while other preclinical studies do report a reduction [359, 361, 362] and still others show no change [363, 364]. Though overall tricyclics mainly block NE and, to a lesser extent, 5HT reuptake, the effects of individual molecules within this class of antidepressants are heterogeneous, enhancing noradrenergic and serotonergic transmission within a different spectrum, as well as each molecule blocks with different affinities M1, α1 and H1 receptors [365], also potentially involved in immuno-modulation. This makes it much more difficult for this class of drugs to discriminate the effects of individual neurotransmitter systems on cytokine production. However, desipramine, a potent NE antidepressant, inhibits TNF-α production and increases the release of IL-10, shifting the balance in favor of Th2 response [366].

Finally, among MonoAmine Oxidase Inhibitors (MAOIs), phenelzine (an irreversible inhibitor of MAO-A and MAO-B) decreases TNF-α production [367], moclobemide (a reversible inhibitor of MAO-A) reduces the production of TNF-α and IL-8 and increases the production of IL-10 [368], and selegiline (a reversible inhibitor of MAO-B) reduces the production of TNF-α and stimulates the biosynthesis of IL-6 and also of IL-1β [369]. MAO-A inhibition is more closely associated with an increase in noradrenergic than in serotonergic system [370], while MAO-B inhibition seems to mainly enhance dopaminergic system [371]; so, in general, MAOIs could shift the cytokine balance towards a Th2 response in relation to the enhancing of catecholamine release.

ANTIDEPRESSANTS AS IMMUNOMODULATORS

In order to conclude, it can be useful to make a nod to some of the currently available data on literature regarding the experimental use of antidepressants as immuno-modulators or their possible immunomodulatory effects in the course of internistic pathologies.

Antidepressants use in Inflammatory and Immune-mediated Diseases

Limited data have shown a reduction of Crohn’s disease activity during treatment with bupropion [372, 373], possibly due to TNF decrease [348], and phenelzine [374], possibly related to β adrenoreceptor agonism due to NE increase [348]. Mirtazapine is not recommended, because of the related TNF increase [348], amitriptyline does not appear to be effective [375], whereas data on paroxetine are controversial [376] (one study reported an activation of Crohn’s disease after paroxetine treatment [377]).

In a study on non-depressed patients with psoriasis, bupropion proved to be effective in reducing skin lesions, as it showed an effectiveness, albeit less incisive, in patients with atopic dermatitis [378]; also MAOIs have shown an efficacy in these conditions [379]. Although SSRIs are generally considered safe in the treatment of psychiatric disorders encountered in dermatological diseases [380], some cases of induction or worsening of psoriasis have been reported during treatment with paroxetine and fluoxetine [381, 382], in which a specific influence of immuno-modulation by 5-HT is assumed [382].

The effectiveness of bupropion and nortriptyline was reported in patients with COPD, a disease associated with cigarette smoking and often depression [383, 384]: nortriptyline treatment was also accompanied by marked improvements in certain respiratory symptoms [384]. Asthma is rather frequently associated with anxiety disorders such as panic disorder [385], for which SSRIs are the treatment of choice [385, 386]; it was noted that some respiratory parameters deteriorated in patients with panic attacks who suspended SSRIs [387], whereas a case of developing of asthma-like reaction during venlafaxine treatment was reported [388]. In this regard, it is noteworthy that glucocorticoid and β2 receptor agonist therapy reduces IL-12 production by antigen presenting cells and greatly suppresses Th2 cytokine synthesis in activated, but not resting T cells, and abolishes eosinophilia; if, however, resting (cytokine-uncommitted) T cells are subsequently activated by antigen presenting cells preexposed to glucocorticoids and β2 receptor agonists, enhanced IL-4 production, but limited IFN-γ synthesis, could be induced, enhancing Th2 response [389]. Thus, while in short term the effects of glucocorticoids and β2 receptors agonists may be beneficial, their long term effects might be to sustain the increased vulnerability of the patient to the allergic condition [1].

In murine T-cell-mediated demyelinating disease model of multiple sclerosis, venlafaxine significantly ameliorated clinical symptoms of the disease in association with a reduction of IL-12, TNF-α and IFN-γ [306]. Treatment with lofepramine (a potent and selective NE uptake blocker [390]) and L-phenylalanine was found to be effective in relieving symptoms of multiple sclerosis, such as pain and fatigue, and in reducing lesion number visible on MRI, hypothetically in relation to noradrenergic activation [391, 392].

MAOIs seem to be effective in rheumatoid arthritis [393, 394], however, SSRIs also significantly inhibited disease progression in an animal model (fluoxetine showing the greatest degree of efficacy at the clinical and histologic levels) [395].

Use of phenelzine and other MAOIs has also been proposed in Behcet’s disease [396] and IgA nephropathy [397].

In animal models of Alzheimer disease, imipramine significantly prevented memory deficit, inhibited the TNF-α increase in frontal cortex and decreased the elevated levels of β-amyloid both in frontal cortex and in hippocampus [398]; prophylactic paroxetine treatment decreased Alzheimer disease like pathology in the animal model [399] and fluoxetine reduced cognitive decline in patients with mild cognitive impairment [400]. Improvement in memory and cognition might be due to hippocampal neurogenesis induced by antidepressants, but the anti-inflammatory properties of antidepressants may be still involved since the inflammation associated with activated microglia has been demonstrated to suppress hippocampal neurogenesis in adult rat [401]. In a mouse model of Parkinson’s disease, paroxetine treatment prevented degeneration of nigrostriatal DA neurons, in association to increased striatal DA levels and reduced expression of IL-1β and TNF-α by activated microglia [402].

Antidepressants use in Metabolic and Cardiovascular Diseases

Emerged evidences suggest that MAOIs such as selegiline and phenelzine do have some anti-atherogenic effects [403, 404]. Also, reboxetine treatment showed beneficial effects on several metabolic parameters [405]. On the other hand, SSRIs instead seem to induce some clinical and biochemical manifestations of metabolic syndrome, such as insulin resistance [406], hypercholesterolemia and visceral obesity [407]. In this respect it should be noted that the increase in plasma 5-HT related to excessive dietary tryptophan may be atherogenic [408]. However, data on lipid dysregulation in depression indicate that, generically, the treatment of this clinical condition would result in an improved metabolic profile [409]. Furthermore, the use of agomelatine in the metabolic syndrome, by virtue of melatonin properties on preventing a number of sequelae in animal models of metabolic syndrome has been proposed [410], considering that melatonergic agonism seems to reorganize the cortisol secretion [356].

Several data show generic efficacy of antidepressants use, including SSRIs, SNRIs, bupropion, mirtazapine and tricyclics, in patients with coronary artery disease and after myocardial infarction, although certain classes of antidepressants are associated with a higher rate of adverse events [411-413]. SSRIs appear to reduce cardiovascular mortality in acute coronary syndromes, especially when associated with depression [414, 415], whereas tricyclics may be related with cardiovascular negative effects such as arrhythmic activity and increased heart rate [415], and bupropion has been associated with myocardial infarction in isolated cases [416-418]. Beyond these general considerations, it is noteworthy that a recent study on patients with heart failure showed lower levels of TNF-α and CPR in patients treated with tricyclics and SNRIs compared to those treated with SSRIs or those without depression, suggesting that the type of antidepressant used may have a significant effect on the underlying inflammatory process of heart failure [419].

Several antidepressants, particularly those with predominantly NE component such as reboxetine, atomoxetine, nortriptyline, venlafaxine, selegiline, amphetamines, and also some SSRIs, have shown benefits in post-stroke recovery (for example, motor function and depression) probably due to the role of NE in enhancing cortical plasticity [420-427], even if it might be also related to the modulation of the underlying inflammatory process.

Antidepressants use in Chronic Infections

In HIV infection models, SSRIs significantly decreased HIV viral replication, suggesting an enhancement of natural killer/CD8 HIV suppression [428, 429]; the use of SSRIs as an adjunctive therapy has also been attempted in the treatment of HIV-infected individuals [430]. In addition, mirtazapine has shown in isolated cases some benefit in the treatment or prophylaxis in HIV patients with progressive multifocal leukoencephalopathy [431, 432]. Also hypericin, another main 5-HT enhancing drug [433], possesses high toxicity towards HIV, as well as towards tumors [434]. On the other hand, MAOIs, such as selegiline, and addictive substances, that increase DA availability, seem to enhance viral replication and infection-related neuronal ultrastructural alterations (spongiform polio-encephalopathy) possibly favoring HIV dementia [435, 436]. SSRIs, particularly paroxetine, are recommended in the treatment of depression in course of HCV infection [437, 438]. In active Mycobacterial disease, it has been proposed that mirtazapine might be the first best preference while bupropion should be avoided [348].

Antidepressants use in Cancer

An association has been found between the use of SSRIs and risk reduction, in some studies it is more evident than in others, of colorectal cancer [439-441], lung cancer [442] and lymphoma/leukemia [443-445]; although the use of SSRIs may stimulate the secretion of prolactin, a potential breast cancer promoter, clinical studies have refuted an increased risk of breast cancer with SSRIs’ use [446-448]. Furthermore, co-administration of ramelteon (MT1 and MT2 agonist) and fluoxetine to increase IL-2 levels in conditions such as metastatic cancer and HIV infection has been proposed [449]. It has also been observed that hypericin seems to inhibit ovarian cancer cells [450]. Mirtazapine, in addition to representing a promising option for the treatment of cancer-related cachexia and anorexia [451, 452], has been proposed as a potential anticancer agent, both as an adjunctive treatment and as a molecule with direct effects itself, in glioblastoma [453], in osteosarcoma [454] and, more generally, during cancer immunotherapy; the drug-induced TNF increase could have a beneficial effect through the enhancement of antigen driven lymphocyte expansion [348]. Also, melatonin and agomelatine have been shown to inhibit the growth of melanoma cells [455]. On the other hand, an association between tricyclics use and increased risk of lung cancer in clinical studies [442], colorectal cancer in preclinical studies (but not in clinical studies) [439, 441] and melanoma has been observed [456]. In a preclinical study on melanoma, pro-metastatic effects and simultaneously inhibitory actions have been demonstrated on the growth of primary tumor (especially in young animals) of desipramine, a potent NE tricyclics, and to a lesser extent also of fluoxetine, which among SSRIs also enhances NE/DA transmission by 5-HT2C blocking [457]. Imipramine instead, a 5-HT tricyclic, appears to inhibit tumor growth in an animal model of stress-related tumoral process [222]. Finally, MAOIs have demonstrated an anticancer effect in hormone-dependent tumors: in animal models selegiline reduced by systemic way the growth of mammary tumors [458, 459], while clorgiline reduced in vitro the growth of advanced prostate cancer (it is noteworthy that a high expression of MAO-A has been identified in high-grade primary prostate cancer) [460].

In summary, literature shows some evidence of immunomodulatory effects of antidepressants; however, specific molecules or classes seem to induce positive immunomodulatory effects in some groups of diseases and possibly negative in others. In diseases such as Crohn's, psoriasis, rheumatoid arthritis and atherosclerosis, in which the pathophysiological process appears to shift towards Th1 response, some drugs such as MAOIs, for example, and bupropion in a few cases, have been used with some benefit on immune imbalance, while other drugs such as SSRIs do not seem to be effective. On the other hand, in diseases such as HIV and some cancers, in which the progression of infection or metastatization seems to correlate with a Th2 switch, drugs such as SSRIs and mirtazapine might favorably affect disease control, while the use of other drugs, such as MAOIs in HIV and possibly tricyclics in neoplastic disease, appears to be associated in some cases with disease progression.

DISCUSSION

Several human chronic diseases seem to be characterized by a stereotyped multiphasic process resulting from complex interactions between predisposing genetic traits, infections, episodic and chronic activation of the neuroendocrine stress system and oscillations in the Th1-Th2 balance system. Changes in cytokine expression often show a mixed pattern. Nevertheless, in the early stages, Th2 activation is often detected in several immune-mediated diseases, correlated with humoral response and allergic reactions or production of autoantibodies, which in some cases appear to be clinically silent, in other determine clinically significant pathophysiological phenomena. In the late stages, the immune response seems to show in several diseases a switch from Th2 to Th1 pattern, correlated with cell-mediated organ inflammation. Also, chronic activation of the Th2-related stress system seems to lead to a Th1 switch with elevation of both Th1 and Th2 inflammatory cytokines that results in chronic systemic inflammation associated with a cluster of metabolic disturbances and cardiovascular diseases. So, episodic activation of the neuroendocrine stress system due to several causes, including stressful events and intercurrent infections together with genetic polymorphisms and epigenetic factors, affects the Th2 response. Conversely, Th2-to-Th1 switch could be related to chronic activation of stress system associated with genetic-epigenetic abnormalities of systemic feedback mechanisms and/or local inflammatory factors, since stress system tolerates phasic rather than chronic activations [46, 461]: a relative hypoactive stress system with defective central and peripheral components (hypothalamic response, adrenal secretion, etc.) may facilitate or sustain the Th1 shift [31, 82-85, 462]. Finally, intercurrent activations of the stress system (due to environmental or internal stressors) on the basis of chronic Th1 hyperactivation could lead to transitory Th1-to-Th2 switches with relative displacements of the immune reserve, allowing a progression of underlying pathologies such as latent viral infections or tumor metastatization. Thus, while acute environmental stress (at large) is associated with Th2 activation, chronic stress, depending on interactions of genetic, epigenetic, infective, tumoral and environmental factors, could lead to a switch towards chronic Th1 activation (at least partially autonomized), possibly in relation to a relative functional impairment of the NEI stress system, as far as superimposed acute stresses can induce new transitory Th2 switches. In addition to systemic oscillations in the Th1-Th2 balance, compensatory systemic adaptations of the cytokine balance related to local inflammatory alterations in organism districts could result in counter-polar alterations of the Th1-Th2 balance in other districts [1]. The neuroendocrine system appears to affect the balance and dynamic switching of cytokine system; however the flow of information appears to be bidirectional, making it a reason of some observations on the relationship between psychopathological alterations and immunological changes. If on one hand bidirectional relationships could be highlighted between oscillations in the Th1-Th2 balance of immune system and oscillations depression-mania in affectivity associated with neurohormonal changes, on the other hand, it is possible that chronic activation of the stress system with relative Th2-to-Th1 switch might associate in the psychopathology with the transition from anxious dimensions to unipolar depressive dimensions. Therefore, the integrated NEI system could correlate, in a bidirectional way, the oscillations in the immune system with affective and behavioral alterations as epiphenomena of systemic pathologies, making reason of eventual integrated activities of drugs such as antidepressants [463].

By correlating the data on the relationship between neurotransmitters and cytokines with those concerning the relationship between antidepressants and neurotransmission and cytokine balance modulation, it is possible to suppose an interaction between the neurotransmitter balance and the Th1/Th2 balance, as basis of the adaptive response of the organism to the environment in human physiology and pathophysiology, whereas the neuroendocrine and immune systems are considered the two major adaptive systems of the body [464]. In the mechanism of action of several antidepressants a 5-HT/NE neuromodulatory balance seems to play an important role, which in turn could influence the Th1/Th2 cytokine balance toward Th1 or Th2 pattern responses. Specifically, it could be hypothesized that as the specific antidepressant increases NE tone, it increases Th2 cytokines production, as it reduces NE tone (or increases 5-HT tone, in turn possibly reducing NE tone), it reduces Th2 cytokines production, shifting the Th1/Th2 balance relatively in favor of Th2 or Th1 response, respectively. Although some data seem at large to correlate neurotransmitter balance with cytokine balance, and in particular the noradrenergic system with the Th2 response [1], it is likely that tentative concept is too simplistic. It is possible that antidepressants do not affect the neurotransmitter and Th1/Th2 balance modulation in an absolute way, but rather in a relative one, ideally compensating a previous imbalance. Antidepressants appear to cause a modulation of neurotransmission and receptor sensitivity with knock-on effect, possibly leading to desensitization of hypersensitive receptors and to hypersensitization of desensitized receptors, thus ideally restoring receptor sensitivity to physiological levels (provided the integrity of neurotransmitter and feedback systems). For example, it has been shown that SSRIs' chronic administration causes a desensitization of 5-HT2A and 5-HT2C receptors, that are found to be hypersensitive in patients with depressive or anxiety disorders [295, 465]. According to this, SSRIs could differently affect cytokine production in different patients who, although classified as depressed by the scales commonly used, could present different symptom clusters related to the alteration of sensitivity of different receptor subtypes. Desensitizing 5-HT1A autoreceptor, SSRIs would mainly increase 5-HT tone and possibly Th1 cytokines; the same SSRIs, however, would also desensitize 5-HT2 receptors, increasing the NE/DA tone and possibly the Th2 cytokines only in those patients presenting with hypersensitivity of these receptors subtypes; thus, different cytokine patterns might depend on neurobiological background of different patients. Likewise, antidepressants would not affect immunomodulation in an absolute way, but would rather compensate in a relative one previous imbalances in Th1/Th2 system. Some studies show that antidepressants can induce differential cytokine changes between responders and nonresponders. In a study on depressed patients, duloxetine increased IL-6 plasma levels only in responders, whereas in nonresponders IL-6 levels remained unchanged; pre-treatment IL-6 levels were lower in those patients who responded to treatment than in those who did not; post-treatment IL-6 levels of responders were normalized by treatment until they reached post-treatment IL-6 levels of nonresponders [318]. Evidences provided by another study gave similar IL-6 results among pre-treatment and post-treatment responders vs. nonresponders, suggesting that IL-6 levels might dichotomize the patients into subsequent responders and nonresponders already at admission [240]. It could be hypothesized that in such cases drugs with high noradrenergic component such as duloxetine lead to clinical response only in depressed patients with a relative hypo-noradrenergic state, as reflected in low basal IL-6 levels (marker of relative deficit of Th2 response); likewise, the same drugs would enhance Th2 response only in those patients with cytokine balance relatively shifted towards Th1 because of Th2 deficit, but not in an absolute way in patients with normal Th2 response or in subjects with compensated Th1/Th2 balance [318].

Several factors therefore seem to affect immuno-modulation, including the pharmacologic one. Among these, the timing in the use of immunomodulators, possibly including antidepressants, could play an important role, according to temporal phases and regional distributions of cytokine balance in the Th1-Th2 dynamic switching and its neurohormonal control.

The considerations presented in this work are intended as preliminary, since most of the reported clinical data are based on small samples studies or on individual observations, while for most of the molecules, studies on uniform and large samples and for adequate times are currently the exception. Moreover, in some cases data are consistent, in other cases less concordant; in this regard, it is to be noted the importance of discussion of clinical studies compared to preclinical (e.g. variations in cytokine levels during 52 week study on depressed patients treated with SSRIs [252] compared to those detected in course of some preclinical studies on 5-HT and cytokines interactions [33]). This also happens because in vitro effects of molecules on cytokine production can have opposite results depending on the dose and according to the time of action [32]. Considering that antidepressant treatment needs at least 10-14 days for any clinical effectiveness to appear, the treatment in vitro cells with antidepressants for 16-24 h appears to reflect only acute effects of the drugs; furthermore, the concentrations used in some in vitro studies seem to be rather higher than clinically relevant concentrations [162]. Several other factors could play an important role, such as systemic mechanisms of mutual influence between neurotransmitter and cytokine systems, receptor sensitivity modulation and feedback effects, that may vary in healthy subjects or in patients affected by different pathologies. For example, opposite results on noradrenergic tone in relation to time of action and receptor sensitivity during NRIs administration have been reported [323], as well as opposite results on Th2 cytokines synthesis have been observed in activated and resting T cells exposed to β2 agonists [1].

Despite broad limits make it difficult to reach conclusions, overall considerations on neuro-immuno-modulation could represent an additional aid in the study of pathophysiology of psychiatric disorders and in the choice of specific antidepressant (and virtually other drugs) in specific clusters of symptoms especially in comorbidity with several internal pathologies. Finally, antidepressants represent promising immunomodulator agents in psychiatric and non-psychiatric diseases, such as cancer and others. Overall, the reported considerations are tentative and require experimental confirmation or refutation by future studies.

ACKNOWLEDGEMENTS

Authors have no acknowledgement to state.

CONFLICT OF INTERESTS

Authors have no conflict of interest to declare.

ROLE OF THE FUNDING SOURCE

The authors did not receive any financial support for this paper.

