SYSTEMATIC REVIEW / META-ANALYSIS

The Role of Brain Structure and Function in the Association Between Inflammation and Depressive Symptoms: A Systematic Review Michelle L. Byrne, PhD, Sarah Whittle, PhD, and Nicholas B. Allen, PhD ABSTRACT Objective: Major depressive disorder and related symptoms have been shown to be associated with inflammation, and this association is likely to be mediated through changes in brain structure and function. This article provides a systematic review of studies that have used brain imaging techniques to identify neural mechanisms linking inflammation and depressive symptoms. Methods: A systematic search of online databases identified 26 studies that fulfilled the inclusion and exclusion criteria. Results: In general, increased peripheral inflammation was associated with differences in function in several subcortical regions, as well as medial and ventral prefrontal regions—both at rest (7 studies) and during exposure to emotional stimuli (14 studies). Also, increased activation in some of these regions was associated with depression (18 studies). Too few studies have measured neuroinflammation markers (three) or brain structure (three), so generalizations about these mechanisms cannot yet be made. Conclusions: This review supports the view that peripheral inflammation is an etiological process that may influence depression via effects on brain function. Several methodological inconsistencies in the extant literature need to be addressed, most notably a lack of formal mediational testing in longitudinal designs and inconsistencies across imaging methods and inflammation induction and measurement techniques. Further work is also required to establish the mechanisms by which basal inflammation levels influence brain function and depressive symptoms in both healthy and clinical samples. Key words: depression, inflammation, brain function, brain structure, neuroimaging.

INTRODUCTION

respond to stress regardless of whether it is physical or psychosocial (12–14). There are several metabolic and behavioral changes that may occur during this acute phase, including fever, loss of appetite, lethargy, and loss of interest in social behaviors. Although these sickness behaviors, which are potentially adaptive in circumstances when an organism is suffering from illness or infection, are not identical to symptoms of MDD, they share some similarities (15,16). Furthermore, injection of cytokines can induce depressive-like behaviors in mice (17), monkeys (18), and

Overview Majordepressive disorder(MDD) isa debilitating neuropsychiatric disease, with lifetime prevalence rates estimated to be 13% to 16% in thegeneralpopulation (1,2). It is predicted tobe one of the highest global causes of disease burden by 2020 (3). MDD shows a strong association with other medical illnesses (4–6), and inflammation has been commonly hypothesized to be a key mechanism explaining this link. Indeed, recent research has shown that depressed persons display elevated levels of inflammatory markers (for reviews, see Refs. (7,8)). Stress is a strong predictor of the development of MDD (9) and a trigger for inflammatory responses, which can lead to depression (10). An acute-phase response to stress is initiated by an activation of immune cells and the release of cytokines (11), which results in a systemic reaction referred to as “inflammation,” Inflammatory processes

ACC = anterior cingulate cortex, CRP = C-reactive protein, fMRI = functional magnetic resonance imaging, IDO = indoleamine 2,3-dioxygenase, IFN = interferon, IL = interleukin, LPS = lipopolysaccharide, MDD = major depressive disorder, MRI = magnetic resonance imaging, MRS = magnetic resonance spectroscopy, OFC = orbitofrontal cortex, PFC = prefrontal cortex, PET = positron emission tomography, sACC = subgenual anterior cingulate cortex, TNF-α = tumor necrosis factor α

Supplemental Content From the Department of Psychology (Byrne, Allen), The University of Oregon, Eugene, Oregon; Melbourne Neuropsychiatry Centre (Whittle), Department of Psychiatry, The University of Melbourne & Melbourne Health, Victoria, Australia; and Melbourne School of Psychological Sciences (Allen), The University of Melbourne, Victoria, Australia. Address correspondence and reprint requests to Michelle L. Byrne, PhD, Department of Psychology 1227 University of Oregon, Eugene, OR 97403. E-mail: [email protected] Received for publication March 10, 2015; revision received November 6, 2015. DOI: 10.1097/PSY.0000000000000311 Copyright © 2016 by the American Psychosomatic Society

Psychosomatic Medicine, V 00 • 00-00

1

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

SYSTEMATIC REVIEW / META-ANALYSIS

research directions that require further investigation. Especially, this may justify the effort involved in conducting longitudinal research to test formal mediation models whereby inflammatory processes influence brain activity and development in areas responsible for emotional processing, which may, in turn, promote risk for the development of depressive disorders. This article briefly summarizes research on brain function and structure in depression and the effects of inflammation on brain function, before providing a comprehensive, systematic review of studies that have used brain imaging techniques to identify neural mechanisms potentially linking inflammation and depressive symptoms. Our aim is to identify specific brain regions and networks that may mediate the relationship between inflammation and depressive symptoms. As such, electroencephalogram studies were not included in our review because they would not be as informative as to the specific anatomical circuits and regions involved.

humans (19). This suggests that there may be an etiological path to MDD that is cytokine induced, especially if the inflammatory response is chronic or reoccurring (20), which can be due to range of factors such as stress and life-style factors such as diet, sleep, smoking, and obesity (for review, see Ref. (21)). For example, administration of interferon-α (IFN-α) to patients with hepatitis C (19,22–25) or interleukin (IL)-2 to patients with cancer (26) is associated with increases in depressive symptoms and fatigue (27). It should be noted that one study found that patients with MDD reported improved mood the next morning after endotoxin administration (28). Nevertheless, a recent review of studies measuring cognitive performance after endotoxin administration found that the data were inconsistent (29), so increases in peripheral inflammation may not always necessarily translate to cytokineinduced depression, especially symptoms related to neurocognitive functioning. Furthermore, cytokine-induced depression may have specific neuroimmune mechanisms (30), mainly through the inflammatory, oxidative, and nitrosative stress pathways (31,32). Via nuclear factor κB, an inflammatory signaling molecule, a peripheral inflammatory response activates brain microglia to produce proinflammatory cytokines (18). This activates expression of indoleamine 2,3-dioxygenase (IDO) in the brain (33), which reduces tryptophan levels and, in turn, activates production of kynurenine (24). This process is thought to lead to depressive-like behaviors, which animal research has shown are different from sickness behaviors (17), and can sometimes contribute to reoccurring depressive illness in humans (34). Other ways that peripheral inflammation has shown to have effects on the brain include vagal activation (20) and, recently, availability of serotonin transporter in the brainstem (35). Therefore, the effect of the immune system on mood and behavioral symptoms of depression is likely to be mediated through observable changes in brain function and structure. However, the neural regions and networks that mediate this effect have not yet been fully identified. Research using neuroimaging to link inflammation and depressive symptoms is a promising approach in this regard, because differences in brain function and structure have been implicated in both depression and (to a lesser extent) inflammation. Emerging research has investigated the neural mechanisms underpinning the association between inflammation and depression, and there may be a link between inflammatory processes and differences in brain function and structure associated with MDD, schizophrenia, and Alzheimer's disease (36). However, to date, there has been no systematic review of research findings investigating the associations between brain function and structure, inflammation, and depression, specifically. A review of the existing research is timely to establish whether there are consistent depressionrelated differences in neural regions or networks that may be influenced by inflammatory processes, and will highlight Psychosomatic Medicine, V 00 • 00-00

