Skip to main content
Physiology logoLink to Physiology
. 2019 Feb 6;34(2):123–133. doi: 10.1152/physiol.00047.2018

Central and Peripheral Inflammation Link Metabolic Syndrome and Major Depressive Disorder

Kenny L Chan 1, Flurin Cathomas 1, Scott J Russo 1
PMCID: PMC6586832  PMID: 30724127

Abstract

Metabolic syndrome and major depression are two of the most common and debilitating disorders worldwide, occurring with significant rates of comorbidity. Recent studies have uncovered that each of these conditions is associated with chronic, low-grade inflammation. This is characterized by increased circulating pro-inflammatory cytokines, altered leukocyte population frequencies in blood, accumulation of immune cells in tissues including the brain, and activation of these immune cells. Cytokines that become elevated during obesity can contribute to the progression of metabolic syndrome by directly causing insulin resistance. During chronic stress, there is evidence that these cytokines promote depression-like behavior by disrupting neurotransmitter synthesis and signal transduction. Animal models of obesity and depression have revealed a bi-directional relationship whereby high-fat feeding and chronic stress synergize and exacerbate metabolic dysregulation and behavioral abnormalities. Although far from conclusive, emerging evidence suggests that inflammation in the central and peripheral immune system may link metabolic syndrome to major depressive disorder. In this review, we will synthesize available data supporting this view and identify critical areas for future investigation.

Introduction

Metabolic syndrome and major depressive disorder (MDD) represent two of the most debilitating disorders worldwide and are often reported with high rates of comorbidity. Although the definition of metabolic syndrome continues to evolve, it is typically characterized by a cluster of conditions, including obesity, hyperglycemia, insulin resistance, dyslipidemia, and hypertension that predispose individuals to cardiovascular disease and Type 2 diabetes (T2D) (54). In 2016, the World Health Organization (WHO) estimated that 650 million adults were obese—a number that tripled since 1975 and continues to rise (136). MDD is a complex psychiatric illness that negatively affects emotion, cognition, and motivation (3). It is estimated that over 300 million people are affected by MDD, with an estimated 16.6% lifetime prevalence (58, 137). In the most severe cases, MDD can lead to suicide, and the WHO cites MDD as the leading cause of worldwide disability (137).

MDD has many comorbidities spanning cardiovascular disease, neurological disorders, and psychiatric illnesses, among others (42, 103, 124). Comorbidity between MDD and metabolic syndrome has been well recognized, although individual risk varies dramatically across cohorts. For example, one study in a diverse adult population found that participants with metabolic syndrome had higher scores on the Hospital Anxiety and Depression Scale (depression subscale), with waist circumference and HDL cholesterol showing significant and independent correlations (33). A separate study found a U-shaped relationship between body weight and depression, with higher depression prevalence in both underweight and obese subjects relative to normal-weight controls (19). In cohorts comprised exclusively of women, severity of depressive symptoms and stressful life events correlate with concurrent metabolic syndrome or predict risk for its development (99, 100). Conversely, some studies including a longitudinal assessment of 5,698 participants as part of the Northern Finland Birth Cohort Project found no associations between metabolic syndrome and depression or anxiety (48). Thus this relationship remains incompletely understood, likely due to the heterogeneity of both metabolic syndrome and MDD as complex, multifactorial disorders involving lifelong interplay between genetics and the environment (76, 98). Moreover, since MDD is diagnosed exclusively by behavioral symptoms, there is a need to identify biological factors that contribute to its pathogenesis. Recent work has highlighted both MDD and metabolic syndrome as inflammatory conditions, involving both systemic and central immune cells. Surprisingly, the literature investigating depression, inflammation, and metabolic syndrome together remains sparse and mostly correlational. Here, we discuss common and divergent inflammatory features between metabolic syndrome and MDD to understand whether inflammation could be a potential mediator behind the comorbidity of these disorders.

Metabolic Inflammation Arises With Obesity

In 1993, Hotamisligil et al. observed that the pro-inflammatory cytokine TNF-α was increased in obese adipose tissue where it directly interfered with the ability of insulin to regulate blood glucose levels (53). Since then, the association between inflammation and insulin resistance, or “metabolic inflammation,” has become the major focus of a large body of research investigating how obesity could lead to T2D. Metabolic inflammation is broadly characterized by altered circulating cytokine profiles, immune cell infiltration into tissues, and activation of inflammatory pathways within tissue parenchyma.

In humans, a vast array of circulating pro-inflammatory cytokines becomes elevated in obese individuals, including monocyte chemoattractant protein 1 (MCP1/CCL2), IL-1β, IL-5, IL-6, IL-8, IL-10, IL-12, IL-18, IFNγ, and TNF-α, and C-reactive protein (CRP) (9, 10, 29, 43, 56, 97, 108). Similarly, in rodents, plasma levels of the cytokines CCL2, CXCL1, CXCL5, IL-1β, IL-6, and TNF-α are higher in genetic or diet-induced models of obesity (22, 60, 92). Many of these cytokines have been implicated in metabolic inflammation and subsequent metabolic dysfunction. For example, IL-1β, IL-6, and TNF-α have been demonstrated to directly contribute to insulin resistance by activating stress kinases, such as IKK, JNK, and p38 MAPK in muscle and fat cells, which phosphorylate inhibitory serine residues on insulin receptor substrate 1 (IRS1), immediately downstream of the insulin receptor, thereby blocking signal transduction (13, 51, 52, 59, 104). The cytokines IL-12 and IFNγ play key roles in activating the immune system; IL-12 promotes Th1 differentiation of naive CD4+ T cells, and IFNγ promotes pro-inflammatory M1 polarization of macrophages (8, 49, 77). Importantly, these immune cell activation states have been correlated with obesity and insulin resistance in both humans and rodents (70, 79). Furthermore, chemokines such as CCL2 and CXCL1 are capable of chemoattraction, leading to immune cell egress from bone marrow and eventual migration into tissues (26, 111).

During metabolic syndrome or obesity, blood leukocyte frequencies become significantly altered, with humans and mice both displaying leukocytosis (89, 125). More specifically, total monocyte, neutrophil and lymphocyte counts increase and positively correlate with body mass index (BMI), body fat percentage, and insulin resistance (105, 143). Diet-induced obese mouse models have additionally revealed increases in the frequencies of these cell types along with their progenitors in the bone marrow (114, 115, 125), implicating mechanisms that drive both differentiation and release from bone marrow stores. Phenotypically, there have been reported increases in non-classical (CD14+ CD16++) and intermediate (CD14++ CD16+) monocytes and decreased classical (CD14++ CD16) monocytes in blood from obese patients (30, 97). Interestingly, although classical monocytes are typically associated with greater expression of CCR2, the receptor for CCL2, surface levels of chemokine receptors, including CCR2 and CCR5, rise with obesity (62). Additionally, monocytes from obese patients appear primed for activation, since they produce higher amounts of cytokines when stimulated ex vivo with lipopolysaccharide (LPS), a component of the outer membrane of gram-negative bacteria (30). In parallel, high fat feeding in mice increases circulating Ly6Chi monocytes (114), the subset which has greater CCR2 expression and which is considered to be more inflammatory in nature (41). Moreover, monocytes from obese patients exhibit a stronger capability to migrate toward the chemokines CCL2 and CCL5 (62). This suggests that obesity could enhance monocyte migration from bone marrow to blood and eventual tissue infiltration.

A defining feature of obesity-linked metabolic inflammation is the infiltration of immune cells into tissues that regulate whole-body glucose metabolism. This phenomenon was first observed as an increase in total macrophages in the adipose tissue from obese patients (130, 141), but a series of subsequent studies have now established that T cells, B cells, eosinophils, mast cells, dendritic cells, natural killer cells, and neutrophils can all infiltrate adipose tissue to varying degrees and contribute to the regulation of insulin sensitivity (23, 69, 123, 131, 132, 138, 139). Substantial attention has been given to the role of tissue macrophages during the development of T2D, since they increase in other metabolic tissues such as liver and skeletal muscle as well, although other leukocyte subtypes such as neutrophils have also been reported to infiltrate these tissues (38, 39, 88, 123). Macrophages that accumulate in tissues during obesity are largely monocyte-derived with their appearance in tissues dependent on CCL2 and CCR2 (22, 129). However, there is also some evidence that obesity triggers local proliferation of tissue resident macrophages in adipose tissue (2, 144). Metabolic status can also influence macrophage differentiation to produce a spectrum of inflammatory states. For example, in adipose tissue from lean humans or mice, macrophages assume an anti- or non-inflammatory M2 polarization, whereas high fat feeding or obesity stimulates a conversion to pro-inflammatory M1 macrophages (70). These M1 macrophages are characterized by elevated CD11c surface expression, as well as heightened production of pro-inflammatory cytokines including CCL2, IL-6, and TNF-α (68, 70). Mechanistically, saturated fatty acids, a major component of obesogenic and atherogenic diets, but not unsaturated fatty acids, are able to directly activate pro-inflammatory pathways in macrophages ex vivo (20, 112). Notably, selectively depleting total or CD11c+ macrophages from adipose tissue or interfering with CCL2/CCR2 alleviates diet-induced insulin resistance (15, 37, 94, 129), demonstrating that macrophages contribute to whole-body glycemic dysregulation. Accompanying the inflammatory response by immune cells is activation of intrinsic pro-inflammatory pathways in parenchymal cells of metabolic tissues such as adipocytes, hepatocytes, and myocytes. For example, exposure of these cells to excess saturated fats or inflammatory stimuli can directly induce insulin resistance or release of chemoattractants, which exacerbate leukocyte tissue infiltration, thus propagating the cycle of metabolic inflammation (96, 121).