REFERENCES

  • 1.Elenkov IJ. Neurohormonal-cytokine interactions: implications for inflammation, common human diseases and well-being. Neurochem. Int. 2008;52(1-2):40–51. doi: 10.1016/j.neuint.2007.06.037. [DOI] [PubMed] [Google Scholar]
  • 2.Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern. Med. Rev. 2003;8(3):223–246. [PubMed] [Google Scholar]
  • 3.Hickie I, Lloyd A. Are cytokines associated with neuropsychiatric syndromes in humans? Int. J. Immunopharmacol. 1995;17(8):677–683. doi: 10.1016/0192-0561(95)00054-6. [DOI] [PubMed] [Google Scholar]
  • 4.Janssen DG, Caniato RN, Verster JC, Baune BT. A psychoneuroimmunological review on cytokines involved in antidepressant treatment response. Hum. Psychopharmacol. 2010;25(3):201–215. doi: 10.1002/hup.1103. [DOI] [PubMed] [Google Scholar]
  • 5.Halaris A. Comorbidity between depression and cardiovascular disease. Int. Angiol. 2009;28(2):92–99. [PubMed] [Google Scholar]
  • 6.Rabi-Zikic T, Zarkov M, Nedic A, Slankamenac P, Zivanovic Z, Dobrenov D. Depression as the cause and consequence of cerebrovascular diseases. Med. Pregl. 2007;60(5-6):255–260. doi: 10.2298/mpns0706255r. [DOI] [PubMed] [Google Scholar]
  • 7.Maunder RG, Levenstein S. The role of stress in the development and clinical course of inflammatory bowel disease: epidemiological evidence. Curr. Mol. Med. 2008;8(4):247–252. doi: 10.2174/156652408784533832. [DOI] [PubMed] [Google Scholar]
  • 8.Kurina LM, Goldacre MJ, Yeates D, Gill LE. Depression and anxiety in people with inflammatory bowel disease. J. Epidemiol. Commun. Health. 2001;55(10):716–720. doi: 10.1136/jech.55.10.716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kurd SK, Troxel AB, Crits-Christoph P, Gelfand JM. The risk of depression, anxiety, and suicidality in patients with psoriasis: a population-based cohort study. Arch. Dermatol. 2010;146(8):891–895. doi: 10.1001/archdermatol.2010.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wilson I. Depression in the patient with COPD. Int. J .Chron Obstruct. Pulmon. Dis. 2006;1(1):61–64. doi: 10.2147/copd.2006.1.1.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bruce TO. Comorbid depression in rheumatoid arthritis: pathophysiology and clinical implications. Curr. Psychiatry Rep. 2008;10(3):258–264. doi: 10.1007/s11920-008-0042-1. [DOI] [PubMed] [Google Scholar]
  • 12.Siegert RJ, Abernethy DA. Depression in multiple sclerosis: a review. J. Neurol. Neurosurg. Psychiatry. 2005;76(4):469–475. doi: 10.1136/jnnp.2004.054635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wekking EM. Psychiatric symptoms in systemic lupus erythematosus: an update. Psychosom. Med. 1993;55(2):219–228. doi: 10.1097/00006842-199303000-00011. [DOI] [PubMed] [Google Scholar]
  • 14.Egede LE, Ellis C. Diabetes and depression: global perspectives. Diabetes Res. Clin. Pract. 2010;87(3):302–312. doi: 10.1016/j.diabres.2010.01.024. [DOI] [PubMed] [Google Scholar]
  • 15.Bunevicius R. Thyroid disorders in mental patients. Curr. Opin. Psychiatry. 2009;22(4):391–395. doi: 10.1097/YCO.0b013e328329e1ae. [DOI] [PubMed] [Google Scholar]
  • 16.Anisman H, Merali Z, Hayley S. Neurotransmitter, peptide and cytokine processes in relation to depressive disorder: comorbidity between depression and neurodegenerative disorders. Prog. Neurobiol. 2008;85(1):1–74. doi: 10.1016/j.pneurobio.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 17.Yates WR, Gleason O. Hepatitis C and depression. Depress Anxiety. 1998;7(4):188–193. doi: 10.1002/(sici)1520-6394(1998)7:4<188::aid-da7>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 18.Benton T. Depression and HIV/AIDS. Curr. Psychiatry Rep. 2008;10(3):280–285. doi: 10.1007/s11920-008-0045-y. [DOI] [PubMed] [Google Scholar]
  • 19.Reich M. [Depression in oncology] Cancer Radiother. 2010;14(6-7):535–538. doi: 10.1016/j.canrad.2010.06.003. [DOI] [PubMed] [Google Scholar]
  • 20.Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu. Rev. Immunol. 1989;7:145–173. doi: 10.1146/annurev.iy.07.040189.001045. [DOI] [PubMed] [Google Scholar]
  • 21.Sanders VM, Baker RA, Ramer-Quinn DS, Kasprowicz DJ, Fuchs BA, Street NE. Differential expression of the beta2-adrenergic receptor by Th1 and Th2 clones: implications for cytokine production and B cell help. J. Immunol. 1997;158(9):4200–4210. [PubMed] [Google Scholar]
  • 22.Gonzalez A, Fazzino F, Castillo M, Mata S, Lima L. Serotonin, 5-HT1A serotonin receptors and proliferation of lymphocytes in major depression patients. Neuroimmunomodulation. 2007;14(1):8–15. doi: 10.1159/000107283. [DOI] [PubMed] [Google Scholar]
  • 23.Lima L, Urbina M. Serotonin transporter modulation in blood lymphocytes from patients with major depression. Cell Mol. Neurobiol. 2002;22(5-6):797–804. doi: 10.1023/a:1021869310702. [DOI] [PubMed] [Google Scholar]
  • 24.Sternberg EM. Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nat. Rev. Immunol. 2006;6(4):318–328. doi: 10.1038/nri1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Farber L, Haus U, Spath M, Drechsler S. Physiology and pathophysiology of the 5-HT3 receptor. Scand. J. Rheumatol. Suppl. 2004;119:2–8. [PubMed] [Google Scholar]
  • 26.Mossner R, Lesch KP. Role of serotonin in the immune system and in neuroimmune interactions. Brain Behav. Immun. 1998;12(4):249–271. doi: 10.1006/brbi.1998.0532. [DOI] [PubMed] [Google Scholar]
  • 27.Okuyama-Tamura M, Mikuni M, Kojima I. Modulation of the human glucocorticoid receptor function by antidepressive compounds. Neurosci. Lett. 2003;342(3):206–210. doi: 10.1016/s0304-3940(03)00261-1. [DOI] [PubMed] [Google Scholar]
  • 28.Paul EW. Fundamental Immunology. Philadelphia: Lippincott Williams & Wilkins; 2003. [Google Scholar]
  • 29.de Kloet ER, Derijk RH, Meijer OC. Therapy Insight: is there an imbalanced response of mineralocorticoid and glucocorticoid receptors in depression? Nat. Clin. Pract. Endocrinol. Metab. 2007;3(2):168–179. doi: 10.1038/ncpendmet0403. [DOI] [PubMed] [Google Scholar]
  • 30.Rook GA. Glucocorticoids and immune function. Baillieres Best Pract. Res. Clin. Endocrinol. Metab. 1999;13(4):567–581. doi: 10.1053/beem.1999.0044. [DOI] [PubMed] [Google Scholar]
  • 31.Elenkov IJ, Chrousos GP. Stress Hormones, Th1/Th2 patterns, Pro/Anti-inflammatory Cytokines and Susceptibility to Disease. Trends Endocrinol. Metab. 1999;10(9):359–368. doi: 10.1016/s1043-2760(99)00188-5. [DOI] [PubMed] [Google Scholar]
  • 32.Kubera M, Maes M, Kenis G, Kim YK, Lason W. Effects of serotonin and serotonergic agonists and antagonists on the production of tumor necrosis factor alpha and interleukin-6. Psychiatry Res. 2005;134(3):251–258. doi: 10.1016/j.psychres.2004.01.014. [DOI] [PubMed] [Google Scholar]
  • 33.Muller T, Durk T, Blumenthal B, Grimm M, Cicko S, Panther E, Sorichter S, Herouy Y, Di Virgilio F, Ferrari D, Norgauer J, Idzko M. 5-hydroxytryptamine modulates migration, cytokine and chemokine release and T-cell priming capacity of dendritic cells in vitro and in vivo. PLoS One. 2009;4(7):e6453. doi: 10.1371/journal.pone.0006453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Jahnova E. [The role of serotonin in the regulation of the immune response] Bratisl Lek Listy. 1994;95(4):181–184. [PubMed] [Google Scholar]
  • 35.Szabo ST, de Montigny C, Blier P. Progressive attenuation of the firing activity of locus coeruleus noradrenergic neurons by sustained administration of selective serotonin reuptake inhibitors. Int. J. Neuropsychopharmacol. 2000;3(1):1–11. doi: 10.1017/S1461145700001772. [DOI] [PubMed] [Google Scholar]
  • 36.Di Giovanni G, Esposito E, Di Matteo V. Role of serotonin in central dopamine dysfunction. CNS Neurosci. Ther. 2010;16(3):179–194. doi: 10.1111/j.1755-5949.2010.00135.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Aosaki T, Miura M, Suzuki T, Nishimura K, Masuda M. Acetylcholine-dopamine balance hypothesis in the striatum: an update. Geriatr. Gerontol. Int. 2010;10(Suppl 1):S148–157. doi: 10.1111/j.1447-0594.2010.00588.x. [DOI] [PubMed] [Google Scholar]
  • 38.Hoebel BG, Avena NM, Rada P. Accumbens dopamine-acetylcholine balance in approach and avoidance. Curr. Opin. Pharmacol. 2007;7(6):617–627. doi: 10.1016/j.coph.2007.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Nakano K, Higashi T, Takagi R, Hashimoto K, Tanaka Y, Matsushita S. Dopamine released by dendritic cells polarizes Th2 differentiation. Int. Immunol. 2009;21(6):645–654. doi: 10.1093/intimm/dxp033. [DOI] [PubMed] [Google Scholar]
  • 40.Ilani T, Strous RD, Fuchs S. Dopaminergic regulation of immune cells via D3 dopamine receptor: a pathway mediated by activated T cells. FASEB J. 2004;18(13):1600–1602. doi: 10.1096/fj.04-1652fje. [DOI] [PubMed] [Google Scholar]
  • 41.Ghosh MC, Mondal AC, Basu S, Banerjee S, Majumder J, Bhattacharya D, Dasgupta PS. Dopamine inhibits cytokine release and expression of tyrosine kinases, Lck and Fyn in activated T cells. Int. Immunopharmacol. 2003;3(7):1019–1026. doi: 10.1016/S1567-5769(03)00100-0. [DOI] [PubMed] [Google Scholar]
  • 42.Kikuchi H, Itoh J, Fukuda S. Chronic nicotine stimulation modulates the immune response of mucosal T cells to Th1-dominant pattern via nAChR by upregulation of Th1-specific transcriptional factor. Neurosci. Lett. 2008;432(3):217–221. doi: 10.1016/j.neulet.2007.12.027. [DOI] [PubMed] [Google Scholar]
  • 43.Kuhlwein E, Irwin M. Melatonin modulation of lymphocyte proliferation and Th1/Th2 cytokine expression. J. Neuroimmunol. 2001;117(1-2):51–57. doi: 10.1016/s0165-5728(01)00325-3. [DOI] [PubMed] [Google Scholar]
  • 44.Garcia-Maurino S, Gonzalez-Haba MG, Calvo JR, Rafii-El-Idrissi M, Sanchez-Margalet V, Goberna R, Guerrero JM. Melatonin enhances IL-2, IL-6, and IFN-gamma production by human circulating CD4+ cells: a possible nuclear receptor-mediated mechanism involving T helper type 1 lymphocytes and monocytes. J. Immunol. 1997;159(2):574–581. [PubMed] [Google Scholar]
  • 45.Levite M. Neurotransmitters activate T-cells and elicit crucial functions via neurotransmitter receptors. Curr. Opin. Pharmacol. 2008;8(4):460–471. doi: 10.1016/j.coph.2008.05.001. [DOI] [PubMed] [Google Scholar]
  • 46.Elenkov IJ, Chrousos GP. Stress system--organization, physiology and immunoregulation. Neuroimmunomodulation. 2006;13(5-6):257–267. doi: 10.1159/000104853. [DOI] [PubMed] [Google Scholar]
  • 47.Lanfumey L, Mongeau R, Cohen-Salmon C, Hamon M. Corticosteroid-serotonin interactions in the neurobiological mechanisms of stress-related disorders. Neurosci. Biobehav. Rev. 2008;32(6):1174–1184. doi: 10.1016/j.neubiorev.2008.04.006. [DOI] [PubMed] [Google Scholar]
  • 48.Wosiski-Kuhn M, Stranahan AM. Opposing effects of positive and negative stress on hippocampal plasticity over the lifespan. Ageing Res. Rev. 2011. [Epub ahead of print] [DOI] [PubMed]
  • 49.Branchi I, Francia N, Alleva E. Epigenetic control of neurobehavioural plasticity: the role of neurotrophins. Behav. Pharmacol. 2004;15(5-6):353–362. doi: 10.1097/00008877-200409000-00006. [DOI] [PubMed] [Google Scholar]
  • 50.Kasakura S. A role for T-helper type 1 and type 2 cytokines in the pathogenesis of various human diseases. Rinsho Byori. 1998;46(9):915–921. [PubMed] [Google Scholar]
  • 51.Segal BM, Dwyer BK, Shevach EM. An interleukin (IL)-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease. J. Exp. Med. 1998;187(4):537–546. doi: 10.1084/jem.187.4.537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Humbert M, Menz G, Ying S, Corrigan CJ, Robinson DS, Durham SR, Kay AB. The immunopathology of extrinsic (atopic) and intrinsic (non-atopic) asthma: more similarities than differences. Immunol. Today. 1999;20(11):528–533. doi: 10.1016/s0167-5699(99)01535-2. [DOI] [PubMed] [Google Scholar]
  • 53.Wills-Karp M. IL-12/IL-13 axis in allergic asthma. J. Allergy Clin. Immunol. 2001;107(1):9–18. doi: 10.1067/mai.2001.112265. [DOI] [PubMed] [Google Scholar]
  • 54.Link H. The cytokine storm in multiple sclerosis. Mult. Scler. 1998;4(1):12–15. doi: 10.1177/135245859800400104. [DOI] [PubMed] [Google Scholar]
  • 55.Kato H, Suzumura A. Cytokines in MS lesion. Nippon Rinsho. 2003;61(8):1428–1434. [PubMed] [Google Scholar]
  • 56.Kang BY, Kim TS. Targeting cytokines of the interleukin- 12 family in autoimmunity. Curr. Med. Chem. 2006;13(10):1149–1156. doi: 10.2174/092986706776360879. [DOI] [PubMed] [Google Scholar]
  • 57.Bennett JL, Stuve O. Update on inflammation, neurodegeneration, and immunoregulation in multiple sclerosis: therapeutic implications. Clin. Neuropharmacol. 2009;32(3):121–132. doi: 10.1097/WNF.0b013e3181880359. [DOI] [PubMed] [Google Scholar]
  • 58.Schmitz T, Chew LJ. Cytokines and myelination in the central nervous system. Sci. World J. 2008;8:1119–1147. doi: 10.1100/tsw.2008.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Welsh CJ, Steelman AJ, Mi W, Young CR, Dean DD, Storts R, Welsh TH, Jr, Meagher MW. Effects of stress on the immune response to Theiler's virus--implications for virus-induced autoimmunity. Neuroimmunomodulation. 2010;17(3):169–172. doi: 10.1159/000258715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Minagar A, Carpenter A, Alexander JS. The destructive alliance: interactions of leukocytes, cerebral endothelial cells, and the immune cascade in pathogenesis of multiple sclerosis. Int. Rev. Neurobiol. 2007;79:1–11. doi: 10.1016/S0074-7742(07)79001-3. [DOI] [PubMed] [Google Scholar]
  • 61.Sospedra M, Martin R. Immunology of multiple sclerosis. Annu. Rev. Immunol. 2005;23:683–747. doi: 10.1146/annurev.immunol.23.021704.115707. [DOI] [PubMed] [Google Scholar]
  • 62.Vervoordeldonk MJ, Tak PP. Cytokines in rheumatoid arthritis. Curr. Rheumatol. Rep. 2002;4(3):208–217. doi: 10.1007/s11926-002-0067-0. [DOI] [PubMed] [Google Scholar]
  • 63.Miossec P. Cytokines in rheumatoid arthritis: is it all TNF-alpha? Cell Mol Biol (Noisy-le-grand): 2001;47(4):675–678. [PubMed] [Google Scholar]
  • 64.Umehara H, Tanaka M, Mimori T. [Interferon-gamma in pathogenesis of rheumatoid arthritis] Nippon Rinsho. 2005;63(Suppl 1):181–184. [PubMed] [Google Scholar]
  • 65.Cornelissen F, van Hamburg JP, Lubberts E. The IL-12/IL-23 axis and its role in Th17 cell development, pathology and plasticity in arthritis. Curr. Opin. Investig. Drugs. 2009;10(5):452–462. [PubMed] [Google Scholar]
  • 66.Malemud CJ, Miller AH. Pro-inflammatory cytokine-induced SAPK/MAPK and JAK/STAT in rheumatoid arthritis and the new anti-depression drugs. Expert Opin. Ther. Targets. 2008;12(2):171–183. doi: 10.1517/14728222.12.2.171. [DOI] [PubMed] [Google Scholar]
  • 67.Knedla A, Neumann E, Muller-Ladner U. Developments in the synovial biology field 2006. Arthritis Res. Ther. 2007;9(2):209. doi: 10.1186/ar2140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Firestein GS. Immunologic mechanisms in the pathogenesis of rheumatoid arthritis. J. Clin. Rheumatol. 2005;11(3 Suppl):S39–44. doi: 10.1097/01.rhu.0000166673.34461.33. [DOI] [PubMed] [Google Scholar]
  • 69.Raza K, Falciani F, Curnow SJ, Ross EJ, Lee CY, Akbar AN, Lord JM, Gordon C, Buckley CD, Salmon M. Early rheumatoid arthritis is characterized by a distinct and transient synovial fluid cytokine profile of T cell and stromal cell origin. Arthritis Res. Ther. 2005;7(4):R784–795. doi: 10.1186/ar1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rabinovitch A. An update on cytokines in the pathogenesis of insulin-dependent diabetes mellitus. Diabetes Metab. Rev. 1998;14(2):129–151. doi: 10.1002/(sici)1099-0895(199806)14:2<129::aid-dmr208>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
  • 71.Costa VS, Mattana TC, da Silva ME. Unregulated IL-23/IL-17 immune response in autoimmune diseases. Diabetes Res. Clin. Pract. 2010;88(3):222–226. doi: 10.1016/j.diabres.2010.03.014. [DOI] [PubMed] [Google Scholar]
  • 72.Bonifacio E, Scirpoli M, Kredel K, Fuchtenbusch M, Ziegler AG. Early autoantibody responses in prediabetes are IgG1 dominated and suggest antigen-specific regulation. J. Immunol. 1999;163(1):525–532. [PubMed] [Google Scholar]
  • 73.Phenekos C, Vryonidou A, Gritzapis AD, Baxevanis CN, Goula M, Papamichail M. Th1 and Th2 serum cytokine profiles characterize patients with Hashimoto's thyroiditis (Th1) and Graves' disease (Th2) Neuroimmunomodulation. 2004;11(4):209–213. doi: 10.1159/000078438. [DOI] [PubMed] [Google Scholar]
  • 74.Lichiardopol C, Mota M. The thyroid and autoimmunity. Rom. J. Intern. Med. 2009;47(3):207–215. [PubMed] [Google Scholar]
  • 75.Ajjan RA, Watson PF, McIntosh RS, Weetman AP. Intrathyroidal cytokine gene expression in Hashimoto's thyroiditis. Clin. Exp. Immunol. 1996;105(3):523–528. doi: 10.1046/j.1365-2249.1996.d01-784.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Guo J, Rapoport B, McLachlan SM. Balance of Th1/Th2 cytokines in thyroid autoantibody synthesis in vitro. Autoimmunity. 1999;30(1):1–9. doi: 10.3109/08916939908994754. [DOI] [PubMed] [Google Scholar]
