Skip to main content

Psychological Factors in Asthma

Abstract

Asthma has long been considered a condition in which psychological factors have a role. As in many illnesses, psychological variables may affect outcome in asthma via their effects on treatment adherence and symptom reporting. Emerging evidence suggests that the relation between asthma and psychological factors may be more complex than that, however. Central cognitive processes may influence not only the interpretation of asthma symptoms but also the manifestation of measurable changes in immune and physiologic markers of asthma. Furthermore, asthma and major depressive disorder share several risk factors and have similar patterns of dysregulation in key biologic systems, including the neuroendocrine stress response, cytokines, and neuropeptides. Despite the evidence that depression is common in people with asthma and exerts a negative impact on outcome, few treatment studies have examined whether improving symptoms of depression do, in fact, result in better control of asthma symptoms or improved quality of life in patients with asthma.

Psychological factors may influence the symptoms and management of asthma, and numerous pathways may contribute to the links between asthma and psychiatric disease states such as depression. The notion that emotional stress can precipitate or exacerbate acute and chronic asthma [1] has been recognized anecdotally for many years. Psychological barriers, such as faulty symptom attribution, adoption or rejection of the sick role, and low self-esteem, may negatively impact treatment adherence. Conversely, the presence of a chronic and potentially life-threatening illness may exert enough stress that an anxiety or depressive disorder emerges in vulnerable patients. As a consequence, epidemiologic associations between major depressive disorder (MDD) and asthma might be apparent but not reflect a shared pathophysiologic vulnerability. Alternatively, there may be aspects of dysregulation in key biologic systems, such as the neuroendocrine stress response or cytokine system, that predispose people to both asthma and psychiatric illness independent of the psychological impact of one chronic illness on the other. More provocatively, perhaps, there may be components of central or peripheral nervous system dysfunction that predispose people to asthma or worsen the course of asthma independent of behavioural response style or the experience of illness-related stress or depression.

The purpose of this review is to summarize the disparate reports in the literature that point toward an association between asthma and psychological factors. The review has four primary components. The first briefly examines the evidence that psychological interventions can be beneficial in the treatment of asthma, ignoring whether the patients involved in the intervention have any a priori evidence of psychological distress or impaired psychosocial function. The second part of the review addresses the limited literature on whether the presence of psychiatric illness, primarily major depression or an anxiety disorder (AD), has a negative impact on asthma outcome and whether treatment of the psychiatric condition improves these outcomes and also considers the epidemiologic evidence of an association between asthma and depression. The third section considers the multiple biologic factors that could contribute to a shared vulnerability for depression and asthma as several key systems share patterns of dysregulation across these illnesses. Finally, we discuss a nascent literature examining the central nervous system (CNS) correlates of an asthmatic response.

Psychological Interventions Aimed at Improving Adherence and Asthma Control

A number of studies have examined the efficacy of psychological therapies at improving various aspects of asthma control or quality of life. These studies have been reviewed for both adults [2] and children [3, 4] and are not discussed in detail here. Because psychotherapy models can be grouped according to their theoretical frameworks or methods of operation, the various approaches are briefly discussed below:

  1. 1.

    Behavioural therapies focus on identifying the processes by which behaviour has been learned via association, reward, or observation and modifying behaviour using methods such as systematic desensitization, selective reinforcement, and positive modeling. The behaviour itself, rather than the underlying motivations, is the focus of behavioural interventions. Dahl found positive results following behavioural therapy when school absenteeism and use of as-needed medications were the outcome measures [5].

  2. 2.

    Cognitive therapies focus on identification and constructive management of incorrect and damaging thoughts, such as perceptions of helplessness or inappropriate fear of asthma attack, that can trigger episodes. Information (eg, about the relationships between anxiety and bronchoconstriction) also targets cognitions.

  3. 3.

    Cognitive behaviour therapy (CBT) incorporates the key elements of both behavioural and cognitive models and is currently used more frequently than either cognitive or behavioural therapies alone. Two studies measuring asthma knowledge as an outcome reported benefits of CBT, [6, 7] and CBT has been reported to have a positive effect on self-efficacy measures.

  4. 4.

    Relaxation techniques are generally conducted with or without biofeedback and were the focus of several earlier studies of psychological interventions in asthma. Relaxation techniques control stress and anxiety, which, in asthma, may improve breathing and respiratory function. Such programs generally include progressive relaxation, autogenic training, which focuses on attending to bodily feelings and mentally controlling them, and hypnosis or deep relaxation, which may be induced using mental imagery. This is often accompanied by autosuggestion to create positive thoughts and feedback of biologic indicators, which the subject must control via relaxation. Alexander and Weingarten measured the effect of relaxation therapy on peak expiratory flow and found effects favouring the treatment group compared with the control group [8, 9]. In addition, self-hypnosisassisted relaxation reduced emergency room visits, again in a single study that also found that self-reports of asthma improved in the self-hypnosis group [10]. In contrast, hospital admission rates were not decreased following biofeedback, [11, 12] nor were self-hypnosis rates or use of as-needed medications, [13] but emergency room visits were in a single study [11]. The results from these studies highlight the variability in outcome measures employed and the difficulty of understanding these studies in a systematic manner given this variability.

  5. 5.

    Psychodynamic psychotherapies attempt to uncover the emotional issues and response styles that drive patients to behave in maladaptive ways. Controlled trials of dynamic therapy are infrequent, and there is little evidence that they are likely to be of utility in a significant number of patients with asthma.

  6. 6.

    Counseling involves talking over problems with a health professional. In supportive counseling, the counselor acts primarily as a good listener who provides emotional support. Supportive therapy sometimes has a problem-solving focus and may be helpful for patients experiencing an acute crisis.

  7. 7.

    Family therapy attempts to understand family dynamics. Gustafsson and colleagues concluded that dysfunctional family interaction seems to be the result rather than the cause of wheezing in children [14]. There is evidence that family therapy may improve symptoms in children with asthma.

  8. 8.

    Educational approaches do not attempt to alter core psychological processes and therefore are not psychological therapies as such. They are already the subject of systematic reviews [15] and are routinely included as necessary components of optimal asthma care.

  9. 9.

    Breathing retraining exercises include a range of techniques for improving breathing control in asthma (eg, Buteyko technique, yoga, and transcendental meditation). These are not regarded as standard psychotherapies, although aspects of breathing retraining may be included in behavioural therapy or CBT. A Cochrane review [16] has previously examined the effectiveness of breathing retraining exercises, suggesting that conclusions must be viewed with caution.

Despite the trials of various psychological approaches in asthma, there are no sufficiently powered studies of any single therapy to draw conclusions regarding the utility of these approaches for improving asthma-related outcome. The systematic review that examined the efficacy of psychological treatments in children with asthma included 12 studies that met inclusion criteria, but the studies were small and the quality was poor. The authors stated that they could draw no conclusions regarding the effectiveness of psychological interventions for children with asthma because of the limited literature and variability among extant studies. Thus, in the aggregate, the benefit of psychological interventions for children and adults with asthma is difficult to assess because of the diversity of techniques used, the variety of outcomes measured, and the absence of appropriately powered trials.

A key issue apparent from these studies is how to select patients with asthma for psychological intervention. It may be that a randomized controlled trial that includes any patient with asthma who is willing to participate is not the most appropriate design as it is roughly analogous to including normal-weight people in a weight loss trial for obesity. Trials in which the population is enriched to have psychosocial distress or stress may more precisely reflect patients who are able to benefit, by virtue of having significant room for improvement, in the way in which they understand the illness and themselves in relation to the illness. Similarly, patients with very mild and well-controlled asthma are unlikely to have much room for improvement following a psychological intervention. It is probable that there is nonrandom overlap between these two groups, so the patients with the worst asthma control will, with some frequency, be those with the worst psychological adjustment to the illness. Examining the benefit of psychological therapies in this group might yield a stronger signal than in many previous trials. Furthermore, access to good psychological therapy is generally limited by therapist availability; therefore, such treatment arguably will be reserved in the clinical setting for patients with the most distress and the most to benefit from intervention. In summary, it is unfortunately possible that there is a reasonably sized subset of patients with poor asthma control related to poor psychological coping but that effective interventions for these people are not being routinely received or even offered because the trials to date do not allow conclusions to be made with any confidence.

Relationships between Asthma and Psychiatric Illness

Epidemiologic Associations between Asthma and Depression

The prevalence of MDD is higher in people with asthma relative to the general population. Individuals with allergic disease also have higher rates of MDD than nonatopic individuals [17, 18]. The presence of atopic disease increases the risk of depression in both men and women, although a more substantial body of evidence exists for the latter, [19] in whom the prevalence of MDD is generally higher. Patients with MDD or the other common mood disorder, bipolar affective disorder, also have an increased risk of developing immunoglobulin (Ig)E-mediated allergic conditions, including asthma, than the general population [20–22]. Asthma and hay fever also occur more frequently in patients with mood disorders and their family members than in those with schizophrenia [23].

Unfortunately, the literature on the prevalence of psychiatric disorders in patients with asthma is complicated by a number of issues, not the least of which is the problem of accurately defining and detecting cases of both disorders. There is significant variation in the rates of MDD in patients with asthma that appears in part secondary to ascertainment issues. Population-based studies have not reported rates of comorbidity as high as studies that evaluated depression in a clinical cohort of patients with asthma, for whom lifetime rates of asthma have been recorded to be as high as 47% [24, 25]. This may represent an accurate reflection of the asthma population as it is possible that the overall rates of psychiatric illness in those with mild and well-controlled asthma are low, with elevated rates observed in patients surveyed in tertiary care clinical settings who are likely to have more severe and chronic asthma. Regardless, the fact that individuals with asthma manifest higher rates of MDD and vice versa suggests that the two conditions may have shared pathogenic elements.

Familial Associations between Asthma and Depression

Further support for a link between asthma and MDD comes from family studies that suggest that the prevalence of one disorder is increased in the family members of index cases with the other. The initial evidence for this link came from mothers whose children had asthma but did not have MDD [26, 27]. In some studies, rates of depression in family members were related to the severity of the child's asthma symptoms, raising the possibility that these were related to the stress of having an ill child [28, 29]. Wamboldt and colleagues reported that mood but not ADs were increased in the relatives of adolescents with severe asthma and that the onset of these problems was equally likely to have occurred before as after the proband's asthma diagnosis [30]. More recent studies provide further proof that the prevalence of mood disorders is increased in the parents of children with asthma [31] even when childhood mental illness is considered [32].

Evidence supporting a genetic link between asthma and depression comes from Wamboldt and colleagues' study of Finnish twin pairs in which they assessed the prevalence of atopic disease and depressive symptomatology [33]. They found a within-person correlation between atopic and depressive symptoms of 0.103 and, using a best-fit model, estimated that 64% of this association was due to shared familial vulnerability, mainly additive genetic factors.

Common Environmental Risk Factors for Asthma and Depression

Obesity

Obesity generates a systemic inflammatory milieu [34] that increases the risk of numerous somatic conditions, including both asthma [35] and MDD. Epidemiologic studies suggest that there is an increased prevalence of asthma in obese adults, that this relationship is dose dependent, and that the link is stronger in women [36]. This association may reflect the direct mechanical effects of obesity, [37] immune system alterations, [38] or the effect of hormones such as leptin [39] imposed by excess weight.

Obese individuals also appear to be at increased risk of developing MDD [40]. The etiology of this seemingly bidirectional relationship is unknown but likely involves genetic and environmental influences, including the psychological experience of being overweight, as well as alterations in various hormones and cytokines. Although iatrogenic and clinical disease factors are most often implicated, it is possible that MDD and obesity share common pathogenic factors, [41] including dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, [42] neurotransmitter systems, [43, 44] and/or immune function [45, 46].

Smoking during Pregnancy

Maternal smoking during pregnancy has been proposed to increase the risk of both MDD [47] and asthma [48]. Adolescents exposed to cigarette smoking in utero have an increased risk of MDD prior to correcting for confounding and selection factors but not after this correction [47, 49]. Smoking in pregnancy is also associated in epidemiologic studies with an increased risk of asthma in children, adolescents, and adults, even when confounding variables are controlled for [48]. Numerous mechanisms have been proposed to account for this relationship, including the effects of smoking on fetal respiratory system development, [50] lung cyclic adenosine monophosphate (cAMP) levels, and phosphodiesterase 4 (PDE4) activity, which together may increase airway hyperresponsiveness [51].

Interestingly, asthmatics who are currently smoking or who have smoked in the past are relatively resistant to the anti-inflammatory effects of glucocorticoids (GCs) [52, 53]. Smoking and the oxidative stress it produces can affect GC receptor nuclear translocation and nuclear cofactors [54, 55]. Cases of severe GC-resistant asthma also manifest an increase in oxidative stress [56]. It is possible therefore that exposure to cigarette smoke in utero has similar effects on these pathways, increasing the risk of GC resistance and diseases associated with GC dysregulation later in life, including asthma and depression.