In addition to peripheral inflammation that occurs with obesity, emerging literature has explored inflammation in the central nervous system (CNS), with many of these studies focusing on the hypothalamus given its role in regulating appetite and satiety (109). Importantly, high saturated fat diets trigger hypothalamic inflammation, leading to central leptin and insulin resistance, resulting in increased food intake and weight gain (84). These findings have prompted further investigation into the involvement of central immune cells in metabolic dysfunction. In obese humans (BMI > 30), accumulation of microglia (CNS-resident macrophages) as defined by expression of ionized calcium-binding adaptor molecule 1 (Iba1) is increased in the hypothalamus compared with healthy (BMI < 25) controls (11). BMI has also been positively correlated with frontal cortex expression of iNOS, a hallmark M1 macrophage marker, and negatively correlated with expression of IL-10, an M2 macrophage marker (65). In mice, high-fat diets induce hypothalamic and hippocampal cytokine expression, including IL-1β, IL-6, and TNF-α (31, 83), as well as CNS macrophage infiltration, with one study proposing that these cells originate from blood monocytes (16). However, the ability of peripheral leukocytes to enter the brain parenchyma is controversial and may be an artifact of experimental techniques such as irradiation (85). Some authors have hypothesized that high-fat diets or diet-associated inflammation can result in the breakdown of the blood-brain barrier (BBB), thus permitting access to the CNS by peripheral immune cells or cytokines. Indeed, one study has suggested that BBB permeability as assessed by the CSF-to-serum albumin ratio is higher in overweight or obese humans compared with healthy controls (45). Obese mice and rats also show BBB breakdown as measured by a sodium fluorescein permeability assay (119), with one report highlighting this occurrence specifically at the hippocampus, but no differences were observed in the prefrontal cortex or striatum (57). Additionally, this permeability is accompanied by decreased gene expression of tight-junction proteins, occludin, claudin-5, and claudin-12 in BBB capillaries and choroid plexus (57). Other studies have shown that saturated fats increase adhesion molecule expression on microvascular endothelial cells, thus promoting monocyte adhesion and transmigration (95). Cytokines may also be produced by endothelial cells, with one study compellingly demonstrating that adiponectin, which stimulates negative energy balance and is decreased in obesity, suppresses adluminal IL-6 secretion from BBB endothelial cells into the brain (116). Furthermore, compromised BBB integrity has been directly correlated with the appearance of CD45+ CD11b+ (myeloid) and CD11b+ Ly6chi (inflammatory monocyte) cells in the forebrain (119). These immune cells may provoke metabolic syndrome, since depleting microglia using a colony-stimulating factor 1 receptor (CSF1R) inhibitor or preventing their activation via genetic manipulation in the mediobasal hypothalamus has beneficial metabolic effects, primarily by suppressing food intake and weight gain (126). Collectively, these studies underscore a critical role for immune cells, along with central and peripheral inflammation in the development of metabolic syndrome, and have given rise to the field of “immunometabolism.”

Major Depressive Disorder is Associated With Central and Peripheral Inflammation

A major revelation in understanding the etiology of mood disorders including MDD has been the discovery of an inflammatory component to these conditions. A key observation was reported in 1987 when a subset of hepatitis C patients treated with the pro-inflammatory cytokine interferon alpha were found to develop depression, which then subsided upon cessation of treatment (102). Further parallels have been observed between immune/inflammatory illness and depression, which share some common behavioral changes, including lethargy, anhedonia, and social withdrawal—a concept termed “sickness behavior” (25). Evolutionarily, these now maladaptive behaviors have been hypothesized to have previously played a role in facilitating recovery while preventing transmission of infectious disease (86). Current research has confirmed that pro-inflammatory cytokines or pathogenic compounds such as LPS can produce depression-like behavior in both humans and rodents (40, 44, 61, 142). These findings have fueled a necessity to elucidate inflammatory changes during MDD and develop potential novel drug targets.

Similar to what is seen in patients with metabolic syndrome, individuals with MDD display a chronic, low-grade inflammation, which can be characterized through circulating cytokine profiles. A number of studies involving patients meeting Diagnostic and Statistical Manual of Mental Disorders (DSM) criteria for MDD have found significant elevations in plasma or serum levels of CCL2, IFNγ, IL-1α, IL-1β, IL-2, IL-6, IL-8, IL-12, and TNF-α, along with CRP (32, 50, 73, 113, 133). The “cytokine hypothesis of depression” postulates that these cytokines play a causative role in MDD progression (107). One study found that, in treatment-resistant MDD patients, administration of the TNF-α antagonist infliximab relieved depressive symptoms in individuals with high baseline inflammatory markers (101). Although human MDD is challenging to model in animals, a variety of paradigms, such as chronic unpredictable stress, learned helplessness, chronic social defeat stress, and witness defeat, have been established to recapitulate stress-induced long-lasting depression-like behavior in rodents (for review, see Ref. 80) that have been used to test causal hypotheses about the role of inflammation in depression. Compared with control mice, stressed mice displaying depression-like behaviors exhibit increased circulating cytokines including CCL2, CXCL1, IL-1β, IL-6, and TNF-α (14, 50, 118, 120). Central infusion of IL-1 receptor antagonist, which inhibits IL-1β signaling, prevents social defeat stress-induced social avoidance in rats (5). Furthermore, antibody-mediated neutralization of IL-6, or genetic depletion of IL-6 from peripheral leukocytes by bone marrow transplantation, attenuates susceptibility to chronic social defeat stress (50). These studies emphasize an important bi-directional interaction between the brain and immune system. However, several key questions remain. First, what are the mechanisms that trigger cytokine production? Studies of individual differences in stress-induced cytokine production among inbred C57BL/6J mice suggest that there may be cell intrinsic differences in leukocyte reactivity that promote inflammation and depressive-like behaviors (50). Next, how does inflammation influence behavior, and how do specific immune cell subtypes or compartments contribute to these processes?

To begin to address these questions, studies have focused predominantly on three compartments of immune cells: CNS resident immune cells, splenic macrophages, and peripheral leukocytes in the systemic circulation that may communicate with the brain. Leukocytosis has been identified in blood from both MDD patients and mouse models of depression (50, 75). When specific populations of immune cells were investigated, monocytes and neutrophils have been reported to increase, whereas lymphocytes decrease, with perceived stress and depression in humans and with chronic unpredictable stress in mice (28, 35, 47, 75). Studies in mice have shown that chronic stress activates hematopoiesis in the bone marrow and promotes an increase in circulating Ly6Chi monocytes (47). Moreover, subthreshold stress following social defeat triggers Ly6Chi monocyte egress from the spleen, and splenectomy before social defeat prevents monocyte traffic to the brain (78). During stressful events, activation of the hypothalamus-pituitary-adrenal (HPA) axis causes release of glucocorticoids such as cortisol from the adrenal glands into the bloodstream (145). Although glucocorticoids are normally immunosuppressive, it is hypothesized that chronic stress elicits HPA hyperactivity, leading to glucocorticoid resistance, which begets pro-inflammatory activation of immune cells (4, 91). Interestingly, peripheral leukocytes from stress-susceptible mice release greater amounts of IL-6 following stimulation with LPS compared with leukocytes from stress-resilient mice (50). Additional studies are required to further evaluate how chronic stress affects immune cell differentiation and function.