  • 77.de la Vega JR, Vilaplana JC, Biro A, Hammond L, Bottazzo GF, Mirakian R. IL-10 expression in thyroid glands: protective or harmful role against thyroid autoimmunity? Clin. Exp. Immunol. 1998;113(1):126–135. doi: 10.1046/j.1365-2249.1998.00628.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yamaguchi Y, Takahashi H, Satoh T, Okazaki Y, Mizuki N, Takahashi K, Ikezawa Z, Kuwana M. Natural killer cells control a T-helper 1 response in patients with Behcet's disease. Arthritis Res. Ther. 2010;12(3):R80. doi: 10.1186/ar3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hamzaoui K, Ben Dhifallah I, Karray E, Sassi FH, Hamzaoui A. Vitamin D modulates peripheral immunity in patients with Behcet's disease. Clin. Exp. Rheumatol. 2010;28(4) Suppl 60:S50–57. [PubMed] [Google Scholar]
  • 80.Suzuki H, Suzuki Y, Aizawa M, Yamanaka T, Kihara M, Pang H, Horikoshi S, Tomino Y. Th1 polarization in murine IgA nephropathy directed by bone marrow-derived cells. Kidney Int. 2007;72(3):319–327. doi: 10.1038/sj.ki.5002300. [DOI] [PubMed] [Google Scholar]
  • 81.Lim CS, Yoon HJ, Kim YS, Ahn C, Han JS, Kim S, Lee JS, Lee HS, Chae DW. Clinicopathological correlation of intrarenal cytokines and chemokines in IgA nephropathy. Nephrology (Carlton) 2003;8(1):21–27. doi: 10.1046/j.1440-1797.2003.00128.x. [DOI] [PubMed] [Google Scholar]
  • 82.Cutolo M, Foppiani L, Prete C, Ballarino P, Sulli A, Villaggio B, Seriolo B, Giusti M, Accardo S. Hypothalamic-pituitary-adrenocortical axis function in premenopausal women with rheumatoid arthritis not treated with glucocorticoids. J. Rheumatol. 1999;26(2):282–288. [PubMed] [Google Scholar]
  • 83.Gudbjornsson B, Skogseid B, Oberg K, Wide L, Hallgren R. Intact adrenocorticotropic hormone secretion but impaired cortisol response in patients with active rheumatoid arthritis. Effect of glucocorticoids. J. Rheumatol. 1996;23(4):596–602. [PubMed] [Google Scholar]
  • 84.Neeck G, Federlin K, Graef V, Rusch D, Schmidt KL. Adrenal secretion of cortisol in patients with rheumatoid arthritis. J. Rheumatol. 1990;17(1):24–29. [PubMed] [Google Scholar]
  • 85.Chikanza IC, Petrou P, Kingsley G, Chrousos G, Panayi GS. Defective hypothalamic response to immune and inflammatory stimuli in patients with rheumatoid arthritis. Arthritis Rheum. 1992;35(11):1281–1288. doi: 10.1002/art.1780351107. [DOI] [PubMed] [Google Scholar]
  • 86.Mok CC, Lau CS. Pathogenesis of systemic lupus erythematosus. J. Clin. Pathol. 2003;56(7):481–490. doi: 10.1136/jcp.56.7.481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Salem ML. Estrogen a double-edged sword: modulation of TH1- and TH2-mediated inflammations by differential regulation of TH1/TH2 cytokine production. Curr. Drug Targets Inflamm. Allergy. 2004;3(1):97–104. doi: 10.2174/1568010043483944. [DOI] [PubMed] [Google Scholar]
  • 88.Mangini AJ, Lafyatis R, Van Seventer JM. Type I interferons inhibition of inflammatory T helper cell responses in systemic lupus erythematosus. Ann. N. Y. Acad. Sci. 2007;1108:11–23. doi: 10.1196/annals.1422.002. [DOI] [PubMed] [Google Scholar]
  • 89.Gomez D, Correa PA, Gomez LM, Cadena J, Molina JF, Anaya JM. Th1/Th2 cytokines in patients with systemic lupus erythematosus: is tumor necrosis factor alpha protective? Semin. Arthritis Rheum. 2004;33(6):404–413. doi: 10.1016/j.semarthrit.2003.11.002. [DOI] [PubMed] [Google Scholar]
  • 90.Mok MY, Wu HJ, Lo Y, Lau CS. The relation of interleukin 17 (IL-17) and IL-23 to Th1/Th2 cytokines and disease activity in systemic lupus erythematosus. J. Rheumatol. 2010;37(10):2046–2052. doi: 10.3899/jrheum.100293. [DOI] [PubMed] [Google Scholar]
  • 91.Hayashi T. Therapeutic strategies for SLE involving cytokines: mechanism-oriented therapies especially IFN-gamma targeting gene therapy. J. Biomed. Biotechnol. 2010;2010 doi: 10.1155/2010/461641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Calvani N, Tucci M, Richards HB, Tartaglia P, Silvestris F. Th1 cytokines in the pathogenesis of lupus nephritis: the role of IL-18. Autoimmun. Rev. 2005;4(8):542–548. doi: 10.1016/j.autrev.2005.04.009. [DOI] [PubMed] [Google Scholar]
  • 93.Nakashima H, Akahoshi M, Masutani K. Th1/Th2 balance of SLE patients with lupus nephritis. Rinsho Byori. 2006;54(7):706–713. [PubMed] [Google Scholar]
  • 94.Werfel T. The role of leukocytes, keratinocytes, and allergen-specific IgE in the development of atopic dermatitis. J. Invest. Dermatol. 2009;129(8):1878–1891. doi: 10.1038/jid.2009.71. [DOI] [PubMed] [Google Scholar]
  • 95.Levine SJ, Wenzel SE. Narrative review: the role of Th2 immune pathway modulation in the treatment of severe asthma and its phenotypes. Ann. Intern. Med. 2010;152(4):232–237. doi: 10.1059/0003-4819-152-4-201002160-00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Renauld JC. New insights into the role of cytokines in asthma. J. Clin. Pathol. 2001;54(8):577–589. doi: 10.1136/jcp.54.8.577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.de Blic J, Tillie-Leblond I, Tonnel AB, Jaubert F, Scheinmann P, Gosset P. Difficult asthma in children: an analysis of airway inflammation. J. Allergy Clin. Immunol. 2004;113(1):94–100. doi: 10.1016/j.jaci.2003.10.045. [DOI] [PubMed] [Google Scholar]
  • 98.Pene J, Chevalier S, Preisser L, Venereau E, Guilleux MH, Ghannam S, Moles JP, Danger Y, Ravon E, Lesaux S, Yssel H, Gascan H. Chronically inflamed human tissues are infiltrated by highly differentiated Th17 lymphocytes. J. Immunol. 2008;180(11):7423–7430. doi: 10.4049/jimmunol.180.11.7423. [DOI] [PubMed] [Google Scholar]
  • 99.Meiler F, Zimmermann M, Blaser K, Akdis CA, Akdis M. T-cell subsets in the pathogenesis of human asthma. Curr. Allergy Asthma Rep. 2006;6(2):91–96. doi: 10.1007/s11882-006-0045-0. [DOI] [PubMed] [Google Scholar]
  • 100.Chaplin DD. Cell cooperation in development of eosinophil-predominant inflammation in airways. Immunol. Res. 2002;26(1-3):55–62. doi: 10.1385/IR:26:1-3:055. [DOI] [PubMed] [Google Scholar]
  • 101.Surendar J, Mohan V, Rao MM, Babu S, Aravindhan V. Increased Levels of Both Th1 and Th2 Cytokines in Subjects with Metabolic Syndrome (CURES-103) Diabetes Technol. Ther. 2011;13(4):477–482. doi: 10.1089/dia.2010.0178. [DOI] [PubMed] [Google Scholar]
  • 102.Elenkov IJ, Iezzoni DG, Daly A, Harris AG, Chrousos GP. Cytokine dysregulation, inflammation and well-being. Neuroimmunomodulation. 2005;12(5):255–269. doi: 10.1159/000087104. [DOI] [PubMed] [Google Scholar]
  • 103.Stumpf C, Auer C, Yilmaz A, Lewczuk P, Klinghammer L, Schneider M, Daniel WG, Schmieder RE, Garlichs CD. Serum levels of the Th1 chemoattractant interferon-gamma-inducible protein (IP) 10 are elevated in patients with essential hypertension. Hypertens Res. 2011;34(4):484–488. doi: 10.1038/hr.2010.258. [DOI] [PubMed] [Google Scholar]
  • 104.Chamontin B. [The best of hypertension 2005] Arch. Mal Coeur Vaiss. 2006;99 Spec No 1(1):35–41. [PubMed] [Google Scholar]
  • 105.Lijnen PJ, Petrov VV, Fagard RH. Association between transforming growth factor-beta and hypertension. Am. J. Hypertens. 2003;16(7):604–611. doi: 10.1016/s0895-7061(03)00847-1. [DOI] [PubMed] [Google Scholar]
  • 106.Millis RM. Epigenetics and hypertension. Curr. Hypertens Rep. 2011;13(1):21–28. doi: 10.1007/s11906-010-0173-8. [DOI] [PubMed] [Google Scholar]
  • 107.Esler M, Eikelis N, Schlaich M, Lambert G, Alvarenga M, Kaye D, El-Osta A, Guo L, Barton D, Pier C, Brenchley C, Dawood T, Jennings G, Lambert E. Human sympathetic nerve biology: parallel influences of stress and epigenetics in essential hypertension and panic disorder. Ann. N. Y. Acad. Sci. 2008;1148:338–348. doi: 10.1196/annals.1410.064. [DOI] [PubMed] [Google Scholar]
  • 108.al'Absi M, Arnett DK. Adrenocortical responses to psychological stress and risk for hypertension. Biomed. Pharmacother. 2000;54(5):234–244. doi: 10.1016/S0753-3322(00)80065-7. [DOI] [PubMed] [Google Scholar]
  • 109.Whitworth JA, Brown MA, Kelly JJ, Williamson PM. Mechanisms of cortisol-induced hypertension in humans. Steroids. 1995;60(1):76–80. doi: 10.1016/0039-128x(94)00033-9. [DOI] [PubMed] [Google Scholar]
  • 110.Shamshiev AT, Ampenberger F, Ernst B, Rohrer L, Marsland BJ, Kopf M. Dyslipidemia inhibits Toll-like receptor-induced activation of CD8alpha-negative dendritic cells and protective Th1 type immunity. J. Exp. Med. 2007;204(2):441–452. doi: 10.1084/jem.20061737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhou X, Paulsson G, Stemme S, Hansson GK. Hypercholesterolemia is associated with a T helper (Th) 1/Th2 switch of the autoimmune response in atherosclerotic apo E-knockout mice. J. Clin. Invest. 1998;101(8):1717–1725. doi: 10.1172/JCI1216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Arner P. Catecholamine-induced lipolysis in obesity. Int. J. Obes. Relat. Metab. Disord. 1999;23(Suppl 1):10–13. doi: 10.1038/sj.ijo.0800789. [DOI] [PubMed] [Google Scholar]
  • 113.Brillon DJ, Zheng B, Campbell RG, Matthews DE. Effect of cortisol on energy expenditure and amino acid metabolism in humans. Am. J. Physiol. 1995;268(3 Pt 1):E501–513. doi: 10.1152/ajpendo.1995.268.3.E501. [DOI] [PubMed] [Google Scholar]
  • 114.Rocha VZ, Folco EJ, Sukhova G, Shimizu K, Gotsman I, Vernon AH, Libby P. Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ. Res. 2008;103(5):467–476. doi: 10.1161/CIRCRESAHA.108.177105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bertola A, Bonnafous S, Anty R, Patouraux S, Saint-Paul MC, Iannelli A, Gugenheim J, Barr J, Mato JM, Le Marchand-Brustel Y, Tran A, Gual P. Hepatic expression patterns of inflammatory and immune response genes associated with obesity and NASH in morbidly obese patients. PLoS One. 2010;5(10):e13577. doi: 10.1371/journal.pone.0013577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Meshkani R, Adeli K. Hepatic insulin resistance, metabolic syndrome and cardiovascular disease. Clin. Biochem. 2009;42(13-14):1331–1346. doi: 10.1016/j.clinbiochem.2009.05.018. [DOI] [PubMed] [Google Scholar]
  • 117.Fain JN. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam. Horm. 2006;74:443–477. doi: 10.1016/S0083-6729(06)74018-3. [DOI] [PubMed] [Google Scholar]
  • 118.Pacifico L, Di Renzo L, Anania C, Osborn JF, Ippoliti F, Schiavo E, Chiesa C. Increased T-helper interferon-gamma-secreting cells in obese children. Eur. J. Endocrinol. 2006;154(5):691–697. doi: 10.1530/eje.1.02138. [DOI] [PubMed] [Google Scholar]
  • 119.Schmidt MV, Czisch M, Sterlemann V, Reinel C, Samann P, Muller MB. Chronic social stress during adolescence in mice alters fat distribution in late life: prevention by antidepressant treatment. Stress. 2009;12(1):89–94. doi: 10.1080/10253890802049343. [DOI] [PubMed] [Google Scholar]
  • 120.Drapeau V, Therrien F, Richard D, Tremblay A. Is visceral obesity a physiological adaptation to stress? Panminerva Med. 2003;45(3):189–195. [PubMed] [Google Scholar]
  • 121.Kyrou I, Chrousos GP, Tsigos C. Stress, visceral obesity, and metabolic complications. Ann. N. Y. Acad. Sci. 2006;1083:77–110. doi: 10.1196/annals.1367.008. [DOI] [PubMed] [Google Scholar]
  • 122.Hamdy O, Porramatikul S, Al-Ozairi E. Metabolic obesity: the paradox between visceral and subcutaneous fat. Curr. Diabetes Rev. 2006;2(4):367–373. doi: 10.2174/1573399810602040367. [DOI] [PubMed] [Google Scholar]
  • 123.Yudkin JS. Inflammation, obesity, and the metabolic syndrome. Horm. Metab. Res. 2007;39(10):707–709. doi: 10.1055/s-2007-985898. [DOI] [PubMed] [Google Scholar]
  • 124.Borst SE. The role of TNF-alpha in insulin resistance. Endocrine. 2004;23(2-3):177–182. doi: 10.1385/ENDO:23:2-3:177. [DOI] [PubMed] [Google Scholar]
  • 125.Jagannathan-Bogdan M, McDonnell ME, Shin H, Rehman Q, Hasturk H, Apovian CM, Nikolajczyk BS. Elevated proinflammatory cytokine production by a skewed T cell compartment requires monocytes and promotes inflammation in type 2 diabetes. J. Immunol. 2011;186(2):1162–1172. doi: 10.4049/jimmunol.1002615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Viardot A, Grey ST, Mackay F, Chisholm D. Potential antiinflammatory role of insulin via the preferential polarization of effector T cells toward a T helper 2 phenotype. Endocrinology. 2007;148(1):346–353. doi: 10.1210/en.2006-0686. [DOI] [PubMed] [Google Scholar]
  • 127.Jawien J. New insights into immunological aspects of atherosclerosis. Pol. Arch. Med. Wewn. 2008;118(3):127–131. [PubMed] [Google Scholar]
  • 128.Rodriguez G, Mago N, Rosa F. [Role of inflammation in atherogenesis] Invest. Clin. 2009;50(1):109–129. [PubMed] [Google Scholar]
  • 129.Hansson GK. Inflammatory mechanisms in atherosclerosis. J. Thromb. Haemost. 2009;7(Suppl 1):328–331. doi: 10.1111/j.1538-7836.2009.03416.x. [DOI] [PubMed] [Google Scholar]
  • 130.Huber SA, Sakkinen P, David C, Newell MK, Tracy RP. T helper-cell phenotype regulates atherosclerosis in mice under conditions of mild hypercholesterolemia. Circulation. 2001;103(21):2610–2616. doi: 10.1161/01.cir.103.21.2610. [DOI] [PubMed] [Google Scholar]
  • 131.Greaves DR, Channon KM. Inflammation and immune responses in atherosclerosis. Trends Immunol. 2002;23(11):535–541. doi: 10.1016/s1471-4906(02)02331-1. [DOI] [PubMed] [Google Scholar]
  • 132.Shinozawa Y, Xie XQ, Endo T, Takuma K, Koike K. [Correlation between intravascular coagulation/fibrinolysis system and cytokines] Nippon Rinsho. 2004;62(12):2253–2261. [PubMed] [Google Scholar]
  • 133.Chu AJ. Blood coagulation as an intrinsic pathway for proinflammation: a mini review. Inflamm. Allergy Drug Targets. 2010;9(1):32–44. doi: 10.2174/187152810791292890. [DOI] [PubMed] [Google Scholar]
  • 134.Devaraj S, Rosenson RS, Jialal I. Metabolic syndrome: an appraisal of the pro-inflammatory and procoagulant status. Endocrinol. Metab. Clin. North Am. table of contents. 2004;33(2):431–453. doi: 10.1016/j.ecl.2004.03.008. table of contents. [DOI] [PubMed] [Google Scholar]
  • 135.Gando S. Microvascular thrombosis and multiple organ dysfunction syndrome. Crit. Care Med. 2010;38(2 Suppl):S35–42. doi: 10.1097/CCM.0b013e3181c9e31d. [DOI] [PubMed] [Google Scholar]
  • 136.De Cicco M. The prothrombotic state in cancer: pathogenic mechanisms. Crit. Rev. Oncol. Hematol. 2004;50(3):187–196. doi: 10.1016/j.critrevonc.2003.10.003. [DOI] [PubMed] [Google Scholar]
  • 137.Cheng X, Liao YH, Li B, Zhang JY, Ge HX, Yuan J, Wang M, Liu Y, Guo ZQ, Chen J, Zhang J, Chen P. [The significance of Th1/Th2 function imbalance in patients with post-infarction cardiac insufficiency] Zhonghua Xin Xue Guan Bing Za Zhi. 2005;33(6):526–528. [PubMed] [Google Scholar]
  • 138.Cheng X, Ding Y, Xia C, Tang T, Yu X, Xie J, Liao M, Yao R, Chen Y, Wang M, Liao YH. Atorvastatin modulates Th1/Th2 response in patients with chronic heart failure. J. Card. Fail. 2009;15(2):158–162. doi: 10.1016/j.cardfail.2008.10.001. [DOI] [PubMed] [Google Scholar]
  • 139.Cheng X, Liao YH, Zhang J, Li B, Ge H, Yuan J, Wang M, Lu B, Liu Y, Cheng Y. Effects of Atorvastatin on Th polarization in patients with acute myocardial infarction. Eur. J. Heart Fail. 2005;7(7):1099–1104. doi: 10.1016/j.ejheart.2005.01.020. [DOI] [PubMed] [Google Scholar]
  • 140.Vlastelica M. Emotional stress as a trigger in sudden cardiac death. Psychiatr. Danub. 2008;20(3):411–414. [PubMed] [Google Scholar]
  • 141.Zupancic ML. Acute psychological stress as a precipitant of acute coronary syndromes in patients with undiagnosed ischemic heart disease: a case report and literature review. Prim. Care Companion J. Clin. Psychiatry. 2009;11(1):21–24. doi: 10.4088/pcc.08r00623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bossowska A, Kiersnowska-Rogowska B, Bossowski A, Galar B, Sowinski P. Cytokines in patients with ischaemic heart disease or myocardial infarction. Kardiol. Pol. 2003;59(8):105–114. [PubMed] [Google Scholar]
  • 143.Hartford M, Wiklund O, Mattsson Hulten L, Perers E, Person A, Herlitz J, Hurt-Camejo E, Karlsson T, Caidahl K. CRP, interleukin-6, secretory phospholipase A2 group IIA, and intercellular adhesion molecule-1 during the early phase of acute coronary syndromes and long-term follow-up. Int. J. Cardiol. 2006;108(1):55–62. doi: 10.1016/j.ijcard.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 144.Riese U, Brenner S, Docke WD, Prosch S, Reinke P, Oppert M, Volk HD, Platzer C. Catecholamines induce IL-10 release in patients suffering from acute myocardial infarction by transactivating its promoter in monocytic but not in T-cells. Mol. Cell Biochem. 2000;212(1-2):45–50. [PubMed] [Google Scholar]
  • 145.Platzer C, Docke W, Volk H, Prosch S. Catecholamines trigger IL-10 release in acute systemic stress reaction by direct stimulation of its promoter/enhancer activity in monocytic cells. J. Neuroimmunol. 2000;105(1):31–38. doi: 10.1016/s0165-5728(00)00205-8. [DOI] [PubMed] [Google Scholar]
  • 146.Wei L. Immunological aspect of cardiac remodeling: T lymphocyte subsets in inflammation-mediated cardiac fibrosis. Exp. Mol. Pathol. 2011;90(1):74–78. doi: 10.1016/j.yexmp.2010.10.004. [DOI] [PubMed] [Google Scholar]
  • 147.Gage JR, Fonarow G, Hamilton M, Widawski M, Martinez-Maza O, Vredevoe DL. Beta blocker and angiotensin-converting enzyme inhibitor therapy is associated with decreased Th1/Th2 cytokine ratios and inflammatory cytokine production in patients with chronic heart failure. Neuroimmunomodulation. 2004;11(3):173–180. doi: 10.1159/000076766. [DOI] [PubMed] [Google Scholar]