Neural Mechanisms in Depression The past decade has seen a wealth of neuroimaging studies providing evidence for structural and functional brain changes associated with depression. A number of recent meta-analyses have been performed that elucidate the most consistent and significant neural differences associated with the disorder (37,38). These meta-analyses suggest significant reductions in gray matter in structures within a corticolimbic circuit, including the amygdala, hippocampus, striatum, dorsomedial and dorsolateral prefrontal cortex (PFC), anterior cingulate cortex (ACC), and orbitofrontal cortex (OFC), which are hypothesized to underlie some of the core cognitive, affective, and behavioral depressive symptoms. Both increased and decreased neural activities associated with emotional and cognitive processing have been found in similar areas. For example, a recent meta-analysis found evidence for opposing effects for negative and positive emotional stimuli processing in the amygdala, striatum, parahippocampus, cerebellum, fusiform, and ACC, with depressed participants displaying hyperactivation to negative stimuli, and hypoactivation to positive stimuli (39). Depression has also been associated with differences in functional connectivity in brain networks that are thought to underlie mood regulation, as well as the default mode network, a network of regions including the medial PFC, posterior cingulate, precuneus, and neighboring regions of the parietal cortex thought to underlie self-referential processing and autobiographical recall (40).

Peripheral Inflammatory Effects on Brain Structure and Function Although the literature is less substantial, neuroimaging studies have also examined the effect of inflammation on brain structure and function. Many have implicated proinflammatory markers such as C-reactive protein (CRP)

2

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

Inflammation and Depression Associations

measures in an attempt to better understand the neural mechanisms through which inflammation might be associated with depression.

(41,42), as well as ILs such as IL-6 (43,44). For example, in healthy adult men, greater CRP levels were associated with cortical thinning in several left hemisphere perisylvian regions (41). Peripheral IL-6 levels have shown to be negatively associated with gray-matter volume in the hippocampus (41). Other studies have used an experimental design by increasing levels of systemic inflammation through either injection of endotoxins or exposure to stress. These studies have found inflammation-associated changes in activity in the substantia nigra (43), and the dorsal ACC (45,46) and anterior insula (45). Patients receiving IFN-α therapy have shown increases in glucose metabolism in the basal ganglia and cerebellum and decreases in glucose metabolism in the dorsal PFC (47). Furthermore, inflammatory responses are associated with increased microglia activity across the whole brain (48,49), as well as with specific changes in activity in the amygdala (17,50,51), hypothalamus (17), and hippocampus (17,52), and damage to white matter during sensitive developmental periods (53). In particular, studies have shown associations between peripheral inflammation and activity in regions relevant to emotional processing such as the ACC and insula (54) and the subgenual ACC (sACC) and OFC (55). Overall, inflammation seems to affect brain development and functioning, especially in emotion-related brain networks, and due to the association of depression with both inflammation and changes in brain structure and function in such networks (refer to Fig. 1 for a summary of the overlapping literature), several research studies have combined these

Systematic Review—Methods The systematic review was conducted according to the PRISMA guidelines (56). Figure 2 presents a flowchart and details of the procedure.

Literature Search Online searches of the PubMed, MEDLINE, and PsycINFO databases were performed in August 2015. Abstracts were reviewed for references to magnetic resonance imaging (MRI), diffusion tensor/weighted imaging, voxelbased morphometry, functional MRI (fMRI), positron emission tomography (PET), magnetic resonance spectroscopy (MRS), inflammation, and depressive symptoms or depression. If the study was relevant, the full text was retrieved. Additional studies were retrieved through other methods, including reviewing bibliographies of articles identified through the search terms.

Inclusion and Exclusion Criteria Journal articles were included if they reported on studies that a) measured peripheral inflammation, induced an inflammatory response through cytokine or endotoxin injection, or measured another marker of inflammatory functioning (e.g., genes); b) measured brain structure using MRI or diffusion methods, brain function using fMRI or PET, or brain

FIGURE 1. Summary of the literature on associations between inflammation and brain structure/function, and depression and brain structure/function. Associations are change in activity or metabolism, except the following: *less gray matter only, **cortical thinning only, and ***increase in activity and less gray matter. CRP = C-reactive protein; IL = interleukin; OFC = orbitofrontal cortex; PFC = prefrontal cortex; ACC = anterior cingulate cortex.

Psychosomatic Medicine, V 00 • 00-00

3

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

SYSTEMATIC REVIEW / META-ANALYSIS

FIGURE 2. Procedure used for study selection. fMRI = functional magnetic resonance imaging.

PET and MRS Studies of Resting Brain Metabolism

metabolism using MRS; c) measured depressive illness or any discrete symptoms relevant to depressive illness such as social rejection, neurovegetation, fatigue, anhedonia, depressed mood, or grief, or was designed to elicit depressed mood; and d) examined associations between these three measures.

Four studies used MRS techniques (57–60) and six used PET (47,61–65). Increased inflammation, whether induced or measured peripherally, was associated with differences in function in several regions at rest, including alterations in dopaminergic function in the caudate, putamen, and ventral striatum (63), and higher glucose metabolism in the insula (64), putamen, and nucleus accumbens (47), although one study found decreased metabolism in the dorsolateral PFC (47). These neural changes were, in turn, associated with depressive symptoms. Three studies measured the neuroinflammation marker translocator protein

Systematic Review—Results Information from each study is presented in Table S1, Supplemental Digital Content 1, http://links.lww.com/PSYMED/A272. Twenty-six studies were identified as relevant and are reviewed later. Results are categorized based on study protocol. Psychosomatic Medicine, V 00 • 00-00

4

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

Inflammation and Depression Associations

in the amygdala (66,75), ACC (46,75), sACC (55,66), dorsal ACC (45,72), basal ganglia, thalamus, hypothalamus (75), anterior insula (45,72), OFC (55), and substantia nigra (43). Some studies also measured associations between depression and neural activity. In general, increased depressive symptoms were associated with increased activity in the medial PFC, hippocampus, posterior cingulate cortex (66), and substantia nigra (43), and depressed patients had more μ-opioid activation associated with inflammation (75). Furthermore, in some studies, inflammation was also associated with depression or depressive symptoms (43,46,75), but not in all (66,72). However, one study reported an association between inflammation and depressed mood in females only (72), and this association was mediated by brain activity.