Asthma and Anxiety

Katon and colleagues conducted a review of the literature on the relationships between asthma and anxiety in children, adolescents, and adults [57]. They concluded that up to one-third of children and adolescents may meet the criteria for a comorbid AD. The rates of AD in adults with asthma ranged from 6 to 24%, although the studies had many of the same limitations as the studies of depression and asthma, including issues with small samples, ascertainment biases, and questionable methods of confirming the diagnosis of asthma or AD.

A study examined not only the rates of depression and anxiety in adolescents but also the likelihood that the comorbid psychiatric condition was recognized and treated [58]. Only about one-third of youth with anxiety had the condition recognized within the last year, and only about one in five youth with MDD had adequate treatment. A commentary accompanying this article concluded that the methods used by Katon and colleagues were probably conservative in the estimates of rates receiving treatment, so the actual rates of treatment of MDD or anxiety in youth with asthma may be even lower than 20% [59]. Thus, there appears to be a significant dissociation between studies that, despite limitations, suggest that anxiety and MDD occur frequently in asthma and studies that suggest that in routine clinical practice comorbid psychiatric conditions are infrequently recognized in patients with asthma and even less frequently treated.

Treatment of Psychiatric Symptoms to Improve Asthma and Health-Related Quality of Life

Pharmacologic Treatment

There is a notable paucity of data examining whether treating MDD in people with asthma will improve asthma outcome. Brown and colleagues randomized 90 patients with asthma and an episode of depression to citalopram, a commonly used antidepressant, or placebo [60]. The impact of this intervention on asthma symptoms was difficult to evaluate between antidepressant- and placebo-treated patients because at end point there was no difference in depression scores between antidepressant- and placebo-treated patients. Nonetheless, antidepressant-treated patients required fewer oral corticosteroids and there was a correlation between asthma symptom severity and depression symptoms. Perhaps the most interesting result in the study was the fact that patients who had substantial improvement in depressive symptoms (regardless of whether they were medication or placebo treated) had greater improvement in a variety of asthma-related scales than patients whose depressive symptoms did not improve significantly. These results do, therefore, support the notion that treating depressive symptoms may improve outcome in patients with asthma.

To our knowledge, only one other trial, conducted several decades ago, has evaluated the impact of antidepressant treatment on asthma outcome. In 1969, Sanger examined whether the antidepressants amitriptyline and doxepin improved depressive and anxiety symptoms in patients with allergic diseases, including some patients with asthma [61]. Doxepin appeared to have a more potent effect than amitriptyline because the particularly potent antihistaminergic properties of doxepin are not known.

Behavioural Treatment

We were unable to find any studies that had focused specifically on using psychological treatment for MDD in patients with asthma. Given that there are time-limited psychotherapies that are acceptable to patients, safe and effective treatments for MDD, it is unfortunate that no information exists on whether use of such therapies would improve asthma as well as depressive symptoms. A recent trial examined the benefit of CBT for patients with somatization disorder, in which patients have a preoccupation with physical symptoms that are disproportionate to any identifiable pathophysiologic process [62]. CBT was effective in this study, and the gains were maintained so that at follow-up months after treatment finished, there was evidence that patients were accessing medical resources less often than those who had not received CBT. These results provide indirect evidence to suggest that patients whose limitations associated with asthma appear greater than that predicted by the physical severity of the illness might benefit from CBT.

Pathophysiologic Links between Asthma and Depression

Stress and GC Resistance

The experience of significant stress early in life is a risk factor for the development of both MDD and asthma and, via GC resistance, may represent the most important link between the two conditions. A subset of patients who are exposed to psychological/emotional stress early in life have subtle dysregulation of the sympathetic and parasympathetic nervous systems and the HPA axis, including GC resistance, which bias the immune system toward a T helper (Th)2 response, [63, 64] immune system hyperactivity, and inflammation. It is possible that increased inflammation brings out a latent genetic risk for both asthma and depression, with the former having either a lower threshold for expression or with developmental factors interacting with inflammation to produce asthma. Depression, which, compared with asthma, is uncommon in prepubertal children, may have a higher threshold for symptom expression, requiring an increased duration of exposure or higher levels of GC resistance.

Immune development may also be influenced by prenatal imprinting or programming [65, 66]. Stress in utero not only results in the overexpression of cortisol in the mother but also stimulates secretion of corticotropin-releasing hormone (CRH) by the placenta. Such exposure appears to alter humoral immune responses and individuals' sensitivity to stress in postnatal life [67]. Postnatal stress has also been implicated in the development and exacerbation of asthma [68]. Parenting difficulties when a child is 3 weeks old were a predictor of early-onset childhood asthma in those predisposed to the disorder [69]. Other studies suggested that parenting difficulties, [70] but not family stress, [71] are associated with asthma.

GCs effectively suppress asthma symptoms in most people; however, a small number of patients fail to respond to exogenous steroids, even when they are given high doses [72]. Although GC-resistant patients exist on a spectrum, they have significant illness burden and present significant management challenges. They have usually had asthma longer than the average patient and manifest irreversible airflow obstruction and a greater inflammatory burden [73]. GC signaling defects are also present in depressed patients [74]. Nearly 50% of persons with depression have elevated cortisol levels, [75] with higher rates of dexamethasone nonsuppression in those with psychotic depression [76] and a higher number of lifetime depressive episodes [77]. Cortisol and CRH levels in cerebrospinal fluid (CSF) are increased in depressed patients, [78, 79] especially dexamethasone nonsuppressors [80]. Somatic treatments such as electroconvulsive therapy and medications normalize elevated CRH levels [81, 82].

Resistance to GCs may occur as a result of a number of factors, with long-term exposure to inflammatory cytokines often proposed as a key factor. The mechanisms through which this occurs may involve mitogen-activated protein kinase (MAPK), nuclear factor κB (NF-κB), and cyclooxygenase (COX) pathways (see Pace and colleagues for a review [83]). Stressful experiences may cause the developing autonomic nervous system (ANS) to be more labile, which can evolve into emotionally triggered asthma symptoms [30].

Cytokines

Cytokines affect inflammatory responses, and the processes they govern are implicated in the pathophysiology of many diseases, including those with CNS manifestations. Peripheral cytokines increase glial cell release of cytokines in the brain via the vagus and glossopharyngeal nerves rather than acting directly on the brain themselves [84]. The intersection of the cytokine and HPA systems is mechanistically relevant to the development of both asthma and MDD.

Depression is characterized by immune activation, particularly the innate immune system [85]. Sickness behaviour, the emotional and behavioural symptoms that develop as a consequence of acute infection or cytokine therapy, appears to be the result of increased levels of the proinflammatory cytokines interleukin (IL)-1 and tumour necrosis factor (TNF) and is the most frequently cited evidence linking cytokine activation with depression. Vital to the development of sickness behaviour is the enzyme indoleamine-2,3-dioxygenase (IDO), which is increased in interferon (IFN)-treated patients who become depressed and degrades tryptophan into the neurotoxic metabolites quinolinic acid and 3-hydroxykyurenine, which cross the blood-brain barrier and bind glutamate receptors. IDO appears to affect brain monoamine neurotransmission, and this may be the mechanism by which it affects mood [86]. Proinflammatory cytokines may also induce tissue resistance to GCs by inhibitory effects on the expression or function of GC receptors, which might contribute to CRH release secondary to reduced feedback inhibition as well as an increase in cytokine release [87].

A number of cytokines are dysregulated in patients with MDD, including IL-6, [88] which participates in the transition from innate to acquired immunity and in the polarization of immune responses from a Th1 to a Th2 type, [89] which is also of relevance to asthma development. IL-1β appears to be increased in those with asthma [90, 91] and depression [88] and in those with depression and asthma [92]. Through IL-5, it results in increased production of intercellular adhesion molecule 1 (ICAM-1) and vascular cellular adhesion molecule 1 (VCAM-1) by endothelial cells (see below) [93]. IL-1β alters behaviour in rodents, inducing anorexia, sleep disturbances, and memory impairment; it also alters monoamine and neuropeptide neurotransmitter metabolism [94].

High levels of TNF can exacerbate inflammatory and pro-oxidative functions [95]. TNF levels are increased in those with MDD [88, 96] and are associated with asthmatic complications. TNF acts preferentially on smooth muscle cells in airways, resulting in damage to bronchial epithelial cells as well as leakage of these and endothelial cells [97]. TNF protein and gene expression levels appear to be increased in the bronchoalveolar lavage fluid of asthmatics, [98] and the TNF receptor-IgG1Fc fusion protein appears to improve lung function in these patients [99].

Thus, despite the complexity of elucidating the role of the cytokine system in either depression or asthma, there is substantive evidence that the diseases share dysregulation of some key cytokines. Whether this overlap reflects a specific relationship or simply common states of inflammatory processes remains to be clarified. Unfortunately, the same dilemma is relevant to most of the systems discussed below.

Immune System Imbalance: Type 1 Th1 versus Th2 Phenotypes

Some propose that a reduction in exposure to microbes is responsible for the increasing prevalence of asthma as a lack of exposure may lead to a polarization of the allergen specific T-cell response toward Th2 instead of Th1 immunity [100]. IL-4 is particularly important in that it regulates IgE isotype switching, VCAM-1 production and Th cell commitment, and allergen-induced eosinophilia in asthmatics [101, 102]. IL-5 plays an important role in eosinophil differentiation and survival. IL-13 is involved in airway hyperresponsiveness in these individuals [100].

The role of Th1-Th2 cytokine balance has, not surprisingly, been much less extensively investigated in those with MDD. Although numerous studies have examined plasma cytokine and immune cell levels in those with depression, few have examined the balance between Th1 and Th2 cytokines in this population. Pavon and colleagues examined the serum levels of cortisol as well as Th1 (IL-2 and IFN-γ) and Th2 (IL-4 and IL-13) cytokines in 33 unmedicated outpatients with MDD and compared them with 33 nondepressed controls [103]. In this study, the depressed patients appeared to have a preference for Th2 immune responses. Given that cortisol was also elevated in this sample, and given the propensity for cortisol to increase Th2 activity, [104] it may be that the immune shift to a Th2 response was driven by altered activity in the HPA axis. Mendlovic and colleagues also demonstrated a predilection for a Th2-like profile of cytokine secretion from the T cells of a small sample of depressed patients compared with controls [105].

Nuclear Factor κB

NF-κB is a major transcription factor that is induced by a large number of factors, including proinflammatory cytokines and other mediators of stress, and plays a role in the development of immunity. Dysregulation, including aberrant activation of the NF-κB pathway, is seen in numerous diseases, including asthma and MDD [106].

NF-κB exists in the cytoplasm of cells in an inactive form bound to its inhibitor IκB. When proinflammatory cytokines such as TNF bind their receptors, it results in NF-κB translocation to the nucleus, which promotes gene expression. It has been hypothesized that activation of this pathway is relevant to the pathophysiology of MDD since certain cytokines appear to contribute to the development of depression in some individuals (see above) and since serotonin-containing neurons, long implicated in the development of depression, also contain NF-κB. One group has hypothesized that cytokines' activation of NF-κB leads to depression via increases in 5HT1A gene expression, which result in decreased firing of serotonin neurons and serotoninergic neurotransmission [107].

Asthma is also characterized by abnormal activation of cytokines and adhesion molecules and is triggered by a number of environmental agents, many of which result in NF-κB activation. NF-κB appears to have an important role in allergic inflammation, [108] and inhaled GCs have demonstrated inhibitory effects on NF-κB [109]. It is possible, then, that a number of factors common to the pathophysiology of asthma and MDD, including alterations in the HPA axis and cytokine dysregulation, converge on NF-κB signaling, which may serve as a final common pathway contributing to the development of these disorders.

Oxidative Stress

Oxidative stress may be relevant to the pathogenesis of asthma [110]. The capacity of the body's natural antioxidant system appears reduced in those with asthma in times of disease stability, [111] as well as exacerbation [112]. Levels of oxidative stress are elevated not only locally in airways but also systemically, [113] and levels of oxidative stress markers appear to correlate with disease severity [114]. Increases in oxidative stress have also been implicated in shifting immune responses to a Th2 phenotype [115].

Psychological stress may affect the body's capability to deal effectively with reactive oxygen species and increase oxidative stress [116, 117]. MDD is associated with increased levels of reactive oxygen species, [118] and depressed people have evidence of excess oxidative damage, [119, 120] independent of other causes of oxidative injury [121]. Those with multiple depressive episodes appear to incur more damage than those with fewer [122].

Increased innate immune responses [123] and inflammation [124] are also associated with MDD and can increase oxidative stress and may contribute to or account for the above findings. Indeed, overstimulation of the enzyme IDO raises levels of metabolites of kynurenine and 3-hydroxykynurenine, which increase oxidative stress [86]. It is currently unknown whether oxidative stress contributes to or is an epiphenomenon of the pathogenesis of depression [125].