The concept of peripheral immune cells infiltrating the brain during chronic stress and MDD, however, is controversial. Some studies have reported stress-induced, CCL2/CCR2-dependent recruitment of peripheral monocytes to the brain (78, 135). Others have shown that, although BBB permeability increases in both humans and mice during depression or chronic stress, respectively, in a manner similar to what has been reported during metabolic syndrome, monocytes may remain in the ventricular space and vasculature without entering the brain parenchyma (81, 90). Importantly, stress-induced BBB breakdown was observed to occur at the nucleus accumbens (NAc), a region of the striatum involved in motivation and reward, along with the hippocampus, but not the prefrontal cortex or hypothalamus (81). Although the specific mechanisms driving stress-induced alterations in the BBB are not well established, there is evidence from a learned helplessness model of depression to suggest that peripheral cytokines, such as TNF-α, may promote the breakdown of the BBB (21). These studies establish a potential interface for peripheral signals to affect brain and behavior. There is also significant evidence to show that resident immune cells in the brain adopt an activated phenotype during chronic stress and depression. In one study, a positron emission tomography approach was applied to measure translocator protein density measured by distribution volume (TSPO VT), a marker of activated microglia, in MDD patients experiencing a major depressive episode. The authors found significantly increased TSPO VT in the prefrontal cortex, anterior cingulate cortex, and insula in subjects with MDD compared with controls (110). Similar microglia activation has been seen following chronic unpredictable stress in the hippocampus of rats (128).

Within the brain, there have been several proposed mechanisms by which cytokines and immune cells could affect behavior. Cytokines such as IL-1β, IL-6, and TNF-α become elevated in the brain during chronic stress or depression, and arise from local production in the CNS or translocation across the BBB from the periphery (81, 93, 134). MDD pathophysiology has been characterized by defective signaling of monoamine neutrotransmitters, such as serotonin (5-HT), dopamine (DA), and norepinephrine (NE). Inflammatory cytokines including IL-6 and TNF-α have been shown to activate indoleamine-2,3-dioxygenase (IDO), consequently reducing 5-HT synthesis by reducing the availability of its precursor tryptophan (74). IDO activation favors conversion of tryptophan into metabolites of the kynurenine pathway, 3-hydroxykynurenine, and quinolinic acid. These metabolites, which are thought to increase with anxiety and depression, are excitotoxic and can also activate monoamine oxidase (MAO), a target of antidepressants that degrades 5-HT, DA, and NE (107). However, a meta-analysis of nine studies found that circulating kynurenine levels decrease in patients with unipolar depression compared with healthy controls (6), indicating that further research is required to elucidate the role of kynurenine in depression. Similarly, cytokines may interfere with DA synthesis by reducing bioavailability of tetrahydrobiopterin (BH4), an important enzyme co-factor involved in metabolism of phenylalanine to DA (36). There is also a growing literature to suggest that cytokines are capable of acting directly on neurons through cytokine receptors within the plasma membrane to change excitability, synaptic strength, and synaptic scaling (12, 67, 117, 127). Furthermore, cytokines such as IL-1β can contribute to heightened activation of the HPA axis, thus compounding the inflammatory response to stress (106). Recent work has suggested that microglia also exhibit enhanced phagocytic activity during chronic stress, which may be involved in synaptic remodeling (66). Together, these studies illustrate multiple pathways whereby chronic stress—through activation of the immune system—can promote depressive-like behavior.

Metabolic Syndrome and Major Depressive Disorder Exhibit a Bi-Directional Relationship

With both metabolic syndrome and MDD showing many inflammatory perturbations in common, it is tempting to speculate that inflammation mediates their frequent comorbidity. One clinical evaluation of MDD patients reported elevated circulating IL-6 and CRP, most notably in patients with the highest BMI (87). This association is often seen in atypical depression, where subjects show increased appetite along with symptoms of depression, which then correlate with greater serum TNF-α and CRP (64). A genome-wide association study also identified that these patients carry genetic risk variants for increased BMI, CRP, and leptin levels (82). In a surgery-induced weight-loss intervention study, formerly obese patients displayed reduced inflammation and significant improvements in emotional status following weight loss (17). Additional studies found positive correlations between CRP levels and depressive mood in obese but not non-obese subjects (27, 63), suggesting that inflammation may mediate a subset of depressive disorders commonly concurrent with obesity and metabolic syndrome. Intriguingly, in a twin study involving 323 pairs of male twins with or without metabolic syndrome, the authors observed that incidence of metabolic syndrome is associated with higher depressive symptoms, with inflammation (circulating IL-6 and CRP) identified as a determinant variable in this relationship (18). However, it is difficult to determine causative effects in humans, so investigation using preclinical rodent models can provide valuable and complementary mechanistic insights.

Animal models of depression and obesity have revealed a bi-directional relationship between these disorders. For example, high fat-fed mice show decreased social interaction, sucrose preference, and grooming during splash test, along with increased immobility time during forced swim and tail-suspension tests, with no differences seen in open-field or the elevated plus maze (46, 55, 140). One study comparing control diet, diet-induced obesity, and isocaloric high-fat diet suggests that diet-induced behavioral changes are affected by diet, independent of weight gain and obesity (122), indicating that specific dietary factors or metabolites can confer depression-like symptoms. Inversely, chronic mild stress in rats has been shown to worsen glucose tolerance (7). The detrimental effects of chronic stress or high-fat diet appear to be additive, as social defeat exacerbates diet-induced elevations in plasma glucose and lipid levels. Moreover, high-fat feeding in the Flinders sensitive line genetic rat model of depression resulted in higher immobility time in forced swim test, interpreted as behavioral despair, and worsened insulin tolerance (1, 24). These observations, however, are mixed, since some studies do not find an effect of diet on stress-induced depression-like behavior (24). Conversely, caloric restriction has been shown to promote resilience to social defeat stress through regulation of a variety of feeding peptides, such as orexin and ghrelin (71, 72). However, to our surprise, there is remarkably little research exploring a functional link between inflammation, metabolic dysfunction, and depression. One recent report by Dutheil et al. (34) found that high-fat diets promoted anxiety- and depression-like behavior assessed by the novelty suppressed feeding and sucrose preference tests, alongside expected increases in glucose intolerance and hippocampal IL-1β, IL-6, and TNF-α levels. Interestingly, when these high-fat-fed rats were treated with a P2X7 receptor antagonist, which blocks inflammasome activation and cytokine production, diet-induced behavioral abnormalities were attenuated (34), indicating that inflammation may indeed be the common link between metabolic and behavioral dysfunction. Future questions examining the role of specific immune cells and cytokines in the relationship between metabolic syndrome and depression remain an open topic.

Conclusions and Future Directions

The prevalence and comorbidity of metabolic syndrome and MDD is a continuously growing epidemic with major health and economic burdens. The revelation that both of these debilitating disorders may have common inflammatory origins, involving both the central and peripheral immune system, has afforded us a greater understanding of the heterogeneity of these conditions (FIGURE 1). Although research to date in the fields of immunometabolism and neuroinflammation has produced encouraging preliminary results, there remains a vast expanse of unexplored questions requiring the interdisciplinary knowledge of metabolism, neuroscience, and immunology. Animal models remain an invaluable tool to establish causal links between inflammation, metabolism, and depression. Although correlational data from human cohorts suggest that inflammation associated with depression is exacerbated by obesity, studies in rodents have yet to determine whether chronic stress and obesity have additive or synergistic effects on whole-body inflammation. Moreover, genetic manipulation or neutralizing antibodies can be used to test whether specific cytokines or leukocyte subsets that increase in the CNS or circulation during obesity contribute to depression-like behavior, or whether those that increase with chronic stress lead to dysregulated appetite and glucose metabolism. Elucidating these mechanisms linking metabolic syndrome, depression, and inflammation could generate potential new therapeutic targets or cell type-specific strategies to combat both metabolic syndrome and MDD.

FIGURE 1.

FIGURE 1.

Chronic, low-grade inflammation during metabolic syndrome and major depressive disorder

In the CNS, indexes of inflammation, including microglia accumulation and activation, cytokine expression, and blood-brain barrier (BBB) permeability, appear in both metabolic syndrome and depression. During obesity and metabolic syndrome, this is observed primarily in the hypothalamus and hippocampus, while the nucleus accumbens (NAc) and hippocampus are afflicted with chronic stress or major depressive disorder. In the periphery, pro-inflammatory cytokines, along with circulating monocytes and neutrophils, increase in both disorders; however, there is a reported decrease in blood lymphocytes during depression. Peripheral tissue infiltration by immune cells occurs during metabolic syndrome, but less is known about the effects of depression on these tissues. These inflammatory changes are associated with increased appetite and glucose intolerance during metabolic syndrome, and anhedonia and reduced reward-seeking behavior during depression. Increases and decreases expressed in text boxes are relative to healthy subjects.

Acknowledgments

This review was supported by National Institute of Health Grants RO1 MH-090264, P50 MH-096890 (to S.J.R.), P50 AT-008661-01 (to S.J.R.), RO1 MH-104559 (to S.J.R.), an Early Postdoc Mobility Fellowship (Swiss National Science Foundation), and a Walter and Gertrud Siegenthaler Postdoctoral Fellowship (to F.C.).