18 kDa (TSPO) using PET—one study did not find any difference between depressed and control participants (62); however, one study measured TSPO density and found elevated levels in depressed patients in the PFC, ACC, and insula compared with controls (61), and another found that within patients with chronic fatigue syndrome, peak TSPO in the thalamus, midbrain, and amygdala was associated with cognitive impairment (65). MRS studies in patients with hepatitis C showed that treatment with proinflammatory cytokines increased glutaminergic metabolism in the dorsal ACC, pregenual ACC, and left basal ganglia, which were, in turn, associated with increased depressive symptoms (60) and reduced motivation (59). Older age was also associated with a greater increase in glutaminergic metabolism in the left basal ganglia, which was also associated with increased peripheral inflammation and depressive symptoms (57). Another MRS study of breast cancer survivors found that fatigue was associated with higher levels of peripheral inflammation and changes in brain metabolic function in the posterior insula, but there were no associations between inflammation and measures of brain metabolites (58).

Other Methods Three studies measured brain structure. One found that patients with depression had smaller hippocampal volumes and increased inflammation compared with controls (76). Another used quantitative magnetization transfer to measure microstructure and found that inflammation induced changes in the ventral striatum which were, in turn, associated with increases in fatigue (77). Finally, one study found that inflammatory genes were associated with volume of the sACC, hippocampus, and caudate, but not with depression (67).

fMRI and PET Studies of Task-Related Activity All studies involving tasks to measure activity used fMRI techniques (43,45,46,55,63,66–74), except one, which used PET to measure μ-opioid receptor activity during a sadness induction task (75). Tasks included exposure to emotional stimuli, reward processing, and emotion or stress induction. During exposure to emotional (particularly negative) stimuli, increased inflammation was associated with activation in a number of brain regions including the amygdala (67,69), ventromedial PFC (66), hippocampus (67), inferior OFC (68), ACC (73,74), and insula (74). Increased activation in some of these regions was associated with depression, either by an effect only in depressed patients (67) or by an increase in depressive symptoms (46,68,69,72–74). One study showed that inflammation was associated with increases in both depressed mood and brain activation while viewing aversive pictures, but did not analyze explicit associations between brain function and mood (68). Only one study found inflammation (measured by a genetic variant associated with increased inflammation) to be associated with decreased brain activity in depression during negative emotional stimuli (70). Two studies used reward tasks to elicit brain activity and showed that inflammation was associated with less activation of the ventral striatum in relation to anticipating the reward (71) and actual receipt of the reward (63). In turn, decreased activity during reward processing was associated with depressive symptoms. Finally, seven studies used tasks that were designed to elicit stress, sadness, or other negative feelings such as social exclusion. In these studies, increases in inflammation were associated with increased activity during these tasks Psychosomatic Medicine, V 00 • 00-00

DISCUSSION Summary Overall, this review suggests that inflammation is associated with changes in brain function and depression, and although not explicitly tested, findings to date provide some preliminary support for a model by which inflammatory effects on brain function might mediate the effects of inflammation on depressive symptoms. As such, this review provides grounds to encourage future research to formally test the hypothesis that inflammation is one of the etiological processes that influences depressed mood and depressive disorders via effects on brain function. Although methodologies differed, inflammation was consistently associated with differences in function in several subcortical, and medial and ventral prefrontal regions, both during rest and in response to aversive and positive/reward stimuli. Below, the extant literature is discussed with respect to four emergent issues: a) the specific brain regions that are emerging as most critical/promising, b) imaging methods, c) inflammation methods, and d) assessment of depression and depressive symptoms.

Brain Regions Subcortical Regions Our review of the literature suggests that at rest, inflammation is associated with differences in dopaminergic function

5

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

SYSTEMATIC REVIEW / META-ANALYSIS

and a recent study has also found that experimentally induced inflammation changes the subjective value of rewards versus punishments, as well as reducing the encoding of reward prediction errors within the ventral striatum (80). Interestingly, another recent study showed that endotoxin administration and the associated increase in inflammation increased activity in the ventral striatum while viewing a personally identified support figure (81), suggesting that inflammation may not uniformly change all types of reward behavior. Overall, this review found that the current literature, albeit limited, suggests that reward processing networks may be involved in the etiology of cytokine-induced depression, and also may play a specific role in IDO pathways. The subcortical brain regions that were identified as having both immune- and depression-related increases in activation in response to emotional stimuli included the amygdala, ventral pallidum, hypothalamus, and hippocampus. Many of these regions are consistently identified as being hyperactive in response to negative emotional stimuli in depressive disorder (82). This review showed that emotionally valenced human faces were particularly salient stimuli that were associated with increased inflammation and depression, suggesting that these regions are part of an important network attending to social cues that may be affected by inflammatory states.

(suggestive of impaired dopamine release or increased dopamine reuptake) in the caudate, putamen, and ventral striatum (e.g., Ref (62)). Although experimentally induced inflammation was associated with significant increases in dopamine uptake in striatal areas, decreased dopamine uptake in these areas at baseline was associated with development of neuropsychiatric symptoms in the context of experimentally induced inflammation. Lower dopamine uptake in the striatum has consistently been implicated in depression (78), suggesting that dopaminergic tone may serve as an important vulnerability factor mediating the impact of cytokines on depression. As noted earlier, IFN-α has been associated with decreased dopamine levels (63). As such, decreased availability of dopamine may indirectly lead to an overactivation of glutamatergic projections to the striatum and basal ganglia output nuclei. Glutamate concentrations (57,59) and metabolic activity (47) were also increased in the basal ganglia (especially striatal areas) in patients treated with cytokine injections, which, in turn, were associated with increased depressive symptoms, and activity in the substantia nigra was associated with peripheral inflammation and depressive symptoms (43) (refer to Fig. 3 for a summary of all brain regions). Two studies found that the ventral striatum was less active in response to inflammation during reward tasks, whether anticipation or actual receipt of reward (63,71). Another found that inflammation was associated with changes in the microstructure of the ventral striatum at rest, which also predicted an increase in fatigue (77). Reduced ventral striatum activation in response to reward is also a well-established finding in the depression literature (79),

Cortical Regions Both at rest and in studies of processing emotional stimuli, activity in the medial PFC (especially ventral region), inferior OFC, and ACC (dorsal and subgenual regions) was associated with increased inflammation and depression.