  • 148.Shenhar-Tsarfaty S, Assayag EB, Bova I, Shopin L, Berliner S, Shapira I, Bornstein NM. Early signaling of inflammation in acute ischemic stroke: clinical and rheological implications. Thromb. Res. 2008;122(2):167–173. doi: 10.1016/j.thromres.2007.10.019. [DOI] [PubMed] [Google Scholar]
  • 149.Vila N, Castillo J, Davalos A, Chamorro A. Proinflammatory cytokines and early neurological worsening in ischemic stroke. Stroke. 2000;31(10):2325–2329. doi: 10.1161/01.str.31.10.2325. [DOI] [PubMed] [Google Scholar]
  • 150.Okazaki K, Nishida A, Kato M, Kozawa K, Uga N, Kimura H. Elevation of cytokine concentrations in asphyxiated neonates. Biol. Neonate. 2006;89(3):183–189. doi: 10.1159/000089180. [DOI] [PubMed] [Google Scholar]
  • 151.Santos M, Kovari E, Gold G, Bozikas VP, Hof PR, Bouras C, Giannakopoulos P. The neuroanatomical model of post-stroke depression: towards a change of focus? J. Neurol. Sci. 2009;283(1-2):158–162. doi: 10.1016/j.jns.2009.02.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Nestle FO, Turka LA, Nickoloff BJ. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. J. Clin. Invest. 1994;94(1):202–209. doi: 10.1172/JCI117308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Vena GA, Vestita M, Cassano N. Psoriasis and cardiovascular disease. Dermatol. Ther. 2010;23(2):144–151. doi: 10.1111/j.1529-8019.2010.01308.x. [DOI] [PubMed] [Google Scholar]
  • 154.Hodge G, Nairn J, Holmes M, Reynolds PN, Hodge S. Increased intracellular T helper 1 proinflammatory cytokine production in peripheral blood, bronchoalveolar lavage and intraepithelial T cells of COPD subjects. Clin. Exp. Immunol. 2007;150(1):22–29. doi: 10.1111/j.1365-2249.2007.03451.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Macario CC, Torres Tajes JP, Cordoba Lanus E. [COPD: bronchial and systemic inflammation] Arch. Bronconeumol. 2010;46(Suppl 4):9–15. doi: 10.1016/S0300-2896(10)70027-2. [DOI] [PubMed] [Google Scholar]
  • 156.Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat. Rev. Immunol. 2003;3(7):521–533. doi: 10.1038/nri1132. [DOI] [PubMed] [Google Scholar]
  • 157.De Martinis M, Di Benedetto MC, Mengoli LP, Ginaldi L. Senile osteoporosis: is it an immune-mediated disease? Inflamm. Res. 2006;55(10):399–404. doi: 10.1007/s00011-006-6034-x. [DOI] [PubMed] [Google Scholar]
  • 158.Sandmand M, Bruunsgaard H, Kemp K, Andersen-Ranberg K, Pedersen AN, Skinhoj P, Pedersen BK. Is ageing associated with a shift in the balance between Type 1 and Type 2 cytokines in humans? Clin. Exp. Immunol. 2002;127(1):107–114. doi: 10.1046/j.1365-2249.2002.01736.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Ginaldi L, De Martinis M, D'Ostilio A, Marini L, Loreto MF, Quaglino D. The immune system in the elderly: III. Innate immunity. Immunol. Res. 1999;20(2):117–126. doi: 10.1007/BF02786468. [DOI] [PubMed] [Google Scholar]
  • 160.Chan EC, Smith R, Lewin T, Brinsmead MW, Zhang HP, Cubis J, Thornton K, Hurt D. Plasma corticotropin-releasing hormone, beta-endorphin and cortisol inter-relationships during human pregnancy. Acta Endocrinol. (Copenh) 1993;128(4):339–344. doi: 10.1530/acta.0.1280339. [DOI] [PubMed] [Google Scholar]
  • 161.Dantzer R, Wollman E, Vitkovic L, Yirmiya R. Cytokines and depression: fortuitous or causative association? Mol. Psychiatry. 1999;4(4):328–332. doi: 10.1038/sj.mp.4000572. [DOI] [PubMed] [Google Scholar]
  • 162.Hashioka S, McGeer PL, Monji A, Kanba S. Anti-inflammatory effects of antidepressants: possibilities for preventives against Alzheimer's disease. Cent. Nerv. Syst. Agents Med.Chem. 2009;9(1):12–19. doi: 10.2174/187152409787601897. [DOI] [PubMed] [Google Scholar]
  • 163.Nagatsu T, Sawada M. Inflammatory process in Parkinson's disease: role for cytokines. Curr. Pharm. Des. 2005;11(8):999–1016. doi: 10.2174/1381612053381620. [DOI] [PubMed] [Google Scholar]
  • 164.Sawada M, Imamura K, Nagatsu T. Role of cytokines in inflammatory process in Parkinson's disease. J. Neural. Transm. Suppl. 2006. pp. 373–381. [DOI] [PubMed]
  • 165.Brodacki B, Staszewski J, Toczylowska B, Kozlowska E, Drela N, Chalimoniuk M, Stepien A. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci. Lett. 2008;441(2):158–162. doi: 10.1016/j.neulet.2008.06.040. [DOI] [PubMed] [Google Scholar]
  • 166.Nagatsu T, Mogi M, Ichinose H, Togari A. Changes in cytokines and neurotrophins in Parkinson's disease. J. Neural. Transm. Suppl. 2000;vol. (60):277–290. doi: 10.1007/978-3-7091-6301-6_19. [DOI] [PubMed] [Google Scholar]
  • 167.Nagatsu T, Mogi M, Ichinose H, Togari A. Cytokines in Parkinson's disease. J. Neural. Transm. Suppl. 2000;vol. (58):143–151. [PubMed] [Google Scholar]
  • 168.Mount MP, Lira A, Grimes D, Smith PD, Faucher S, Slack R, Anisman H, Hayley S, Park DS. Involvement of interferon-gamma in microglial-mediated loss of dopaminergic neurons. J. Neurosci. 2007;27(12):3328–3337. doi: 10.1523/JNEUROSCI.5321-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Mihara T, Nakashima M, Kuroiwa A, Akitake Y, Ono K, Hosokawa M, Yamada T, Takahashi M. Natural killer cells of Parkinson's disease patients are set up for activation: a possible role for innate immunity in the pathogenesis of this disease. Parkinsonism Relat. Disord. 2008;14(1):46–51. doi: 10.1016/j.parkreldis.2007.05.013. [DOI] [PubMed] [Google Scholar]
  • 170.Baba Y, Kuroiwa A, Uitti RJ, Wszolek ZK, Yamada T. Alterations of T-lymphocyte populations in Parkinson disease. Parkinsonism Relat. Disord. 2005;11(8):493–498. doi: 10.1016/j.parkreldis.2005.07.005. [DOI] [PubMed] [Google Scholar]
  • 171.Arimoto T, Choi DY, Lu X, Liu M, Nguyen XV, Zheng N, Stewart CA, Kim HC, Bing G. Interleukin-10 protects against inflammation-mediated degeneration of dopaminergic neurons in substantia nigra. Neurobiol. Aging. 2006;28(6):894–906. doi: 10.1016/j.neurobiolaging.2006.04.011. [DOI] [PubMed] [Google Scholar]
  • 172.Johnston LC, Su X, Maguire-Zeiss K, Horovitz K, Ankoudinova I, Guschin D, Hadaczek P, Federoff HJ, Bankiewicz K, Forsayeth J. Human interleukin-10 gene transfer is protective in a rat model of Parkinson's disease. Mol. Ther. 2008;16(8):1392–1399. doi: 10.1038/mt.2008.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Engler H, Doenlen R, Riether C, Engler A, Niemi MB, Besedovsky HO, del Rey A, Pacheco-Lopez G, Feldon J, Schedlowski M. Time-dependent alterations of peripheral immune parameters after nigrostriatal dopamine depletion in a rat model of Parkinson's disease. Brain Behav. Immun. 2009;23(4):518–526. doi: 10.1016/j.bbi.2009.01.018. [DOI] [PubMed] [Google Scholar]
  • 174.Kluter H, Vieregge P, Stolze H, Kirchner H. Defective production of interleukin-2 in patients with idiopathic Parkinson's disease. J. Neurol. Sci. 1995;133(1-2):134–139. doi: 10.1016/0022-510x(95)00180-a. [DOI] [PubMed] [Google Scholar]
  • 175.Reale M, Iarlori C, Feliciani C, Gambi D. Peripheral chemokine receptors, their ligands, cytokines and Alzheimer's disease. J. Alzheimers Dis. 2008;14(2):147–159. doi: 10.3233/jad-2008-14203. [DOI] [PubMed] [Google Scholar]
  • 176.Cao C, Arendash GW, Dickson A, Mamcarz MB, Lin X, Ethell DW. Abeta-specific Th2 cells provide cognitive and pathological benefits to Alzheimer's mice without infiltrating the CNS. Neurobiol. Dis. 2009;34(1):63–70. doi: 10.1016/j.nbd.2008.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kim HD, Tahara K, Maxwell JA, Lalonde R, Fukuiwa T, Fujihashi K, Van Kampen KR, Kong FK, Tang DC, Fukuchi K. Nasal inoculation of an adenovirus vector encoding 11 tandem repeats of Abeta1-6 upregulates IL-10 expression and reduces amyloid load in a Mo/Hu APPswe PS1dE9 mouse model of Alzheimer's disease. J. Gene Med. 2007;9(2):88–98. doi: 10.1002/jgm.993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Gadient RA, Otten UH. Interleukin-6 (IL-6)--a molecule with both beneficial and destructive potentials. Prog. Neurobiol. 1997;52(5):379–390. doi: 10.1016/s0301-0082(97)00021-x. [DOI] [PubMed] [Google Scholar]
  • 179.Fitzgerald PJ. Is elevated norepinephrine an etiological factor in some cases of Alzheimer's disease? Curr. Alzheimer Res. 2010;7(6):506–516. doi: 10.2174/156720510792231775. [DOI] [PubMed] [Google Scholar]
  • 180.Clerici M, Shearer GM. A TH1-->TH2 switch is a critical step in the etiology of HIV infection. Immunol. Today. 1993;14(3):107–111. doi: 10.1016/0167-5699(93)90208-3. [DOI] [PubMed] [Google Scholar]
  • 181.Capitanio JP, Abel K, Mendoza SP, Blozis SA, McChesney MB, Cole SW, Mason WA. Personality and serotonin transporter genotype interact with social context to affect immunity and viral set-point in simian immunodeficiency virus disease. Brain Behav. Immun. 2008;22(5):676–689. doi: 10.1016/j.bbi.2007.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Capitanio JP, Lerche NW. Psychosocial factors and disease progression in simian AIDS: a preliminary report. AIDS. 1991;5(9):1103–1106. doi: 10.1097/00002030-199109000-00007. [DOI] [PubMed] [Google Scholar]
  • 183.Reilly KH, Clark RA, Schmidt N, Benight CC, Kissinger P. The effect of post-traumatic stress disorder on HIV disease progression following hurricane Katrina. AIDS Care. 2009;21(10):1298–1305. doi: 10.1080/09540120902732027. [DOI] [PubMed] [Google Scholar]
  • 184.Gore-Felton C, Koopman C. Behavioral mediation of the relationship between psychosocial factors and HIV disease progression. Psychosom. Med. 2008;70(5):569–574. doi: 10.1097/PSY.0b013e318177353e. [DOI] [PubMed] [Google Scholar]
  • 185.Fleshner M, Brennan FX, Nguyen K, Watkins LR, Maier SF. RU-486 blocks differentially suppressive effect of stress on in vivo anti-KLH immunoglobulin response. Am. J. Physiol. 1996;271(5 Pt 2):R1344–1352. doi: 10.1152/ajpregu.1996.271.5.R1344. [DOI] [PubMed] [Google Scholar]
  • 186.Pacifici R, Zuccaro P, Hernandez Lopez C, Pichini S, Di Carlo S, Farre M, Roset PN, Ortuno J, Segura J, Torre RL. Acute effects of 3,4-methylenedioxymethamphetamine alone and in combination with ethanol on the immune system in humans. J. Pharmacol. Exp. Ther. 2001;296(1):207–215. [PubMed] [Google Scholar]
  • 187.Miller G, Chen E, Cole SW. Health psychology: developing biologically plausible models linking the social world and physical health. Annu. Rev. Psychol. 2009;60:501–524. doi: 10.1146/annurev.psych.60.110707.163551. [DOI] [PubMed] [Google Scholar]
  • 188.Becker Y. The changes in the T helper 1 (Th1) and T helper 2 (Th2) cytokine balance during HIV-1 infection are indicative of an allergic response to viral proteins that may be reversed by Th2 cytokine inhibitors and immune response modifiers--a review and hypothesis. Virus Genes. 2004;28(1):5–18. doi: 10.1023/B:VIRU.0000012260.32578.72. [DOI] [PubMed] [Google Scholar]
  • 189.Rook GA, Onyebujoh P, Stanford JL. TH1/TH2 switching and loss of CD4+ T cells in chronic infections: an immuno- endocrinological hypothesis not exclusive to HIV. Immunol. Today. 1993;14(11):568–569. doi: 10.1016/0167-5699(93)90190-V. [DOI] [PubMed] [Google Scholar]
  • 190.Pawlowska M. [HCV: immune response and its modifiers in clinical development] Przegl. Epidemiol. 2005;59(2):519–523. [PubMed] [Google Scholar]
  • 191.Par G, Berki T, Palinkas L, Balogh P, Szereday L, Halasz M, Szekeres-Bartho J, Miseta A, Hegedus G, Mozsik G, Hunyady B, Par A. [Immunology of HCV infection: the causes of impaired cellular immune response and the effect of antiviral treatment] Orv. Hetil. 2006;147(13):591–600. [PubMed] [Google Scholar]
  • 192.Trapero M, Garcia-Buey L, Munoz C, Viton M, Moreno-Monteagudo JA, Borque MJ, Quintana NE, Moreno-Otero R. Maintenance of T1 response as induced during PEG-IFNalpha plus ribavirin therapy controls viral replication in genotype-1 patients with chronic hepatitis C. Rev. Esp. Enferm. Dig. 2005;97(7):481–490. doi: 10.4321/s1130-01082005000700003. [DOI] [PubMed] [Google Scholar]
  • 193.Hukkanen V, Broberg E, Salmi A, Eralinna JP. Cytokines in experimental herpes simplex virus infection. Int. Rev. Immunol. 2002;21(4-5):355–371. doi: 10.1080/08830180213276. [DOI] [PubMed] [Google Scholar]
  • 194.Miller RL, Tomai MA, Harrison CJ, Bernstein DI. Immunomodulation as a treatment strategy for genital herpes: review of the evidence. Int. Immunopharmacol. 2002;2(4):443–451. doi: 10.1016/s1567-5769(01)00184-9. [DOI] [PubMed] [Google Scholar]
  • 195.Hengge UR, Benninghoff B, Ruzicka T, Goos M. Topical immunomodulators--progress towards treating inflammation, infection, and cancer. Lancet Infect. Dis. 2001;1(3):189–198. doi: 10.1016/s1473-3099(01)00095-0. [DOI] [PubMed] [Google Scholar]
  • 196.Reiner SL, Locksley RM. Cytokines in the differentiation of Th1/Th2 CD4+ subsets in leishmaniasis. J. Cell Biochem. 1993;53(4):323–328. doi: 10.1002/jcb.240530409. [DOI] [PubMed] [Google Scholar]
  • 197.Fitzgerald TJ. The Th1/Th2-like switch in syphilitic infection: is it detrimental? Infect. Immun. 1992;60(9):3475–3479. doi: 10.1128/iai.60.9.3475-3479.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.He XY, Xiao L, Chen HB, Hao J, Li J, Wang YJ, He K, Gao Y, Shi BY. T regulatory cells and Th1/Th2 cytokines in peripheral blood from tuberculosis patients. Eur. J. Clin. Microbiol. Infect. Dis. 2010;29(6):643–650. doi: 10.1007/s10096-010-0908-0. [DOI] [PubMed] [Google Scholar]
  • 199.Lienhardt C, Azzurri A, Amedei A, Fielding K, Sillah J, Sow OY, Bah B, Benagiano M, Diallo A, Manetti R, Manneh K, Gustafson P, Bennett S, D'Elios MM, McAdam K, Del Prete G. Active tuberculosis in Africa is associated with reduced Th1 and increased Th2 activity in vivo. Eur. J. Immunol. 2002;32(6):1605–1613. doi: 10.1002/1521-4141(200206)32:6<1605::AID-IMMU1605>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
  • 200.Garcia M, Vargas JA, Castejon R, Navas E, Durantez A. Flow-cytometric assessment of lymphocyte cytokine production in tuberculosis. Tuberculosis (Edinb) 2002;82(1):37–41. doi: 10.1054/tube.2001.0321. [DOI] [PubMed] [Google Scholar]
  • 201.Kelestimur F. The endocrinology of adrenal tuberculosis: the effects of tuberculosis on the hypothalamo-pituitary-adrenal axis and adrenocortical function. J. Endocrinol. Invest. 2004;27(4):380–386. doi: 10.1007/BF03351067. [DOI] [PubMed] [Google Scholar]
  • 202.Patel SA, Ndabahaliye A, Lim PK, Milton R, Rameshwar P. Challenges in the development of future treatments for breast cancer stem cells. Breast Cancer (London) 2010;2:1–11. [PMC free article] [PubMed] [Google Scholar]
  • 203.Moeller M, Kershaw MH, Cameron R, Westwood JA, Trapani JA, Smyth MJ, Darcy PK. Sustained antigen-specific antitumor recall response mediated by gene-modified CD4+ T helper-1 and CD8+ T cells. Cancer Res. 2007;67(23):11428–11437. doi: 10.1158/0008-5472.CAN-07-1141. [DOI] [PubMed] [Google Scholar]
  • 204.Scola L, Giacalone A, Marasa L, Mirabile M, Vaccarino L, Forte GI, Giannitrapani L, Caruso C, Montalto G, Lio D. Genetic determined downregulation of both type 1 and type 2 cytokine pathways might be protective against pancreatic cancer. Ann. N. Y. Acad. Sci. 2009;1155:284–288. doi: 10.1111/j.1749-6632.2008.03686.x. [DOI] [PubMed] [Google Scholar]
  • 205.Lissoni P, Rovelli F, Tisi E, Brivio F, Ardizzoia A, Barni S, Tancini G, Saudelli M, Cesana E, Vigano MG. Relation between macrophage and T helper-2 lymphocyte functions in human neoplasms: neopterin, interleukin-10 and interleukin-6 blood levels in early or advanced solid tumors. J. Biol. Regul. Homeost. Agents. 1995;9(4):146–149. [PubMed] [Google Scholar]
  • 206.Budhu A, Forgues M, Ye QH, Jia HL, He P, Zanetti KA, Kammula US, Chen Y, Qin LX, Tang ZY, Wang XW. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell. 2006;10(2):99–111. doi: 10.1016/j.ccr.2006.06.016. [DOI] [PubMed] [Google Scholar]
  • 207.Budhu A, Wang XW. The role of cytokines in hepatocellular carcinoma. J. Leukoc. Biol. 2006;80(6):1197–1213. doi: 10.1189/jlb.0506297. [DOI] [PubMed] [Google Scholar]
  • 208.Sparano A, Lathers DM, Achille N, Petruzzelli GJ, Young MR. Modulation of Th1 and Th2 cytokine profiles and their association with advanced head and neck squamous cell carcinoma. Otolaryngol. Head Neck Surg. 2004;131(5):573–576. doi: 10.1016/j.otohns.2004.03.016. [DOI] [PubMed] [Google Scholar]
  • 209.Pries R, Wollenberg B. Cytokines in head and neck cancer. Cytokine Growth Factor Rev. 2006;17(3):141–146. doi: 10.1016/j.cytogfr.2006.02.001. [DOI] [PubMed] [Google Scholar]
  • 210.Chen X, Xu X, Zhao J, Zhu L, Xue A, Zhang D. [The gene expression of Th1/Th2 cytokines and its role in hypopharyngeal carcinoma] Lin Chuang Er Bi Yan Hou Ke Za Zhi. 2004;18(6):327–328. [PubMed] [Google Scholar]
  • 211.Wei YC, Hu FB, Shen Y, Xu LH, Lin LS. [Postoperative Th1 and Th2 type cytokine changes in patients with esophageal squamous cell carcinoma and their clinical significance] Di Yi Jun Yi Da Xue Xue Bao. 2004;24(11):1271–1273. [PubMed] [Google Scholar]
  • 212.Yamazaki K, Yano T, Kameyama T, Suemitsu R, Yoshino I, Sugio K. Clinical significance of serum TH1/TH2 cytokines in patients with pulmonary adenocarcinoma. Surgery. 2002;131(1 Suppl):S236–241. doi: 10.1067/msy.2002.119795. [DOI] [PubMed] [Google Scholar]