Intracellular Adhesion Molecule 1

Intracellular adhesion molecule 1 (ICAM-1) is involved in the leukocyte adhesion, persistent inflammation, and cellular recruitment critical to the pathogenesis of asthma. ICAM-1 initiates intracellular signaling events and modulates the activation and proliferation of inflammatory cells as well as cytokine production, [126] leading to bronchial hyperresponsiveness and airway inflammation [127]. Increases in soluble ICAM-1 are apparent in asthma exacerbations [128] after allergen provocation [129] and correlate with asthma severity [130].

ICAM-1 appears to be expressed in increased amounts in the brains and serum of depressed patients [131–134] and remains elevated even after adjustment for potential confounders [135]. It also appears that soluble ICAM-1 levels play a role in the development of depression in IFN-treated patients. Patients with malignant melanoma who developed depression on this treatment had higher soluble ICAM-1 levels than those who did not, and the levels correlated with depression severity. These results have been interpreted as suggesting that increases in soluble ICAM-1 reflect the breakdown of the blood-brain barrier, which might then allow cytokines to enter and affect mood changes by modulating neurotransmission [136].

Whether increased levels of soluble adhesion molecules are involved in the pathogenesis of MDD or merely reflect a state of persistent, low-grade inflammation is not known, but this may represent another link between depression and asthma. Alternatively, this finding may be related to a primary immune dysfunction with increased cytokines and HPA axis abnormalities, which increased levels of soluble ICAM may reflect.

Prostaglandins and COX-2

COX-2 and its metabolites exert complex effects in the lung as some act as pro- and others as anti-inflammatory mediators [137]. COX-2 gene expression is increased in asthmatic patients' airways; however, increased COX-2 activity suppresses the asthmatic response. That prostaglandin (PG) levels appear to be increased in those with depression suggests that COX-2 activity is increased in these individuals as well. Unlike most tissues, COX-2 is constitutively expressed in the brain [138] and interacts with immune and neurotransmitter systems there. COX-2 may exert its effects by increasing PGE2 levels to stimulate IL-6 production. These findings may account for why treatment with COX-2 inhibitors has been associated in a few studies with reduced depressive symptomatology [139, 140]. Activation of COX-2 increases PGE2 concentrations, which can stimulate the HPA axis. The COX pathway also appears to interact with GC signaling and may modulate GC receptor responses. Thus, it is possible that COX-2 exerts its influence on affect via this mechanism [83].

PGs may also be involved in the pathogenesis of asthma [141] and MDD. They are produced by almost all cell types and participate in the inflammatory cascade that occurs in airways [142]. PGs D2, E2, and F2 have a variety of effects on airway physiology, including polarizing immune cells to a Th2 phenotype, attracting immune cells, stimulating proinflammatory cytokines, increasing mucus production and vascular leakage, and causing constriction of bronchial smooth muscle [142].

PGE2 is increased in the CSF, [143] serum, [144] and saliva [145] of patients with MDD and correlates with the severity of depression [146]. Mastocytosis, a disorder in which there is overproduction of PGD2, often manifests depressive symptomatology,147 and PGs influence behaviour, [148] sleep, [149] and appetite [150]. PGE2 also appears to have a direct effect on the promotion of sickness behaviour [151].

Phosphodiesterase 4

PDE4 is found in a number of cell types, including neurons and immune and airway cells. Both asthma and MDD may involve overactivity of PDE4 [152]. For example, the main gene involved in mucin secretion, MUC5AC, is overexpressed in those with asthma, [153] and PDE4 inhibition may ameliorate this. Rolipram, a PDE4 inhibitor, inhibits neutrophilic and eosinophilic inflammation and the release of cytokines from Th1 and Th2 cells, as well as airway epithelium, basophils, monocytes, and macrophages [154]. Also of relevance to asthma is the fact that PDE4 inhibitors reduce fibrosis and remodeling in the airway via inhibition of certain matrix metalloproteinases (MMPs). Clinically, PDE4 inhibitors reduce early and late inflammatory response to allergens in mild to moderate asthmatics and may produce small improvements in forced expiratory volume in 1 second in asthmatics [155].

Second-messenger impairments affecting cell survival and neuroplasticity are also believed to underlie MDD, [156] and cAMP-mediated signaling is implicated in the pathophysiology of MDD [7]. PDE4 is expressed in neurons in the hippocampus, striatum, substantia nigra, and cerebral cortex, as well as in astrocytes and, of relevance to depression, in the areas of the brain that are involved in reward and affect [157]. PDE4 also participates in cAMP pathways affected by known antidepressants [158]. Rolipram, a PDE4 inhibitor, has antidepressant-like effects in preclinical animal models and plays a role in induction of hippocampal neurogenesis, [159] which may be necessary for antidepressants to effect behavioural change [160]. Moreover, reduced expression of PDE4 appears to protect mice against depressive symptomatology [161].

Matrix Metalloproteinases

MMPs are proteolytic enzymes that degrade extracellular matrix components [162]. The production and function of MMPs are regulated by molecules such as the tissue inhibitors of matrix metalloproteinases (TIMPs), cytokines (eg, TNF, IL-1β), and growth factors. It is speculated that cytokines and MMPs interact in complex ways as a means of producing some of the symptoms of asthma [163].

MMPs may participate in airway remodeling, and increased levels of MMP-9 have been detected in asthma, related to elevated numbers of neutrophils and eosinophils in the airways [162] and correlated with asthma severity. In mouse models of asthma, MMP-9 absence is associated with a decrease in airway infiltration by inflammatory cells, [164] perhaps by decreasing dendritic cell migration [165].

A number of MMPs are not detectable in nonpathologic CNS states but are found in diseases of the CNS [166]. Certain MMPs can convert TNF and IL-6 to their active forms, a mechanism by which MMPs might promote an inflammatory milieu in the CNS [167]. Psychological stress, mediated by activation of the HPA and sympathetic-adrenal medullary axes, as well as cytokine alterations, affect MMP and TIMP levels [168].

Histaminergic System

Histamine is made and released by inflammatory cells and neurons and participates in the regulation of inflammatory responses in several conditions, including asthma. Histamine enhances secretion of proinflammatory cytokines, including IL-1α and -1β, IL-6, and a number of chemokines [169]. Histamine acts as a chemoattractant for eosinophils and mast cells and is released from mast cells during allergic reactions. Moreover, it appears to shift the immune response to Th2 dominance [170]. Histamine exposure causes bronchoconstriction in all humans, although asthmatics are more sensitive to this effect than nonasthmatics, and treatment with H1 receptor antagonists has been shown to improve symptoms and pulmonary function and may delay asthma onset in high-risk individuals [171–173].

Histamine also acts as a neurotransmitter in the brain and has been proposed to be involved in the pathogenesis of depression [174] as histamine type 3 receptor blockers may have antidepressant effects [175]. Alterations in histaminergic activity may also contribute to the experience of mental and physical fatigue experienced by depressed patients [25, 176].

Adenosine

Adenosine is an endogenous nucleoside present at low levels under normal conditions; however, its concentrations increase in the setting of stress and inflammation [177]. Adenosine has proinflammatory and immunomodulatory effects and may be involved in the pathogenesis of asthma [178–180].

Increased adenosine levels may result in depressive symptoms. The involvement of adenosine in the pathophysiology of mood disorders was first proposed when increases in endogenous adenosine levels led to behaviour consistent with learned helplessness and behavioural despair in laboratory animals [181, 182]. Antagonists to adenosine receptors, particularly A2A antagonists, appear to have antidepressant properties, [183] which may be mediated by increases in dopaminergic transmission in the frontal cortex [184].

Nitric Oxide

Nitric oxide (NO) is the only molecule in the body that acts as a hormone, reactive oxygen species, and neurotransmitter. The neurotransmitter and vasodilatory actions of NO are mediated mainly by guanylate cyclase activation in cells, which leads to an increase in the production of cyclic guanosine monophosphate and its dependent kinases [185]. Some evidence suggests that NO may be involved in the pathogenesis of asthma [186]. Evidence supports the role of NO in the pathogenesis of depression and in a number of the symptoms of this syndrome, including cognitive difficulties, sleep, and alterations in appetite [185, 187]. In the brain, neuronal nitric oxide synthase (NOS) produces NO after activation of the N-methyl-D-aspartate receptor by glutamate [185] and acts as a modulator of the HPA axis [188]. Neuronal NOS production is also regulated by GCs in the hippocampus, suggesting that it has a role in the body's response to stress [187]. It appears to be colocalized with a number of neuropeptides in the hypothalamus, including arginine vasopressin, CRH, and oxytocin. Neurons in the prefrontal cortex, amygdala, [189] and the serotoninergic cells of the dorsal raphe nucleus also contain NOS [190].

Neuropeptides

Many neuropeptides exist and have been implicated in the pathophysiology of inflammatory diseases, although we limit our discussion to those mediators that appear to be of relevance to both asthma and MDD. The airway is innervated not only by sympathetic and parasympathetic nerves but also by sensory nerves referred to as the noncholinergic-nonadrenergic that originate mainly from the vagal ganglia. Not surprisingly, a bidirectional relationship exists between the airway surface and the nerves that innervate it, and neuropeptides appear to mediate this relationship [191].

Tachykinins are proinflammatory neuropeptides of which substance P (SP) and neurokinin A (NKA) are members. They regulate neurogenic inflammation in the airway [192]. SP binds NK1 receptors located mainly in the airway epithelium, submucosal glands, and vessels, whereas NKA binds NK2 receptors found predominantly on smooth muscle cells [193]. NKA constricts airway smooth muscle cells with particularly potent effects in smaller airways, producing bronchoconstriction in asthmatics, [194] and SP causes mucus secretion. When aerosolized, SP induces inflammation and hyperresponsiveness of airways [195]. Despite the theoretical appeal of blocking tachykinin receptors, human testing with antagonists has been met with mixed results [191]. However, this may be in part due to difficulties with drug delivery.

Neuropeptides function as neurotransmitters and neuromodulators and are involved in the regulation of emotion and responses to stress [196]. Thus, they have become attractive targets for manipulation with regard to mood disorders. Indeed, SP receptor antagonists have been demonstrated to possess antidepressant effects in double-blind randomized controlled trials. Antagonists to NK1, the main tachykinin receptor in the human brain, appear to have some antidepressant efficacy in treating humans with depression and anxiety [197].

ANS (Parasympathetic Division)

Efferent parasympathetic fibres of the vagus regulate numerous functions, whereas afferent fibres (comprising 80% of the nerve) carry sensory information from the head, neck, abdomen, and chest. Messages are carried to the dorsal medullary complex, particularly the nucleus tractus solitarius, which relays information to other brain regions, including the locus ceruleus and raphe nucleus, as well as limbic, paralimbic, and cortical regions. The parabrachial nucleus also relays information to the hypothalamus, amygdala, and thalamus [198].

Some have suggested that depression produces a state that favours airway constriction in those with asthma. Depression appears to be a state of cholinergic dominance and asthma a condition marked by cholinergic dysregulation [199]. This hypothesis is supported by evidence that shows that some antidepressants result in bronchodilation in laboratory animals [200]. In animals in which hopelessness is induced, cholinergic tone in the ANS increases [201]. Another study reported that children who died of asthma had states of hopelessness in the days preceding their deaths, postulated to have contributed to mortality via ANS dysregulation manifested as increased cholinergic/vagal activation in sad and hopeless individuals [202]. In 1997, Miller and Wood reported that higher levels of induced sadness were associated with greater vagal and presumably cholinergic activation, reflected by increased heart rate variability (HRV) and oxygen saturation variability than happiness in 24 children aged 7 to 18 years [203].

They suggested that this supported the theory that sadness could evoke autonomic patterns that could mediate airway constriction. This work supported previous findings of increased cholinergic/parasympathetic tone in those experiencing hopelessness/depression [201] and Miller and Wood's previously hypothesized model implicating mood-associated vagal mediation of pulmonary function [203]. The increased reactivity of asthmatic patients' airways may be secondary to abnormal ANS control [204]. The parasympathetic/vagal component in particular appears to be relevant to asthma pathogenesis as it is involved in bronchoconstriction secondary to exercise and alterations in airway surface temperature. Asthma is related to abnormal ANS function, including both bronchial hyperreactivity to cholinergic drugs and reduced sensitivity to adrenergic dilators. Alterations in autonomic function have also been noted in asthmatics following exercise relative to nonasthmatic individuals. Enhanced cholinergic airway responsivity has also been postulated to contribute to the development of asthma [205].