No conflicts of interest, financial or otherwise, are declared by the author(s).

K.L.C., F.C., and S.J.R. conception and design of research; K.L.C., F.C., and S.J.R. prepared figures; K.L.C., F.C., and S.J.R. drafted manuscript; K.L.C., F.C., and S.J.R. edited and revised manuscript; K.L.C., F.C., and S.J.R. approved final version of manuscript.

We thank all authors who have contributed to the work summarized in this review.

References

  • 1.Abildgaard A, Solskov L, Volke V, Harvey BH, Lund S, Wegener G. A high-fat diet exacerbates depressive-like behavior in the Flinders Sensitive Line (FSL) rat, a genetic model of depression. Psychoneuroendocrinology 36: 623–633, 2011. doi: 10.1016/j.psyneuen.2010.09.004. [DOI] [PubMed] [Google Scholar]
  • 2.Amano SU, Cohen JL, Vangala P, Tencerova M, Nicoloro SM, Yawe JC, Shen Y, Czech MP, Aouadi M. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab 19: 162–171, 2014. doi: 10.1016/j.cmet.2013.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders (5th ed.). Arlington, VA: American Psychiatric Publishing, 2013. [Google Scholar]
  • 4.Anacker C, Zunszain PA, Carvalho LA, Pariante CM. The glucocorticoid receptor: pivot of depression and of antidepressant treatment? Psychoneuroendocrinology 36: 415–425, 2011. doi: 10.1016/j.psyneuen.2010.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Arakawa H, Blandino P Jr, Deak T. Central infusion of interleukin-1 receptor antagonist blocks the reduction in social behavior produced by prior stressor exposure. Physiol Behav 98: 139–146, 2009. doi: 10.1016/j.physbeh.2009.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Arnone D, Saraykar S, Salem H, Teixeira AL, Dantzer R, Selvaraj S. Role of Kynurenine pathway and its metabolites in mood disorders: a systematic review and meta-analysis of clinical studies. Neurosci Biobehav Rev 92: 477–485, 2018. doi: 10.1016/j.neubiorev.2018.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Aslani S, Vieira N, Marques F, Costa PS, Sousa N, Palha JA. The effect of high-fat diet on rat’s mood, feeding behavior and response to stress. Transl Psychiatry 5: e684, 2015. doi: 10.1038/tp.2015.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Athie-Morales V, Smits HH, Cantrell DA, Hilkens CMU. Sustained IL-12 signaling is required for Th1 development. J Immunol 172: 61–69, 2004. doi: 10.4049/jimmunol.172.1.61. [DOI] [PubMed] [Google Scholar]
  • 9.Aygun AD, Gungor S, Ustundag B, Gurgoze MK, Sen Y. Proinflammatory cytokines and leptin are increased in serum of prepubertal obese children. Mediators Inflamm 2005: 180–183, 2005. doi: 10.1155/MI.2005.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bastard JP, Jardel C, Bruckert E, Blondy P, Capeau J, Laville M, Vidal H, Hainque B. Elevated levels of interleukin 6 are reduced in serum and subcutaneous adipose tissue of obese women after weight loss. J Clin Endocrinol Metab 85: 3338–3342, 2000. doi: 10.1210/jc.85.9.3338. [DOI] [PubMed] [Google Scholar]
  • 11.Baufeld C, Osterloh A, Prokop S, Miller KR, Heppner FL. High-fat diet-induced brain region-specific phenotypic spectrum of CNS resident microglia. Acta Neuropathol 132: 361–375, 2016. doi: 10.1007/s00401-016-1595-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK, Von Zastrow M, Beattie MS, Malenka RC. Control of synaptic strength by glial TNFalpha. Science 295: 2282–2285, 2002. doi: 10.1126/science.1067859. [DOI] [PubMed] [Google Scholar]
  • 13.Broussard SR, McCusker RH, Novakofski JE, Strle K, Shen WH, Johnson RW, Dantzer R, Kelley KW. IL-1beta impairs insulin-like growth factor i-induced differentiation and downstream activation signals of the insulin-like growth factor i receptor in myoblasts. J Immunol 172: 7713–7720, 2004. doi: 10.4049/jimmunol.172.12.7713. [DOI] [PubMed] [Google Scholar]
  • 14.Brydon L, Edwards S, Jia H, Mohamed-Ali V, Zachary I, Martin JF, Steptoe A. Psychological stress activates interleukin-1beta gene expression in human mononuclear cells. Brain Behav Immun 19: 540–546, 2005. doi: 10.1016/j.bbi.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 15.Bu L, Gao M, Qu S, Liu D. Intraperitoneal injection of clodronate liposomes eliminates visceral adipose macrophages and blocks high-fat diet-induced weight gain and development of insulin resistance. AAPS J 15: 1001–1011, 2013. doi: 10.1208/s12248-013-9501-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Buckman LB, Hasty AH, Flaherty DK, Buckman CT, Thompson MM, Matlock BK, Weller K, Ellacott KLJ. Obesity induced by a high-fat diet is associated with increased immune cell entry into the central nervous system. Brain Behav Immun 35: 33–42, 2014. doi: 10.1016/j.bbi.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Capuron L, Poitou C, Machaux-Tholliez D, Frochot V, Bouillot J-L, Basdevant A, Layé S, Clément K. Relationship between adiposity, emotional status and eating behaviour in obese women: role of inflammation. Psychol Med 41: 1517–1528, 2011. doi: 10.1017/S0033291710001984. [DOI] [PubMed] [Google Scholar]
  • 18.Capuron L, Su S, Miller AH, Bremner JD, Goldberg J, Vogt GJ, Maisano C, Jones L, Murrah NV, Vaccarino V. Depressive symptoms and metabolic syndrome: is inflammation the underlying link? Biol Psychiatry 64: 896–900, 2008. doi: 10.1016/j.biopsych.2008.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Carey M, Small H, Yoong SL, Boyes A, Bisquera A, Sanson-Fisher R. Prevalence of comorbid depression and obesity in general practice: a cross-sectional survey. Br J Gen Pract 64: e122–e127, 2014. doi: 10.3399/bjgp14X677482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chan KL, Pillon NJ, Sivaloganathan DM, Costford SR, Liu Z, Théret M, Chazaud B, Klip A. Palmitoleate reverses high fat-induced proinflammatory macrophage polarization via AMP-activated protein kinase (AMPK). J Biol Chem 290: 16979–16988, 2015. doi: 10.1074/jbc.M115.646992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chandler S, Miller KM, Clements JM, Lury J, Corkill D, Anthony DC, Adams SE, Gearing AJ. Matrix metalloproteinases, tumor necrosis factor and multiple sclerosis: an overview. J Neuroimmunol 72: 155–161, 1997. doi: 10.1016/S0165-5728(96)00179-8. [DOI] [PubMed] [Google Scholar]
  • 22.Chen A, Mumick S, Zhang C, Lamb J, Dai H, Weingarth D, Mudgett J, Chen H, MacNeil DJ, Reitman ML, Qian S. Diet induction of monocyte chemoattractant protein-1 and its impact on obesity. Obes Res 13: 1311–1320, 2005. doi: 10.1038/oby.2005.159. [DOI] [PubMed] [Google Scholar]
  • 23.Cho KW, Zamarron BF, Muir LA, Singer K, Porsche CE, DelProposto JB, Geletka L, Meyer KA, O’Rourke RW, Lumeng CN. Adipose tissue dendritic cells are independent contributors to obesity-induced inflammation and insulin resistance. J Immunol 197: 3650–3661, 2016. doi: 10.4049/jimmunol.1600820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chuang J-C, Cui H, Mason BL, Mahgoub M, Bookout AL, Yu HG, Perello M, Elmquist JK, Repa JJ, Zigman JM, Lutter M. Chronic social defeat stress disrupts regulation of lipid synthesis. J Lipid Res 51: 1344–1353, 2010. doi: 10.1194/jlr.M002196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Dantzer R, 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 9: 46–56, 2008. doi: 10.1038/nrn2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Delano MJ, Kelly-Scumpia KM, Thayer TC, Winfield RD, Scumpia PO, Cuenca AG, Harrington PB, O’Malley KA, Warner E, Gabrilovich S, Mathews CE, Laface D, Heyworth PG, Ramphal R, Strieter RM, Moldawer LL, Efron PA. Neutrophil mobilization from the bone marrow during polymicrobial sepsis is dependent on CXCL12 signaling. J Immunol 187: 911–918, 2011. doi: 10.4049/jimmunol.1100588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Delgado I, Huet L, Dexpert S, Beau C, Forestier D, Ledaguenel P, Aubert A, Sauvant J, Aouizerate B, Magne E, Capuron L. Depressive symptoms in obesity: Relative contribution of low-grade inflammation and metabolic health. Psychoneuroendocrinology 91: 55–61, 2018. doi: 10.1016/j.psyneuen.2018.02.030. [DOI] [PubMed] [Google Scholar]