  • 213.Teschendorff AE, Gomez S, Arenas A, El-Ashry D, Schmidt M, Gehrmann M, Caldas C. Improved prognostic classification of breast cancer defined by antagonistic activation patterns of immune response pathway modules. BMC Cancer. 2010;10:604. doi: 10.1186/1471-2407-10-604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Kobayashi M, Kobayashi H, Pollard RB, Suzuki F. A pathogenic role of Th2 cells and their cytokine products on the pulmonary metastasis of murine B16 melanoma. J. Immunol. 1998;160(12):5869–5873. [PubMed] [Google Scholar]
  • 215.Weng AP, Shahsafaei A, Dorfman DM. CXCR4/CD184 immunoreactivity in T-cell non-Hodgkin lymphomas with an overall Th1- Th2+ immunophenotype. Am. J. Clin. Pathol. 2003;119(3):424–430. doi: 10.1309/rf4pvceggn2xaf86. [DOI] [PubMed] [Google Scholar]
  • 216.Rico MJ, Matar P, Gervasoni SI, Bonfil RD, Calcaterra N, Scharovsky OG. The transition to the metastatic phenotype of rat lymphoma cells involves up-regulation of IL-10 receptor expression and IL-10 secretion. Clin. Exp. Metastasis. 2005;22(2):127–135. doi: 10.1007/s10585-005-5140-4. [DOI] [PubMed] [Google Scholar]
  • 217.Bellone G, Turletti A, Artusio E, Mareschi K, Carbone A, Tibaudi D, Robecchi A, Emanuelli G, Rodeck U. Tumor-associated transforming growth factor-beta and interleukin-10 contribute to a systemic Th2 immune phenotype in pancreatic carcinoma patients. Am. J. Pathol. 1999;155(2):537–547. doi: 10.1016/s0002-9440(10)65149-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Feurino LW, Zhang Y, Bharadwaj U, Zhang R, Li F, Fisher WE, Brunicardi FC, Chen C, Yao Q, Min L. IL-6 stimulates Th2 type cytokine secretion and upregulates VEGF and NRP-1 expression in pancreatic cancer cells. Cancer Biol. Ther. 2007;6(7):1096–1100. doi: 10.4161/cbt.6.7.4328. [DOI] [PubMed] [Google Scholar]
  • 219.Tassi E, Braga M, Longhi R, Gavazzi F, Parmiani G, Di Carlo V, Protti MP. Non-redundant role for IL-12 and IL-27 in modulating Th2 polarization of carcinoembryonic antigen specific CD4 T cells from pancreatic cancer patients. PLoS One. 2009;4(10):e7234. doi: 10.1371/journal.pone.0007234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Reiche EM, Morimoto HK, Nunes SM. Stress and depression-induced immune dysfunction: implications for the development and progression of cancer. Int. Rev. Psychiatry. 2005;17(6):515–527. doi: 10.1080/02646830500382102. [DOI] [PubMed] [Google Scholar]
  • 221.Yang M, Ma C, Liu S, Sun J, Shao Q, Gao W, Zhang Y, Li Z, Xie Q, Dong Z, Qu X. Hypoxia skews dendritic cells to a T helper type 2-stimulating phenotype and promotes tumour cell migration by dendritic cell-derived osteopontin. Immunology. 2009;128(1 Suppl):e237–249. doi: 10.1111/j.1365-2567.2008.02954.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Basso AM, Depiante-Depaoli M, Molina VA. Chronic variable stress facilitates tumoral growth: reversal by imipramine administration. Life Sci. 1992;50(23):1789–1796. doi: 10.1016/0024-3205(92)90063-u. [DOI] [PubMed] [Google Scholar]
  • 223.Palesh O, Butler LD, Koopman C, Giese-Davis J, Carlson R, Spiegel D. Stress history and breast cancer recurrence. J. Psychosom. Res. 2007;63(3):233–239. doi: 10.1016/j.jpsychores.2007.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Katz JB, Muller AJ, Prendergast GC. Indoleamine 2,3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunol. Rev. 2008;222:206–221. doi: 10.1111/j.1600-065X.2008.00610.x. [DOI] [PubMed] [Google Scholar]
  • 225.Schrocksnadel K, Wirleitner B, Winkler C, Fuchs D. Monitoring tryptophan metabolism in chronic immune activation. Clin. Chim. Acta. 2006;364(1-2):82–90. doi: 10.1016/j.cca.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • 226.Tanke MA, Kema IP, Dijck-Brouwer J, Doornbos B, De Vries EG, Korf J. Low plasma tryptophan in carcinoid patients is associated with increased urinary cortisol excretion. Psychoneuroendocrinology. 2008;33(9):1297–1301. doi: 10.1016/j.psyneuen.2008.07.005. [DOI] [PubMed] [Google Scholar]
  • 227.Brandacher G, Winkler C, Schroecksnadel K, Margreiter R, Fuchs D. Antitumoral activity of interferon-gamma involved in impaired immune function in cancer patients. Curr. Drug Metab. 2006;7(6):599–612. doi: 10.2174/138920006778017768. [DOI] [PubMed] [Google Scholar]
  • 228.Zeng J, Cai S, Yi Y, He Y, Wang Z, Jiang G, Li X, Du J. Prevention of spontaneous tumor development in a ret transgenic mouse model by ret peptide vaccination with indoleamine 2,3-dioxygenase inhibitor 1-methyl tryptophan. Cancer Res. 2009;69(9):3963–3970. doi: 10.1158/0008-5472.CAN-08-2476. [DOI] [PubMed] [Google Scholar]
  • 229.Boasso A, Shearer GM. How does indoleamine 2,3-dioxygenase contribute to HIV-mediated immune dysregulation. Curr. Drug Metab. 2007;8(3):217–223. doi: 10.2174/138920007780362527. [DOI] [PubMed] [Google Scholar]
  • 230.Opitz CA, Wick W, Steinman L, Platten M. Tryptophan degradation in autoimmune diseases. Cell Mol. Life Sci. 2007;64(19-20):2542–2563. doi: 10.1007/s00018-007-7140-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Criado G, Simelyte E, Inglis JJ, Essex D, Williams RO. Indoleamine 2,3 dioxygenase-mediated tryptophan catabolism regulates accumulation of Th1/Th17 cells in the joint in collagen-induced arthritis. Arthritis Rheum. 2009;60(5):1342–1351. doi: 10.1002/art.24446. [DOI] [PubMed] [Google Scholar]
  • 232.Pertovaara M, Hasan T, Raitala A, Oja SS, Yli-Kerttula U, Korpela M, Hurme M. Indoleamine 2,3-dioxygenase activity is increased in patients with systemic lupus erythematosus and predicts disease activation in the sunny season. Clin. Exp. Immunol. 2007;150(2):274–278. doi: 10.1111/j.1365-2249.2007.03480.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Harvie MN, Campbell IT, Howell A, Thatcher N. Acceptability and tolerance of a low tyrosine and phenylalanine diet in patients with advanced cancer -- a pilot study. J. Hum. Nutr. Diet. 2002;15(3):193–202. doi: 10.1046/j.1365-277x.2002.00365.x. [DOI] [PubMed] [Google Scholar]
  • 234.Matar P, Rozados VR, Gervasoni SI, Scharovsky GO. Th2/Th1 switch induced by a single low dose of cyclophosphamide in a rat metastatic lymphoma model. Cancer Immunol. Immunother. 2002;50(11):588–596. doi: 10.1007/s00262-001-0237-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Fishman MA, Perelson AS. Th1/Th2 cross regulation. J. Theor. Biol. 1994;170(1):25–56. doi: 10.1006/jtbi.1994.1166. [DOI] [PubMed] [Google Scholar]
  • 236.Ader R, Felten DL, Cohen N. Psychoneuroimmunology. San Diego California, USA: Academic Press; 2001. [Google Scholar]
  • 237.Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, Lanctot KL. A meta-analysis of cytokines in major depression. Biol. Psychiatry. 2010;67(5):446–457. doi: 10.1016/j.biopsych.2009.09.033. [DOI] [PubMed] [Google Scholar]
  • 238.Schlatter J, Ortuno F, Cervera-Enguix S. Monocytic parameters in patients with dysthymia versus major depression. J. Affect. Disord. 2004;78(3):243–247. doi: 10.1016/S0165-0327(02)00316-6. [DOI] [PubMed] [Google Scholar]
  • 239.Maes M. Major depression and activation of the inflammatory response system. Adv. Exp. Med. Biol. 1999;461:25–46. doi: 10.1007/978-0-585-37970-8_2. [DOI] [PubMed] [Google Scholar]
  • 240.Lanquillon S, Krieg JC, Bening-Abu-Shach U, Vedder H. Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology. 2000;22(4):370–379. doi: 10.1016/S0893-133X(99)00134-7. [DOI] [PubMed] [Google Scholar]
  • 241.Axelson DA, Doraiswamy PM, McDonald WM, Boyko OB, Tupler LA, Patterson LJ, Nemeroff CB, Ellinwood EH, Jr, Krishnan KR. Hypercortisolemia and hippocampal changes in depression. Psychiatry Res. 1993;47(2):163–173. doi: 10.1016/0165-1781(93)90046-j. [DOI] [PubMed] [Google Scholar]
  • 242.Cleare AJ. Reduced whole blood serotonin in major depression. Depress Anxiety. 1997;5(2):108–111. doi: 10.1002/(sici)1520-6394(1997)5:2<108::aid-da8>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  • 243.Leonard BE. Psychopathology of depression. Drugs Today (Barc) 2007;43(10):705–716. doi: 10.1358/dot.2007.43.10.1122223. [DOI] [PubMed] [Google Scholar]
  • 244.Schiepers OJ, Wichers MC, Maes M. Cytokines and major depression. Prog Neuropsychopharmacol. Biol. Psychiatry. 2005;29(2):201–217. doi: 10.1016/j.pnpbp.2004.11.003. [DOI] [PubMed] [Google Scholar]
  • 245.Aihara M, Ida I, Yuuki N, Oshima A, Kumano H, Takahashi K, Fukuda M, Oriuchi N, Endo K, Matsuda H, Mikuni M. HPA axis dysfunction in unmedicated major depressive disorder and its normalization by pharmacotherapy correlates with alteration of neural activity in prefrontal cortex and limbic/paralimbic regions. Psychiatry Res. 2007;155(3):245–256. doi: 10.1016/j.pscychresns.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 246.Holsboer F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology. 2000;23(5):477–501. doi: 10.1016/S0893-133X(00)00159-7. [DOI] [PubMed] [Google Scholar]
  • 247.Pariante CM, Miller AH. Glucocorticoid receptors in major depression: relevance to pathophysiology and treatment. Biol. Psychiatry. 2001;49(5):391–404. doi: 10.1016/s0006-3223(00)01088-x. [DOI] [PubMed] [Google Scholar]
  • 248.Claes S. Glucocorticoid receptor polymorphisms in major depression. Ann. N. Y. Acad. Sci. 2009;1179:216–228. doi: 10.1111/j.1749-6632.2009.05012.x. [DOI] [PubMed] [Google Scholar]
  • 249.Sapolsky RM, Plotsky PM. Hypercortisolism and its possible neural bases. Biol. Psychiatry. 1990;27(9):937–952. doi: 10.1016/0006-3223(90)90032-w. [DOI] [PubMed] [Google Scholar]
  • 250.Elenkov IJ. Glucocorticoids and the Th1/Th2 balance. Ann. N. Y. Acad. Sci. 2004;1024:138–146. doi: 10.1196/annals.1321.010. [DOI] [PubMed] [Google Scholar]
  • 251.Pavon L, Sandoval-Lopez G, Eugenia Hernandez M, Loria F, Estrada I, Perez M, Moreno J, Avila U, Leff P, Anton B, Heinze G. Th2 cytokine response in Major Depressive Disorder patients before treatment. J. Neuroimmunol. 2006;172(1-2):156–165. doi: 10.1016/j.jneuroim.2005.08.014. [DOI] [PubMed] [Google Scholar]
  • 252.Hernandez ME, Mendieta D, Martinez-Fong D, Loria F, Moreno J, Estrada I, Bojalil R, Pavon L. Variations in circulating cytokine levels during 52 week course of treatment with SSRI for major depressive disorder. Eur. Neuropsychopharmacol. 2008;18(12):917–924. doi: 10.1016/j.euroneuro.2008.08.001. [DOI] [PubMed] [Google Scholar]
  • 253.Legros S, Mendlewicz J, Wybran J. Immunoglobulins, autoantibodies and other serum protein fractions in psychiatric disorders. Eur. Arch. Psychiatry Neurol. Sci. 1985;235(1):9–11. doi: 10.1007/BF00380962. [DOI] [PubMed] [Google Scholar]
  • 254.Zorrilla EP, Luborsky L, McKay JR, Rosenthal R, Houldin A, Tax A, McCorkle R, Seligman DA, Schmidt K. The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav. Immun. 2001;15(3):199–226. doi: 10.1006/brbi.2000.0597. [DOI] [PubMed] [Google Scholar]
  • 255.Cohen S, Janicki-Deverts D, Miller GE. Psychological stress and disease. JAMA. 2007;298(14):1685–1687. doi: 10.1001/jama.298.14.1685. [DOI] [PubMed] [Google Scholar]
  • 256.Lopez JF, Chalmers DT, Little KY, Watson SJ. Bennett Research Award. Regulation of serotonin1A, glucocorticoid, and mineralocorticoid receptor in rat and human hippocampus: implications for the neurobiology of depression. Biol. Psychiatry. 1998;43(8):547–573. doi: 10.1016/s0006-3223(97)00484-8. [DOI] [PubMed] [Google Scholar]
  • 257.Gorman JM, Docherty JP. A hypothesized role for dendritic remodeling in the etiology of mood and anxiety disorders. J. Neuropsychiatry Clin. Neurosci. 2010;22(3):256–264. doi: 10.1176/jnp.2010.22.3.256. [DOI] [PubMed] [Google Scholar]
  • 258.Fernandez SP, Gaspar P. Investigating anxiety and depressive-like phenotypes in genetic mouse models of serotonin depletion. Neuropharmacology. 2012;62(1):144–154. doi: 10.1016/j.neuropharm.2011.08.049. [DOI] [PubMed] [Google Scholar]
  • 259.Gill J, Vythilingam M, Page GG. Low cortisol, high DHEA, and high levels of stimulated TNF-alpha, and IL-6 in women with PTSD. J. Trauma Stress. 2008;21(6):530–539. doi: 10.1002/jts.20372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Sutherland AG, Alexander DA, Hutchison JD. Disturbance of pro-inflammatory cytokines in post-traumatic psychopathology. Cytokine. 2003;24(5):219–225. doi: 10.1016/j.cyto.2003.09.004. [DOI] [PubMed] [Google Scholar]
  • 261.Konuk N, Tekin IO, Ozturk U, Atik L, Atasoy N, Bektas S, Erdogan A. Plasma levels of tumor necrosis factor-alpha and interleukin-6 in obsessive compulsive disorder. Mediat. Inflamm. 2007;2007:65704. doi: 10.1155/2007/65704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Rook GA, Zumla A. Gulf War syndrome: is it due to a systemic shift in cytokine balance towards a Th2 profile? Lancet. 1997;349(9068):1831–1833. doi: 10.1016/S0140-6736(97)01164-1. [DOI] [PubMed] [Google Scholar]
  • 263.Cramer AO, Waldorp LJ, van der Maas HL, Borsboom D. Comorbidity: a network perspective. Behav. Brain Sci. 2010;33(2-3):137–150. doi: 10.1017/S0140525X09991567. discussion 150-193. [DOI] [PubMed] [Google Scholar]
  • 264.Boden JM, Fergusson DM, Horwood LJ. Cigarette smoking and depression: tests of causal linkages using a longitudinal birth cohort. Br. J. Psychiatry. 2010;196(6):440–446. doi: 10.1192/bjp.bp.109.065912. [DOI] [PubMed] [Google Scholar]
  • 265.Coryell W, Young E, Carroll B. Hyperactivity of the hypothalamic-pituitary-adrenal axis and mortality in major depressive disorder. Psychiatry Res. 2006;142(1):99–104. doi: 10.1016/j.psychres.2005.08.009. [DOI] [PubMed] [Google Scholar]
  • 266.Vgontzas AN, Papanicolaou DA, Bixler EO, Hopper K, Lotsikas A, Lin HM, Kales A, Chrousos GP. Sleep apnea and daytime sleepiness and fatigue: relation to visceral obesity, insulin resistance, and hypercytokinemia. J. Clin. Endocrinol. Metab. 2000;85(3):1151–1158. doi: 10.1210/jcem.85.3.6484. [DOI] [PubMed] [Google Scholar]
  • 267.Maes M. Depression is an inflammatory disease, but cell-mediated immune activation is the key component of depression. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2011;35(3):664–75. doi: 10.1016/j.pnpbp.2010.06.014. [DOI] [PubMed] [Google Scholar]
  • 268.Gabbay V, Klein RG, Alonso CM, Babb JS, Nishawala M, De Jesus G, Hirsch GS, Hottinger-Blanc PM, Gonzalez CJ. Immune system dysregulation in adolescent major depressive disorder. J. Affect. Disord. 2009;115(1-2):177–182. doi: 10.1016/j.jad.2008.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Wong ML, Kling MA, Munson PJ, Listwak S, Licinio J, Prolo P, Karp B, McCutcheon IE, Geracioti TD, Jr, DeBellis MD, Rice KC, Goldstein DS, Veldhuis JD, Chrousos GP, Oldfield EH, McCann SM, Gold PW. Pronounced and sustained central hypernoradrenergic function in major depression with melancholic features: relation to hypercortisolism and corticotropin-releasing hormone. Proc. Natl. Acad. Sci. U. S. A. 2000;97(1):325–330. doi: 10.1073/pnas.97.1.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Gold PW, Chrousos GP. The endocrinology of melancholic and atypical depression: relation to neurocircuitry and somatic consequences. Proc. Assoc. Am. Phys. 1999;111(1):22–34. doi: 10.1046/j.1525-1381.1999.09423.x. [DOI] [PubMed] [Google Scholar]
  • 271.Gold PW, Chrousos GP. Organization of the stress system and its dysregulation in melancholic and atypical depression: high vs low CRH/NE states. Mol. Psychiatry. 2002;7(3):254–275. doi: 10.1038/sj.mp.4001032. [DOI] [PubMed] [Google Scholar]
  • 272.Perugi G, Fornaro M, Akiskal HS. Are atypical depression, borderline personality disorder and bipolar II disorder overlapping manifestations of a common cyclothymic diathesis? World Psychiatry. 2011;10(1):45–51. doi: 10.1002/j.2051-5545.2011.tb00013.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Brietzke E, Stertz L, Fernandes BS, Kauer-Sant'anna M, Mascarenhas M, Escosteguy Vargas A, Chies JA, Kapczinski F. Comparison of cytokine levels in depressed, manic and euthymic patients with bipolar disorder. J. Affect. Disord. 2009;116(3):214–217. doi: 10.1016/j.jad.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 274.Liu HC, Yang YY, Chou YM, Chen KP, Shen WW, Leu SJ. Immunologic variables in acute mania of bipolar disorder. J. Neuroimmunol. 2004;150(1-2):116–122. doi: 10.1016/j.jneuroim.2004.01.006. [DOI] [PubMed] [Google Scholar]
  • 275.Kunz M, Cereser KM, Goi PD, Fries GR, Teixeira AL, Fernandes BS, Belmonte-de-Abreu PS, Kauer-Sant'anna M, Kapczinski F, Gama CS. Serum levels of IL-6, IL-10 and TNF-alpha in patients with bipolar disorder and schizophrenia: differences in pro- and anti-inflammatory balance. Rev. Bras Psiquiatr. 2011;33(3):368–74. doi: 10.1590/s1516-44462011000300010. [DOI] [PubMed] [Google Scholar]
  • 276.Garver DL, Tamas RL, Holcomb JA. Elevated interleukin-6 in the cerebrospinal fluid of a previously delineated schizophrenia subtype. Neuropsychopharmacology. 2003;28(8):1515–1520. doi: 10.1038/sj.npp.1300217. [DOI] [PubMed] [Google Scholar]
  • 277.Mittleman BB, Castellanos FX, Jacobsen LK, Rapoport JL, Swedo SE, Shearer GM. Cerebrospinal fluid cytokines in pediatric neuropsychiatric disease. J. Immunol. 1997;159(6):2994–2999. [PubMed] [Google Scholar]