The literature examining HRV in patients with depression has been mixed, with some [206, 207] but not all [208, 209] studies suggesting that HRV is lower in depressed patients, in keeping with excessive sympathetic modulation of the heart rate or inadequate parasympathetic tone. Moreover, vagal nerve stimulation (VNS), an experimental treatment for depression in which the vagus is stimulated, sheds some doubt on whether excess parasympathetic stimulation contributes to depressive symptoms. There is some evidence, however, that VNS therapy may have effects on the airways of certain individuals [210].

Thus, it is possible that frequent experience of the emotional states of sadness and hopelessness, common in those with MDD, may mediate, via increased cholinergic activity, an increased risk of asthma in some individuals, although it has been proposed that the enhanced cholinergic responses may be secondary to asthma rather than a pathogenetic contributor.

Risk of Treatment?

Little attention has been paid to the effects that treatments for either asthma or MDD have on the risk of development of the other. Serotonin has been controversially implicated in the pathophysiology of asthma, and patients with symptomatic asthma display increased plasma serotonin levels relative to asymptomatic individuals [211]. Serotoninergic receptors present in human airways, when activated, appear to stimulate IL-6 release in these cells [212]. Moreover, serotonin may have immunomodulatory effects [213]. Reports requiring replication suggested that tianeptine, a selective serotonin reuptake enhancer, reduces respiratory symptoms in asthmatics [214]. Although it is therefore conceivable that selective serotonin reuptake inhibitors could trigger or worsen asthmatic symptoms via release of IL-6, there are no clinical data to support this. However, tianeptine also appears to have antidepressant effects and may modulate asthma symptoms via this mechanism.

The long-term treatment of asthma and not the experience of asthma itself may also contribute to the risk of developing depression in asthmatics. It has been suggested that corticosteroid treatment taken for a number of indications is associated with depressive symptoms, [215, 216] although these results are limited by the fact that the indications and route of steroid treatment were not known. Thus, it is also possible that in a subset of patients with asthma, perhaps those treated recurrently with oral corticosteroids, that treatment contributes to or accounts for an increase in the rates of MDD seen in this population.

CNS Correlates of Asthma

It is increasingly accepted that psychological stress can modulate asthma symptoms [217, 218]. There is anecdotal and empirical evidence that the stable variable of nonhypnotic suggestibility can determine the susceptibility of asthmatic patients to suggestion of bronchoconstriction, providing a construct for understanding how some, but not all, patients with asthma might be particularly influenced by asthma-related cues [219]. Until recently, however, there were no studies that directly imaged the brain during exposure to asthma-related stimuli.

In a seminal study, Rosenkranz and colleagues used functional magnetic resonance imaging (fMRI) to examine activity in the anterior cingulate cortex (ACC) and insula during exposure to asthma-related words when patients with asthma were exposed to allergen [220]. Although there was a small sample of patients, the results provide provocative evidence that these brain regions were hyperresponsive to asthma-related emotional cues and afferent physiologic signals. The ACC receives input regarding key physical symptoms (eg, shortness of breath) of relevance to asthma (for an extensive review of the ACC, see Devinsky and colleagues [221]). Along with the insula, the ACC is also crucial for the processing of emotional stimuli and is implicated in the pathophysiology of MDD. Rosenkranz and colleagues contextualized their study by stating that "despite the compelling support for a model integrating psychological and physiological factors in asthma, the brain has been largely absent from any discussion of its mechanistic underpinnings." [220]

Capuron and colleagues also used fMRI in patients receiving IFN therapy and found that IFN-treated patients had activation of the dorsal ACC during a visuospatial task that was not present in control subjects [222]. Interestingly, IFN-treated patients performed well on the task but appeared to require more extensive involvement of the ACC than was necessary from control subjects. Although indirect, this study supports the hypothesis that the ACC may be important for understanding the interface of cognition, emotion, and peripheral inflammation. Furthermore, a study of patients with damage to the ACC found that they had impaired autonomic cardiovascular responses in response to mental stress [223]. Studies such as these, which integrate brain imaging with physiologic symptoms or inflammatory markers, are complex to undertake but represent extraordinary opportunities to reveal the role of the brain in modulating various components of the asthmatic response.

Conclusions

Studies of psychological intervention in patients with asthma are limited in their interpretation by the heterogeneity of patient samples, intervention technique, and outcome measures. It is possible that more focused trials of patients with measurable degrees of stress, depression, or psychosocial dysfunction would yield a more definitive answer regarding whether targeting psychological factors in at-risk patients can improve asthma outcome.

Asthma and stress-related psychiatric disorders share a number of environmental risk factors and pathophysiologic mechanisms. Perhaps the most persuasive of these is the early experience of stress and its effects on GC resistance as a vulnerability and prognostic factor for both depression and asthma. There are many physiologic points of intersection between asthma and MDD, however, and the specificity of these associations remains to be determined. Preliminary but promising studies are using functional imaging modalities to examine the CNS response to bronchoconstriction and allergen challenge. Further studies that also examine respiratory, immune, and neuronal responses to challenge may uncover relations between central and peripheral effects that clarify the relationships between cognitive and emotional events and asthma and point toward pharmacologic and nonpharmacologic strategies for improving the outcome of asthma.

References

  1. Sandberg S, Paton JY, Ahola S: The role of acute and chronic stress in asthma attacks in children. Lancet. 2000, 356: 982-7. 10.1016/S0140-6736(00)02715-X.

    Article  CAS  PubMed  Google Scholar 

  2. Yorke J, Fleming SL, Shuldham CM: Psychological interventions for adults with asthma. Cochrane Database Syst Rev. 2006, 1: CD002982-

    PubMed  Google Scholar 

  3. Yorke J, Fleming S, Shuldham C: Psychological interventions for children with asthma. Cochrane Database Syst Rev. 2005, CD003272-

    Google Scholar 

  4. Yorke J, Fleming SL, Shuldham C: A systematic review of psychological interventions for children with asthma. Pediatr Pulmonol. 2007, 42: 114-24. 10.1002/ppul.20464.

    Article  PubMed  Google Scholar 

  5. Dahl J, Gustafsson D, Melin L: Effects of a behavioral treatment program on children with asthma. J Asthma. 1990, 27: 41-6. 10.3109/02770909009073293.

    Article  CAS  PubMed  Google Scholar 

  6. Colland VT: Learning to cope with asthma: a behavioural self-management program for children. Patient Educ Couns. 1993, 22: 141-52. 10.1016/0738-3991(93)90094-D.

    Article  CAS  PubMed  Google Scholar 

  7. Perez J, Tardito D, Racagni G: Protein kinase A and Rap1 levels in platelets of untreated patients with major depression. Mol Psychiatry. 2001, 6: 44-9. 10.1038/sj.mp.4000795.

    Article  CAS  PubMed  Google Scholar 

  8. Alexander AB, Miklich DR, Hershkoff H: The immediate effects of systematic relaxation training on peak expiratory flow rates in asthmatic children. Psychosom Med. 1972, 34: 388-94.

    Article  CAS  PubMed  Google Scholar 

  9. Weingarten MA, Goldberg J, Teperberg Y: A pilot study of the multidisciplinary management of childhood asthma in a family practice. J Asthma. 1985, 22: 261-5. 10.3109/02770908509077458.

    Article  CAS  PubMed  Google Scholar 

  10. Kohen DP: Hypnotherapeutic management of pediatric and adolescent trichotillomania. J Dev Behav Pediatr. 1996, 17: 328-34. 10.1097/00004703-199610000-00007.

    Article  CAS  PubMed  Google Scholar 

  11. Khan AU, Staerk M, Bonk C: Role of counter-conditioning in the treatment of asthma. J Psychosom Res. 1973, 17: 389-92. 10.1016/0022-3999(73)90049-4.

    Article  CAS  PubMed  Google Scholar 

  12. Khan AU: Effectiveness of biofeedback and counter-conditioning in the treatment of bronchial asthma. J Psychosom Res. 1977, 21: 97-104. 10.1016/0022-3999(77)90078-2.

    Article  CAS  PubMed  Google Scholar 

  13. Kotses H, Harver A, Segreto J: Long-term effects of biofeedback-induced facial relaxation on measures of asthma severity in children. Biofeedback Self Regul. 1991, 16: 1-21. 10.1007/BF01000442.

    Article  CAS  PubMed  Google Scholar 

  14. Gustafsson PA, Bjorksten B, Kjellman NI: Family dysfunction in asthma: a prospective study of illness development. J Pediatr. 1994, 125: 493-8. 10.1016/S0022-3476(05)83306-2.

    Article  CAS  PubMed  Google Scholar 

  15. Wolf FM, Guevera JP, Grum CM: Educational interventions for asthma in children. Cochrane Database Syst Rev. 2003, CD000326-

    Google Scholar 

  16. Holloway E, Ram FSF: Breathing exercises for asthma. Cochrane Database Syst Rev. 2004, CD001277-DOI: 10.1002/14651858.CD001277., 1

  17. Centanni S, Di Marco F, Castagna F: Psychological issues in the treatment of asthmatic patients. Respir Med. 2000, 94: 742-9. 10.1053/rmed.1999.0766.

    Article  CAS  PubMed  Google Scholar 

  18. Timonen M, Jokelainen J, Hakko H: Atopy and depression: results from the Northern Finland 1966 Birth Cohort Study. Mol Psychiatry. 2003, 8: 738-44. 10.1038/sj.mp.4001274.

    Article  CAS  PubMed  Google Scholar 

  19. Timonen M, Hakko H, Miettunen J: Association between atopic disorders and depression: findings from the Northern Finland 1966 birth cohort study. Am J Med Genet. 2001, 105: 216-7. 10.1002/ajmg.1199.

    Article  CAS  PubMed  Google Scholar 

  20. Bell IR, Jasnoski ML, Kagan J, King DS: Depression and allergies: survey of a nonclinical population. Psychother Psychosom. 1991, 55: 24-31. 10.1159/000288404.

    Article  CAS  PubMed  Google Scholar 

  21. Matussek P, Agerer D, Seibt G: Allergic disorders in depressive patients. Compr Psychiatry. 1983, 24: 25-34. 10.1016/0010-440X(83)90047-0.

    Article  CAS  PubMed  Google Scholar 

  22. Yatham LN, Kennedy SH, O'Donovan C: Canadian Network for Mood and Anxiety Treatments (CANMAT) guidelines for the management of patients with bipolar disorder: update 2007. Bipolar Disord. 2006, 8: 721-39. 10.1111/j.1399-5618.2006.00432.x.

    Article  CAS  PubMed  Google Scholar 

  23. Nasr S, Altman EG, Meltzer HY: Concordance of atopic and affective disorders. J Affect Disord. 1981, 3: 291-6. 10.1016/0165-0327(81)90030-6.

    Article  CAS  PubMed  Google Scholar 

  24. Nejtek VA, Brown ES, Khan DA: Prevalence of mood disorders and relationship to asthma severity in patients at an inner-city asthma clinic. Ann Allergy Asthma Immunol. 2001, 87: 129-33. 10.1016/S1081-1206(10)62206-5.

    Article  CAS  PubMed  Google Scholar 

  25. Brown RE, Stevens DR, Haas HL: The physiology of brain histamine. Prog Neurobiol. 2001, 63: 637-72. 10.1016/S0301-0082(00)00039-3.

    Article  CAS  PubMed  Google Scholar 

  26. Davis JB: Neurotic illness in the families of children with asthma and wheezy bronchitis: a general practice population study. Psychol Med. 1977, 7: 305-10. 10.1017/S0033291700029408.

    Article  CAS  PubMed  Google Scholar 

  27. Jessop DJ, Riessman CK, Stein RE: Chronic childhood illness and maternal mental health. J Dev Behav Pediatr. 1988, 9: 147-56. 10.1097/00004703-198806000-00006.

    Article  CAS  PubMed  Google Scholar 

  28. Leigh D, Marley E: Bronchial asthma: a genetic, population and psychiatric study. 1967, Oxford: Pergamon Press

    Google Scholar 

  29. Meijer A: A controlled study on asthmatic children and their families. Synopsis of findings. Isr J Psychiatry Relat Sci. 1981, 18: 197-208.

    CAS  PubMed  Google Scholar 

  30. Wamboldt MZ, Weintraub P, Krafchick D, Wamboldt FS: Psychiatric family history in adolescents with severe asthma. J Am Acad Child AdolescPsychiatry. 1996, 35: 1042-9. 10.1097/00004583-199608000-00015.

    Article  CAS  Google Scholar 

  31. Brown ES, Gan V, Jeffress J: Psychiatric symptomatology and disorders in caregivers of children with asthma. Pediatrics. 2006, 118: e1715-20. 10.1542/peds.2006-1119.

    Article  PubMed  Google Scholar 

  32. Ortega AN, Goodwin RD, McQuaid EL, Canino G: Parental mental health, childhood psychiatric disorders, and asthma attacks in island Puerto Rican youth. Ambul Pediatr. 2004, 4: 308-15. 10.1367/A03-169R1.1.