  • 28.Demir S, Atli A, Bulut M, İbiloğlu AO, Güneş M, Kaya MC, Demirpençe Ö, Sır A. Neutrophil-lymphocyte ratio in patients with major depressive disorder undergoing no pharmacological therapy. Neuropsychiatr Dis Treat 11: 2253–2258, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res 29: 313–326, 2009. doi: 10.1089/jir.2008.0027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Devêvre EF, Renovato-Martins M, Clément K, Sautès-Fridman C, Cremer I, Poitou C. Profiling of the three circulating monocyte subpopulations in human obesity. J Immunol 194: 3917–3923, 2015. doi: 10.4049/jimmunol.1402655. [DOI] [PubMed] [Google Scholar]
  • 31.Dinel A-L, André C, Aubert A, Ferreira G, Layé S, Castanon N. Cognitive and emotional alterations are related to hippocampal inflammation in a mouse model of metabolic syndrome. PLoS One 6: e24325, 2011. doi: 10.1371/journal.pone.0024325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, Lanctôt KL. A meta-analysis of cytokines in major depression. Biol Psychiatry 67: 446–457, 2010. doi: 10.1016/j.biopsych.2009.09.033. [DOI] [PubMed] [Google Scholar]
  • 33.Dunbar JA, Reddy P, Davis-Lameloise N, Philpot B, Laatikainen T, Kilkkinen A, Bunker SJ, Best JD, Vartiainen E, Kai Lo S, Janus ED. Depression: an important comorbidity with metabolic syndrome in a general population. Diabetes Care 31: 2368–2373, 2008. doi: 10.2337/dc08-0175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dutheil S, Ota KT, Wohleb ES, Rasmussen K, Duman RS. High-fat diet induced anxiety and anhedonia: impact on brain homeostasis and inflammation. Neuropsychopharmacology 41: 1874–1887, 2016. doi: 10.1038/npp.2015.357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Engler H, Bailey MT, Engler A, Sheridan JF. Effects of repeated social stress on leukocyte distribution in bone marrow, peripheral blood and spleen. J Neuroimmunol 148: 106–115, 2004. doi: 10.1016/j.jneuroim.2003.11.011. [DOI] [PubMed] [Google Scholar]
  • 36.Felger JC, Miller AH. Cytokine effects on the basal ganglia and dopamine function: the subcortical source of inflammatory malaise. Front Neuroendocrinol 33: 315–327, 2012. doi: 10.1016/j.yfrne.2012.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Feng B, Jiao P, Nie Y, Kim T, Jun D, van Rooijen N, Yang Z, Xu H. Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS One 6: e24358, 2011. doi: 10.1371/journal.pone.0024358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fink LN, Costford SR, Lee YS, Jensen TE, Bilan PJ, Oberbach A, Blüher M, Olefsky JM, Sams A, Klip A. Pro-inflammatory macrophages increase in skeletal muscle of high fat-fed mice and correlate with metabolic risk markers in humans. Obesity (Silver Spring) 22: 747–757, 2014. doi: 10.1002/oby.20615. [DOI] [PubMed] [Google Scholar]
  • 39.Fink LN, Oberbach A, Costford SR, Chan KL, Sams A, Blüher M, Klip A. Expression of anti-inflammatory macrophage genes within skeletal muscle correlates with insulin sensitivity in human obesity and type 2 diabetes. Diabetologia 56: 1623–1628, 2013. doi: 10.1007/s00125-013-2897-x. [DOI] [PubMed] [Google Scholar]
  • 40.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 32: 516–531, 2007. doi: 10.1016/j.psyneuen.2007.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19: 71–82, 2003. doi: 10.1016/S1074-7613(03)00174-2. [DOI] [PubMed] [Google Scholar]
  • 42.Glassman AH. Depression and cardiovascular comorbidity. Dialogues Clin Neurosci 9: 9–17, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Goyal R, Faizy AF, Siddiqui SS, Singhai M. Evaluation of TNF-α and IL-6 levels in obese and non-obese diabetics: pre- and postinsulin effects. N Am J Med Sci 4: 180–184, 2012. doi: 10.4103/1947-2714.94944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Grigoleit J-S, Kullmann JS, Wolf OT, Hammes F, Wegner A, Jablonowski S, Engler H, Gizewski E, Oberbeck R, Schedlowski M. Dose-dependent effects of endotoxin on neurobehavioral functions in humans. PLoS One 6: e28330, 2011. doi: 10.1371/journal.pone.0028330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gustafson DR, Karlsson C, Skoog I, Rosengren L, Lissner L, Blennow K. Mid-life adiposity factors relate to blood-brain barrier integrity in late life. J Intern Med 262: 643–650, 2007. doi: 10.1111/j.1365-2796.2007.01869.x. [DOI] [PubMed] [Google Scholar]
  • 46.Hassan AM, Mancano G, Kashofer K, Fröhlich EE, Matak A, Mayerhofer R, Reichmann F, Olivares M, Neyrinck AM, Delzenne NM, Claus SP, Holzer P. High-fat diet induces depression-like behaviour in mice associated with changes in microbiome, neuropeptide Y, and brain metabolome. Nutr Neurosci 1–17, 2018. doi: 10.1080/1028415X.2018.1465713. [DOI] [PubMed] [Google Scholar]
  • 47.Heidt T, Sager HB, Courties G, Dutta P, Iwamoto Y, Zaltsman A, von Zur Muhlen C, Bode C, Fricchione GL, Denninger J, Lin CP, Vinegoni C, Libby P, Swirski FK, Weissleder R, Nahrendorf M. Chronic variable stress activates hematopoietic stem cells. Nat Med 20: 754–758, 2014. doi: 10.1038/nm.3589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Herva A, Räsänen P, Miettunen J, Timonen M, Läksy K, Veijola J, Laitinen J, Ruokonen A, Joukamaa M. Co-occurrence of metabolic syndrome with depression and anxiety in young adults: the Northern Finland 1966 Birth Cohort Study. Psychosom Med 68: 213–216, 2006. doi: 10.1097/01.psy.0000203172.02305.ea. [DOI] [PubMed] [Google Scholar]
  • 49.Heufler C, Koch F, Stanzl U, Topar G, Wysocka M, Trinchieri G, Enk A, Steinman RM, Romani N, Schuler G. Interleukin-12 is produced by dendritic cells and mediates T helper 1 development as well as interferon-gamma production by T helper 1 cells. Eur J Immunol 26: 659–668, 1996. doi: 10.1002/eji.1830260323. [DOI] [PubMed] [Google Scholar]
  • 50.Hodes GE, Pfau ML, Leboeuf M, Golden SA, Christoffel DJ, Bregman D, Rebusi N, Heshmati M, Aleyasin H, Warren BL, Lebonté B, Horn S, Lapidus KA, Stelzhammer V, Wong EHF, Bahn S, Krishnan V, Bolaños-Guzman CA, Murrough JW, Merad M, Russo SJ. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci USA 111: 16136–16141, 2014. doi: 10.1073/pnas.1415191111. A correction for this article is available at . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hotamisligil GS, Murray DL, Choy LN, Spiegelman BM. Tumor necrosis factor alpha inhibits signaling from the insulin receptor. Proc Natl Acad Sci USA 91: 4854–4858, 1994. doi: 10.1073/pnas.91.11.4854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science 271: 665–668, 1996. doi: 10.1126/science.271.5249.665. [DOI] [PubMed] [Google Scholar]
  • 53.Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259: 87–91, 1993. doi: 10.1126/science.7678183. [DOI] [PubMed] [Google Scholar]
  • 54.Huang PL. A comprehensive definition for metabolic syndrome. Dis Model Mech 2: 231–237, 2009. doi: 10.1242/dmm.001180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Isingrini E, Camus V, Le Guisquet A-M, Pingaud M, Devers S, Belzung C. Association between repeated unpredictable chronic mild stress (UCMS) procedures with a high fat diet: a model of fluoxetine resistance in mice. PLoS One 5: e10404, 2010. doi: 10.1371/journal.pone.0010404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jung C, Gerdes N, Fritzenwanger M, Figulla HR. Circulating levels of interleukin-1 family cytokines in overweight adolescents. Mediators Inflamm 2010: 958403, 2010. doi: 10.1155/2010/958403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kanoski SE, Zhang Y, Zheng W, Davidson TL. The effects of a high-energy diet on hippocampal function and blood-brain barrier integrity in the rat. J Alzheimers Dis 21: 207–219, 2010. doi: 10.3233/JAD-2010-091414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 62: 593–602, 2005. doi: 10.1001/archpsyc.62.6.593. [DOI] [PubMed] [Google Scholar]