FIGURE 3. Brain regions identified from systematic review. vmPFC = ventromedial prefrontal cortex; OFC = orbitofrontal cortex; PFC = prefrontal cortex.

Psychosomatic Medicine, V 00 • 00-00

6

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

Inflammation and Depression Associations

of guiding future research, is summarized in Figure 3. However, several other methodological issues need to be addressed to advance the field to enable stronger conclusions about these relationships.

Again, activity in response to negative stimuli in these regions is implicated in depression (82). Furthermore, neural models of depression posit that changes in function within a network that comprises these regions (along with the subcortical regions discussed above, such as the amygdala, ventral pallidum, hypothalamus, hippocampus, and insula), known as the “extended medial network,” underlie the differences in emotional behavior and cognition that characterize depressive disorder (79). Because this network is understood to exert forebrain modulation over visceral responses mediated via the hypothalamus and brainstem, dysfunction within this circuit may account for the differences in autonomic (83,84) and neuroendocrine (85) function that have been associated with depression. The reviewed research suggests that this network is also relevant in inflammatory pathways that may lead to depression, and that areas within this network may be particularly susceptible to inflammation through the IDO and tryptophan/kynurenine pathways. Of relevance is that the sACC was identified in four independent emotion/stress elicitation studies (55,66,73,75). This region has been one of the most consistently implicated regions in depression (86). Therefore, future research should continue to examine the role of inflammation as a moderator of activity in this region. Two studies (59,60) found that IFN-α treatment was associated with an increase in glutamine or Glu/Cr levels in the ACC, which, in turn, was associated with increased depressive symptoms. However it is of note that, generally, in patients with current unipolar depression, MRS levels of total glutamate and glutamine are reported to be lower than controls, whereas elevated levels have been associated with bipolar depression (87). It is thus possible that the neurochemical changes in the ACC produced by IFN-α as measured by MRS may be associated with risk for bipolar as opposed to unipolar depression, although this is yet to be explicitly tested. It should also be noted that a few studies found decreases in activity in the dorsolateral PFC (47), ACC, and amygdala (70) associated with depression and inflammation, suggesting that more research examining these brain regions is needed to establish which pattern of findings is valid. Furthermore, inflammation and depressive symptoms were associated with greater neuronal activity in the insula (45,58,64,72,74). Interestingly, in one study (64), increased activity in the insula was associated with increased social interest and lower peak cytokine levels. It was suggested that in those with higher activity in the insula, serum cytokine levels may have been lower because of an active inhibition of the systemic inflammatory response. Lower cytokine levels may, in turn, have caused less reduction in social interest. In sum, patterns are beginning to emerge in the literature showing changes in activity in certain brain regions and networks, especially the extended medial network. This pattern of findings, which will clearly be important in terms Psychosomatic Medicine, V 00 • 00-00

Imaging Techniques Six of the reviewed studies used PET imaging techniques (47,61,62,64,66,75), 4 used MRS (57–60), 13 examined brain activity with fMRI (43,45,46,55,66–74), and 1 study used both PET and fMRI techniques (63). Although all measure brain function, the nature of the function that is assayed and its interpretation differ significantly between techniques. As such, fMRI, PET, and MRS studies are not directly comparable. Two studies measured neuroinflammation markers but did not measure peripheral inflammation (61,62), or did not make direct comparisons between peripheral inflammation and depressive symptoms or brain activity (65). Only two studies examined brain structure using MRI (76,77), so clearly more research is needed before generalizations can be made about the relationships between inflammation, brain structure, and depression. Furthermore, studies differed in their analytical approach, especially with regard to whole brain versus region of interest approaches, and the statistical thresholds used. A more formal meta-analytic approach will be extremely useful once there are a greater number of published studies. Finally, studies that used tasks to elicit emotion did so in different ways. For example, there may be differences in neural processing of a sadness-induction task compared with a stress-induction task, or task designed to elicit social rejection, or simply viewing emotionally expressive faces. A greater number of studies that use each of these kinds of tasks are needed before strong conclusions can be made about neural mechanisms involved in this kind of processing.

Inflammation Techniques and Measurements Peripheral inflammation was measured in many of the studies reviewed, but in a variety of ways. Although IL-6 was the most commonly examined cytokine, inflammatory markers including IFN-α, TNF-α, IL-1ra, IL-1β, IL-6, IL-10, IL-18, and CRP were all variably measured across studies. Three studies examined expression and variants of inflammatory genes (67,70,76). The results from one of these studies (70) were contrary to expectation, suggesting that the effect of inflammatory genes may need to be examined separately from basal levels of circulating cytokines. Relatedly, studies differed in their experimental technique. Some studies induced an inflammatory response through stress (45,66), or injection of lipopolysaccharide (LPS) (68), whereas others used endotoxins (64,69,71,72), vaccines (43,73,74), injected cytokines as part of a treatment (46,47,57,59,60,63,77), or measured basal levels of inflammatory markers (55,58,75,76). Administration of LPS produces a stronger immune response than a typhoid

7

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

SYSTEMATIC REVIEW / META-ANALYSIS

patients with the former report more psychomotor disturbances and less feelings of guilt (90). Whether this suggests that depressive conditions with a more directly endogenous etiology might have a greater emphasis on vegetative symptoms, and less on cognitive and affective symptoms, than do those that are associated with psychosocial stress is a hypothesis that may require investigation of the common and distinctive neural basis of these putative etiological paths. Future neuroimaging research may be able to play a particularly important role in understanding whether there are specific neural and inflammatory processes that distinguish types of depressive disorder. Furthermore, it will also be important to measure discrete dimensions of depressive symptoms that may be specific to inflammation and differentiate between functional and emotional aspects of depression.

vaccine, and even a small dose of LPS at 0.8 ng/kg can affect memory performance (for review, see Ref. (29)). Typhoid vaccine (43) activates only a low-grade inflammatory response and avoids the confounding effects of fever triggered by LPS administration, but still shows increases in negative mood (88). Clear identification of mechanisms in depression is not possible until more research is conducted with similar inflammation inductions and measurement methods. For example, one study measured inflammation at baseline and after a stress test and found that baseline inflammation levels were not associated with brain activity, whereas levels of inflammation in response to stress were (45), suggesting that different mechanisms might be associated with each of these measures. Finally, three studies (61,62,65) measured neuroinflammation (binding to TSPO). Results from these studies were conflicting in that one found that TSPO was elevated in the PFC, ACC, and insula in patients with depression (61), but another found no differences between patients and controls (62). Another used a sample of patients with chronic fatigue syndrome and found that peak TSPO was associated with cognitive impairment in the thalamus and amygdala (65). It should be noted that within the two studies of patients with depression, the study that found significant results had twice the sample size of the earlier study, suggesting that greater power may be required to observe these effects. Further research with adequate sample sizes, therefore, is needed to determine mechanisms of neuroinflammation in depression.