    Article  PubMed  Google Scholar 

  33. Wamboldt MZ, Hewitt JK, Schmitz S: Familial association between allergic disorders and depression in adult Finnish twins. Am J Med Genet. 2000, 96: 146-53. 10.1002/(SICI)1096-8628(20000403)96:2<146::AID-AJMG4>3.0.CO;2-J.

    Article  CAS  PubMed  Google Scholar 

  34. Fantuzzi G: Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol. 2005, 115: 911-9. 10.1016/j.jaci.2005.02.023.

    Article  CAS  PubMed  Google Scholar 

  35. Weiss ST, Shore S: Obesity and asthma: directions for research. Am J Respir Crit Care Med. 2004, 169: 963-8. 10.1164/rccm.200303-403WS.

    Article  PubMed  Google Scholar 

  36. Ford ES: The epidemiology of obesity and asthma. J Allergy Clin Immunol. 2005, 115: 897-909. 10.1016/j.jaci.2004.11.050.

    Article  PubMed  Google Scholar 

  37. Fredberg JJ, Jones KA, Nathan M: Friction in airway smooth muscle: mechanism, latch, and implications in asthma. J Appl Physiol. 1996, 81: 2703-12.

    CAS  PubMed  Google Scholar 

  38. Beuther DA, Weiss ST, Sutherland ER: Obesity and asthma. Am J Respir Crit Care Med. 2006, 174: 112-9. 10.1164/rccm.200602-231PP.

    Article  PubMed Central  PubMed  Google Scholar 

  39. Sierra-Honigmann MR, Nath AK, Murakami C: Biological action of leptin as an angiogenic factor. Science. 1998, 281: 1683-6. 10.1126/science.281.5383.1683.

    Article  CAS  PubMed  Google Scholar 

  40. McElroy SL, Kotwal R, Malhotra S: Are mood disorders and obesity related? A review for the mental health professional. J Clin Psychiatry. 2004, 65: 634-51. 10.4088/JCP.v65n0507.

    Article  PubMed  Google Scholar 

  41. Faith MS, Matz PE, Jorge MA: Obesity-depression associations in the population. J Psychosom Res. 2002, 53: 935-42. 10.1016/S0022-3999(02)00308-2.

    Article  PubMed  Google Scholar 

  42. Bjorntorp P, Rosmond R: Obesity and cortisol. Nutrition. 2000, 16: 924-36. 10.1016/S0899-9007(00)00422-6.

    Article  CAS  PubMed  Google Scholar 

  43. Lambert GW, Vaz M, Cox HS: Human obesity is associated with a chronic elevation in brain 5-hydroxytryptamine turnover. Clin Sci (Lond). 1999, 96: 191-7. 10.1042/CS19980197.

    Article  CAS  Google Scholar 

  44. Wang GJ, Volkow ND, Logan J: Brain dopamine and obesity. Lancet. 2001, 357: 354-7. 10.1016/S0140-6736(00)03643-6.

    Article  CAS  PubMed  Google Scholar 

  45. Das UN: Is obesity an inflammatory condition?. Nutrition. 2001, 17: 953-66. 10.1016/S0899-9007(01)00672-4.

    Article  CAS  PubMed  Google Scholar 

  46. Kop WJ, Gottdiener JS, Tangen CM: Inflammation and coagulation factors in persons >65 years of age with symptoms of depression but without evidence of myocardial ischemia. Am J Cardiol. 2002, 89: 419-24. 10.1016/S0002-9149(01)02264-0.

    Article  CAS  PubMed  Google Scholar 

  47. Fergusson DM, Woodward LJ, Horwood LJ: Maternal smoking during pregnancy and psychiatric adjustment in late adolescence. Arch Gen Psychiatry. 1998, 55: 721-7. 10.1001/archpsyc.55.8.721.

    Article  CAS  PubMed  Google Scholar 

  48. Ng SP, Zelikoff JT: Smoking during pregnancy: subsequent effects on offspring immune competence and disease vulnerability in later life. Reprod Toxicol. 2006

    Google Scholar 

  49. Wu LT, Anthony JC: Tobacco smoking and depressed mood in late childhood and early adolescence. Am J Public Health. 1999, 89: 1837-40. 10.2105/AJPH.89.12.1837.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Maritz GS, Morley CJ, Harding R: Early developmental origins of impaired lung structure and function. Early Hum Dev. 2005, 81: 763-71. 10.1016/j.earlhumdev.2005.07.002.

    Article  PubMed  Google Scholar 

  51. Singh SP, Barrett EG, Kalra R: Prenatal cigarette smoke decreases lung cAMP and increases airway hyperresponsiveness. Am J Respir Crit Care Med. 2003, 168: 342-7. 10.1164/rccm.200211-1262OC.

    Article  PubMed  Google Scholar 

  52. Chalmers GW, MacLeod KJ, Little SA: Influence of cigarette smoking on inhaled corticosteroid treatment in mild asthma. Thorax. 2002, 57: 226-30. 10.1136/thorax.57.3.226.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  53. Chaudhuri R, Livingston E, McMahon AD: Cigarette smoking impairs the therapeutic response to oral corticosteroids in chronic asthma. Am J Respir Crit Care Med. 2003, 168: 1308-11. 10.1164/rccm.200304-503OC.

    Article  PubMed  Google Scholar 

  54. Okamoto K, Tanaka H, Ogawa H: Redox-dependent regulation of nuclear import of the glucocorticoid receptor. J Bio Chem. 1999, 274: 10363-71. 10.1074/jbc.274.15.10363.

    Article  CAS  Google Scholar 

  55. Ito K, Hanazawa T, Tomita K: Oxidative stress reduces histone deacetylase 2 activity and enhances IL-8 gene expression: role of tyrosine nitration. Biochem Biophys Res Commun. 2004, 315: 240-5. 10.1016/j.bbrc.2004.01.046.

    Article  CAS  PubMed  Google Scholar 

  56. Katsoulis K, Kontakiotis T, Leonardopoulos I: Serum total antioxidant status in severe exacerbation of asthma: correlation with the severity of the disease. J Asthma. 2003, 40: 847-54. 10.1081/JAS-120023575.

    Article  CAS  PubMed  Google Scholar 

  57. Katon WJ, Richardson L, Lozano P, McCauley E: The relationship of asthma and anxiety disorders. Psychosom Med. 2004, 66: 349-55. 10.1097/01.psy.0000126202.89941.ea.

    PubMed  Google Scholar 

  58. Katon WJ, Richardson L, Russo J: Quality of mental health care for youth with asthma and comorbid anxiety and depression. Med Care. 2006, 44: 1064-72. 10.1097/01.mlr.0000237421.17555.8f.

    Article  PubMed  Google Scholar 

  59. Kelleher KJ, Horwitz SM: Quality of mental health care for children: a familiar storyline. Med Care. 2006, 44: 1061-3. 10.1097/01.mlr.0000250626.81967.62.

    Article  PubMed  Google Scholar 

  60. Brown ES, Vigil L, Khan DA: A randomized trial of citalopram versus placebo in outpatients with asthma and major depressive disorder: a proof of concept study. Biol Psychiatry. 2005, 58: 865-70. 10.1016/j.biopsych.2005.04.030.

    Article  CAS  PubMed  Google Scholar 

  61. Sanger MD: Depression and allergic dermatologic disease. Psychosomatics. 1969, 10: 25-7.

    CAS  PubMed  Google Scholar 

  62. Allen LA, Woolfolk RL, Escobar JL: Cognitive-behavioral therapy for somatization disorder: a randomized controlled trial. Arch Intern Med. 2006, 166: 1512-8. 10.1001/archinte.166.14.1512.

    Article  PubMed  Google Scholar 

  63. Wright RJ, Kindlon D, Tollerud D: Does maternal stress influence polarization of TH2-dominated cytokine production and total IgE in maternal and cord blood?. Am J Resp Crit Care Med. 1999, 159: A103-

    Google Scholar 

  64. Elenkov IJ, Webster EL, Torpy DJ, Chrousos GP: Stress, corticotropin-releasing hormone, glucocorticoids, and the immune/inflammatory response: acute and chronic effects. Ann N Y Acad Sci. 1999, 876: 1-11. 10.1111/j.1749-6632.1999.tb07618.x.

    Article  CAS  PubMed  Google Scholar 

  65. Shanks N, Lightman SL: The maternal-neonatal neuro-immune interface: are there long-term implications for inflammatory or stress-related disease?. J Clin Invest. 2001, 108: 1567-73.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  66. Gluckman PD, Cutfield W, Hofman P, Hanson MA: The fetal, neonatal, and infant environments--the long-term consequences for disease risk. Early Hum Dev. 2005, 81: 51-9. 10.1016/j.earlhumdev.2004.10.003.

    Article  PubMed  Google Scholar 

  67. Barker DJ: A new model for the origins of chronic disease. Med Health Care Philos. 2001, 4: 31-5. 10.1023/A:1009934412988.

    Article  CAS  PubMed  Google Scholar 

  68. Morgan WJ, Martinez FD: Risk factors for developing wheezing and asthma in childhood. Pediatr Clin North Am. 1992, 39: 1185-203.

    CAS  PubMed  Google Scholar 

  69. Klinnert MD, Mrazek PJ, Mrazek DA: Early asthma onset: the interaction between family stressors and adaptive parenting. Psychiatry. 1994, 57: 51-61.

    CAS  PubMed  Google Scholar 

  70. Klinnert MD, Nelson HS, Price MR: Onset and persistence of childhood asthma: predictors from infancy. Pediatrics. 2001, 108: E69-10.1542/peds.108.4.e69.

    Article  CAS  PubMed  Google Scholar 

  71. Weil CM, Wade SL, Bauman LJ: The relationship between psychosocial factors and asthma morbidity in inner-city children with asthma. Pediatrics. 1999, 104: 1274-80. 10.1542/peds.104.6.1274.

    Article  CAS  PubMed  Google Scholar 

  72. Leung DY, Spahn JD, Szefler SJ: Immunologic basis and management of steroid-resistant asthma. Allergy Asthma Proc. 1999, 20: 9-14. 10.2500/108854199778681512.

    Article  CAS  PubMed  Google Scholar 

  73. Bumbacea D, Campbell D, Nguyen L: Parameters associated with persistent airflow obstruction in chronic severe asthma. Eur Respir J. 2004, 24: 122-8. 10.1183/09031936.04.00077803.

    Article  CAS  PubMed  Google Scholar 

  74. Pariante CM, Miller AH: Glucocorticoid receptors in major depression: relevance to pathophysiology and treatment. Biol Psychiatry. 2001, 49: 391-404. 10.1016/S0006-3223(00)01088-X.

    Article  CAS  PubMed  Google Scholar 

  75. Brown ES, Varghese FP, McEwen BS: Association of depression with medical illness: does cortisol play a role?. Biol Psychiatry. 2004, 55: 1-9. 10.1016/S0006-3223(03)00473-6.

    Article  CAS  PubMed  Google Scholar 

  76. Arana GW, Baldessarini RJ, Ornsteen M: The dexamethasone suppression test for diagnosis and prognosis in psychiatry. Commentary and review. Arch Gen Psychiatry. 1985, 42: 1193-204.

    Article  CAS  PubMed  Google Scholar 

  77. Lenox RH, Peyser JM, Rothschild B: Failure to normalize the dexamethasone suppression test: association with length of illness. Biol Psychiatry. 1985, 20: 333-7. 10.1016/0006-3223(85)90064-2.

    Article  CAS  PubMed  Google Scholar 

  78. Banki CM, Karmacsi L, Bissette G, Nemeroff CB: CSF corticotropin-releasing hormone and somatostatin in major depression: response to antidepressant treatment and relapse. Eur Neuropsychopharmacol. 1992, 2: 107-13. 10.1016/0924-977X(92)90019-5.

    Article  CAS  PubMed  Google Scholar 

  79. Wong ML, Kling MA, Munson PJ: Pronounced and sustained central hypernoradrenergic function in major depression with melancholic features: relation to hypercortisolism and corticotropin-releasing hormone. Proc Natl Acad Sci USA. 2000, 97: 325-30. 10.1073/pnas.97.1.325.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  80. Roy A, Pickar D, Paul S: CSF corticotropin-releasing hormone in depressed patients and normal control subjects. Am J Psychiatry. 1987, 144: 641-5.

    Article  CAS  PubMed  Google Scholar 

  81. Nemeroff CB, Bissette G, Akil H, Fink M: Neuropeptide concentrations in the cerebrospinal fluid of depressed patients treated with electroconvulsive therapy. Corticotrophin-releasing factor, beta-endorphin and somatostatin. Br J Psychiatry. 1991, 158: 59-63. 10.1192/bjp.158.1.59.