  • 59.Kim H-J, Higashimori T, Park S-Y, Choi H, Dong J, Kim Y-J, Noh H-L, Cho Y-R, Cline G, Kim Y-B, Kim JK. Differential effects of interleukin-6 and -10 on skeletal muscle and liver insulin action in vivo. Diabetes 53: 1060–1067, 2004. doi: 10.2337/diabetes.53.4.1060. [DOI] [PubMed] [Google Scholar]
  • 60.Kim K-A, Gu W, Lee I-A, Joh E-H, Kim D-H. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One 7: e47713, 2012. doi: 10.1371/journal.pone.0047713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Konsman JP, Parnet P, Dantzer R. Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci 25: 154–159, 2002. doi: 10.1016/S0166-2236(00)02088-9. [DOI] [PubMed] [Google Scholar]
  • 62.Krinninger P, Ensenauer R, Ehlers K, Rauh K, Stoll J, Krauss-Etschmann S, Hauner H, Laumen H. Peripheral monocytes of obese women display increased chemokine receptor expression and migration capacity. J Clin Endocrinol Metab 99: 2500–2509, 2014. doi: 10.1210/jc.2013-2611. [DOI] [PubMed] [Google Scholar]
  • 63.Ladwig K-H, Marten-Mittag B, Löwel H, Döring A, Koenig W. Influence of depressive mood on the association of CRP and obesity in 3205 middle aged healthy men. Brain Behav Immun 17: 268–275, 2003. doi: 10.1016/S0889-1591(03)00056-4. [DOI] [PubMed] [Google Scholar]
  • 64.Lamers F, Milaneschi Y, de Jonge P, Giltay EJ, Penninx BWJH. Metabolic and inflammatory markers: associations with individual depressive symptoms. Psychol Med 48: 1102–1110, 2018. doi: 10.1017/S0033291717002483. [DOI] [PubMed] [Google Scholar]
  • 65.Lauridsen JK, Olesen RH, Vendelbo J, Hyde TM, Kleinman JE, Bibby BM, Brock B, Rungby J, Larsen A. High BMI levels associate with reduced mRNA expression of IL10 and increased mRNA expression of iNOS (NOS2) in human frontal cortex. Transl Psychiatry 7: e1044, 2017. doi: 10.1038/tp.2016.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lehmann ML, Cooper HA, Maric D, Herkenham M. Social defeat induces depressive-like states and microglial activation without involvement of peripheral macrophages. J Neuroinflammation 13: 224, 2016. doi: 10.1186/s12974-016-0672-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lewitus GM, Konefal SC, Greenhalgh AD, Pribiag H, Augereau K, Stellwagen D. Microglial TNF-α suppresses cocaine-induced plasticity and behavioral sensitization. Neuron 90: 483–491, 2016. doi: 10.1016/j.neuron.2016.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li P, Lu M, Nguyen MTA, Bae EJ, Chapman J, Feng D, Hawkins M, Pessin JE, Sears DD, Nguyen A-K, Amidi A, Watkins SM, Nguyen U, Olefsky JM. Functional heterogeneity of CD11c-positive adipose tissue macrophages in diet-induced obese mice. J Biol Chem 285: 15333–15345, 2010. doi: 10.1074/jbc.M110.100263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liu J, Divoux A, Sun J, Zhang J, Clément K, Glickman JN, Sukhova GK, Wolters PJ, Du J, Gorgun CZ, Doria A, Libby P, Blumberg RS, Kahn BB, Hotamisligil GS, Shi G-P. Genetic deficiency and pharmacological stabilization of mast cells reduce diet-induced obesity and diabetes in mice. Nat Med 15: 940–945, 2009. doi: 10.1038/nm.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175–184, 2007. doi: 10.1172/JCI29881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lutter M, Krishnan V, Russo SJ, Jung S, McClung CA, Nestler EJ. Orexin signaling mediates the antidepressant-like effect of calorie restriction. J Neurosci 28: 3071–3075, 2008. doi: 10.1523/JNEUROSCI.5584-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lutter M, Sakata I, Osborne-Lawrence S, Rovinsky SA, Anderson JG, Jung S, Birnbaum S, Yanagisawa M, Elmquist JK, Nestler EJ, Zigman JM. The orexigenic hormone ghrelin defends against depressive symptoms of chronic stress. Nat Neurosci 11: 752–753, 2008. doi: 10.1038/nn.2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Maes M, Bosmans E, De Jongh R, Kenis G, Vandoolaeghe E, Neels H. Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine 9: 853–858, 1997. doi: 10.1006/cyto.1997.0238. [DOI] [PubMed] [Google Scholar]
  • 74.Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new ‘5-HT’ hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 35: 702–721, 2011. doi: 10.1016/j.pnpbp.2010.12.017. [DOI] [PubMed] [Google Scholar]
  • 75.Maes M, Van der Planken M, Stevens WJ, Peeters D, DeClerck LS, Bridts CH, Schotte C, Cosyns P. Leukocytosis, monocytosis and neutrophilia: hallmarks of severe depression. J Psychiatr Res 26: 125–134, 1992. doi: 10.1016/0022-3956(92)90004-8. [DOI] [PubMed] [Google Scholar]
  • 76.Mandelli L, Serretti A. Gene environment interaction studies in depression and suicidal behavior: An update. Neurosci Biobehav Rev 37: 2375–2397, 2013. doi: 10.1016/j.neubiorev.2013.07.011. [DOI] [PubMed] [Google Scholar]
  • 77.Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6: 13, 2014. doi: 10.12703/P6-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.McKim DB, Weber MD, Niraula A, Sawicki CM, Liu X, Jarrett BL, Ramirez-Chan K, Wang Y, Roeth RM, Sucaldito AD, Sobol CG, Quan N, Sheridan JF, Godbout JP. Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Mol Psychiatry 23: 1421–1431, 2018. doi: 10.1038/mp.2017.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.McLaughlin T, Liu L-F, Lamendola C, Shen L, Morton J, Rivas H, Winer D, Tolentino L, Choi O, Zhang H, Hui Yen Chng M, Engleman E. T-cell profile in adipose tissue is associated with insulin resistance and systemic inflammation in humans. Arterioscler Thromb Vasc Biol 34: 2637–2643, 2014. doi: 10.1161/ATVBAHA.114.304636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ménard C, Hodes GE, Russo SJ. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 321: 138–162, 2016. doi: 10.1016/j.neuroscience.2015.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Menard C, Pfau ML, Hodes GE, Kana V, Wang VX, Bouchard S, Takahashi A, Flanigan ME, Aleyasin H, LeClair KB, Janssen WG, Labonté B, Parise EM, Lorsch ZS, Golden SA, Heshmati M, Tamminga C, Turecki G, Campbell M, Fayad ZA, Tang CY, Merad M, Russo SJ. Social stress induces neurovascular pathology promoting depression. Nat Neurosci 20: 1752–1760, 2017. doi: 10.1038/s41593-017-0010-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Milaneschi Y, Lamers F, Peyrot WJ, Baune BT, Breen G, Dehghan A, Forstner AJ, Grabe HJ, Homuth G, Kan C, Lewis C, Mullins N, Nauck M, Pistis G, Preisig M, Rivera M, Rietschel M, Streit F, Strohmaier J, Teumer A, Van der Auwera S, Wray NR, Boomsma DI, Penninx BWJH; CHARGE Inflammation Working Group and the Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium . Genetic association of major depression with atypical features and obesity-related immunometabolic dysregulations. JAMA Psychiatry 74: 1214–1225, 2017. doi: 10.1001/jamapsychiatry.2017.3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Milanski M, Arruda AP, Coope A, Ignacio-Souza LM, Nunez CE, Roman EA, Romanatto T, Pascoal LB, Caricilli AM, Torsoni MA, Prada PO, Saad MJ, Velloso LA. Inhibition of hypothalamic inflammation reverses diet-induced insulin resistance in the liver. Diabetes 61: 1455–1462, 2012. doi: 10.2337/db11-0390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, Tsukumo DML, Anhe G, Amaral ME, Takahashi HK, Curi R, Oliveira HC, Carvalheira JBC, Bordin S, Saad MJ, Velloso LA. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: implications for the pathogenesis of obesity. J Neurosci 29: 359–370, 2009. doi: 10.1523/JNEUROSCI.2760-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch U-K, Mack M, Heikenwalder M, Brück W, Priller J, Prinz M. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci 10: 1544–1553, 2007. doi: 10.1038/nn2015. [DOI] [PubMed] [Google Scholar]