Limitations of the Extant Literature Several methodological discrepancies across the studies limit our ability to draw firm conclusions. As of yet, there are few studies that conduct formal statistical tests of mediation to determine if brain activity mediates the association between inflammation and depression. This type of analysis would be especially beneficial in longitudinal research. Only one longitudinal study has been conducted, which found that patients receiving IFN-α treatment for 4 to 6 weeks had increased 18F-DOPA uptake and less 18F-Dopa turnover compared with baseline, and this was associated with increased depressive symptoms (63). Further longitudinal research examining naturally occurring levels of cytokines within individuals over longer periods is justified to elucidate the temporal sequence and potential causal relationships between inflammation, neural biomarkers, and the emergence of depression. Another issue to be considered is the possibility of publication bias, where studies that found significant associations between inflammation, brain function, and depression have been more likely to be published. However, it should be noted that some null findings have been reviewed here, including a study showing no differences in TSPO binding between people with depression and controls (62). Finally, causal mechanisms relevant to depressive disorders cannot yet be inferred from these data. Although some studies used an experimental design by injecting participants with cytokines, these studies did so in healthy adults, not persons diagnosed as having a depressive disorder. As discussed earlier, the pathophysiology of cytokine-induced depression may differ to depressed mood in healthy persons (90).

Assessment of Depressive Disorders and Symptoms Studies also differed in their measurement of depression. Although some included samples of patients with MDD, others only measured symptoms, which may not have reached levels of clinical significance. Some studies measured changes in depressed mood in healthy participants. Although research should be careful not to conflate these results without examining them separately, elevated levels of depressive symptoms do predict the emergence of MDD later (89), so this can shed some light on the pathophysiology of cytokine-induced depression. This systematic review reveals an emerging pattern showing that, with some exceptions, when brain activity and/or increased inflammation is associated with depression, the relationship is observed regardless of whether depression is measured by a clinical diagnosis or an increase in subclinical symptoms. A critical question that arises from this review is whether increased inflammation is one of many etiological processes that contribute to the neural differences in persons with depression, or whether increased inflammation represents a pathway leading to a specific type of neurological change and depression. There is some support for the latter view. For example, symptoms of cytokine-induced depression seem to differ from idiopathic major depression in that Psychosomatic Medicine, V 00 • 00-00

Conclusions and Future Directions This review highlights the small but growing number of research studies showing associations between increased inflammation, brain activity, and measures of depression. It seems that specific neural mechanisms are likely to be implicated in the immune-depression relationship, including

8

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

Inflammation and Depression Associations

10. Slavich GM, Irwin MR. From stress to inflammation and major depressive disorder: a social signal transduction theory of depression. Psychol Bull 2014;140:774–815. 11. Heinrich PC, Castell JV, Andus T. Interleukin-6 and the acute phase response. Biochem J 1990;265:621–36. 12. Hennessy MB, Deak T, Schiml-Webb PA, Wilson SE, Greenlee TM, McCall E. Responses of guinea pig pups during isolation in a novel environment may represent stress-induced sickness behaviors. Physiol Behav 2004;81:5–13. 13. Kaufman JC, Rosenblum LA. The reaction to separation in infant monkeys: anaclitic depression and conservation withdrawal. Psychosom Med 1967;29:648–75. 14. Selye H. The Stress of Life. New York: McGraw-Hill; 1956. 15. Hennessy MB, Schiml-Webb PA, Deak T. Separation, sickness, and depression: a new perspective on an old animal model. Curr Dir Psychol Sci 2009;18:227–31. 16. Hickie I, Lloyd A. Are cytokines associated with neuropsychiatric syndromes in humans? Int J Immunopharmacol 1995;17:677–83. 17. Frenois F, Moreau M, O'Connor J, Lawson M, Micon C, Lestage J, Kelley KW, Dantzer R, Castanon N. Lipopolysaccharide induces delayed FosB/DeltaFosB immunostaining within the mouse extended amygdala, hippocampus and hypothalamus, that parallel the expression of depressive-like behavior. Psychoneuroendocrinology 2007;32:516–31. 18. Felger JC, Alagbe O, Hu F, Mook D, Freeman AA, Sanchez MM, Kalin NH, Ratti E, Nemeroff CB, Miller AH. Effects of interferon-alpha on rhesus monkeys: a nonhuman primate model of cytokine-induced depression. Biol Psychiatry 2007;62:1324–33. 19. Lotrich FE, Rabinovitz M, Gironda P, Pollock BG. Depression following pegylated interferon-alpha: characteristics and vulnerability. J Psychosom Res 2007;63:131–5. 20. Dantzer R, O'Connor JC, Freund GG, Johnson RW, Kelley KW, O'Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci 2008;9: 46–56. 21. Berk M, Williams LJ, Jacka FN, O'Neil A, Pasco JA, Moylan S, Allen NB, Stuart AL, Hayley AC, Byrne ML, Maes M. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med 2013;11:200. 22. Bonaccorso S, Marino V, Puzella A, Pasquini M, Biondi M, Artini M, Almerighi C, Verkerk RRT, Meltzer H, Maes M. Increased depressive ratings in patients with hepatitis C receiving interferon-[alpha]–based immunotherapy are related to interferon-[alpha]–induced changes in the serotonergic system. J Clin Psychopharmacol 2002;22:86–90. 23. Lotrich FE, Ferrell RE, Rabinovitz M, Pollock BG. Risk for depression during interferon-alpha treatment is affected by the serotonin transporter polymorphism. Biol Psychiatry 2009;65:344–8. 24. Capuron L, Neurauter G, Musselman DL, Lawson DH, Nemeroff CB, Fuchs D, Miller AH. Interferon-alpha– induced changes in tryptophan metabolism: relationship to depression and paroxetine treatment. Biol Psychiatry 2003; 54:906–14. 25. Capuron L, Raison CL, Musselman DL, Lawson DH, Nemeroff CB, Miller AH. Association of exaggerated HPA axis response to the initial injection of interferon-alpha with development of depression during interferon-alpha therapy. Am J Psychiatry 2003;160:1342–5. 26. Capuron L, Ravaud A, Dantzer R. Early depressive symptoms in cancer patients receiving interleukin 2 and/or interferon alpha-2b therapy. J Clin Oncol 2000;18:2143–51.

basal dopaminergic functioning, activity in the extended medial network (especially the sACC) while processing negative affective stimuli, and inhibited activity of the ventral striatum during reward tasks. However, further neuroimaging research is needed to determine if cytokineinduced depression has a separate etiological pathway that is different from idiopathic MDD. Importantly, several methodological inconsistencies need to be addressed, most notably a lack of formal mediational testing in longitudinal designs. Furthermore, more studies that use the same inflammation induction techniques, especially stress induction, are required to make generalizations across the type of inflammatory response. Finally, studies that use PET techniques to measure neuroinflammation should also measure peripheral inflammation to enhance our understanding of the relationships between these types of processes and markers. Ultimately, identification of these causal mechanisms will assist treatment, early intervention, and preventative efforts aimed at reducing the burden of depression and its psychosomatic correlates. Source of Funding and Conflicts of Interest: Sarah Whittle was supported by a National Health and Medial Council Career Development Fellowship (ID: 1007716). The authors declare no conflicts of interest.

REFERENCES 1. Hasin DS, Goodwin RD, Stinson FS, Grant BF. Epidemiology of major depressive disorder: results from the National Epidemiologic Survey on Alcoholism and Related Conditions. Arch Gen Psychiatry 2005;62:1097–106. 2. Kessler RC, Berglund PA, Demler O, Jin R, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication (NCS-R). Arch Gen Psychiatry 2005;62:593–602. 3. Murray CJ, Lopez AD. Global Burden of Disease: A Comprehensive Assessment of Mortality and Disability From Diseases, Injuries, and Risk Factors in 1990 and Projected to 2020. Cambridge: Harvard School of Public Health; 1996. 4. Anderson RJ, Freedland KE, Lustman PJ. The prevalence of comorbid depression in adults with diabetes: a metaanalysis. Diabetes Care 2001;24:1069–78. 5. Carney RM, Rich MW, Freedland KE, Saini J, teVelde A, Simeone C, Clark K. Major depressive disorder predicts cardiac events in patients with coronary artery disease. Psychosom Med 1988;50:627–33. 6. McDaniel JS, Musselman DL, Porter MR, Reed DA, Nemeroff CB. Depression in patients with cancer: diagnosis, biology, and treatment. Arch Gen Psychiatry 1995;52:89–99. 7. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, Lanctôt KL. A meta-analysis of cytokines in major depression. Biol Psychiatry 2010;67:446–57. 8. Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med 2009;71:171–86. 9. Gillespie CF, Nemeroff CB. Early life stress and depression: childhood trauma may lead to neurobiologically unique mood disorders. Curr Psychiatr 2005;4:15–30.

Psychosomatic Medicine, V 00 • 00-00

9

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

SYSTEMATIC REVIEW / META-ANALYSIS

27. Raison CL, Rye DB, Woolwine BJ, Vogt GJ, Bautista BM, Spivey JR, Miller AH. Chronic interferon-alpha administration disrupts sleep continuity and depth in patients with hepatitis C: association with fatigue, motor slowing, and increased evening cortisol. Biol Psychiatry 2010;68:942–9. 28. Bauer J, Hohagen F, Gimmel E, Bruns F, Lis S, Krieger S, Ambach W, Guthmann A, Grunze H, Fritsch-Montero R, Weissbach A, Ganter U, Frommberger U, Riemann D, Berger M. Induction of cytokine synthesis and fever suppresses REM sleep and improves mood in patients with major depression. Biol Psychiatry 1995;38:611–21. 29. Schedlowski M, Engler H, Grigoleit JS. Endotoxin-induced experimental systemic inflammation in humans: a model to disentangle immune-to-brain communication. Brain Behav Immun 2014;35:1–8. 30. Loftis JM, Huckans M, Morasco BJ. Neuroimmune mechanisms of cytokine-induced depression: current theories and novel treatment strategies. Neurobiol Dis 2010;37: 519–33. 31. Maes M, Berk M, Goehler L, Song C, Anderson G, Galecki P, Leonard B. Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med 2012; 10:66. 32. Maes M, Kubera M, Obuchowiczwa E, Goehler L, Brzeszcz J. Depression's multiple comorbidities explained by (neuro) inflammatory and oxidative & nitrosative stress pathways. Neuro Endocrinol Lett 2011;32:7–24. 33. O'Connor JC, Lawson MA, Andre C, Moreau M, Lestage J, Castanon N, Kelley KW, Dantzer R. Lipopolysaccharideinduced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Mol Psychiatry 2009;14: 511–22. 34. Maier SF, Watkins LR. Cytokines for psychologists: implications for bidirectional immune-to-brain communication for understanding behavior, mood, and cognition. Psychol Rev 1998;105:83–107. 35. Krishnadas R, Nicol A, Sassarini J, Puri N, Burden AD, Leman J, Combet E, Pimlott S, Hadley D, McInnes IB, Cavanagh J. Circulating tumour necrosis factor is highly correlated with brainstem serotonin transporter availability in humans. Brain Behav Immun 2016;51:29–38. 36. Frodl T, Amico F. Is there an association between peripheral immune markers and structural/functional neuroimaging findings? Prog Neuro Psychopharmacol Biol Psychiatry 2014;48: 295–303. 37. Bora E, Fornito A, Pantelis C, Yücel M. Gray matter abnormalities in major depressive disorder: a meta-analysis of voxel based morphometry studies. J Affect Disord 2012; 138:9–18. 38. Sacher J, Neumann J, Fünfstück T, Soliman A, Villringer A, Schroeter ML. Mapping the depressed brain: a meta-analysis of structural and functional alterations in major depressive disorder. J Affect Disord 2012;140:142–8. 39. Groenewold NA, Opmeer EM, de Jonge P, Aleman A, Costafreda SG. Emotional valence modulates brain functional abnormalities in depression: evidence from a meta-analysis of fMRI studies. Neurosci Biobehav Rev 2012;37:152–63. 40. Wang L, Hermens DF, Hickie IB, Lagopoulos J. A systematic review of resting-state functional-MRI studies in major depression. J Affect Disord 2012;142:6–12. 41. Krishnadas R, McLean J, Batty DG, Burns H, Deans KA, Ford I, McConnachie A, McGinty A, McLean JS, Millar K, Sattar N, Shiels PG, Velupillai YN, Packard CJ, Cavanagh J. Cardio-metabolic risk factors and cortical thickness in a neurologically healthy male population: results from the psychological,