    Article  CAS  PubMed  Google Scholar 

  82. Heuser I, Bissette G, Dettling M: Cerebrospinal fluid concentrations of corticotropin-releasing hormone, vasopressin, and somatostatin in depressed patients and healthy controls: response to amitriptyline treatment. Depress Anxiety. 1998, 8: 71-9. 10.1002/(SICI)1520-6394(1998)8:2<71::AID-DA5>3.0.CO;2-N.

    Article  CAS  PubMed  Google Scholar 

  83. Pace TW, Hu F, Miller AH: Cytokine effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav Immun. 2007, 21: 9-19. 10.1016/j.bbi.2006.08.009.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Watkins LR, Maier SF: Immune regulation of central nervous system functions: from sickness responses to pathological pain. J Intern Med. 2005, 257: 139-55. 10.1111/j.1365-2796.2004.01443.x.

    Article  CAS  PubMed  Google Scholar 

  85. Raison CL, Capuron L, Miller AH: Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol. 2006, 27: 24-31. 10.1016/j.it.2005.11.006.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  86. Wichers MC, Maes M: The role of indoleamine 2,3-dioxygenase (IDO) in the pathophysiology of interferon-alpha-induced depression. J Psychiatry Neurosci. 2004, 29: 11-7.

    PubMed Central  PubMed  Google Scholar 

  87. Raison CL, Miller AH: Depression in cancer: new developments regarding diagnosis and treatment. Biol Psychiatry. 2003, 54: 283-94. 10.1016/S0006-3223(03)00413-X.

    Article  PubMed  Google Scholar 

  88. Zorrilla EP, Luborsky L, McKay JR: The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav Immun. 2001, 15: 199-226. 10.1006/brbi.2000.0597.

    Article  CAS  PubMed  Google Scholar 

  89. Doganci A, Eigenbrod T, Krug N: The IL-6R alpha chain controls lung CD4+CD25+ Treg development and function during allergic airway inflammation in vivo. J Clin Invest. 2005, 115: 313-25.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  90. Dinarello CA: Biologic basis for interleukin-1 in disease. Blood. 1996, 87: 2095-147.

    CAS  PubMed  Google Scholar 

  91. Whelan R, Kim C, Chen M: Role and regulation of interleukin-1 molecules in pro-asthmatic sensitised airway smooth muscle. Eur Respir J. 2004, 24: 559-67. 10.1183/09031936.04.00133803.

    Article  CAS  PubMed  Google Scholar 

  92. Johnson VJ, Yucesoy B, Luster MI: Prevention of IL-1 signaling attenuates airway hyperresponsiveness and inflammation in a murine model of toluene diisocyanate-induced asthma. J Allergy Clin Immunol. 2005, 116: 851-8. 10.1016/j.jaci.2005.07.008.

    Article  CAS  PubMed  Google Scholar 

  93. Dinarello CA: The IL-1 family and inflammatory diseases. Clin Exp Rheumatol. 2002, 20: S1-13.

    CAS  PubMed  Google Scholar 

  94. Song C, Horrobin DF, Leonard BE: The comparison of changes in behavior, neurochemistry, endocrine, and immune functions after different routes, doses and durations of administrations of IL-1beta in rats. Pharmacopsychiatry. 2006, 39: 88-99. 10.1055/s-2006-941557.

    Article  CAS  PubMed  Google Scholar 

  95. Witkamp R, Monshouwer M: Signal transduction in inflammatory processes, current and future therapeutic targets: a mini review. Vet Q. 2000, 22: 11-6.

    Article  CAS  PubMed  Google Scholar 

  96. Raison CL, Miller AH: When not enough is too much: the role of insufficient glucocorticoid signaling in the pathophysiology of stress-related disorders. Am J Psychiatry. 2003, 160: 1554-65. 10.1176/appi.ajp.160.9.1554.

    Article  PubMed  Google Scholar 

  97. Kampf C, Relova AJ, Sandler S, Roomans GM: Effects of TNF-alpha, IFN-gamma and IL-beta on normal human bronchial epithelial cells. Eur Respir J. 1999, 14: 84-91. 10.1034/j.1399-3003.1999.14a15.x.

    Article  CAS  PubMed  Google Scholar 

  98. Howarth PH, Babu KS, Arshad HS: Tumour necrosis factor (TNFalpha) as a novel therapeutic target in symptomatic corticosteroid dependent asthma. Thorax. 2005, 60: 1012-8. 10.1136/thx.2005.045260.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  99. Berry MA, Hargadon B, Shelley M: Evidence of a role of tumor necrosis factor alpha in refractory asthma. N Engl J Med. 2006, 354: 697-708. 10.1056/NEJMoa050580.

    Article  CAS  PubMed  Google Scholar 

  100. O'Byrne PM: Cytokines or their antagonists for the treatment of asthma. Chest. 2006, 130: 244-50. 10.1378/chest.130.1.244.

    Article  PubMed  CAS  Google Scholar 

  101. Bentley A, Ying S, Gaga M: Tissue eosinophilia and increased numbers of cell expressing mRNA for IL-4 and IL-5 occur in asthma but not bronchiectasis. J Allergy Clin Immunol. 1998, 101: S107-44.

    Google Scholar 

  102. Zangrilli JG, Shaver JR, Cirelli RA: sVCAM-1 levels after segmental antigen challenge correlate with eosinophil influx, IL-4 and IL-5 production, and the late phase response. Am J Respir Crit Care Med. 1995, 151: 1346-53.

    Article  CAS  PubMed  Google Scholar 

  103. Pavon L, Sandoval-Lopez G, Eugenia HM: Th2 cytokine response in major depressive disorder patients before treatment. J Neuroimmunol. 2006, 172: 156-65. 10.1016/j.jneuroim.2005.08.014.

    Article  CAS  PubMed  Google Scholar 

  104. Padgett DA, Sheridan JF, Loria RM: Steroid hormone regulation of a polyclonal TH2 immune response. Ann N Y Acad Sci. 1995, 774: 323-5. 10.1111/j.1749-6632.1995.tb17398.x-i1.

    Article  CAS  PubMed  Google Scholar 

  105. Mendlovic S, Mozes E, Eilat E: Immune activation in nontreated suicidal major depression. Immunol Lett. 1999, 67: 105-8. 10.1016/S0165-2478(98)00145-X.

    Article  CAS  PubMed  Google Scholar 

  106. Kumar A, Takada Y, Boriek AM, Aggarwal BB: Nuclear factor-kappaB: its role in health and disease. J Mol Med. 2004, 82: 434-48. 10.1007/s00109-004-0555-y.

    Article  CAS  PubMed  Google Scholar 

  107. Bethea CL, Reddy AP, Smith LJ: Nuclear factor kappa B in the dorsal raphe of macaques: an anatomical link for steroids, cytokines and serotonin. J Psychiatry Neurosci. 2006, 31: 105-14.

    PubMed Central  PubMed  Google Scholar 

  108. Christman JW, Sadikot RT, Blackwell TS: The role of nuclear factor-kappa B in pulmonary diseases. Chest. 2000, 117: 1482-7. 10.1378/chest.117.5.1482.

    Article  CAS  PubMed  Google Scholar 

  109. Hancox RJ, Stevens DA, Adcock IM: Effects of inhaled beta agonist and corticosteroid treatment on nuclear transcription factors in bronchial mucosa in asthma. Thorax. 1999, 54: 488-92. 10.1136/thx.54.6.488.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  110. Bowler RP: Oxidative stress in the pathogenesis of asthma. Curr Allergy Asthma Rep. 2004, 4: 116-22. 10.1007/s11882-004-0056-7.

    Article  PubMed  Google Scholar 

  111. Liao MF, Chen CC, Hsu MH: Evaluation of the serum antioxidant status in asthmatic children. Acta Paediatr Taiwan. 2004, 45: 213-7.

    PubMed  Google Scholar 

  112. Nadeem A, Raj HG, Chhabra SK: Increased oxidative stress in acute exacerbations of asthma. J Asthma. 2005, 42: 45-50. 10.1081/JAS-200044774.

    Article  CAS  PubMed  Google Scholar 

  113. Monteseirin J, Bonilla I, Camacho J: Elevated secretion of myeloperoxidase by neutrophils from asthmatic patients: the effect of immunotherapy. J Allergy Clin Immunol. 2001, 107: 623-6. 10.1067/mai.2001.113566.

    Article  CAS  PubMed  Google Scholar 

  114. Mak JC, Chan-Yeung MM: Reactive oxidant species in asthma. Curr Opin Pulm Med. 2006, 12: 7-11. 10.1097/01.mcp.0000198067.50457.71.

    Article  CAS  PubMed  Google Scholar 

  115. Wright RJ, Cohen RT, Cohen S: The impact of stress on the development and expression of atopy. Curr Opin Allergy Clin Immunol. 2005, 5: 23-9. 10.1097/00130832-200502000-00006.

    Article  PubMed  Google Scholar 

  116. Yamaguchi T, Shioji I, Sugimoto A, Yamaoka M: Psychological stress increases bilirubin metabolites in human urine. Biochem Biophys Res Commun. 2002, 293: 517-20. 10.1016/S0006-291X(02)00233-4.

    Article  CAS  PubMed  Google Scholar 

  117. Cernak I, Savic V, Kotur J: Alterations in magnesium and oxidative status during chronic emotional stress. Magnes Res. 2000, 13: 29-36.

    CAS  PubMed  Google Scholar 

  118. McDaniel JS, Musselman DL, Porter MR: Depression in patients with cancer. diagnosis, biology, and treatment. Arch Gen Psychiatry. 1995, 52: 89-99.

    Article  CAS  PubMed  Google Scholar 

  119. Tsuboi H, Shimoi K, Kinae N: Depressive symptoms are independently correlated with lipid peroxidation in a female population: comparison with vitamins and carotenoids. J Psychosom Res. 2004, 56: 53-8. 10.1016/S0022-3999(03)00567-1.

    Article  PubMed  Google Scholar 

  120. Peet M, Murphy B, Shay J, Horrobin D: Depletion of omega-3 fatty acid levels in red blood cell membranes of depressive patients. Biol Psychiatry. 1998, 43: 315-9. 10.1016/S0006-3223(97)00206-0.

    Article  CAS  PubMed  Google Scholar 

  121. Irie M, Miyata M, Kasai H: Depression and possible cancer risk due to oxidative DNA damage. J Psychiatr Res. 2005, 39: 553-60. 10.1016/j.jpsychires.2005.01.009.

    Article  PubMed  Google Scholar 

  122. Forlenza MJ, Miller GE: Increased serum levels of 8-hydroxy-2'-deoxyguanosine in clinical depression. Psychosom Med. 2006, 68: 1-7. 10.1097/01.psy.0000195780.37277.2a.

    Article  CAS  PubMed  Google Scholar 

  123. Herbert TB, Cohen S: Depression and immunity: a meta-analytic review. Psychol Bull. 1993, 113: 472-86. 10.1037/0033-2909.113.3.472.

    Article  CAS  PubMed  Google Scholar 

  124. Miller GE, Freedland KE, Carney RM: Pathways linking depression, adiposity, and inflammatory markers in healthy young adults. Brain Behav Immun. 2003, 17: 276-85. 10.1016/S0889-1591(03)00057-6.

    Article  CAS  PubMed  Google Scholar 

  125. Yao JK, Reddy RD: Metabolic investigation in psychiatric disorders. Mol Neurobiol. 2005, 31: 193-203. 10.1385/MN:31:1-3:193.

    Article  CAS  PubMed  Google Scholar 

  126. Stanciu LA, Djukanovic R: The role of ICAM-1 on T-cells in the pathogenesis of asthma. Eur Respir J. 1998, 11: 949-57. 10.1183/09031936.98.11040949.

    Article  CAS  PubMed  Google Scholar 

  127. Tang ML, Fiscus LC: Important roles for L-selectin and ICAM-1 in the development of allergic airway inflammation in asthma. Pulm Pharmacol Ther. 2001, 14: 203-10. 10.1006/pupt.2001.0293.

    Article  CAS  PubMed  Google Scholar 

  128. Montefort S, Lai CK, Kapahi P: Circulating adhesion molecules in asthma. Am J Respir Crit Care Med. 1994, 149: 1149-52.

    Article  CAS  PubMed  Google Scholar 

  129. Gonokami Y, Konno S, Kurokawa M: Circulating intracellular adhesion molecule-1 concentrations following bronchial provocation in atopic asthma. Int Arch Allergy Immunol. 1997, 112: 386-91. 10.1159/000237485.

    Article  CAS  PubMed  Google Scholar 

  130. Gorska-Ciebiada M, Ciebiada M, Gorska MM: Intercellular adhesion molecule 1 and tumor necrosis factor alpha in asthma and persistent allergic rhinitis: relationship with disease severity. Ann Allergy Asthma Immunol. 2006, 97: 66-72. 10.1016/S1081-1206(10)61372-5.

    Article  CAS  PubMed  Google Scholar 

  131. Thomas AJ, Ferrier IN, Kalaria RN: Elevation in late-life depression of intercellular adhesion molecule-1 expression in the dorsolateral prefrontal cortex. Am J Psychiatry. 2000, 157: 1682-4. 10.1176/appi.ajp.157.10.1682.

    Article  CAS  PubMed  Google Scholar 

  132. Rajagopalan S, Brook R, Rubenfire M: Abnormal brachial artery flow-mediated vasodilation in young adults with major depression. Am J Cardiol. 2001, 88: 196-8. 10.1016/S0002-9149(01)01623-X. A7.

    Article  CAS  PubMed  Google Scholar 

  133. Lesperance F, Frasure-Smith N, Theroux P, Irwin M: The association between major depression and levels of soluble intercellular adhesion molecule 1, interleukin-6, and C-reactive protein in patients with recent acute coronary syndromes. Am J Psychiatry. 2004, 161: 271-7. 10.1176/appi.ajp.161.2.271.

    Article  PubMed  Google Scholar 

  134. Empana JP, Sykes DH, Luc G: Contributions of depressive mood and circulating inflammatory markers to coronary heart disease in healthy European men: the Prospective Epidemiological Study of Myocardial Infarction (PRIME). Circulation. 2005, 111: 2299-305. 10.1161/01.CIR.0000164203.54111.AE.

    Article  CAS  PubMed  Google Scholar 

  135. Dimopoulos N, Piperi C, Salonicioti A: Elevation of plasma concentration of adhesion molecules in late-life depression. Int J Geriatr Psychiatry. 2006, 21: 965-71. 10.1002/gps.1592.

    Article  PubMed  Google Scholar 

  136. Schaefer M, Horn M, Schmidt F: Correlation between sICAM-1 and depressive symptoms during adjuvant treatment of melanoma with interferon-alpha. Brain Behav Immun. 2004, 18: 555-62. 10.1016/j.bbi.2004.02.002.

    Article  CAS  PubMed  Google Scholar 

  137. Park GY, Christman JW: Involvement of cyclooxygenase-2 and prostaglandins in the molecular pathogenesis of inflammatory lung diseases. Am J Physiol Lung Cell Mol Physiol. 2006, 290: L797-805. 10.1152/ajplung.00513.2005.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  138. Yaksh TL, Dirig DM, Conway CM: The acute antihyperalgesic action of nonsteroidal, anti-inflammatory drugs and release of spinal prostaglandin E2 is mediated by the inhibition of constitutive spinal cyclooxygenase-2 (COX-2) but not COX-1. J Neurosci. 2001, 21: 5847-53.

    CAS  PubMed  Google Scholar 

  139. Muller N, Schwarz MJ, Dehning S: The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: results of a double-blind, randomized, placebo controlled, add-on pilot study to reboxetine. Mol Psychiatry. 2006, 11: 680-4. 10.1038/sj.mp.4001805.

    Article  CAS  PubMed  Google Scholar 

  140. Collantes-Esteves E, Fernandez-Perrez Ch: Improved self-control of ostheoarthritis pain and self-reported health status in nonresponders to celecoxib switched to rofecoxib: results of PAVIA, an open-label post-marketing survey in Spain. Curr Med Res Opin. 2003, 19: 402-10. 10.1185/030079903125001938.

    Article  CAS  Google Scholar 

  141. Devillier P, Bessard G: Thromboxane A2 and related prostaglandins in airways. Fundam Clin Pharmacol. 1997, 11: 2-18. 10.1111/j.1472-8206.1997.tb00163.x.

    Article  CAS  PubMed  Google Scholar 

  142. Rolin S, Masereel B, Dogne JM: Prostanoids as pharmacological targets in COPD and asthma. Eur J Pharmacol. 2006, 533: 89-100. 10.1016/j.ejphar.2005.12.058.

    Article  CAS  PubMed  Google Scholar 

  143. Linnoila M, Whorton AR, Rubinow DR: CSF prostaglandin levels in depressed and schizophrenic patients. Arch Gen Psychiatry. 1983, 40: 405-6.

    Article  CAS  PubMed  Google Scholar 

  144. Calabrese JR, Skwerer RG, Barna B: Depression, immunocompetence, and prostaglandins of the E series. Psychiatry Res. 1986, 17: 41-7. 10.1016/0165-1781(86)90040-5.

    Article  CAS  PubMed  Google Scholar 

  145. Ohishi K, Ueno R, Nishino S: Increased level of salivary prostaglandins in patients with major depression. Biol Psychiatry. 1988, 23: 326-34. 10.1016/0006-3223(88)90283-1.

    Article  CAS  PubMed  Google Scholar 

  146. Nishino S, Ueno R, Ohishi K: Salivary prostaglandin concentrations: possible state indicators for major depression. Am J Psychiatry. 1989, 146: 365-8.

    Article  CAS  PubMed  Google Scholar 

  147. Roberts LJ, Sweetman BJ, Lewis RA: Increased production of prostaglandin D2 in patients with systemic mastocytosis. N Engl J Med. 1980, 303: 1400-4. 10.1056/NEJM198012113032405.

    Article  PubMed  Google Scholar 

  148. Forstermann U, Heldt R, Hertting G: Effects of intracerebroventricular administration of prostaglandin D2 on behaviour, blood pressure and body temperature as compared to prostaglandins E2 and F2 alpha. Psychopharmacology (Berl). 1983, 80: 365-70. 10.1007/BF00432122.

    Article  CAS  Google Scholar 

  149. Ueno R, Ishikawa Y, Nakayama T, Hayaishi O: Prostaglandin D2 induces sleep when microinjected into the preoptic area of conscious rats. Biochem Biophys Res Commun. 1982, 109: 576-82. 10.1016/0006-291X(82)91760-0.

    Article  CAS  PubMed  Google Scholar 

  150. Coceani F: Prostaglandins and the central nervous system. Arch Intern Med. 1974, 133: 119-29. 10.1001/archinte.133.1.119.

    Article  CAS  PubMed  Google Scholar 

  151. Yirmiya R, Barak O, Avitsur R: Intracerebral administration of Mycoplasma fermentans produces sickness behavior: role of prostaglandins. Brain Res. 1997, 749: 71-81. 10.1016/S0006-8993(96)01295-4.

    Article  CAS  PubMed  Google Scholar 

  152. Houslay M, Shafer P, Zhang K: Phosphodiesterase-4 as a therapeutic target. Drug Discov Today. 2005, 10: 1503-19. 10.1016/S1359-6446(05)03622-6.

    Article  CAS  PubMed  Google Scholar 

  153. Mata M, Sarria B, Buenestado A: Phosphodiesterase 4 inhibition decreases MUC5AC expression induced by epidermal growth factor in human airway epithelial cells. Thorax. 2005, 60: 144-52. 10.1136/thx.2004.025692.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  154. Sanz MJ, Cortijo J, Morcillo EJ: PDE4 inhibitors as new anti-inflammatory drugs: effects on cell trafficking and cell adhesion molecules expression. Pharmacol Ther. 2005, 106: 269-97. 10.1016/j.pharmthera.2004.12.001.

    Article  CAS  PubMed  Google Scholar 

  155. Chang K: Phosphodiesterase inhibitors in airways disease. Eur J Pharmacol. 2006, 533: 110-7. 10.1016/j.ejphar.2005.12.059.

    Article  CAS  Google Scholar 

  156. Manji HK, Drevets WC, Charney DS: The cellular neurobiology of depression. Nat Med. 2001, 7: 541-7. 10.1038/87865.

    Article  CAS  PubMed  Google Scholar 

  157. Cherry JA, Davis RL: Cyclic AMP phosphodiesterases are localized in regions of the mouse brain associated with reinforcement, movement, and affect. J Comp Neurol. 1999, 407: 287-301. 10.1002/(SICI)1096-9861(19990503)407:2<287::AID-CNE9>3.0.CO;2-R.

    Article  CAS  PubMed  Google Scholar 

  158. O'Donnell JM, Zhang HT: Antidepressant effects of inhibitors of cAMP phosphodiesterase (PDE4). Trends Pharmacol Sci. 2004, 25: 158-63. 10.1016/j.tips.2004.01.003.

    Article  PubMed  CAS  Google Scholar 

  159. Nakagawa S, Kim JE, Lee R: Regulation of neurogenesis in adult mouse hippocampus by cAMP and the cAMP response element-binding protein. J Neurosci. 2002, 22: 3673-82.

    CAS  PubMed  Google Scholar 

  160. Santarelli L, Saxe M, Gross C: Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003, 301: 805-9. 10.1126/science.1083328.

    Article  CAS  PubMed  Google Scholar 

  161. Zhang HT, Huang Y, Jin SL: Antidepressant-like profile and reduced sensitivity to rolipram in mice deficient in the PDE4D phosphodiesterase enzyme. Neuropsychopharmacology. 2002, 27: 587-95.

    CAS  PubMed  Google Scholar 

  162. Handsley MM, Edwards DR: Metalloproteinases and their inhibitors in tumor angiogenesis. Int J Cancer. 2005, 115: 849-60. 10.1002/ijc.20945.

    Article  CAS  PubMed  Google Scholar 

  163. Gueders MM, Foidart JM, Noel A, Cataldo DD: Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the respiratory tract: potential implications in asthma and other lung diseases. Eur J Pharmacol. 2006, 533: 133-44. 10.1016/j.ejphar.2005.12.082.

    Article  CAS  PubMed  Google Scholar 

  164. Cataldo DD, Tournoy KG, Vermaelen K: Matrix metalloproteinase-9 deficiency impairs cellular infiltration and bronchial hyperresponsiveness during allergen-induced airway inflammation. Am J Pathol. 2002, 161: 491-8. 10.1016/S0002-9440(10)64205-8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  165. Vermaelen KY, Cataldo D, Tournoy K: Matrix metalloproteinase-9-mediated dendritic cell recruitment into the airways is a critical step in a mouse model of asthma. J Immunol. 2003, 171: 1016-22.

    Article  CAS  PubMed  Google Scholar 

  166. Yong VW: Metalloproteinases: mediators of pathology and regeneration in the CNS. Nat Rev Neurosci. 2005, 6: 931-44. 10.1038/nrn1807.

    Article  CAS  PubMed  Google Scholar 

  167. Yong VW, Power C, Forsyth P, Edwards DR: Metalloproteinases in biology and pathology of the nervous system. Nat Rev Neurosci. 2001, 2: 502-11. 10.1038/35081571.

    Article  CAS  PubMed  Google Scholar 

  168. Yang EV, Bane CM, MacCallum RC: Stress-related modulation of matrix metalloproteinase expression. J Neuroimmunol. 2002, 133: 144-50. 10.1016/S0165-5728(02)00270-9.

    Article  CAS  PubMed  Google Scholar 

  169. Akdis CA, Simons FE: Histamine receptors are hot in immunopharmacology. Eur J Pharmacol. 2006, 533: 69-76. 10.1016/j.ejphar.2005.12.044.

    Article  CAS  PubMed  Google Scholar 

  170. Elenkov IJ, Chrousos GP: Stress hormones, Th1/Th2 patterns, pro/anti-inflammatory cytokines and susceptibility to disease. Trends Endocrinol Metab. 1999, 10: 359-68. 10.1016/S1043-2760(99)00188-5.

    Article  CAS  PubMed  Google Scholar 

  171. Baena-Cagnani CE, Berger WE, DuBuske LM: Comparative effects of desloratadine versus montelukast on asthma symptoms and use of beta 2-agonists in patients with seasonal allergic rhinitis and asthma. Int Arch Allergy Immunol. 2003, 130: 307-13. 10.1159/000070218.

    Article  CAS  PubMed  Google Scholar 

  172. Simons FE: Advances in H1-antihistamines. N Engl J Med. 2004, 351: 2203-17. 10.1056/NEJMra033121.

    Article  CAS  PubMed  Google Scholar 

  173. Warner JO: A double-blinded, randomized, placebo-controlled trial of cetirizine in preventing the onset of asthma in children with atopic dermatitis: 18 months' treatment and 18 months' posttreatment follow-up. J Allergy Clin Immunol. 2001, 108: 929-37. 10.1067/mai.2001.120015.

    Article  CAS  PubMed  Google Scholar 

  174. Ito C, Shen H, Toyota H: Effects of the acute and chronic restraint stresses on the central histaminergic neuron system of Fischer rat. Neurosci Lett. 1999, 262: 143-5. 10.1016/S0304-3940(99)00052-X.

    Article  CAS  PubMed  Google Scholar 

  175. Ito C: The role of brain histamine in acute and chronic stresses. Biomed Pharmacother. 2000, 54: 263-7. 10.1016/S0753-3322(00)80069-4.

    Article  CAS  PubMed  Google Scholar 

  176. Stahl SM: The psychopharmacology of energy and fatigue. J Clin Psychiatry. 2002, 63: 7-8.

    Article  PubMed  Google Scholar 

  177. Russo C, Arcidiacono G, Polosa R: Adenosine receptors: promising targets for the development of novel therapeutics and diagnostics for asthma. Fundam Clin Pharmacol. 2006, 20: 9-19. 10.1111/j.1472-8206.2005.00388.x.

    Article  CAS  PubMed  Google Scholar 

  178. Cushley MJ, Tattersfield AE, Holgate ST: Inhaled adenosine and guanosine on airway resistance in normal and asthmatic subjects. Br J Clin Pharmacol. 1983, 15: 161-5.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  179. Huszar E, Vass G, Vizi E: Adenosine in exhaled breath condensate in healthy volunteers and in patients with asthma. Eur Respir J. 2002, 20: 1393-8. 10.1183/09031936.02.00005002.

    Article  CAS  PubMed  Google Scholar 

  180. Forsythe P, McGarvey LP, Heaney LG: Adenosine induces histamine release from human bronchoalveolar lavage mast cells. Clin Sci (Lond). 1999, 96: 349-55. 10.1042/CS19980228.

    Article  CAS  Google Scholar 

  181. Porsolt RD, Bertin A, Jalfre M: Behavioral despair in mice: a primary screening test for antidepressants. Arch Int Pharmacodyn Ther. 1977, 229: 327-36.

    CAS  PubMed  Google Scholar 

  182. Woodson JC, Minor TR, Job RF: Inhibition of adenosine deaminase by erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) mimics the effect of inescapable shock on escape learning in rats. Behav Neurosci. 1998, 112: 399-409. 10.1037/0735-7044.112.2.399.

    Article  CAS  PubMed  Google Scholar 

  183. El Yacoubi M, Ledent C, Parmentier M: Adenosine A2A receptor antagonists are potential antidepressants: evidence based on pharmacology and A2A receptor knockout mice. Br J Pharmacol. 2001, 134: 68-77. 10.1038/sj.bjp.0704240.

    Article  CAS  PubMed  Google Scholar 

  184. El Yacoubi M, Costentin J, Vaugeois JM: Adenosine A2A receptors and depression. Neurology. 2003, 61: S82-7.

    Article  CAS  PubMed  Google Scholar 

  185. Akyol O, Zoroglu SS, Armutcu F: Nitric oxide as a physiopathological factor in neuropsychiatric disorders. In Vivo. 2004, 18: 377-90.

    CAS  PubMed  Google Scholar 

  186. Redington AE: Modulation of nitric oxide pathways: therapeutic potential in asthma and chronic obstructive pulmonary disease. Eur J Pharmacol. 2006, 533: 263-76. 10.1016/j.ejphar.2005.12.069.

    Article  CAS  PubMed  Google Scholar 

  187. McLeod TM, Lopez-Figueroa AL, Lopez-Figueroa MO: Nitric oxide, stress, and depression. Psychopharmacol Bull. 2001, 35: 24-41.

    CAS  PubMed  Google Scholar 

  188. van Amsterdam JG, Opperhuizen A: Nitric oxide and biopterin in depression and stress. Psychiatry Res. 1999, 85: 33-8. 10.1016/S0165-1781(98)00140-1.

    Article  CAS  PubMed  Google Scholar 

  189. Bredt DS, Hwang PM, Snyder SH: Localization of nitric oxide synthase indicating a neural role for nitric oxide. Nature. 1990, 347: 768-70. 10.1038/347768a0.

    Article  CAS  PubMed  Google Scholar 

  190. Xu ZQ, Hokfelt T: Expression of galanin and nitric oxide synthase in subpopulations of serotonin neurons of the rat dorsal raphe nucleus. J Chem Neuroanat. 1997, 13: 169-87. 10.1016/S0891-0618(97)00043-4.

    Article  CAS  PubMed  Google Scholar 

  191. Groneberg DA, Quarcoo D, Frossard N, Fischer A: Neurogenic mechanisms in bronchial inflammatory diseases. Allergy. 2004, 59: 1139-52. 10.1111/j.1398-9995.2004.00665.x.

    Article  CAS  PubMed  Google Scholar 

  192. Fischer A, McGregor GP, Saria A: Induction of tachykinin gene and peptide expression in guinea pig nodose primary afferent neurons by allergic airway inflammation. J Clin Invest. 1996, 98: 2284-91. 10.1172/JCI119039.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  193. Komatsu T, Yamamoto M, Shimokata K, Nagura H: Distribution of substance P-immunoreactive and calcitonin gene-related peptide-immunoreactive nerves in normal human lungs. Int Arch Allergy Appl Immunol. 1991, 95: 23-8. 10.1159/000235449.

    Article  CAS  PubMed  Google Scholar 

  194. Crimi N, Oliveri R, Polosa R: Effect of oral terfenadine on bronchoconstrictor response to inhaled neurokinin A and histamine in asthmatic subjects. Eur Respir J. 1993, 6: 1462-7.

    CAS  PubMed  Google Scholar 

  195. Boichot E, Lagente V, Paubert-Braquet M, Frossard N: Inhaled substance P induces activation of alveolar macrophages and increases airway responses in the guinea-pig. Neuropeptides. 1993, 25: 307-13. 10.1016/0143-4179(93)90048-F.

    Article  CAS  PubMed  Google Scholar 

  196. Hokfelt T, Broberger C, Xu ZQ: Neuropeptides--an overview. Neuropharmacology. 2000, 39: 1337-56. 10.1016/S0028-3908(00)00010-1.

    Article  CAS  PubMed  Google Scholar 

  197. Kramer MS, Cutler N, Feighner J: Distinct mechanism for antidepressant activity by blockade of central substance P receptors. Science. 1998, 281: 1640-5. 10.1126/science.281.5383.1640.

    Article  CAS  PubMed  Google Scholar 

  198. Nemeroff CB, Mayberg HS, Krahl SE: VNS therapy in treatment-resistant depression: clinical evidence and putative neurobiological mechanisms. Neuropsychopharmacology. 2006, 31: 1345-55. 10.1038/sj.npp.1301082.

    Article  PubMed  Google Scholar 

  199. Galil N: Depression and asthma in children. Curr Opin Pediatr. 2000, 12: 331-5. 10.1097/00008480-200008000-00008.

    Article  CAS  PubMed  Google Scholar 

  200. Avni J, Bruderman I: The effect of amitriptyline on pulmonary ventilation and the mechanics of breathing. Psychopharmacologia. 1969, 14: 184-92. 10.1007/BF00404216.

    Article  CAS  PubMed  Google Scholar 

  201. Hofer MA: Cardiac and respiratory function during sudden prolonged immobility in wild rodents. Psychosom Med. 1970, 32: 633-47.

    Article  CAS  PubMed  Google Scholar 

  202. Miller BD, Strunk RC: Circumstances surrounding the deaths of children due to asthma. A case-control study. Am J Dis Child. 1989, 143: 1294-9.

    Article  CAS  PubMed  Google Scholar 

  203. Miller BD, Wood BL: Influence of specific emotional states on autonomic reactivity and pulmonary function in asthmatic children. J Am Acad Child Adolesc Psychiatry. 1997, 36: 669-77. 10.1097/00004583-199705000-00018.

    Article  CAS  PubMed  Google Scholar 

  204. Lewis MJ, Short AL, Lewis KE: Autonomic nervous system control of the cardiovascular and respiratory systems in asthma. Respir Med. 2006, 100: 1688-705. 10.1016/j.rmed.2006.01.019.

    Article  CAS  PubMed  Google Scholar 

  205. Kallenbach JM, Webster T, Dowdeswell R: Reflex heart rate control in asthma. Evidence of parasympathetic overactivity. Chest. 1985, 87: 644-8. 10.1378/chest.87.5.644.

    Article  CAS  PubMed  Google Scholar 

  206. Rechlin T, Weis M, Spitzer A, Kaschka WP: Are affective disorders associated with alterations of heart rate variability?. J Affect Disord. 1994, 32: 271-5. 10.1016/0165-0327(94)90091-4.

    Article  CAS  PubMed  Google Scholar 

  207. Dalack GW, Roose SP: Perspectives on the relationship between cardiovascular disease and affective disorder. J Clin Psychiatry. 1990, 51 (Suppl): 4-9.

    PubMed  Google Scholar 

  208. Yeragani VK, Pohl R, Balon R: Heart rate variability in patients with major depression. Psychiatry Res. 1991, 37: 35-46. 10.1016/0165-1781(91)90104-W.

    Article  CAS  PubMed  Google Scholar 

  209. Gehi A, Mangano D, Pipkin S: Depression and heart rate variability in patients with stable coronary heart disease: findings from the Heart and Soul Study. Arch Gen Psychiatry. 2005, 62: 661-6. 10.1001/archpsyc.62.6.661.

    Article  PubMed Central  PubMed  Google Scholar 

  210. Lotvall J, Lunde H, Augustinson LE: Airway effects of direct left-sided cervical vagal stimulation in patients with complex partial seizures. Epilepsy Res. 1994, 18: 149-54. 10.1016/0920-1211(94)90007-8.

    Article  CAS  PubMed  Google Scholar 

  211. Lechin F, Dijs van der B, Orozco B: Increased levels of free serotonin in plasma of symptomatic asthmatic patients. Ann Allergy Asthma Immunol. 1996, 77: 245-53. 10.1016/S1081-1206(10)63263-2.

    Article  CAS  PubMed  Google Scholar 

  212. Bayer H, Muller T, Myrtek D: Serotoninergic receptors on human airway epithelial cells. Am J Respir Cell Mol Biol. 2007, 36: 85-93. 10.1165/rcmb.2006-0151OC.

    Article  CAS  PubMed  Google Scholar 

  213. Young MR, Matthews JP: Serotonin regulation of T-cell subpopulations and of macrophage accessory function. Immunology. 1995, 84: 148-52.

    PubMed Central  CAS  PubMed  Google Scholar 

  214. Lechin F, van der DB, Orozco B: Neuropharmacologic treatment of bronchial asthma with the antidepressant tianeptine: a double-blind, crossover placebo-controlled study. Clin Pharmacol Ther. 1998, 64: 223-32. 10.1016/S0009-9236(98)90156-4.

    Article  CAS  PubMed  Google Scholar 

  215. Patten SB, Lavorato DH: Medication use and major depressive syndrome in a community population. Compr Psychiatry. 2001, 42: 124-31. 10.1053/comp.2001.21218.

    Article  CAS  PubMed  Google Scholar 

  216. Patten SB: Exogenous corticosteroids and major depression in the general population. J Psychosom Res. 2000, 49: 447-9. 10.1016/S0022-3999(00)00187-2.

    Article  CAS  PubMed  Google Scholar 

  217. Liu LY, Coe CL, Swenson CA: School examinations enhance airway inflammation to antigen challenge. Am J Respir Crit Care Med. 2002, 165: 1062-7.

    Article  PubMed  Google Scholar 

  218. Hoglund CO, Axen J, Kemi C: Changes in immune regulation in response to examination stress in atopic and healthy individuals. Clin Exp Allergy. 2006, 36: 982-92. 10.1111/j.1365-2222.2006.02529.x.

    Article  PubMed  Google Scholar 

  219. Leigh R, MacQueen G, Tougas G: Change in forced expiratory volume in 1 second after sham bronchoconstrictor in suggestible but not suggestion-resistant asthmatic subjects: a pilot study. Psychosom Med. 2003, 65: 791-5. 10.1097/01.PSY.0000079454.48714.1B.

    Article  PubMed  Google Scholar 

  220. Rosenkranz MA, Busse WW, Johnstone T: Neural circuitry underlying the interaction between emotion and asthma symptom exacerbation. Proc Natl Acad Sci USA. 2005, 102: 13319-24. 10.1073/pnas.0504365102.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  221. Devinsky O, Morrell MJ, Vogt BA: Contributions of anterior cingulate cortex to behaviour. Brain. 1995, 118 (Pt 1): 279-306. 10.1093/brain/118.1.279.

    Article  PubMed  Google Scholar 

  222. Capuron L, Pagnoni G, Demetrashvili M: Anterior cingulate activation and error processing during interferon-alpha treatment. Biol Psychiatry. 2005, 58: 190-6. 10.1016/j.biopsych.2005.03.033.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  223. Critchley HD, Mathias CJ, Josephs O: Human cingulate cortex and autonomic control: converging neuroimaging and clinical evidence. Brain. 2003, 126: 2139-52. 10.1093/brain/awg216.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Glenda MacQueen.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Van Lieshout, R.J., MacQueen, G. Psychological Factors in Asthma. All Asth Clin Immun 4, 12 (2008). https://doi.org/10.1186/1710-1492-4-1-12

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/1710-1492-4-1-12

Keywords