  • 278.Liberzon I, King AP, Britton JC, Phan KL, Abelson JL, Taylor SF. Paralimbic and medial prefrontal cortical involvement in neuroendocrine responses to traumatic stimuli. Am. J. Psychiatry. 2007;164(8):1250–1258. doi: 10.1176/appi.ajp.2007.06081367. [DOI] [PubMed] [Google Scholar]
  • 279.Cerullo MA, Adler CM, Delbello MP, Strakowski SM. The functional neuroanatomy of bipolar disorder. Int. Rev. Psychiatry. 2009;21(4):314–322. doi: 10.1080/09540260902962107. [DOI] [PubMed] [Google Scholar]
  • 280.Fountoulakis KN, Giannakopoulos P, Kovari E, Bouras C. Assessing the role of cingulate cortex in bipolar disorder: neuropathological, structural and functional imaging data. Brain Res. Rev. 2008;59(1):9–21. doi: 10.1016/j.brainresrev.2008.04.005. [DOI] [PubMed] [Google Scholar]
  • 281.Chen CH, Suckling J, Lennox BR, Ooi C, Bullmore ET. A quantitative meta-analysis of fMRI studies in bipolar disorder. Bipolar Disord. 2011;13(1):1–15. doi: 10.1111/j.1399-5618.2011.00893.x. [DOI] [PubMed] [Google Scholar]
  • 282.MacKinnon DF, Zamoiski R. Panic comorbidity with bipolar disorder: what is the manic-panic connection? Bipolar Disord. 2006;8(6):648–664. doi: 10.1111/j.1399-5618.2006.00356.x. [DOI] [PubMed] [Google Scholar]
  • 283.Dedovic K, Duchesne A, Andrews J, Engert V, Pruessner JC. The brain and the stress axis: the neural correlates of cortisol regulation in response to stress. Neuroimage. 2009;47(3):864–871. doi: 10.1016/j.neuroimage.2009.05.074. [DOI] [PubMed] [Google Scholar]
  • 284.Kenis G, Maes M. Effects of antidepressants on the production of cytokines. Int. J. Neuropsychopharmacol. 2002;5(4):401–412. doi: 10.1017/S1461145702003164. [DOI] [PubMed] [Google Scholar]
  • 285.Basterzi AD, Aydemir C, Kisa C, Aksaray S, Tuzer V, Yazici K, Goka E. IL-6 levels decrease with SSRI treatment in patients with major depression. Hum. Psychopharmacol. 2005;20(7):473–476. doi: 10.1002/hup.717. [DOI] [PubMed] [Google Scholar]
  • 286.Diamond M, Kelly JP, Connor TJ. Antidepressants suppress production of the Th1 cytokine interferon-gamma, independent of monoamine transporter blockade. Eur. Neuropsychopharmacol. 2006;16(7):481–490. doi: 10.1016/j.euroneuro.2005.11.011. [DOI] [PubMed] [Google Scholar]
  • 287.Leo R, Di Lorenzo G, Tesauro M, Razzini C, Forleo GB, Chiricolo G, Cola C, Zanasi M, Troisi A, Siracusano A, Lauro R, Romeo F. Association between enhanced soluble CD40 ligand and proinflammatory and prothrombotic states in major depressive disorder: pilot observations on the effects of selective serotonin reuptake inhibitor therapy. J. Clin. Psychiatry. 2006;67(11):1760–1766. doi: 10.4088/jcp.v67n1114. [DOI] [PubMed] [Google Scholar]
  • 288.Tsao CW, Lin YS, Chen CC, Bai CH, Wu SR. Cytokines and serotonin transporter in patients with major depression. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2006;30(5):899–905. doi: 10.1016/j.pnpbp.2006.01.029. [DOI] [PubMed] [Google Scholar]
  • 289.Nemeroff CB, Owens MJ. Pharmacologic differences among the SSRIs: focus on monoamine transporters and the HPA axis. CNS Spectr. 2004;9(6) Suppl 4:23–31. doi: 10.1017/s1092852900025475. [DOI] [PubMed] [Google Scholar]
  • 290.Blardi P, De Lalla A, Leo A, Auteri A, Iapichino S, Di Muro A, Dell'Erba A, Castrogiovanni P. Serotonin and fluoxetine levels in plasma and platelets after fluoxetine treatment in depressive patients. J. Clin. Psychopharmacol. 2002;22(2):131–136. doi: 10.1097/00004714-200204000-00005. [DOI] [PubMed] [Google Scholar]
  • 291.Castrogiovanni P, Blardi P, De Lalla A, Dell'Erba A, Auteri A. Can serotonin and fluoxetine levels in plasma and platelets predict clinical response in depression? Psychopharmacol. Bull. 2003;37(2):102–108. [PubMed] [Google Scholar]
  • 292.Blier P. Pharmacology of rapid-onset antidepressant treatment strategies. J. Clin. Psychiatry. 2001;62 (Suppl 15):12–17. [PubMed] [Google Scholar]
  • 293.El Mansari M, Sanchez C, Chouvet G, Renaud B, Haddjeri N. Effects of acute and long-term administration of escitalopram and citalopram on serotonin neurotransmission: an in vivo electrophysiological study in rat brain. Neuropsychopharmacology. 2005;30(7):1269–1277. doi: 10.1038/sj.npp.1300686. [DOI] [PubMed] [Google Scholar]
  • 294.Hensler JG. Differential regulation of 5-HT1A receptor-G protein interactions in brain following chronic antidepressant administration. Neuropsychopharmacology. 2002;26(5):565–573. doi: 10.1016/S0893-133X(01)00395-5. [DOI] [PubMed] [Google Scholar]
  • 295.Yamauchi M, Miyara T, Matsushima T, Imanishi T. Desensitization of 5-HT2A receptor function by chronic administration of selective serotonin reuptake inhibitors. Brain Res. 2006;1067(1):164–169. doi: 10.1016/j.brainres.2005.10.075. [DOI] [PubMed] [Google Scholar]
  • 296.Yamauchi M, Tatebayashi T, Nagase K, Kojima M, Imanishi T. Chronic treatment with fluvoxamine desensitizes 5-HT2C receptor-mediated hypolocomotion in rats. Pharmacol. Biochem. Behav. 2004;78(4):683–689. doi: 10.1016/j.pbb.2004.05.003. [DOI] [PubMed] [Google Scholar]
  • 297.Antonijevic IA. Depressive disorders -- is it time to endorse different pathophysiologies? Psychoneuroendocrinology. 2006;31(1):1–15. doi: 10.1016/j.psyneuen.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 298.Nikisch G, Agren H, Eap CB, Czernik A, Baumann P, Mathe AA. Neuropeptide Y and corticotropin-releasing hormone in CSF mark response to antidepressive treatment with citalopram. Int. J. Neuropsychopharmacol. 2005;8(3):403–410. doi: 10.1017/S1461145705005158. [DOI] [PubMed] [Google Scholar]
  • 299.Papiol S, Arias B, Gasto C, Gutierrez B, Catalan R, Fananas L. Genetic variability at HPA axis in major depression and clinical response to antidepressant treatment. J. Affect Disord. 2007;104(1-3):83–90. doi: 10.1016/j.jad.2007.02.017. [DOI] [PubMed] [Google Scholar]
  • 300.Taler M, Gil-Ad I, Lomnitski L, Korov I, Baharav E, Bar M, Zolokov A, Weizman A. Immunomodulatory effect of selective serotonin reuptake inhibitors (SSRIs) on human T lymphocyte function and gene expression. Eur. Neuropsychopharmacol. 2007;17(12):774–780. doi: 10.1016/j.euroneuro.2007.03.010. [DOI] [PubMed] [Google Scholar]
  • 301.Padol IT, Hunt RH. Association of myocardial infarctions with COX-2 inhibition may be related to immunomodulation towards a Th1 response resulting in atheromatous plaque instability: an evidence-based interpretation. Rheumatology (Oxford) 2010;49(5):837–843. doi: 10.1093/rheumatology/kep225. [DOI] [PubMed] [Google Scholar]
  • 302.De Berardis D, Conti CM, Serroni N, Moschetta FS, Olivieri L, Carano A, Salerno RM, Cavuto M, Farina B, Alessandrini M, Janiri L, Pozzi G, Di Giannantonio M. The effect of newer serotonin-noradrenalin antidepressants on cytokine production: a review of the current literature. Int. J. Immunopathol. Pharmacol. 2010;23(2):417–422. doi: 10.1177/039463201002300204. [DOI] [PubMed] [Google Scholar]
  • 303.Kubera M, Lin AH, Kenis G, Bosmans E, van Bockstaele D, Maes M. Anti-Inflammatory effects of antidepressants through suppression of the interferon-gamma/interleukin-10 production ratio. J. Clin. Psychopharmacol. 2001;21(2):199–206. doi: 10.1097/00004714-200104000-00012. [DOI] [PubMed] [Google Scholar]
  • 304.Lee KM, Kim YK. The role of IL-12 and TGF-beta1 in the pathophysiology of major depressive disorder. Int. Immunopharmacol. 2006;6(8):1298–1304. doi: 10.1016/j.intimp.2006.03.015. [DOI] [PubMed] [Google Scholar]
  • 305.Vollmar P, Haghikia A, Dermietzel R, Faustmann PM. Venlafaxine exhibits an anti-inflammatory effect in an inflammatory co-culture model. Int. J. Neuropsychopharmacol. 2008;11(1):111–117. doi: 10.1017/S1461145707007729. [DOI] [PubMed] [Google Scholar]
  • 306.Vollmar P, Nessler S, Kalluri SR, Hartung HP, Hemmer B. The antidepressant venlafaxine ameliorates murine experimental autoimmune encephalomyelitis by suppression of pro-inflammatory cytokines. Int. J. Neuropsychopharmacol. 2009;12(4):525–536. doi: 10.1017/S1461145708009425. [DOI] [PubMed] [Google Scholar]
  • 307.Diehl S, Rincon M. The two faces of IL-6 on Th1/Th2 differentiation. Mol. Immunol. 2002;39(9):531–536. doi: 10.1016/s0161-5890(02)00210-9. [DOI] [PubMed] [Google Scholar]
  • 308.Piletz JE, Halaris A, Iqbal O, Hoppensteadt D, Fareed J, Zhu H, Sinacore J, Lindsay Devane C. Pro-inflammatory biomakers in depression: Treatment with venlafaxine. World J. Biol. Psychiatry. 2008. pp. 1–11. [DOI] [PubMed]
  • 309.Kubera M, Kenis G, Bosmans E, Kajta M, Basta-Kaim A, Scharpe S, Budziszewska B, Maes M. Stimulatory effect of antidepressants on the production of IL-6. Int. Immunopharmacol. 2004;4(2):185–192. doi: 10.1016/j.intimp.2003.11.006. [DOI] [PubMed] [Google Scholar]
  • 310.Beique J, de Montigny C, Blier P, Debonnel G. Effects of sustained administration of the serotonin and norepinephrine reuptake inhibitor venlafaxine: I. in vivo electrophysiological studies in the rat. Neuropharmacology. 2000;39(10):1800–1812. doi: 10.1016/s0028-3908(00)00017-4. [DOI] [PubMed] [Google Scholar]
  • 311.Beique J, de Montigny C, Blier P, Debonnel G. Effects of sustained administration of the serotonin and norepinephrine reuptake inhibitor venlafaxine: II. In vitro studies in the rat. Neuropharmacology. 2000;39(10):1813–1822. doi: 10.1016/s0028-3908(00)00018-6. [DOI] [PubMed] [Google Scholar]
  • 312.Nalepa I, Manier DH, Gillespie DD, Rossby SP, Schmidt DE, Sulser F. Lack of beta adrenoceptor desensitization in brain following the dual noradrenaline and serotonin reuptake inhibitor venlafaxine. Eur. Neuropsychopharmacol. 1998;8(3):227–232. doi: 10.1016/s0924-977x(97)00078-3. [DOI] [PubMed] [Google Scholar]
  • 313.Debonnel G, Saint-Andre E, Hebert C, de Montigny C, Lavoie N, Blier P. Differential physiological effects of a low dose and high doses of venlafaxine in major depression. Int. J. Neuropsychopharmacol. 2007;10(1):51–61. doi: 10.1017/S1461145705006413. [DOI] [PubMed] [Google Scholar]
  • 314.Trivedi MH, Desaiah D, Ossanna MJ, Pritchett YL, Brannan SK, Detke MJ. Clinical evidence for serotonin and norepinephrine reuptake inhibition of duloxetine. Int. Clin. Psychopharmacol. 2008;23(3):161–169. doi: 10.1097/YIC.0b013e3282f41d7e. [DOI] [PubMed] [Google Scholar]
  • 315.Rueter LE, Kasamo K, de Montigny C, Blier P. Effect of long-term administration of duloxetine on the function of serotonin and noradrenaline terminals in the rat brain. Naunyn Schmiedebergs Arch. Pharmacol. 1998;357(6):600–610. doi: 10.1007/pl00005214. [DOI] [PubMed] [Google Scholar]
  • 316.Rueter LE, De Montigny C, Blier P. Electrophysiological characterization of the effect of long-term duloxetine administration on the rat serotonergic and noradrenergic systems. J. Pharmacol. Exp. Ther. 1998;285(2):404–412. [PubMed] [Google Scholar]
  • 317.Invernizzi RW, Garattini S. Role of presynaptic alpha2-adrenoceptors in antidepressant action: recent findings from microdialysis studies. Prog Neuropsychopharmacol. Biol. Psychiatry. 2004;28(5):819–827. doi: 10.1016/j.pnpbp.2004.05.026. [DOI] [PubMed] [Google Scholar]
  • 318.Fornaro M, Martino M, Battaglia F, Colicchio S, Perugi G. Increase in IL-6 levels among major depressive disorder patients after a 6-week treatment with duloxetine 60 mg/day: a preliminary observation. Neuropsychiatr. Dis. Treat. 2011;7:51–56. doi: 10.2147/NDT.S16382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.McNamee EN, Ryan KM, Griffin EW, Gonzalez-Reyes RE, Ryan KJ, Harkin A, Connor TJ. Noradrenaline acting at central beta-adrenoceptors induces interleukin-10 and suppressor of cytokine signaling-3 expression in rat brain: implications for neurodegeneration. Brain Behav. Immun. 2010;24(4):660–671. doi: 10.1016/j.bbi.2010.02.005. [DOI] [PubMed] [Google Scholar]
  • 320.McNamee EN, Griffin EW, Ryan KM, Ryan KJ, Heffernan S, Harkin A, Connor TJ. Noradrenaline acting at beta-adrenoceptors induces expression of IL-1beta and its negative regulators IL-1ra and IL-1RII, and drives an overall anti-inflammatory phenotype in rat cortex. Neuropharmacology. 2010;59(1-2):37–48. doi: 10.1016/j.neuropharm.2010.03.014. [DOI] [PubMed] [Google Scholar]
  • 321.Hashioka S, Klegeris A, Monji A, Kato T, Sawada M, McGeer PL, Kanba S. Antidepressants inhibit interferon-gamma-induced microglial production of IL-6 and nitric oxide. Exp. Neurol. 2007;206(1):33–42. doi: 10.1016/j.expneurol.2007.03.022. [DOI] [PubMed] [Google Scholar]
  • 322.O'Sullivan JB, Ryan KM, Curtin NM, Harkin A, Connor TJ. Noradrenaline reuptake inhibitors limit neuroinflammation in rat cortex following a systemic inflammatory challenge: implications for depression and neurodegeneration. Int. J. Neuropsychopharmacol. 2009;12(5):687–699. doi: 10.1017/S146114570800967X. [DOI] [PubMed] [Google Scholar]
  • 323.Agelink MW, Ullrich H, Baumann B, Strum S, Majewski T. Effects of reboxetine, a selective norepinephrine reuptake inhibitor, on sympathetic and parasympathetic outflow to the heart: preliminary data. Psychopharmacology (Berl) 2002;163(2):151–156. doi: 10.1007/s00213-002-1146-7. [DOI] [PubMed] [Google Scholar]
  • 324.Parini S, Renoldi G, Battaglia A, Invernizzi RW. Chronic reboxetine desensitizes terminal but not somatodendritic alpha2-adrenoceptors controlling noradrenaline release in the rat dorsal hippocampus. Neuropsychopharmacology. 2005;30(6):1048–1055. doi: 10.1038/sj.npp.1300661. [DOI] [PubMed] [Google Scholar]
  • 325.Rogoz Z, Margas W, Skuza G, Solich J, Kusmider M, Dziedzicka-Wasylewska M. Effect of repeated treatment with reboxetine on the central alpha 1-adrenergic and dopaminergic receptors. Pol. J. Pharmacol. 2002;54(6):593–603. [PubMed] [Google Scholar]
  • 326.Rogoz Z, Kolasiewicz W. Effect of repeated treatment with reboxetine on the central alpha1-adrenergic system. Pol. J. Pharmacol. 2001;53(6):663–667. [PubMed] [Google Scholar]
  • 327.Gould GG, Pardon MC, Morilak DA, Frazer A. Regulatory effects of reboxetine treatment alone, or following paroxetine treatment, on brain noradrenergic and serotonergic systems. Neuropsychopharmacology. 2003;28(9):1633–1641. doi: 10.1038/sj.npp.1300236. [DOI] [PubMed] [Google Scholar]
  • 328.Holoubek G, Noldner M, Treiber K, Muller WE. Effect of chronic antidepressant treatment on beta-receptor coupled signal transduction cascade. Which effect matters most? Pharmacopsychiatry. 2004;37(Suppl 2):S113–119. doi: 10.1055/s-2004-832664. [DOI] [PubMed] [Google Scholar]
  • 329.Linner L, Endersz H, Ohman D, Bengtsson F, Schalling M, Svensson TH. Reboxetine modulates the firing pattern of dopamine cells in the ventral tegmental area and selectively increases dopamine availability in the prefrontal cortex. J. Pharmacol. Exp. Ther. 2001;297(2):540–546. [PubMed] [Google Scholar]
  • 330.Page ME, Lucki I. Effects of acute and chronic reboxetine treatment on stress-induced monoamine efflux in the rat frontal cortex. Neuropsychopharmacology. 2002;27(2):237–247. doi: 10.1016/S0893-133X(02)00301-9. [DOI] [PubMed] [Google Scholar]
  • 331.Szabo ST, Blier P. Effects of the selective norepinephrine reuptake inhibitor reboxetine on norepinephrine and serotonin transmission in the rat hippocampus. Neuropsychopharmacology. 2001;25(6):845–857. doi: 10.1016/S0893-133X(01)00284-6. [DOI] [PubMed] [Google Scholar]
  • 332.Miller DK, Wong EH, Chesnut MD, Dwoskin LP. Reboxetine: functional inhibition of monoamine transporters and nicotinic acetylcholine receptors. J. Pharmacol. Exp. Ther. 2002;302(2):687–695. doi: 10.1124/jpet.302.2.687. [DOI] [PubMed] [Google Scholar]
  • 333.Tzavara ET, Bymaster FP, Overshiner CD, Davis RJ, Perry KW, Wolff M, McKinzie DL, Witkin JM, Nomikos GG. Procholinergic and memory enhancing properties of the selective norepinephrine uptake inhibitor atomoxetine. Mol. Psychiatry. 2006;11(2):187–195. doi: 10.1038/sj.mp.4001763. [DOI] [PubMed] [Google Scholar]
  • 334.Schule C, Baghai T, Schmidbauer S, Bidlingmaier M, Strasburger CJ, Laakmann G. Reboxetine acutely stimulates cortisol, ACTH, growth hormone and prolactin secretion in healthy male subjects. Psychoneuroendocrinology. 2004;29(2):185–200. doi: 10.1016/s0306-4530(03)00022-2. [DOI] [PubMed] [Google Scholar]
  • 335.Schule C, Baghai T, Laakmann G. Mirtazapine decreases stimulatory effects of reboxetine on cortisol, adrenocorticotropin and prolactin secretion in healthy male subjects. Neuroendocrinology. 2004;79(1):54–62. doi: 10.1159/000076046. [DOI] [PubMed] [Google Scholar]
  • 336.Brustolim D, Ribeiro-dos-Santos R, Kast RE, Altschuler EL, Soares MB. A new chapter opens in anti-inflammatory treatments: the antidepressant bupropion lowers production of tumor necrosis factor-alpha and interferon-gamma in mice. Int. Immunopharmacol. 2006;6(6):903–907. doi: 10.1016/j.intimp.2005.12.007. [DOI] [PubMed] [Google Scholar]
  • 337.Eller T, Vasar V, Shlik J, Maron E. Effects of bupropion augmentation on pro-inflammatory cytokines in escitalopram-resistant patients with major depressive disorder. J. Psychopharmacol. 2009;23(7):854–858. doi: 10.1177/0269881108091077. [DOI] [PubMed] [Google Scholar]