  • 86.Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol 16: 22–34, 2016. doi: 10.1038/nri.2015.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Miller GE, Stetler CA, Carney RM, Freedland KE, Banks WA. Clinical depression and inflammatory risk markers for coronary heart disease. Am J Cardiol 90: 1279–1283, 2002. doi: 10.1016/S0002-9149(02)02863-1. [DOI] [PubMed] [Google Scholar]
  • 88.Morinaga H, Mayoral R, Heinrichsdorff J, Osborn O, Franck N, Hah N, Walenta E, Bandyopadhyay G, Pessentheiner AR, Chi TJ, Chung H, Bogner-Strauss JG, Evans RM, Olefsky JM, Oh DY. Characterization of distinct subpopulations of hepatic macrophages in HFD/obese mice. Diabetes 64: 1120–1130, 2015. doi: 10.2337/db14-1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nanji AA, Freeman JB. Relationship between body weight and total leukocyte count in morbid obesity. Am J Clin Pathol 84: 346–347, 1985. doi: 10.1093/ajcp/84.3.346. [DOI] [PubMed] [Google Scholar]
  • 90.Niklasson F, Agren H. Brain energy metabolism and blood-brain barrier permeability in depressive patients: analyses of creatine, creatinine, urate, and albumin in CSF and blood. Biol Psychiatry 19: 1183–1206, 1984. [PubMed] [Google Scholar]
  • 91.Niraula A, Wang Y, Godbout JP, Sheridan JF. Corticosterone production during repeated social defeat causes monocyte mobilization from the bone marrow, glucocorticoid resistance, and neurovascular adhesion molecule expression. J Neurosci 38: 2328–2340, 2018. doi: 10.1523/JNEUROSCI.2568-17.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Nunemaker CS, Chung HG, Verrilli GM, Corbin KL, Upadhye A, Sharma PR. Increased serum CXCL1 and CXCL5 are linked to obesity, hyperglycemia, and impaired islet function. J Endocrinol 222: 267–276, 2014. doi: 10.1530/JOE-14-0126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Pandey GN, Rizavi HS, Ren X, Fareed J, Hoppensteadt DA, Roberts RC, Conley RR, Dwivedi Y. Proinflammatory cytokines in the prefrontal cortex of teenage suicide victims. J Psychiatr Res 46: 57–63, 2012. doi: 10.1016/j.jpsychires.2011.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Patsouris D, Li P-P, Thapar D, Chapman J, Olefsky JM, Neels JG. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab 8: 301–309, 2008. doi: 10.1016/j.cmet.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Pillon NJ, Azizi PM, Li YE, Liu J, Wang C, Chan KL, Hopperton KE, Bazinet RP, Heit B, Bilan PJ, Lee WL, Klip A. Palmitate-induced inflammatory pathways in human adipose microvascular endothelial cells promote monocyte adhesion and impair insulin transcytosis. Am J Physiol Endocrinol Metab 309: E35–E44, 2015. doi: 10.1152/ajpendo.00611.2014. [DOI] [PubMed] [Google Scholar]
  • 96.Pillon NJ, Li YE, Fink LN, Brozinick JT, Nikolayev A, Kuo M-S, Bilan PJ, Klip A. Nucleotides released from palmitate-challenged muscle cells through pannexin-3 attract monocytes. Diabetes 63: 3815–3826, 2014. doi: 10.2337/db14-0150. [DOI] [PubMed] [Google Scholar]
  • 97.Poitou C, Dalmas E, Renovato M, Benhamo V, Hajduch F, Abdennour M, Kahn J-F, Veyrie N, Rizkalla S, Fridman W-H, Sautès-Fridman C, Clément K, Cremer I. CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss: relationships with fat mass and subclinical atherosclerosis. Arterioscler Thromb Vasc Biol 31: 2322–2330, 2011. doi: 10.1161/ATVBAHA.111.230979. [DOI] [PubMed] [Google Scholar]
  • 98.Qi L, Cho YA. Gene-environment interaction and obesity. Nutr Rev 66: 684–694, 2008. doi: 10.1111/j.1753-4887.2008.00128.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Räikkönen K, Matthews KA, Kuller LH. The relationship between psychological risk attributes and the metabolic syndrome in healthy women: antecedent or consequence? Metabolism 51: 1573–1577, 2002. doi: 10.1053/meta.2002.36301. [DOI] [PubMed] [Google Scholar]
  • 100.Räikkönen K, Matthews KA, Kuller LH. Depressive symptoms and stressful life events predict metabolic syndrome among middle-aged women: a comparison of World Health Organization, Adult Treatment Panel III, and International Diabetes Foundation definitions. Diabetes Care 30: 872–877, 2007. doi: 10.2337/dc06-1857. [DOI] [PubMed] [Google Scholar]
  • 101.Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, Haroon E, Miller AH. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry 70: 31–41, 2013. doi: 10.1001/2013.jamapsychiatry.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Renault PF, Hoofnagle JH, Park Y, Mullen KD, Peters M, Jones DB, Rustgi V, Jones EA. Psychiatric complications of long-term interferon alfa therapy. Arch Intern Med 147: 1577–1580, 1987. doi: 10.1001/archinte.1987.00370090055011. [DOI] [PubMed] [Google Scholar]
  • 103.Rickards H. Depression in neurological disorders: Parkinson’s disease, multiple sclerosis, and stroke. J Neurol Neurosurg Psychiatry 76, Suppl 1: i48–i52, 2005. doi: 10.1136/jnnp.2004.060426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rui L, Aguirre V, Kim JK, Shulman GI, Lee A, Corbould A, Dunaif A, White MF. Insulin/IGF-1 and TNF-alpha stimulate phosphorylation of IRS-1 at inhibitory Ser307 via distinct pathways. J Clin Invest 107: 181–189, 2001. doi: 10.1172/JCI10934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ryder E, Diez-Ewald M, Mosquera J, Fernández E, Pedreañez A, Vargas R, Peña C, Fernández N. Association of obesity with leukocyte count in obese individuals without metabolic syndrome. Diabetes Metab Syndr 8: 197–204, 2014. doi: 10.1016/j.dsx.2014.09.002. [DOI] [PubMed] [Google Scholar]
  • 106.Sapolsky R, Rivier C, Yamamoto G, Plotsky P, Vale W. Interleukin-1 stimulates the secretion of hypothalamic corticotropin-releasing factor. Science 238: 522–524, 1987. doi: 10.1126/science.2821621. [DOI] [PubMed] [Google Scholar]
  • 107.Schiepers OJG, Wichers MC, Maes M. Cytokines and major depression. Prog Neuropsychopharmacol Biol Psychiatry 29: 201–217, 2005. doi: 10.1016/j.pnpbp.2004.11.003. [DOI] [PubMed] [Google Scholar]
  • 108.Schmidt FM, Weschenfelder J, Sander C, Minkwitz J, Thormann J, Chittka T, Mergl R, Kirkby KC, Faßhauer M, Stumvoll M, Holdt LM, Teupser D, Hegerl U, Himmerich H. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS One 10: e0121971, 2015. doi: 10.1371/journal.pone.0121971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature 404: 661–671, 2000. doi: 10.1038/35007534. [DOI] [PubMed] [Google Scholar]
  • 110.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 72: 268–275, 2015. doi: 10.1001/jamapsychiatry.2014.2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol 11: 762–774, 2011. doi: 10.1038/nri3070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116: 3015–3025, 2006. doi: 10.1172/JCI28898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Simon NM, McNamara K, Chow CW, Maser RS, Papakostas GI, Pollack MH, Nierenberg AA, Fava M, Wong KK. A detailed examination of cytokine abnormalities in Major Depressive Disorder. Eur Neuropsychopharmacol 18: 230–233, 2008. doi: 10.1016/j.euroneuro.2007.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Singer K, DelProposto J, Morris DL, Zamarron B, Mergian T, Maley N, Cho KW, Geletka L, Subbaiah P, Muir L, Martinez-Santibanez G, Lumeng CN. Diet-induced obesity promotes myelopoiesis in hematopoietic stem cells. Mol Metab 3: 664–675, 2014. doi: 10.1016/j.molmet.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Singer K, Maley N, Mergian T, DelProposto J, Cho KW, Zamarron BF, Martinez-Santibanez G, Geletka L, Muir L, Wachowiak P, Demirjian C, Lumeng CN. Differences in hematopoietic stem cells contribute to sexually dimorphic inflammatory responses to high fat diet-induced obesity. J Biol Chem 290: 13250–13262, 2015. doi: 10.1074/jbc.M114.634568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Spranger J, Verma S, Göhring I, Bobbert T, Seifert J, Sindler AL, Pfeiffer A, Hileman SM, Tschöp M, Banks WA. Adiponectin does not cross the blood-brain barrier but modifies cytokine expression of brain endothelial cells. Diabetes 55: 141–147, 2006. doi: 10.2337/diabetes.55.01.06.db05-1077. [DOI] [PubMed] [Google Scholar]
  • 117.Stellwagen D, Malenka RC. Synaptic scaling mediated by glial TNF-alpha. Nature 440: 1054–1059, 2006. doi: 10.1038/nature04671. [DOI] [PubMed] [Google Scholar]
  • 118.Stewart AM, Roy S, Wong K, Gaikwad S, Chung KM, Kalueff AV. Cytokine and endocrine parameters in mouse chronic social defeat: implications for translational ‘cross-domain’ modeling of stress-related brain disorders. Behav Brain Res 276: 84–91, 2015. doi: 10.1016/j.bbr.2014.08.037. [DOI] [PubMed] [Google Scholar]
  • 119.Stranahan AM, Hao S, Dey A, Yu X, Baban B. Blood-brain barrier breakdown promotes macrophage infiltration and cognitive impairment in leptin receptor-deficient mice. J Cereb Blood Flow Metab 36: 2108–2121, 2016. doi: 10.1177/0271678X16642233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Tagliari B, Tagliari AP, Schmitz F, da Cunha AA, Dalmaz C, Wyse ATS. Chronic variable stress alters inflammatory and cholinergic parameters in hippocampus of rats. Neurochem Res 36: 487–493, 2011. doi: 10.1007/s11064-010-0367-0. [DOI] [PubMed] [Google Scholar]
  • 121.Takahashi K, Yamaguchi S, Shimoyama T, Seki H, Miyokawa K, Katsuta H, Tanaka T, Yoshimoto K, Ohno H, Nagamatsu S, Ishida H. JNK- and IkappaB-dependent pathways regulate MCP-1 but not adiponectin release from artificially hypertrophied 3T3-L1 adipocytes preloaded with palmitate in vitro. Am J Physiol Endocrinol Metab 294: E898–E909, 2008. doi: 10.1152/ajpendo.00131.2007. [DOI] [PubMed] [Google Scholar]
  • 122.Takase K, Tsuneoka Y, Oda S, Kuroda M, Funato H. High-fat diet feeding alters olfactory-, social-, and reward-related behaviors of mice independent of obesity. Obesity (Silver Spring) 24: 886–894, 2016. doi: 10.1002/oby.21441. [DOI] [PubMed] [Google Scholar]
  • 123.Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, Lu M, Li P, Yan Q, Zhu Y, Ofrecio J, Lin M, Brenner MB, Olefsky JM. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med 18: 1407–1412, 2012. doi: 10.1038/nm.2885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Thaipisuttikul P, Ittasakul P, Waleeprakhon P, Wisajun P, Jullagate S. Psychiatric comorbidities in patients with major depressive disorder. Neuropsychiatr Dis Treat 10: 2097–2103, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Trottier MD, Naaz A, Li Y, Fraker PJ. Enhancement of hematopoiesis and lymphopoiesis in diet-induced obese mice. Proc Natl Acad Sci USA 109: 7622–7629, 2012. doi: 10.1073/pnas.1205129109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Valdearcos M, Douglass JD, Robblee MM, Dorfman MD, Stifler DR, Bennett ML, Gerritse I, Fasnacht R, Barres BA, Thaler JP, Koliwad SK. Microglial Inflammatory Signaling Orchestrates the Hypothalamic Immune Response to Dietary Excess and Mediates Obesity Susceptibility. Cell Metab 26: 185–197.e3, 2017. doi: 10.1016/j.cmet.2017.05.015 . A correction for this article is available at https://doi.org/10.1016/j.cmet.2018.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Vezzani A, Maroso M, Balosso S, Sanchez M-A, Bartfai T. IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures. Brain Behav Immun 25: 1281–1289, 2011. doi: 10.1016/j.bbi.2011.03.018. [DOI] [PubMed] [Google Scholar]
  • 128.Wang Y-L, Han Q-Q, Gong W-Q, Pan D-H, Wang L-Z, Hu W, Yang M, Li B, Yu J, Liu Q. Microglial activation mediates chronic mild stress-induced depressive- and anxiety-like behavior in adult rats. J Neuroinflammation 15: 21, 2018. doi: 10.1186/s12974-018-1054-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, Charo I, Leibel RL, Ferrante AW Jr. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest 116: 115–124, 2006. doi: 10.1172/JCI24335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112: 1796–1808, 2003. doi: 10.1172/JCI200319246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wensveen FM, Jelenčić V, Valentić S, Šestan M, Wensveen TT, Theurich S, Glasner A, Mendrila D, Štimac D, Wunderlich FT, Brüning JC, Mandelboim O, Polić B. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol 16: 376–385, 2015. doi: 10.1038/ni.3120. [DOI] [PubMed] [Google Scholar]
  • 132.Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, Tsui H, Wu P, Davidson MG, Alonso MN, Leong HX, Glassford A, Caimol M, Kenkel JA, Tedder TF, McLaughlin T, Miklos DB, Dosch H-M, Engleman EG. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17: 610–617, 2011. doi: 10.1038/nm.2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wium-Andersen MK, Ørsted DD, Nielsen SF, Nordestgaard BG. Elevated C-reactive protein levels, psychological distress, and depression in 73, 131 individuals. JAMA Psychiatry 70: 176–184, 2013. doi: 10.1001/2013.jamapsychiatry.102. [DOI] [PubMed] [Google Scholar]
  • 134.Wohleb ES, Hanke ML, Corona AW, Powell ND, Stiner LM, Bailey MT, Nelson RJ, Godbout JP, Sheridan JF. β-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J Neurosci 31: 6277–6288, 2011. doi: 10.1523/JNEUROSCI.0450-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wohleb ES, Powell ND, Godbout JP, Sheridan JF. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J Neurosci 33: 13820–13833, 2013. doi: 10.1523/JNEUROSCI.1671-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.World Health Organization Obesity and overweight: Fact sheet [Online]. Geneva, Switzerland: World Health Organization, 2018. http://www.who.int/en/news-room/fact-sheets/detail/obesity-and-overweight. [Google Scholar]
  • 137.World Health Organization Depression: Fact sheet [Online]. Geneva, Switzerland: World Health Organization, 2018. http://www.who.int/news-room/fact-sheets/detail/depression. [Google Scholar]
  • 138.Wu D, Molofsky AB, Liang H-E, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, Chawla A, Locksley RM. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science 332: 243–247, 2011. doi: 10.1126/science.1201475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wu H, Ghosh S, Perrard XD, Feng L, Garcia GE, Perrard JL, Sweeney JF, Peterson LE, Chan L, Smith CW, Ballantyne CM. T-cell accumulation and regulated on activation, normal T cell expressed and secreted upregulation in adipose tissue in obesity. Circulation 115: 1029–1038, 2007. doi: 10.1161/CIRCULATIONAHA.106.638379. [DOI] [PubMed] [Google Scholar]
  • 140.Wu H, Liu Q, Kalavagunta PK, Huang Q, Lv W, An X, Chen H, Wang T, Heriniaina RM, Qiao T, Shang J. Normal diet Vs High fat diet - A comparative study: Behavioral and neuroimmunological changes in adolescent male mice. Metab Brain Dis 33: 177–190, 2018. doi: 10.1007/s11011-017-0140-z. [DOI] [PubMed] [Google Scholar]
  • 141.Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112: 1821–1830, 2003. doi: 10.1172/JCI200319451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Yirmiya R. Endotoxin produces a depressive-like episode in rats. Brain Res 711: 163–174, 1996. doi: 10.1016/0006-8993(95)01415-2. [DOI] [PubMed] [Google Scholar]
  • 143.Yoshimura A, Ohnishi S, Orito C, Kawahara Y, Takasaki H, Takeda H, Sakamoto N, Hashino S. Association of peripheral total and differential leukocyte counts with obesity-related complications in young adults. Obes Facts 8: 1–16, 2015. doi: 10.1159/000373881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zamarron BF, Mergian TA, Cho KW, Martinez-Santibanez G, Luan D, Singer K, DelProposto JL, Geletka LM, Muir LA, Lumeng CN. Macrophage proliferation sustains adipose tissue inflammation in formerly obese mice. Diabetes 66: 392–406, 2017. doi: 10.2337/db16-0500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zunszain PA, Anacker C, Cattaneo A, Carvalho LA, Pariante CM. Glucocorticoids, cytokines and brain abnormalities in depression. Prog Neuropsychopharmacol Biol Psychiatry 35: 722–729, 2011. doi: 10.1016/j.pnpbp.2010.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiology are provided here courtesy of American Physiological Society

RESOURCES