Psychosomatic Medicine, V 00 • 00-00

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

10

social and biological determinants of ill health (pSoBid) study. Neuroimage Clin 2013;2:646–57. Taki Y, Thyreau B, Kinomura S, Sato K, Goto R, Wu K, Kakizaki M, Tsuji I, Kawashima R, Fukuda H. Correlation Between High-Sensitivity C-Reactive Protein and Brain Gray Matter Volume in Healthy Elderly Subjects. Hum Brain Mapp 2013;34:2418–24. Brydon L, Harrison NA, Walker C, Steptoe A, Critchley HD. Peripheral inflammation is associated with altered substantia nigra activity and psychomotor slowing in humans. Biol Psychiatry 2008;63:1022–9. Marsland AL, Gianaros PJ, Abramowitch SM, Manuck SB, Hariri AR. Interleukin-6 covaries inversely with hippocampal grey matter volume in middle-aged adults. Biol Psychiatry 2008;64:484–90. Slavich GM, Way BM, Eisenberger NI, Taylor SE. Neural sensitivity to social rejection is associated with inflammatory responses to social stress. Proc Natl Acad Sci 2010; 107:14817–22. Capuron L, Pagnoni G, Demetrashvili M, Woolwine BJ, Nemeroff CB, Berns GS, Miller AH. Anterior cingulate activation and error processing during interferon-alpha treatment. Biol Psychiatry 2005;58:190–6. Capuron L, Pagnoni G, Demetrashvili MF, Lawson DH, Fornwalt FB, Woolwine B, Berns GS, Nemeroff CB, Miller AH. Basal ganglia hypermetabolism and symptoms of fatigue during interferon-alpha therapy. Neuropsychopharmacology 2007;32:2384–92. Hannestad J, Gallezot JD, Schafbauer T, Lim K, Kloczynski T, Morris ED, Carson RE, Ding YS, Cosgrove KP. Endotoxin-induced systemic inflammation activates microglia: [11C]PBR28 positron emission tomography in nonhuman primates. Neuroimage 2012;63:232–9. Sandiego CM, Gallezot JD, Pittman B, Nabulsi N, Lim K, Lin S-F, Matuskey D, Lee J-Y, O'Connor KC, Huang Y, Carson RE, Hannestad J, Cosgrove KP. Imaging robust microglial activation after lipopolysaccharide administration in humans with PET. Proc Natl Acad Sci 2015;112:12468–73. Engler H, Doenlen R, Engler A, Riether C, Prager G, Niemi MB, Pacheco-López G, Krügel U, Schedlowski M. Acute amygdaloid response to systemic inflammation. Brain Behav Immun 2011;25:1384–92. Prager G, Hadamitzky M, Engler A, Doenlen R, Wirth T, Pacheco-López G, Krügel U, Schedlowski M, Engler H. Amygdaloid signature of peripheral immune activation by bacterial lipopolysaccharide or staphylococcal enterotoxin B. J Neuroimmune Pharmacol 2013;8:42–50. Murray C, Lynch MA. Evidence that increased hippocampal expression of the cytokine interleukin-1β is a common trigger for age- and stress-induced impairments in long-term potentiation. J Neurosci 1998;18:2974–81. Pang Y, Cai Z, Rhodes PG. Disturbance of oligodendrocyte development, hypomyelination and white matter injury in the neonatal rat brain after intracerebral injection of lipopolysaccharide. Brain Res Dev Brain Res 2003; 140:205–14. Rosenkranz MA, Busse WW, Johnstone T, Swenson CA, Crisafi GM, Jackson MM, Bosch JA, Sheridan JF, Davidson RJ. Neural circuitry underlying the interaction between emotion and asthma symptom exacerbation. Proc Natl Acad Sci 2005;102:13319–24. O'Connor MF, Irwin MR, Wellisch DK. When grief heats up: proinflammatory cytokines predict regional brain activation. Neuroimage 2009;47:891–6.

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

Inflammation and Depression Associations

56. Moher D, Liberati A, Tetzlaff J, Altman DG. The PRISMA Group. PLoS Med 2009;6:e1000097. 57. Haroon E, Felger JC, Woolwine BJ, Chen X, Parekh S, Spivey JR, Hu XP, Miller AH. Age-related increases in basal ganglia glutamate are associated with TNF, reduced motivation and decreased psychomotor speed during IFN-alpha treatment: preliminary findings. Brain Behav Immun 2015; 46:17–22. 58. Zick SM, Zwickey H, Wood L, Foerster B, Khabir T, Wright B, Ichesco E, Sen A, Harris RE. Preliminary differences in peripheral immune markers and brain metabolites between fatigued and non-fatigued breast cancer survivors: a pilot study. Brain Imaging Behav 2014;8:506–16. 59. Haroon E, Woolwine BJ, Chen X, Pace TW, Parekh S, Spivey JR, Hu XP, Miller AH. IFN-alpha–induced cortical and subcortical glutamate changes assessed by magnetic resonance spectroscopy. Neuropsychopharmacology 2014;39: 1777–85. 60. Taylor MJ, Godlewska B, Near J, Christmas D, Potokar J, Collier J, Klenerman P, Barnes E, Cowen PJ. Effect of interferon-alpha on cortical glutamate in patients with hepatitis C: a proton magnetic resonance spectroscopy study. Psychol Med 2014;44:789–95. 61. Setiawan E, Wilson AA, Mizrahi R, Rusjan PM, Miler L, Rajkowska G, Suridjan I, Kennedy JL, Rekkas PV, Houle S, Meyer JH. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry 2015;72:268–75. 62. Hannestad J, DellaGioia N, Gallezot JD, Lim K, Nabulsi N, Esterlis I, Pittman B, Lee JY, O'Connor KC, Pelletier D, Carson RE. The neuroinflammation marker translocator protein is not elevated in individuals with mild-to-moderate depression: a [11C]PBR28 PET study. Brain Behav Immun 2013; 33:131–8. 63. Capuron L, Pagnoni G, Drake DF, Woolwine BJ, Spivey JR, Crowe RJ, Votaw JR, Goodman MM, Miller AH. Dopaminergic mechanisms of reduced basal ganglia responses to hedonic reward during interferon alfa administration. Arch Gen Psychiatry 2012;69:1044–53. 64. Hannestad J, Subramanyam K, DellaGioia N, Planeta-Wilson B, Weinzimmer D, Pittman B, Carson RE. Glucose metabolism in the insula and cingulate is affected by systemic inflammation in humans. J Nucl Med 2012;53:601–7. 65. Nakatomi Y, Mizuno K, Ishii A, Wada Y, Tanaka M, Tazawa S, Onoe K, Fukuda S, Kawabe J, Takahashi K, Kataoka Y, Shiomi S, Yamaguti K, Inaba M, Kuratsune H, Watanabe Y. Neuroinflammation in patients with chronic fatigue syndrome/ myalgic encephalomyelitis: an 11C-(R)-PK11195 PET study. J Nucl Med 2014;55:945–50. 66. Muscatell KA, Dedovic K, Slavich GM, Jarcho MR, Breen EC, Bower JE, Irwin MR, Eisenberger NI. Greater amygdala activity and dorsomedial prefrontal–amygdala coupling are associated with enhanced inflammatory responses to stress. Brain Behav Immun 2015;43:46–53. 67. Savitz J, Frank MB, Victor T, Bebak M, Marino JH, Bellgowan PS, McKinney BA, Bodurka J, Kent Teague T, Drevets WC. Inflammation and neurological disease–related genes are differentially expressed in depressed patients with mood disorders and correlate with morphometric and functional imaging abnormalities. Brain Behav Immun 2013;31: 161–71. 68. Kullmann JS, Grigoleit JS, Lichte P, Kobbe P, Rosenberger C, Banner C, Wolf OT, Engler H, Oberbeck R, Elsenbruch S, Bingel U, Forsting M, Gizewski ER, Schedlowski M. Neural response to emotional stimuli

Psychosomatic Medicine, V 00 • 00-00

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79. 80.

81.

82.

11

during experimental human endotoxemia. Hum Brain Mapp 2013;34:2217–27. Inagaki TK, Muscatell KA, Irwin MR, Cole SW, Eisenberger NI. Inflammation selectively enhances amygdala activity to socially threatening images. Neuroimage 2012;59:3222–6. Baune BT, Dannlowski U, Domschke K, Janssen DG, Jordan MA, Ohrmann P, Bauer J, Biros E, Arolt V, Kugel H, Baxter AG, Suslow T. The interleukin 1 beta (IL1B) gene is associated with failure to achieve remission and impaired emotion processing in major depression. Biol Psychiatry 2010;67: 543–9. Eisenberger NI, Berkman ET, Inagaki TK, Rameson LT, Mashal NM, Irwin MR. Inflammation-induced anhedonia: endotoxin reduces ventral striatum responses to reward. Biol Psychiatry 2010;68:748–54. Eisenberger NI, Inagaki TK, Rameson LT, Mashal NM, Irwin MR. An fMRI study of cytokine-induced depressed mood and social pain: the role of sex differences. Neuroimage 2009;47: 881–90. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol Psychiatry 2009;66:407–14. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Dolan RJ, Critchley HD. Neural origins of human sickness in interoceptive responses to inflammation. Biol Psychiatry 2009;66:415–22. Prossin AR, Koch AE, Campbell PL, McInnis MG, Zalcman SS, Zubieta JK. Association of plasma interleukin-18 levels with emotion regulation and mu-opioid neurotransmitter function in major depression and healthy volunteers. Biol Psychiatry 2011;69:808–12. Frodl T, Carballedo A, Hughes MM, Saleh K, Fagan A, Skokauskas N, Mcloughlin DM, Meaney J, Keane VO, Connor TJ. Reduced expression of glucocorticoid-inducible genes GILZ and SGK-1: high IL-6 levels are associated with reduced hippocampal volumes in major depressive disorder. Transl Psychiatry 2012;2:e88. Dowell NG, Cooper EA, Tibbl EJ, Voon V, Critchley HD, Cercignani M, Harrison NA. Acute changes in striatal microstructure predict the development of interferon-alpha induced fatigue. Biol Psychiatry 2015:1–9. Bragulat V, Paillere-Martinot ML, Artiges E, Frouin V, Poline JB, Martinot JL. Dopaminergic function in depressed patients with affective flattening or with impulsivity: F-18 fluoro-L-dopa positron emission tomography study with voxel-based analysis. Psychiatry Res 2007;154: 115–24. Price JL, Drevets WC. Neurocircuitry of mood disorders. Neuropsychopharmacology 2010;35:192–216. Harrison NA, Voon V, Cercignani M, Cooper EA, Pessiglione M, Critchley HD. A neurocomputational account of how inflammation enhances sensitivity to punishments versus rewards. Biol Psychiatry 2015:1–9. Inagaki TK, Muscatell K, Irwin MR, Moieni M, Dutcher JM, Jevtic I, Breen EC, Eisenberger NI. The role of the ventral striatum in inflammatory-induced approach toward support figures. Brain Behav Immun 2015;44: 247–52. Hamilton JP, Etkin A, Furman DJ, Lemus MG, Johnson RF, Gotlib IH. Functional neuroimaging of major depressive disorder: a meta-analysis and new integration of baseline activation and neural response data. Am J Psychiatry 2012;169: 693–703.

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

SYSTEMATIC REVIEW / META-ANALYSIS

87. Yüksel C, Öngür D. Magnetic resonance spectroscopy studies of glutamate-related abnormalities in mood disorders. Biol Psychiatry 2010;68:785–94. 88. Wright CE, Strike PC, Brydon L, Steptoe A. Acute inflammation and negative mood: mediation by cytokine activation. Brain Behav Immun 2005;19:345–50. 89. Pine DS, Cohen E, Cohen P, Brook J. Adolescent depressive symptoms as predictors of adult depression: moodiness or mood disorder? Am J Psychiatry 1999;156: 133–5. 90. Capuron L, Fornwalt FB, Knight BT, Harvey PD, Ninan PT, Miller AH. Does cytokine-induced depression differ from idiopathic major depression in medically healthy individuals? J Affect Disord 2009;119:181–5.

83. Veith RC, Lewis N, Linares OA, Barnes RF, Raskind MA, Villacres EC, Murburg M, Ashleigh A, Castillo S, Peskind ER, Pascualy M, Halter JB. Sympathetic nervous system activity in major depression. Arch Gen Psychiatry 1994; 51:411. 84. 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. 85. Parker KJ, Schatzberg AF, Lyons DM. Neuroendocrine aspects of hypercortisolism in major depression. Horm Behav 2003;43:60–6. 86. Mayberg HS, Voon AMLV, McNeely HE, Seminowicz D, Kennedy SH. Deep brain stimulation for treatment-resistant depression. Neuron 2005;45:651–60.

Psychosomatic Medicine, V 00 • 00-00

12

Month 2016

Copyright © 2016 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

The Role of Brain Structure and Function in the Association Between Inflammation and Depressive Symptoms: A Systematic Review.

Major depressive disorder and related symptoms have been shown to be associated with inflammation, and this association is likely to be mediated throu...
2MB Sizes 3 Downloads 7 Views