  • 338.Stahl SM, Pradko JF, Haight BR, Modell JG, Rockett CB, Learned-Coughlin S. A Review of the Neuropharmacology of Bupropion, a Dual Norepinephrine and Dopamine Reuptake Inhibitor. Prim. Care Companion J. Clin. Psychiatry. 2004;6(4):159–166. doi: 10.4088/pcc.v06n0403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Ferris RM, Beaman OJ. Bupropion: a new antidepressant drug, the mechanism of action of which is not associated with down-regulation of postsynaptic beta-adrenergic, serotonergic (5-HT2), alpha 2-adrenergic, imipramine and dopaminergic receptors in brain. Neuropharmacology. 1983;22(11):1257–1267. doi: 10.1016/0028-3908(83)90198-3. [DOI] [PubMed] [Google Scholar]
  • 340.Arias HR. Is the inhibition of nicotinic acetylcholine receptors by bupropion involved in its clinical actions? Int. J. Biochem. Cell Biol. 2009;41(11):2098–2108. doi: 10.1016/j.biocel.2009.05.015. [DOI] [PubMed] [Google Scholar]
  • 341.Arias HR, Gumilar F, Rosenberg A, Targowska-Duda KM, Feuerbach D, Jozwiak K, Moaddel R, Wainer IW, Bouzat C. Interaction of bupropion with muscle-type nicotinic acetylcholine receptors in different conformational states. Biochemistry. 2009;48(21):4506–4518. doi: 10.1021/bi802206k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Kraus T, Haack M, Schuld A, Hinze-Selch D, Koethe D, Pollmacher T. Body weight, the tumor necrosis factor system, and leptin production during treatment with mirtazapine or venlafaxine. Pharmacopsychiatry. 2002;35(6):220–225. doi: 10.1055/s-2002-36390. [DOI] [PubMed] [Google Scholar]
  • 343.Kubera M, Roman A, Basta-Kaim A, Budziszewska B, Zajicova A, Holan V, Rogoz Z, Skuza G, Leskiewicz M, Regulska M, Jagla G, Nowak W, Lason W. Effect of acute and repeated treatment with mirtazapine on the immunity of noradrenaline transporter knockout C57BL/6J mice. Pharmacol. Biochem. Behav. 2006;85(4):813–819. doi: 10.1016/j.pbb.2006.11.017. [DOI] [PubMed] [Google Scholar]
  • 344.Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 2001;7(3):249–264. doi: 10.1111/j.1527-3458.2001.tb00198.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Haddjeri N, Blier P, de Montigny C. Effects of long-term treatment with the alpha 2-adrenoceptor antagonist mirtazapine on 5-HT neurotransmission. Naunyn Schmiedebergs Arch. Pharmacol. 1997;355(1):20–29. doi: 10.1007/pl00004913. [DOI] [PubMed] [Google Scholar]
  • 346.Haddjeri N, Blier P, de Montigny C. Acute and long-term actions of the antidepressant drug mirtazapine on central 5-HT neurotransmission. J. Affect. Disord. 1998;51(3):255–266. doi: 10.1016/s0165-0327(98)00223-7. [DOI] [PubMed] [Google Scholar]
  • 347.de Boer T. The effects of mirtazapine on central noradrenergic and serotonergic neurotransmission. Int. Clin. Psychopharmacol. 1995;10(Suppl 4):19–23. doi: 10.1097/00004850-199512004-00004. [DOI] [PubMed] [Google Scholar]
  • 348.Kast RE. Anti- and pro-inflammatory considerations in antidepressant use during medical illness: bupropion lowers and mirtazapine increases circulating tumor necrosis factor-alpha levels. Gen. Hosp. Psychiatry. 2003;25(6):495–496. doi: 10.1016/s0163-8343(03)00093-8. [DOI] [PubMed] [Google Scholar]
  • 349.Millan MJ, Gobert A, Rivet JM, Adhumeau-Auclair A, Cussac D, Newman-Tancredi A, Dekeyne A, Nicolas JP, Lejeune F. Mirtazapine enhances frontocortical dopaminergic and corticolimbic adrenergic, but not serotonergic, transmission by blockade of alpha2-adrenergic and serotonin2C receptors: a comparison with citalopram. Eur. J. Neurosci. 2000;12(3):1079–1095. doi: 10.1046/j.1460-9568.2000.00982.x. [DOI] [PubMed] [Google Scholar]
  • 350.Schmid DA, Wichniak A, Uhr M, Ising M, Brunner H, Held K, Weikel JC, Sonntag A, Steiger A. Changes of sleep architecture, spectral composition of sleep EEG, the nocturnal secretion of cortisol, ACTH, GH, prolactin, melatonin, ghrelin, and leptin, and the DEX-CRH test in depressed patients during treatment with mirtazapine. Neuropsychopharmacology. 2006;31(4):832–844. doi: 10.1038/sj.npp.1300923. [DOI] [PubMed] [Google Scholar]
  • 351.Stahl SM. Mechanism of action of trazodone: a multifunctional drug. CNS Spectr. 2009;14(10):536–546. doi: 10.1017/s1092852900024020. [DOI] [PubMed] [Google Scholar]
  • 352.Shen KP, Lo YC, Yang RC, Liu HW, Chen IJ, Wu BN. Antioxidant eugenosedin-A protects against lipopolysaccharide-induced hypotension, hyperglycaemia and cytokine immunoreactivity in rats and mice. J. Pharm. Pharmacol. 2005;57(1):117–125. doi: 10.1211/0022357055137. [DOI] [PubMed] [Google Scholar]
  • 353.Maestro S, Casella C, Milone A, Muratori F, Palacio-Espasa F. Study of the onset of autism through home movies. Psychopathology. 1999;32(6):292–300. doi: 10.1159/000029102. [DOI] [PubMed] [Google Scholar]
  • 354.San L, Arranz B. Agomelatine: a novel mechanism of antidepressant action involving the melatonergic and the serotonergic system. Eur. Psychiatry. 2008;23(6):396–402. doi: 10.1016/j.eurpsy.2008.04.002. [DOI] [PubMed] [Google Scholar]
  • 355.Carrillo-Vico A, Garcia-Maurino S, Calvo JR, Guerrero JM. Melatonin counteracts the inhibitory effect of PGE2 on IL-2 production in human lymphocytes via its mt1 membrane receptor. FASEB J. 2003;17(6):755–757. doi: 10.1096/fj.02-0501fje. [DOI] [PubMed] [Google Scholar]
  • 356.Leproult R, Van Onderbergen A, L'Hermite-Baleriaux M, Van Cauter E, Copinschi G. Phase-shifts of 24-h rhythms of hormonal release and body temperature following early evening administration of the melatonin agonist agomelatine in healthy older men. Clin. Endocrinol. (Oxf) 2005;63(3):298–304. doi: 10.1111/j.1365-2265.2005.02341.x. [DOI] [PubMed] [Google Scholar]
  • 357.Hanoun N, Mocaer E, Boyer PA, Hamon M, Lanfumey L. Differential effects of the novel antidepressant agomelatine (S 20098) versus fluoxetine on 5-HT1A receptors in the rat brain. Neuropharmacology. 2004;47(4):515–526. doi: 10.1016/j.neuropharm.2004.06.003. [DOI] [PubMed] [Google Scholar]
  • 358.Ying SW, Rusak B, Mocaer E. Chronic exposure to melatonin receptor agonists does not alter their effects on suprachiasmatic nucleus neurons. Eur. J. Pharmacol. 1998;342(1):29–37. doi: 10.1016/s0014-2999(97)01443-x. [DOI] [PubMed] [Google Scholar]
  • 359.Xia Z, DePierre JW, Nassberger L. Tricyclic antidepressants inhibit IL-6, IL-1 beta and TNF-alpha release in human blood monocytes and IL-2 and interferon-gamma in T cells. Immunopharmacology. 1996;34(1):27–37. doi: 10.1016/0162-3109(96)00111-7. [DOI] [PubMed] [Google Scholar]
  • 360.Braeckman L, De Bacquer D, Maes L, De Backer G. Effects of a low-intensity worksite-based nutrition intervention. Occup. Med. (Lond) 1999;49(8):549–555. doi: 10.1093/occmed/49.8.549. [DOI] [PubMed] [Google Scholar]
  • 361.Huang YY, Peng CH, Yang YP, Wu CC, Hsu WM, Wang HJ, Chan KH, Chou YP, Chen SJ, Chang YL. Desipramine activated Bcl-2 expression and inhibited lipopolysaccharide-induced apoptosis in hippocampus-derived adult neural stem cells. J. Pharmacol. Sci. 2007;104(1):61–72. doi: 10.1254/jphs.fp0061255. [DOI] [PubMed] [Google Scholar]
  • 362.Tai YH, Wang YH, Wang JJ, Tao PL, Tung CS, Wong CS. Amitriptyline suppresses neuroinflammation and up-regulates glutamate transporters in morphine-tolerant rats. Pain. 2006;124(1-2):77–86. doi: 10.1016/j.pain.2006.03.018. [DOI] [PubMed] [Google Scholar]
  • 363.Budziszewska B, Basta-Kaim A, Kubera M, Jaworska L, Leskiewicz M, Tetich M, Otczyk M, Zajicova A, Holan V, Lason W. Effect of lipopolysaccharide and antidepressant drugs on glucocorticoid receptor-mediated gene transcription. Pharmacol. Rep. 2005;57(4):540–544. [PubMed] [Google Scholar]
  • 364.Wang W, Danielsson A, Svanberg E, Lundholm K. Lack of effects by tricyclic antidepressant and serotonin inhibitors on anorexia in MCG 101 tumor-bearing mice with eicosanoid-related cachexia. Nutrition. 2003;19(1):47–53. doi: 10.1016/s0899-9007(02)00921-8. [DOI] [PubMed] [Google Scholar]
  • 365.Stahl SM. Basic psychopharmacology of antidepressants, part 1: Antidepressants have seven distinct mechanisms of action. J. Clin. Psychiatry. 1998;59(Suppl 4):5–14. [PubMed] [Google Scholar]
  • 366.Shen Y, Connor TJ, Nolan Y, Kelly JP, Leonard BE. Differential effect of chronic antidepressant treatments on lipopolysaccharide-induced depressive-like behavioural symptoms in the rat. Life Sci. 1999;65(17):1773–1786. doi: 10.1016/s0024-3205(99)00430-0. [DOI] [PubMed] [Google Scholar]
  • 367.Ekuni D, Firth JD, Nayer T, Tomofuji T, Sanbe T, Irie K, Yamamoto T, Oka T, Liu Z, Vielkind J, Putnins EE. Lipopolysaccharide-induced epithelial monoamine oxidase mediates alveolar bone loss in a rat chronic wound model. Am. J. Pathol. 2009;175(4):1398–1409. doi: 10.2353/ajpath.2009.090108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Lin A, Song C, Kenis G, Bosmans E, De Jongh R, Scharpe S, Maes M. The in vitro immunosuppressive effects of moclobemide in healthy volunteers. J. Affect. Disord. 2000;58(1):69–74. doi: 10.1016/s0165-0327(99)00076-2. [DOI] [PubMed] [Google Scholar]
  • 369.Muller T, Kuhn W, Kruger R, Przuntek H. Selegiline as immunostimulant--a novel mechanism of action? J. Neural. Transm. Suppl. 1998;52:321–328. doi: 10.1007/978-3-7091-6499-0_33. [DOI] [PubMed] [Google Scholar]
  • 370.Garrick NA, Scheinin M, Chang WH, Linnoila M, Murphy DL. Differential effects of clorgyline on catecholamine and indoleamine metabolites in the cerebrospinal fluid of rhesus monkeys. Biochem. Pharmacol. 1984;33(9):1423–1427. doi: 10.1016/0006-2952(84)90408-8. [DOI] [PubMed] [Google Scholar]
  • 371.Gerlach M, Youdim MB, Riederer P. Pharmacology of selegiline. Neurology. 1996;47(6) Suppl 3:S137–145. doi: 10.1212/wnl.47.6_suppl_3.137s. [DOI] [PubMed] [Google Scholar]
  • 372.Kast RE, Altschuler EL. Remission of Crohn's disease on bupropion. Gastroenterology. 2001;121(5):1260–1261. doi: 10.1053/gast.2001.29467. [DOI] [PubMed] [Google Scholar]
  • 373.Kane S, Altschuler EL, Kast RE. Crohn's disease remission on bupropion. Gastroenterology. 2003;125(4):1290. doi: 10.1016/j.gastro.2003.02.004. [DOI] [PubMed] [Google Scholar]
  • 374.Kast RE. Crohn's disease remission with phenelzine treatment. Gastroenterology. 1998;115(4):1034–1035. doi: 10.1016/s0016-5085(98)70292-6. [DOI] [PubMed] [Google Scholar]
  • 375.Scott MA, Letrent KJ, Hager KL, Burch JL. Use of transdermal amitriptyline gel in a patient with chronic pain and depression. Pharmacotherapy. 1999;19(2):236–239. doi: 10.1592/phco.19.3.236.30922. [DOI] [PubMed] [Google Scholar]
  • 376.Mikocka-Walus AA, Turnbull DA, Moulding NT, Wilson IG, Andrews JM, Holtmann GJ. Antidepressants and inflammatory bowel disease: a systematic review. Clin. Pract. Epidemiol. Ment. Health. 2006;2:24. doi: 10.1186/1745-0179-2-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Torras BMT, Pardo FC, Bernad G JA, Roig GI, Pedregosa DMM, Plans SE. Three cases of intestinal inflammatory disease caused by anti-depressant treatment with paroxetine. Aten Primaria. 2003;31(2):135. doi: 10.1016/S0212-6567(03)79153-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Modell JG, Boyce S, Taylor E, Katholi C. Treatment of atopic dermatitis and psoriasis vulgaris with bupropion-SR: a pilot study. Psychosom. Med. 2002;64(5):835–840. doi: 10.1097/01.psy.0000021954.59258.9b. [DOI] [PubMed] [Google Scholar]
  • 379.Alpsoy E, Ozcan E, Cetin L, Ozgur O, Er H, Yilmaz E, Karaman T. Is the efficacy of topical corticosteroid therapy for psoriasis vulgaris enhanced by concurrent moclobemide therapy? A double-blind, placebo-controlled study. J. Am. Acad. Dermatol. 1998;38(2 Pt 1):197–200. doi: 10.1016/s0190-9622(98)70240-3. [DOI] [PubMed] [Google Scholar]
  • 380.Gupta MA, Gupta AK. The use of psychotropic drugs in dermatology. Dermatol. Clin. 2000;18(4):711–725. doi: 10.1016/s0733-8635(05)70222-9. x. [DOI] [PubMed] [Google Scholar]
  • 381.Osborne SF, Stafford L, Orr KG. Paroxetine-associated psoriasis. Am. J. Psychiatry. 2002;159(12):2113. doi: 10.1176/appi.ajp.159.12.2113. [DOI] [PubMed] [Google Scholar]
  • 382.Tamer E, Gur G, Polat M, Alli N. Flare-up of pustular psoriasis with fluoxetine: possibility of a serotoninergic influence? J. Dermatol. Treat. 2009;20(3):1–3. doi: 10.1080/09546630802449096. [DOI] [PubMed] [Google Scholar]
  • 383.Wagena EJ, Huibers MJ, van Schayck CP. Antidepressants in the treatment of patients with COPD: possible associations between smoking cigarettes, COPD and depression. Thorax. 2001;56(8):587–588. doi: 10.1136/thorax.56.8.587a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Borson S, McDonald GJ, Gayle T, Deffebach M, Lakshminarayan S, VanTuinen C. Improvement in mood, physical symptoms, and function with nortriptyline for depression in patients with chronic obstructive pulmonary disease. Psychosomatics. 1992;33(2):190–201. doi: 10.1016/S0033-3182(92)71995-1. [DOI] [PubMed] [Google Scholar]
  • 385.Smoller JW, Otto MW. Panic, dyspnea, and asthma. Curr. Opin. Pulm. Med. 1998;4(1):40–45. doi: 10.1097/00063198-199801000-00008. [DOI] [PubMed] [Google Scholar]
  • 386.Brown ES, Vigil L, Khan DA, Liggin JD, Carmody TJ, Rush AJ. A randomized trial of citalopram versus placebo in outpatients with asthma and major depressive disorder: a proof of concept study. Biol. Psychiatry. 2005;58(11):865–870. doi: 10.1016/j.biopsych.2005.04.030. [DOI] [PubMed] [Google Scholar]
  • 387.Nardi AE. Where are the guidelines for the treatment of asthma with panic spectrum symptoms? Am. J. Respir. Crit. Care Med. 2005;172(8):1055–1056. doi: 10.1164/ajrccm.172.8.952. author reply 1056. [DOI] [PubMed] [Google Scholar]
  • 388.Melien O, Skaali T, Myhr K, Brors O. Venlafaxine and asthma. Nord. J. Psychiatry. 2005;59(6):538–540. doi: 10.1080/08039480500363595. [DOI] [PubMed] [Google Scholar]
  • 389.DeKruyff RH, Fang Y, Umetsu DT. Corticosteroids enhance the capacity of macrophages to induce Th2 cytokine synthesis in CD4+ lymphocytes by inhibiting IL-12 production. J. Immunol. 1998;160(5):2231–2237. [PubMed] [Google Scholar]
  • 390.Bolden-Watson C, Richelson E. Blockade by newly-developed antidepressants of biogenic amine uptake into rat brain synaptosomes. Life Sci. 1993;52(12):1023–1029. doi: 10.1016/0024-3205(93)90194-8. [DOI] [PubMed] [Google Scholar]
  • 391.Loder C, Allawi J, Horrobin DF. Treatment of multiple sclerosis with lofepramine, L-phenylalanine and vitamin B(12): mechanism of action and clinical importance: roles of the locus coeruleus and central noradrenergic systems. Med. Hypotheses. 2002;59(5):594–602. doi: 10.1016/s0306-9877(02)00261-x. [DOI] [PubMed] [Google Scholar]
  • 392.Puri BK, Bydder GM, Chaudhuri KR, Al Saffar BY, Curati WL, White SJ, Mitchell L, Hajnal JV, Horrobin DF. MRI changes in multiple sclerosis following treatment with lofepramine and L-phenylalanine. Neuroreport. 2001;12(9):1821–1824. doi: 10.1097/00001756-200107030-00012. [DOI] [PubMed] [Google Scholar]
  • 393.Altschuler EL. Monoamine oxidase inhibitors in rheumatoid arthritis-anti-tumor necrosis factor? Int. J. Immunopharmacol. 2000;22(11):1007–1008. doi: 10.1016/s0192-0561(00)00047-3. [DOI] [PubMed] [Google Scholar]
  • 394.Lieb J. Remission of rheumatoid arthritis and other disorders of immunity in patients taking monoamine oxidase inhibitors. Int. J. Immunopharmacol. 1983;5(4):353–357. doi: 10.1016/0192-0561(83)90039-5. [DOI] [PubMed] [Google Scholar]
  • 395.Sacre S, Medghalchi M, Gregory B, Brennan F, Williams R. Fluoxetine and citalopram exhibit potent antiinflammatory activity in human and murine models of rheumatoid arthritis and inhibit toll-like receptors. Arthritis Rheum. 2010;62(3):683–693. doi: 10.1002/art.27304. [DOI] [PubMed] [Google Scholar]
  • 396.Altschuler EL. Phenelzine as adjuvant treatment for Behcet's disease? Int. J. Dermatol. 1999;38(12):955. [PubMed] [Google Scholar]
  • 397.Altschuler EL. Monoamine oxidase inhibitors for IgA nephropathy. Med. Hypotheses. 2001;56(2):225–226. doi: 10.1054/mehy.2000.1158. [DOI] [PubMed] [Google Scholar]
  • 398.Chavant F, Deguil J, Pain S, Ingrand I, Milin S, Fauconneau B, Perault-Pochat MC, Lafay-Chebassier C. Imipramine, in part through tumor necrosis factor alpha inhibition, prevents cognitive decline and beta-amyloid accumulation in a mouse model of Alzheimer's disease. J. Pharmacol. Exp. Ther. 2010;332(2):505–514. doi: 10.1124/jpet.109.162164. [DOI] [PubMed] [Google Scholar]
  • 399.Nelson RL, Guo Z, Halagappa VM, Pearson M, Gray AJ, Matsuoka Y, Brown M, Martin B, Iyun T, Maudsley S, Clark RF, Mattson MP. Prophylactic treatment with paroxetine ameliorates behavioral deficits and retards the development of amyloid and tau pathologies in 3xTgAD mice. Exp. Neurol. 2007;205(1):166–176. doi: 10.1016/j.expneurol.2007.01.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400.Mowla A, Mosavinasab M, Pani A. Does fluoxetine have any effect on the cognition of patients with mild cognitive impairment? A double-blind, placebo-controlled, clinical trial. J. Clin. Psychopharmacol. 2007;27(1):67–70. doi: 10.1097/JCP.0b013e31802e0002. [DOI] [PubMed] [Google Scholar]
  • 401.Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc. Natl. Acad. Sci. U. S. A. 2003;100(23):13632–13637. doi: 10.1073/pnas.2234031100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Chung YC, Kim SR, Jin BK. Paroxetine prevents loss of nigrostriatal dopaminergic neurons by inhibiting brain inflammation and oxidative stress in an experimental model of Parkinson's disease. J. Immunol. 2010;185(2):1230–1237. doi: 10.4049/jimmunol.1000208. [DOI] [PubMed] [Google Scholar]
  • 403.Thomas T, Bhavnani BR, Thomas P. Inhibition of human LDL oxidation by the neuroprotective drug l-deprenyl. Neurol. Res. 2002;24(2):169–173. doi: 10.1179/016164102101199729. [DOI] [PubMed] [Google Scholar]
  • 404.Galvani S, Coatrieux C, Elbaz M, Grazide MH, Thiers JC, Parini A, Uchida K, Kamar N, Rostaing L, Baltas M, Salvayre R, Negre-Salvayre A. Carbonyl scavenger and antiatherogenic effects of hydrazine derivatives. Free Radic. Biol. Med. 2008;45(10):1457–1467. doi: 10.1016/j.freeradbiomed.2008.08.026. [DOI] [PubMed] [Google Scholar]
  • 405.Paslakis G, Gilles M, Lederbogen F, Schilling C, Scharnholz B, Deuschle M. The effect of a 4-week treatment with reboxetine on metabolic parameters of depressed inpatients. Eur. Arch. Psychiatry Clin. Neurosci. 2011;261(3):173–177. doi: 10.1007/s00406-010-0164-4. [DOI] [PubMed] [Google Scholar]
  • 406.Levkovitz Y, Ben-Shushan G, Hershkovitz A, Isaac R, Gil-Ad I, Shvartsman D, Ronen D, Weizman A, Zick Y. Antidepressants induce cellular insulin resistance by activation of IRS-1 kinases. Mol. Cell Neurosci. 2007;36(3):305–312. doi: 10.1016/j.mcn.2007.05.009. [DOI] [PubMed] [Google Scholar]
  • 407.Raeder MB, Bjelland I, Emil Vollset S, Steen VM. Obesity, dyslipidemia, and diabetes with selective serotonin reuptake inhibitors: the Hordaland Health Study. J. Clin. Psychiatry. 2006;67(12):1974–1982. doi: 10.4088/jcp.v67n1219. [DOI] [PubMed] [Google Scholar]
  • 408.Aviram M, Cogan U, Mokady S. Excessive dietary tryptophan enhances plasma lipid peroxidation in rats. Atherosclerosis. 1991;88(1):29–34. doi: 10.1016/0021-9150(91)90254-z. [DOI] [PubMed] [Google Scholar]
  • 409.Kopf D, Westphal S, Luley CW, Ritter S, Gilles M, Weber-Hamann B, Lederbogen F, Lehnert H, Henn FA, Heuser I, Deuschle M. Lipid metabolism and insulin resistance in depressed patients: significance of weight, hypercortisolism, and antidepressant treatment. J. Clin. Psychopharmacol. 2004;24(5):527–531. doi: 10.1097/01.jcp.0000138762.23482.63. [DOI] [PubMed] [Google Scholar]
  • 410.Cardinali DP, Cano P, Jimenez-Ortega V, Esquifino AI. Melatonin and the metabolic syndrome: physiopathologic and therapeutical implications. Neuroendocrinology. 2011;93(3):133–142. doi: 10.1159/000324699. [DOI] [PubMed] [Google Scholar]
  • 411.Taylor D. Antidepressant drugs and cardiovascular pathology: a clinical overview of effectiveness and safety. Acta Psychiatr. Scand. 2008;118(6):434–442. doi: 10.1111/j.1600-0447.2008.01260.x. [DOI] [PubMed] [Google Scholar]
  • 412.Roose SP, Laghrissi-Thode F, Kennedy JS, Nelson JC, Bigger JT, Jr, Pollock BG, Gaffney A, Narayan M, Finkel MS, McCafferty J, Gergel I. Comparison of paroxetine and nortriptyline in depressed patients with ischemic heart disease. JAMA. 1998;279(4):287–291. doi: 10.1001/jama.279.4.287. [DOI] [PubMed] [Google Scholar]
  • 413.Regan KL. Depression treatment with selective serotonin reuptake inhibitors for the postacute coronary syndrome population: a literature review. J. Cardiovasc. Nurs. 2008;23(6):489–496. doi: 10.1097/01.JCN.0000338929.89210.af. [DOI] [PubMed] [Google Scholar]
  • 414.Somberg TC, Arora RR. Depression and heart disease: therapeutic implications. Cardiology. 2008;111(2):75–81. doi: 10.1159/000119692. [DOI] [PubMed] [Google Scholar]
  • 415.Jiang W, Davidson JR. Antidepressant therapy in patients with ischemic heart disease. Am. Heart J. 2005;150(5):871–881. doi: 10.1016/j.ahj.2005.01.041. [DOI] [PubMed] [Google Scholar]
  • 416.George AK, Kunwar AR, Awasthi A. Acute myocardial infarction in a young male on methylphenidate, bupropion, and erythromycin. J. Child Adolesc. Psychopharmacol. 2005;15(4):693–695. doi: 10.1089/cap.2005.15.693. [DOI] [PubMed] [Google Scholar]
  • 417.Pederson KJ, Kuntz DH, Garbe GJ. Acute myocardial ischemia associated with ingestion of bupropion and pseudoephedrine in a 21-year-old man. Can. J. Cardiol. 2001;17(5):599–601. [PubMed] [Google Scholar]
  • 418.Patterson RN, Herity NA. Acute myocardial infarction following bupropion (Zyban) QJM. 2002;95(1):58–59. doi: 10.1093/qjmed/95.1.58. [DOI] [PubMed] [Google Scholar]
  • 419.Tousoulis D, Drolias A, Antoniades C, Vasiliadou C, Marinou K, Latsios G, Stefanadi E, Gounari P, Siasos G, Papageorgiou N, Trikas A, Stefanadis C. Antidepressive treatment as a modulator of inflammatory process in patients with heart failure: effects on proinflammatory cytokines and acute phase protein levels. Int. J. Cardiol. 2009;134(2):238–243. doi: 10.1016/j.ijcard.2008.02.013. [DOI] [PubMed] [Google Scholar]
  • 420.Zittel S, Weiller C, Liepert J. Reboxetine improves motor function in chronic stroke. A pilot study. J. Neurol. 2007;254(2):197–201. doi: 10.1007/s00415-006-0326-5. [DOI] [PubMed] [Google Scholar]
  • 421.Wang LE, Fink GR, Diekhoff S, Rehme AK, Eickhoff SB, Grefkes C. Noradrenergic enhancement improves motor network connectivity in stroke patients. Ann. Neurol. 2011;69(2):375–388. doi: 10.1002/ana.22237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.Foster DJ, Good DC, Fowlkes A, Sawaki L. Atomoxetine enhances a short-term model of plasticity in humans. Arch. Phys. Med. Rehabil. 2006;87(2):216–221. doi: 10.1016/j.apmr.2005.08.131. [DOI] [PubMed] [Google Scholar]
  • 423.Goldstein LB. Effects of amphetamines and small related molecules on recovery after stroke in animals and man. Neuropharmacology. 2000;39(5):852–859. doi: 10.1016/s0028-3908(99)00249-x. [DOI] [PubMed] [Google Scholar]
  • 424.Rampello L, Alvano A, Chiechio S, Raffaele R, Vecchio I, Malaguarnera M. An evaluation of efficacy and safety of reboxetine in elderly patients affected by "retarded" post-stroke depression. A random, placebo-controlled study. Arch. Gerontol. Geriatr. 2005;40(3):275–285. doi: 10.1016/j.archger.2004.09.004. [DOI] [PubMed] [Google Scholar]
  • 425.Kucukalic A, Bravo-Mehmedbasic A, Kulenovic AD, Suljic-Mehmedika E. Venlafaxine efficacy and tolerability in the treatment of post-stroke depression. Psychiatr. Danub. 2007;19(1-2):56–60. [PubMed] [Google Scholar]
  • 426.Sivenius J, Sarasoja T, Aaltonen H, Heinonen E, Kilkku O, Reinikainen K. Selegiline treatment facilitates recovery after stroke. Neurorehabil. Neural Repair. 2001;15(3):183–190. doi: 10.1177/154596830101500305. [DOI] [PubMed] [Google Scholar]
  • 427.Starkstein SE, Mizrahi R, Power BD. Antidepressant therapy in post-stroke depression. Expert Opin. Pharmacother. 2008;9(8):1291–1298. doi: 10.1517/14656566.9.8.1291. [DOI] [PubMed] [Google Scholar]
  • 428.Benton T, Lynch K, Dube B, Gettes DR, Tustin NB, Ping Lai J, Metzger DS, Blume J, Douglas SD, Evans DL. Selective serotonin reuptake inhibitor suppression of HIV infectivity and replication. Psychosom. Med. 2010;72(9):925–932. doi: 10.1097/PSY.0b013e3181f883ce. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 429.Evans DL, Lynch KG, Benton T, Dube B, Gettes DR, Tustin NB, Lai JP, Metzger D, Douglas SD. Selective serotonin reuptake inhibitor and substance P antagonist enhancement of natural killer cell innate immunity in human immunodeficiency virus/acquired immunodeficiency syndrome. Biol. Psychiatry. 2008;63(9):899–905. doi: 10.1016/j.biopsych.2007.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 430.Ances BM, Letendre SL, Alexander T, Ellis RJ. Role of psychiatric medications as adjunct therapy in the treatment of HIV associated neurocognitive disorders. Int. Rev. Psychiatry. 2008;20(1):89–93. doi: 10.1080/09540260701877670. [DOI] [PubMed] [Google Scholar]
  • 431.Cettomai D, McArthur JC. Mirtazapine use in human immunodeficiency virus-infected patients with progressive multifocal leukoencephalopathy. Arch Neurol. 2009;66(2):255–258. doi: 10.1001/archneurol.2008.557. [DOI] [PubMed] [Google Scholar]
  • 432.Lanzafame M, Ferrari S, Lattuada E, Corsini F, Deganello R, Vento S, Concia E. Mirtazapine in an HIV-1 infected patient with progressive multifocal leukoencephalopathy. Infez. Med. 2009;17(1):35–37. [PubMed] [Google Scholar]
  • 433.Butterweck V, Bockers T, Korte B, Wittkowski W, Winterhoff H. Long-term effects of St. John's wort and hypericin on monoamine levels in rat hypothalamus and hippocampus. Brain Res. 2002;930(1-2):21–29. doi: 10.1016/s0006-8993(01)03394-7. [DOI] [PubMed] [Google Scholar]
  • 434.Miskovsky P. Hypericin--a new antiviral and antitumor photosensitizer: mechanism of action and interaction with biological macromolecules. Curr. Drug Targets. 2002;3(1):55–84. doi: 10.2174/1389450023348091. [DOI] [PubMed] [Google Scholar]
  • 435.Czub S, Koutsilieri E, Sopper S, Czub M, Stahl-Hennig C, Muller JG, Pedersen V, Gsell W, Heeney JL, Gerlach M, Gosztonyi G, Riederer P, ter Meulen V. Enhancement of central nervous system pathology in early simian immunodeficiency virus infection by dopaminergic drugs. Acta Neuropathol. 2001;101(2):85–91. doi: 10.1007/s004010000313. [DOI] [PubMed] [Google Scholar]
  • 436.Czub S, Czub M, Koutsilieri E, Sopper S, Villinger F, Muller JG, Stahl-Hennig C, Riederer P, Ter Meulen V, Gosztonyi G. Modulation of simian immunodeficiency virus neuropathology by dopaminergic drugs. Acta Neuropathol. 2004;107(3):216–226. doi: 10.1007/s00401-003-0801-3. [DOI] [PubMed] [Google Scholar]
  • 437.Sockalingam S, Abbey SE. Managing depression during hepatitis C treatment. Can. J. Psychiatry. 2009;54(9):614–625. doi: 10.1177/070674370905400906. [DOI] [PubMed] [Google Scholar]
  • 438.Stauber RE, Stadlbauer V. Novel approaches for therapy of chronic hepatitis C. J. Clin. Virol. 2006;36(2):87–94. doi: 10.1016/j.jcv.2006.02.003. [DOI] [PubMed] [Google Scholar]
  • 439.Coogan PF, Strom BL, Rosenberg L. Antidepressant use and colorectal cancer risk. Pharmacoepidemiol. Drug Saf. 2009;18(11):1111–1114. doi: 10.1002/pds.1808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 440.Gil-Ad I, Zolokov A, Lomnitski L, Taler M, Bar M, Luria D, Ram E, Weizman A. Evaluation of the potential anti-cancer activity of the antidepressant sertraline in human colon cancer cell lines and in colorectal cancer-xenografted mice. Int. J. Oncol. 2008;33(2):277–286. [PubMed] [Google Scholar]
  • 441.Xu W, Tamim H, Shapiro S, Stang MR, Collet JP. Use of antidepressants and risk of colorectal cancer: a nested case-control study. Lancet Oncol. 2006;7(4):301–308. doi: 10.1016/S1470-2045(06)70622-2. [DOI] [PubMed] [Google Scholar]
  • 442.Toh S, Rodriguez LA, Hernandez-Diaz S. Use of antidepressants and risk of lung cancer. Cancer Causes Control. 2007;18(10):1055–1064. doi: 10.1007/s10552-007-9045-1. [DOI] [PubMed] [Google Scholar]
  • 443.Amit BH, Gil-Ad I, Taler M, Bar M, Zolokov A, Weizman A. Proapoptotic and chemosensitizing effects of selective serotonin reuptake inhibitors on T cell lymphoma/leukemia (Jurkat) in vitro. Eur. Neuropsychopharmacol. 2009;19(10):726–734. doi: 10.1016/j.euroneuro.2009.06.003. [DOI] [PubMed] [Google Scholar]
  • 444.Serafeim A, Holder MJ, Grafton G, Chamba A, Drayson MT, Luong QT, Bunce CM, Gregory CD, Barnes NM, Gordon J. Selective serotonin reuptake inhibitors directly signal for apoptosis in biopsy-like Burkitt lymphoma cells. Blood. 2003;101(8):3212–3219. doi: 10.1182/blood-2002-07-2044. [DOI] [PubMed] [Google Scholar]
  • 445.Cloonan SM, Drozgowska A, Fayne D, Williams DC. The antidepressants maprotiline and fluoxetine have potent selective antiproliferative effects against Burkitt lymphoma independently of the norepinephrine and serotonin transporters. Leuk. Lymphoma. 2010;51(3):523–539. doi: 10.3109/10428190903552112. [DOI] [PubMed] [Google Scholar]
  • 446.Ashbury JE, Levesque LE, Beck PA, Aronson KJ. A population-based case-control study of Selective Serotonin Reuptake Inhibitors (SSRIs) and breast cancer: the impact of duration of use, cumulative dose and latency. BMC Med. 2010;8:90. doi: 10.1186/1741-7015-8-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 447.Chien C, Li CI, Heckbert SR, Malone KE, Boudreau DM, Daling JR. Antidepressant use and breast cancer risk. Breast Cancer Res Treat. 2006;95(2):131–140. doi: 10.1007/s10549-005-9056-0. [DOI] [PubMed] [Google Scholar]
  • 448.Lash TL, Cronin-Fenton D, Ahern TP, Rosenberg CL, Lunetta KL, Silliman RA, Hamilton-Dutoit S, Garne JP, Ewertz M, Sorensen HT, Pedersen L. Breast cancer recurrence risk related to concurrent use of SSRI antidepressants and tamoxifen. Acta Oncol. 2010;49(3):305–312. doi: 10.3109/02841860903575273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Kast RE, Altschuler EL. Co-administration of ramelton and fluvoxamine to increase levels of interleukin-2. Med. Hypotheses. 2006;67(6):1389–1390. doi: 10.1016/j.mehy.2006.05.050. [DOI] [PubMed] [Google Scholar]
  • 450.Hwang MS, Yum YN, Joo JH, Kim S, Lee KK, Gee SW, Kang HI, Kim OH. Inhibition of c-erbB-2 expression an activity in human ovarian carcinoma cells by hypericin. Anticancer Res. 2001;21(4A):2649–2655. [PubMed] [Google Scholar]
  • 451.Riechelmann RP, Burman D, Tannock IF, Rodin G, Zimmermann C. Phase II trial of mirtazapine for cancer-related cachexia and anorexia. Am. J. Hosp. Palliat. Care. 2010;27(2):106–110. doi: 10.1177/1049909109345685. [DOI] [PubMed] [Google Scholar]
  • 452.Jia L, Shang YY, Li YY. Effect of antidepressants on body weight, ethology and tumor growth of human pancreatic carcinoma xenografts in nude mice. World J. Gastroenterol. 2008;14(27):4377–4382. doi: 10.3748/wjg.14.4377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 453.Kast RE. Profound blockage of CXCR4 signaling at multiple points using the synergy between plerixafor, mirtazapine, and clotrimazole as a new glioblastoma treatment adjunct. Turk. Neurosurg. 2010;20(4):425–429. doi: 10.5137/1019-5149.JTN.3334-10.0. [DOI] [PubMed] [Google Scholar]
  • 454.Pan CC, Cheng HH, Huang CJ, Lu YC, Chen IS, Liu SI, Hsu SS, Chang HT, Huang JK, Chen JS, Jan CR. The antidepressant mirtazapine-induced cytosolic Ca2+ elevation and cytotoxicity in human osteosarcoma cells. Chin. J. Physiol. 2006;49(6):290–297. [PubMed] [Google Scholar]
  • 455.Roberts JE, Wiechmann AF, Hu DN. Melatonin receptors in human uveal melanocytes and melanoma cells. J. Pineal Res. 2000;28(3):165–171. doi: 10.1034/j.1600-079x.2001.280306.x. [DOI] [PubMed] [Google Scholar]
  • 456.Kubera M, Grygier B, Arteta B, Urbanska K, Basta-Kaim A, Budziszewska B, Leskiewicz M, Kolaczkowska E, Maes M, Szczepanik M, Majewska M, Lason W. Age-dependent stimulatory effect of desipramine and fluoxetine pretreatment on metastasis formation by B16F10 melanoma in male C57BL/6 mice. Pharmacol. Rep. 2009;61(6):1113–1126. doi: 10.1016/s1734-1140(09)70174-4. [DOI] [PubMed] [Google Scholar]
  • 457.Ni YG, Miledi R. Blockage of 5HT2C serotonin receptors by fluoxetine (Prozac) Proc. Natl. Acad. Sci. U. S. A. 1997;94(5):2036–2040. doi: 10.1073/pnas.94.5.2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 458.Thyaga R S, Felten DL. Modulation of neuroendocrine--immune signaling by L-deprenyl and L-desmethyldeprenyl in aging and mammary cancer. Mech. Ageing Dev. 2002;123(8):1065–1079. doi: 10.1016/s0047-6374(01)00390-6. [DOI] [PubMed] [Google Scholar]
  • 459.Thyaga R S, Madden KS, Stevens SY, Felten DL. Anti-tumor effect of L-deprenyl is associated with enhanced central and peripheral neurotransmission and immune reactivity in rats with carcinogen-induced mammary tumors. J. Neuroimmunol. 2000;109(2):95–104. doi: 10.1016/s0165-5728(00)00305-2. [DOI] [PubMed] [Google Scholar]
  • 460.Flamand V, Zhao H, Peehl DM. Targeting monoamine oxidase A in advanced prostate cancer. J. Cancer Res. Clin. Oncol. 2010;136(11):1761–1771. doi: 10.1007/s00432-010-0835-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 461.Cutolo M, Straub RH. Stress as a risk factor in the pathogenesis of rheumatoid arthritis. Neuroimmunomodulation. 2006;13(5-6):277–282. doi: 10.1159/000104855. [DOI] [PubMed] [Google Scholar]
  • 462.Elenkov IJ, Chrousos GP. Stress hormones, proinflammatory and antiinflammatory cytokines, and autoimmunity. Ann. N. Y. Acad. Sci. 2002;966:290–303. doi: 10.1111/j.1749-6632.2002.tb04229.x. [DOI] [PubMed] [Google Scholar]
  • 463.Torta R. Depression as systemic disease: the antidepressants spectrum of actions. Psycho-Oncology. 2006;15(S2) [Google Scholar]
  • 464.Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve--an integrative interface between two supersystems: the brain and the immune system. Pharmacol. Rev. 2000;52(4):595–638. [PubMed] [Google Scholar]
  • 465.Yamauchi M, Hiraoka S, Imanishi T. Role of the serotonergic nervous system in anxiety disorders and the anxiolytic mechanism of selective serotonin reuptake inhibitors. Nihon Shinkei Seishin Yakurigaku Zasshi. 2006;26(5-6):193–198. [PubMed] [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES