Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
Previous Article in Journal
Human Stem Cell Therapy for the Cure of Type 1 Diabetes Mellitus (T1D): A Hurdle Course between Lights and Shadows
Previous Article in Special Issue
How Different Treatments for Acromegaly Modulate Sleep Quality: A Psychometric Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Bidirectional Link between Major Depressive Disorder and Type 2 Diabetes: The Role of Inflammation

by
Alexandra M. Bodnaruc
1,2,†,
Mathilde Roberge
1,3,†,
Isabelle Giroux
1,2 and
Céline Aguer
1,3,4,*
1
Institut du Savoir Montfort, Ottawa, ON K1K 0T2, Canada
2
School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
3
Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
4
Department of Physiology, Faculty of Medicine and Health Sciences, McGill University—Campus Outaouais, Gatineau, QC J8V 3T4, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Endocrines 2024, 5(4), 478-500; https://doi.org/10.3390/endocrines5040035 (registering DOI)
Submission received: 23 April 2024 / Revised: 6 September 2024 / Accepted: 27 September 2024 / Published: 9 October 2024
(This article belongs to the Special Issue Feature Papers in Endocrines: 2024)

Abstract

:
Background/Objectives: There is a bidirectional relationship between major depressive disorder (MDD) and type 2 diabetes (T2D), as MDD increases the risk of T2D by 38% to 67%, and T2D increases the risk of MDD by 15% to 33%. Many factors contribute to the occurrence of comorbid MDD and T2D, including converging pathophysiological pathways like inflammation. The objective of this review was to comprehensively summarize available evidence on the relationship between MDD, T2D, and inflammation. Results: Although the precise mechanisms linking T2D and MDD are still not fully understood, shared inflammatory mechanisms likely contributes to the heightened risk of developing this comorbidity. To date, the evidence supports that chronic low-grade inflammation is a feature of both MDD and T2D and has been shown to interact with pathways that are relevant to the development of both chronic disorders, including the hypothalamic–pituitary–adrenal (HPA) axis, neuroplastic processes, gut microbiome, insulin resistance, and adipose tissue dysfunction. Through their impact on inflammation, dietary and physical activity interventions can play a role in the risk and management of MDD and T2D. Conclusions: Deepening our understanding of the mechanisms underlying the augmented inflammatory responses observed in individuals with the MDD and T2D comorbidity is essential for tailoring appropriate therapeutic strategies.

1. Introduction

Major depressive disorder (MDD) and type 2 diabetes (T2D) are heterogeneous and complex disorders that are responsible for considerable proportions of disability and mortality worldwide [1,2]. Affecting over 250 million individuals globally and with a prevalence that is still on the rise, MDD ranks among the most common and burdensome mental health disorders [1]. Similarly, with global prevalence having nearly quadrupled over the past three decades, T2D affects approximately 537 million adults, making it one of the most widespread cardiometabolic disorders and one of the biggest public health challenges of the century [2].
Over the past three decades, the prevalence of comorbid mental health and cardiometabolic disorders has drastically increased, reaching epidemic proportions in numerous countries [3]. Meta-analyses of prospective cohort studies have reported a 38% to 67% higher risk of developing T2D in individuals with MDD, and a 15% to 33% higher risk of developing MDD among individuals with T2D [4,5,6]. Comorbid MDD and T2D have been associated with poorer health outcomes [7,8,9], lower quality of life [10,11], higher mortality [12,13], as well as increased healthcare-related costs [14,15].
Many factors can contribute to the occurrence of comorbid MDD and T2D, such as shared genetic susceptibility, converging pathophysiological pathways, as well as psychosocial and environmental factors [3]. In this regard, the involvement of inflammation in the pathophysiology of both MDD and T2D has gained increased attention over the past two decades [16]. The objective of this review was to comprehensively summarize available evidence on the relationship between MDD, T2D, and inflammation.

2. MDD

According to the fifth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-V), the core symptoms of MDD are persistent low mood and anhedonia, which are accompanied by alterations in emotion regulation (e.g., excessive feelings of guilt, unworthiness), cognitive abilities (e.g., decreased concentration, memory), and physiological functions (e.g., dysregulated sleeping patterns and appetite, hypo- or hyper-locomotion) [17]. One meets the diagnostic criteria for MDD when there is evidence supporting (1) the manifestation of five or more depressive symptoms, of which at least one is a core symptom, and that these symptoms (2) are present for at least two consecutive weeks and (3) are associated with clinically significant psychological distress and/or disabilities in essential areas of functioning, including activities of daily living, professional activities, and social interactions [17].
Despite significant advances in the understanding of MDD, there are currently no established mechanisms that can explain all facets of the disease. The etiology of MDD involves genetic predispositions, which explain 30–40% of MDD risk [18] and interact with a wide range of psychological (e.g., negative self-concept, sensitivity to rejection, negative emotionality) and environmental factors (e.g., adverse life events, social isolation, low socioeconomic status, poor lifestyle) [19]. These etiological factors are at the origin of pathophysiological alterations affecting, among others, the immune system [20,21,22,23,24,25,26,27,28,29,30,31,32,33,34], monoamine neurotransmitters’ metabolism [35,36,37,38], the hypothalamic-pituitary-adrenal (HPA) axis [39,40,41], neuroplasticity [42,43,44,45], and the gut microbiome [46,47,48].
MDD is associated with a significant increase in morbidity and mortality, often resulting in higher levels of disability and impairment than most physical conditions [49]. MDD increases the risk of several physical disorders, including obesity [50] and T2D [51]. MDD also increases the risk of other mental disorders, such as anxiety [52]. MDD is associated with premature mortality, with a significant proportion attributable to elevated rates of comorbid conditions [49]. Suicide also plays a crucial role in increasing mortality rates, with MDD elevating the risk of suicide death nearly 20-fold [53].

3. T2D

T2D accounts for 90% of all cases of diabetes and is characterized by a chronic state of hyperglycemia resulting from insulin resistance and/or impaired insulin secretion [54]. According to Diabetes Canada, T2D occurs when one has a fasting blood glucose ≥ 7.0 mmol/L, a 2 h post-prandial or random blood glucose ≥ 11.1 mmol/L, or glycated hemoglobin (A1c) ≥ 6.5% [55].
Following a meal, the hormone insulin is secreted into the bloodstream by the pancreatic β-cells, allowing for glucose uptake into the cells [56]. Early in T2D development, the target tissues of insulin, mainly the liver, adipose tissues, and skeletal muscles, become less sensitive to its action, ultimately leading to decreased glucose uptake [56]. This decreased ability of target tissues to respond to insulin is called insulin resistance [56]. The proposed contributors to β-cell dysfunction and insulin resistance include oxidative stress, endoplasmic reticulum stress, amyloid deposition in the pancreas, ectopic lipid deposition, as well as lipotoxicity and glucotoxicity [57]. Although pinpointing the most significant T2D pathophysiological mechanism has proven to be challenging, each of these cellular stressors is believed to either trigger an inflammatory response or be worsened by inflammation. As such, emerging evidence suggests that dysregulation of inflammatory markers and abnormalities in immune function play significant roles in T2D pathogenesis [58].
T2D is closely linked with a plethora of comorbidities, including cardiovascular diseases, chronic kidney disease, osteoporosis, vision impairment, as well as mental health comorbidities such as depression [59]. Estimates indicate that adults with T2D tend to have a life expectancy of around six years shorter than those without the condition [59].

4. Inflammation

4.1. Acute vs. Chronic Inflammation

The term inflammation encompasses a broad range of immune-related processes occurring within the body. Acute inflammation is a protective reaction typically triggered by the recognition of extracellular stimuli by nearby immune-responsive cells and regulated by cytokines, chemokines, and various other signaling molecules [60]. These stimuli can involve damage-associated molecular patterns (DAMPs) or pathogen-associated molecular patterns (PAMPs), activating receptors on or within cells present in the affected tissues [60]. In response, these cells induce and regulate varying degrees of inflammation, chemotaxis, cell growth, and tissue repair locally and throughout the body [60].
After the threat or damage has been addressed, the resolution of acute inflammation is crucial for restoring homeostasis [60,61]. If the inflammatory response is not resolved properly, it can transition into chronic inflammation [60,61]. The feedback loops required for a robust acute inflammatory response may instead exacerbate inflammation, sustaining immune responses and causing tissue damage, leading to tissue fibrosis and destruction [60,61]. While acute inflammation is a normal and essential step in healing, chronic inflammation is detrimental and contributes to the development of several pathologies [60,61].
As shown in Table 1, acute and chronic inflammation differ based on their onset, duration, cause, cells and mediators involved, tissue damage, and outcomes [60,61]. Acute inflammation has an immediate onset in response to pathogens and tissue damage and lasts from days to weeks [60,61]. In contrast, chronic inflammation is due to persistent foreign bodies, persistent acute inflammation, and autoimmune reactions, has a delayed onset and lasts from months to years [60,61]. Acute inflammation promotes tissue repair and primarily involves neutrophils, vasoactive amines, and eicosanoids [60,61]. Conversely, chronic inflammation promotes tissue damage and primarily involves monocytes, macrophages, lymphocytes, fibroblasts, as well as cytokines, growth factors, hydrolytic enzymes, and reactive oxygen species [60,61].

4.2. Central Inflammation

Central inflammation, also called neuroinflammation, refers to inflammatory processes occurring within the central nervous system (CNS), namely in the brain and the spinal cord [62,63]. Central inflammation involves inflammatory mediators that are commonly used by the immune system for inflammatory processes occurring in the periphery, such as cytokines and chemokines [62,63]. Exchanges between the periphery and the CNS are tightly regulated by the blood–brain barrier (BBB), a semi-permeable membrane situated at the interface between the circulatory system and the brain [62,63]. There are several pathways through which peripheral inflammatory mediators can access the CNS, such as (1) passage through leaky regions of the BBB at circumventricular organs, (2) active transport via mediator-specific saturable transporters, (3) neural afferent fiber activation transducing signals to the brain, (4) activation of endothelial cells and perivascular macrophages in the cerebral vasculature to produce local inflammatory mediators such as cytokines, chemokines, prostaglandins, and nitric oxide, and (5) attraction of activated peripheral immune cell including monocytes and T cells to the meninges and the brain parenchyma by chemokines released by activated microglia and adhesion molecules expressed in the CNS [62,63]. Chronic peripheral inflammation also has the potential to disrupt the integrity of the BBB, namely by increasing solute permeability and leukocyte traffic as well as by inducing signaling changes and structural damages [64]. Disruption of the BBB integrity can further facilitate the entry of inflammatory molecules [64]. Inflammatory molecules are also synthesized within the CNS, mainly by microglia, which are the resident macrophages of the brain [65]. Although peripheral inflammatory mediators that migrate or relay information to the brain contribute to the central inflammatory response, the primary features of neuroinflammation include the abnormal activation of resident immune cells, namely microglia, and subsequent involvement of astrocytes [62,63].

4.3. Inflammation and MDD

Extensive evidence stemming from both observational and experimental studies supports the involvement of inflammation in the pathophysiology of MDD. As evidenced by multiple meta-analyses of observational studies, individuals with MDD exhibit key features of an inflammatory response, including increased peripheral levels of proinflammatory cytokines (e.g., interleukin-1 receptor antagonist (IL-1RA), IL-6, IL-12, IL-13, IL-18, tumor necrosis factor alpha (TNF-α) [20]), proinflammatory cytokines receptors (e.g., soluble interleukin-2 receptor (sIL2R), soluble tumor necrosis factor receptor 2 (sTNFR2) [20]), acute phase reactants (e.g., C-reactive protein (CRP), albumin), and chemokines (e.g., C-C motif chemokine ligand 2 (CCL2), C-X-C motif chemokine ligand 4 (CXCL4), CXCL7 [21]). Elevated inflammatory markers, including IL-6 and TNF-α, have also been observed in the cerebrospinal fluid and post-mortem brains of individuals with MDD [22]. Similarly, translocator protein, a positron emission tomography marker of central inflammation, has also been shown to be elevated in the anterior cingulate cortex and temporal cortex of individuals with MDD [22]. These brain areas are involved in processing sensory input, memory functions and complex cognitive functions such as emotionality, empathy, impulse control, and decision-making [24]. While inconsistent, some studies have also reported increased markers of microglial activation and decreased expression of astrocyte-specific markers [22]. Adding to these findings, observational studies have shown that both prior severe infections and autoimmune diseases increase the risk of subsequently developing MDD [25].
The implication of the immune system in the development of depressive symptoms and MDD has also been confirmed by numerous experimental and drug vigilance studies. In animal models, studies have shown that the administration of immune challenges (e.g., lipopolysaccharide (LPS)) can lead to depressive-like behavior and that stress-induced depression is partly mediated by inflammatory changes [27]. Conversely, the administration of antidepressants to animals with inflammation or stress-induced depression decreases depressive-like behaviors partly through a reduction in inflammation [29]. In humans, it has been shown that individuals who receive cytokine treatments (i.e., IL-2 or interferon-alpha (INF-α)) as part of their treatment for hepatitis or cancer often develop depressive symptoms [30]. Several meta-analyses also support that cytokine treatment-induced depression can be prevented with the prophylactic administration of antidepressants [31]. Further supporting the involvement of the immune system in the development of MDD, meta-analyses of randomized controlled trials have shown that treatment with antidepressants reduces inflammation and that treatment with anti-cytokine drugs is effective in diminishing depressive symptoms in individuals with MDD. More specifically, treatment with antidepressants has been shown to decrease circulating levels of IL-4, IL-6, and IL-10 [32]. A decrease in peripheral levels of IL-1β was also observed, but only among individuals treated with SSRI antidepressants [32]. A meta-analysis of 16 clinical trials assessing the antidepressant activity of anti-cytokine drugs reported small-to-moderate beneficial effects for all agents, which targeted TNF-α (i.e., Etanercept, Adalimumab, Infliximab, Eisenberg), IL-6 receptor (i.e., Toclizumab), IL-4 receptor-α (e.g., Dupilumab), and IL-12/23 (i.e., Ustekinumab) [33]. Similarly, in a meta-analysis, the use of the non-steroidal anti-inflammatory drug Celebrex as an adjunctive treatment was found to be associated with significantly higher mean changes in depression scores as well as significantly higher remission rates than placebo in individuals with MDD [34]. As described in the sections below, inflammation plays a role in several pathophysiological alterations associated with MDD, namely alterations within monoamine neurotransmitter systems, the HPA axis, neuroplasticity, and the gut microbiome.

4.3.1. Inflammation and Monoamine Neurotransmitters in MDD

The decreased availability of monoamine neurotransmitters (i.e., serotonin (5-HT), noradrenaline (NA), and dopamine (DA)) is one of the most widely known pathophysiological features of MDD. While results remain somehow inconsistent, studies have reported that individuals with MDD have decreased cerebrospinal fluid levels of monoamine neurotransmitter metabolites (e.g., 5-hydroxyindoleacetic acid, homovanillic acid) and monoamine-degrading enzymes (e.g., monoamine oxidase A (MAO-A)) [37,38]. Furthermore, acute depletion of the 5-HT precursor tryptophan, leading to temporarily lowered 5-HT levels in the CNS, has been shown to influence emotional regulation and contribute to lowering mood, particularly in individuals with a family history of MDD [35,36]. Nonetheless, the most robust proof of the implication of monoamine neurotransmitters in the pathophysiology of MDD stems from the effectiveness of monoamine-targeting antidepressants (e.g., tricyclic antidepressants (TCAs), selective serotonin reuptake inhibitors (SSRIs), selective serotonin and norepinephrine reuptake inhibitors (SSNRIs)) in diminishing depressive symptoms among individuals with MDD [66,67].
There are several pathways through which inflammatory cytokines can lead to reduced synaptic availability of monoamines, including by increasing their reuptake into pre-synaptic neurons and by decreasing their synthesis. In this context, IL-1β and TNF induction of p38 mitogen-activated protein kinase (p38MAPK) has been shown to increase the expression and activity of 5-HT reuptake pumps, leading to decreased synaptic availability of 5-HT and depressive-like behavior in laboratory animals [68]. Inflammatory cytokine administration and chronic low-grade inflammation have also been shown to increase indoleamine 2,3-dioxygenase (IDO) activity, the latter decreasing the conversion of tryptophan into the 5-HT precursor 5-hydroxytryptophan (5-HTP) and increasing the conversion of tryptophan into kynurenine (KYN), which can be further converted into the neurotoxic metabolite quinolinic acid (QUIN) by activated microglia [69,70]. In this regard, correlations between INF-α-induced MDD, decreased plasma tryptophan, and increased KYN have been reported [69,70]. Similarly, INF-α-induced MDD has been associated with increased cerebrospinal concentration of KYN and QUIN [71]. Inflammatory cytokines have also been shown to reduce the synthesis of monoamine neurotransmitters by decreasing the availability of tetrahydrobiopterin (BH4), a key enzyme co-factor in the synthesis of all monoamines [72].

4.3.2. Inflammation and the HPA Axis in MDD

Another widely confirmed biological feature of MDD is the abnormal activity of the HPA axis, which is the principal regulator of physiological processes in response to physical and psychological stress [73]. A recent meta-analysis indicates that cortisol levels, measured in saliva, cerebrospinal fluid, urine, or hair, are slightly higher in individuals with MDD than in healthy controls [39]. More specifically, one meta-analysis has shown that higher morning cortisol levels specifically are associated with a higher prospective risk of MDD in adult populations [40]. Recent findings from a meta-analysis also suggest that high cortisol levels in individuals with MDD are resistant to feedback inhibition by the HPA axis [41]. Further supporting the involvement of the HPA axis in the pathophysiology of MDD, almost all currently available anti-depressants are known to influence cortisol levels [74].
There is a bidirectional relationship between inflammation and the HPA axis whereby, in normal physiological conditions, inflammation activates the HPA axis, and glucocorticoids decrease inflammation [73]. In individuals with MDD, however, evidence suggests that elevated levels of glucocorticoids may co-exist with high levels of inflammatory cytokines [75]. Several inflammation-related mechanisms have been proposed to account for this phenomenon, such as disruption of negative feedback via glucocorticoid receptors (GRs) [41], variations in GR expression and function [76,77], and changes in glucocorticoid bioavailability [78,79]. Studies conducted in individuals with MDD have found positive associations between cytokine production and glucocorticoid resistance, as assessed by the dexamethasone suppression test [41]. Proinflammatory cytokines have also been shown to impact the relative expression of the α and β isoforms of GRs, with a shift towards the inactive β isoform in individuals with MDD [76]. Along the same lines, proinflammatory cytokines affect GR function by inhibiting their translocation from the cytoplasm to the nucleus as well as by altering their interactions with nuclear proteins, including Nuclear Factor kappa B (NF-κB) [77]. Additional potential impacts of cytokines on glucocorticoid homeostasis encompass the activation of 11 beta-hydroxysteroid dehydrogenase 2 (11β-HSD-2), leading to cortisol inactivation [78], as well as the augmentation of activity and expression of the multidrug resistance P-glycoprotein pump, prompting the expulsion of glucocorticoids from the cell [79]. These mechanisms collectively result in reduced bioavailability of glucocorticoids at the cellular level [78,79].

4.3.3. Inflammation and Neuroplasticity in MDD

Several indicators of reduced neuroplasticity, which denotes the ability of the CNS to reorganize its connections (i.e., synaptic plasticity), structure (i.e., structural plasticity), and function (i.e., functional plasticity) in response to various stimuli, have been associated with MDD [80]. Numerous studies indicate that individuals experiencing MDD may exhibit reduced neurogenesis, as evidenced by decreased volumes in several areas of the brain, including the basal ganglia and frontal regions [45]. Interestingly, antidepressants have demonstrated the ability to enhance hippocampal neurogenesis [42]. Along the same lines, brain-derived neurotrophic factor (BDNF), a neurotrophin promoting nerve cell survival, differentiation, and maintenance, has been observed to be diminished in individuals with MDD, with partial restoration observed following antidepressant administration [43,44].
Inflammation can impact neuroplastic processes through several mechanisms. Elevated levels of inflammatory cytokines, along with their signaling pathways, such as NF-κB, have the potential to diminish the expression and efficacy of the excitatory amino acid transporter 2 (EAAT2) present on astrocytes, leading to the reversal of glutamate efflux [81,82]. Conversion of KYN to QUIN by microglia further contributes to excessive glutamate signaling, notably through N-Methyl-D-Aspartate (NMDA) receptors, by decreasing astrocytic glutamate reuptake and stimulating glutamate release [82,83]. The overflow of glutamate into the extrasynaptic space triggers an activation of extrasynaptic NMDA receptors, which can result in decreased levels of neurotrophic factors, including BDNF [82,84]. When combined with excessive synaptic NMDA receptor signaling, which creates a swift influx of calcium ions that accumulate in the mitochondria, triggering apoptotic cascades, including caspase activation and heightened production of reactive oxygen species, this process ultimately leads to excitotoxicity and synaptic damage [82].

4.3.4. Inflammation and the Gut Microbiome in MDD

Systematic review and meta-analyses have shown that the gut microbiota of individuals with MDD differ from those of healthy individuals [46,47]. Differences in the representation of up to 50 bacterial taxa have been reported, with individuals with MDD having a higher abundance of pro-inflammatory bacterial species (e.g., Enterobacteriaceae and Desulfovibrio) and a lower abundance of short-chain fatty acid (SCFA)-producing bacterial species (e.g., Faecalibacterium) [46,47]. While results have been inconsistent, there is also some evidence of disparities in the α-diversity (differences in the types of species living in a given gut area) and β-diversity (differences in the types of species living in multiple gut areas) of the microbiota of individuals with MDD compared to healthy controls [46,47]. Supporting a more causal relationship between the gut microbiome and MDD, fecal transplantation from adults with MDD to germ-free mice has been shown to increase depressive-like behavior in recipients [48].
Several gut microbiota alterations observed in individuals with MDD have been linked with inflammation. For example, a higher abundance of pro-inflammatory bacterial species has been associated with “leaky gut”, promoting macrophage infiltration and active LPS translocation into the systemic circulation [85]. Macrophage infiltration produces and activates pro-inflammatory cytokines, leading to local inflammation, while LPS binds to Toll-Like Receptor 4 (TLR-4) expressed on immune cells, thereby activating pro-inflammatory cascades both locally and systemically [85]. The lower abundance of SCFA-producing bacteria can also directly influence inflammation, as SCFAs have been shown to reduce the secretion of pro-inflammatory cytokines, increase the secretion of the anti-inflammatory cytokine IL-10, and induce the development of regulatory T (Treg) cells [86]. Gut microbiome α and β-diversity have also been linked with the severity of systemic and local gut inflammation following trauma exposure [87].

4.4. Inflammation and T2D

T2D is increasingly recognized as an inflammatory disorder, with substantial evidence linking inflammation directly to insulin resistance. In states of overnutrition, such as obesity, hyperglycemia and hyperlipemia occur, promoting insulin resistance and persistent inflammation. In this context, pancreatic β-cells are exposed to various forms of stress, including inflammation, endoplasmic reticulum stress, oxidative stress, and amyloid stress, which can ultimately compromise their integrity [88]. Activation of pro-inflammatory macrophages and their accumulation in metabolic tissues play a pivotal role in sustaining the inflammatory state. While macrophages are the primary effector cells involved in this chronic state of low-grade inflammation, various other immune cell types are also at play [89].

4.4.1. Obesity and Adipose Tissue Inflammation in T2D

It is estimated that 80 to 90% of individuals who develop T2D are either overweight or have obesity [90]. Excessive body fat accumulation after the onset of T2D also further promotes hyperglycemia, which contributes to increasing the risks for T2D comorbidities and overall mortality rates [90]. The association between obesity and chronic low-grade inflammation is pivotal in the onset and progression of obesity-related metabolic disorders, notably insulin resistance, in the context of T2D [91]. Indeed, inflammation within adipose tissue is deemed a critical precipitating metabolic dysfunction. Studies have consistently demonstrated the accumulation of macrophages in both murine and human adipose tissue during obesity, with adipose tissue macrophages significantly contributing to the insulin-resistant state [92,93]. Consistent with the aforementioned findings, preventing the accumulation of adipose tissue macrophages has been shown to protect obese mice from glucose intolerance and insulin resistance [94]. Various other immune cell types contribute to the inflammatory state in adipose tissue during obesity, such as specific T and B cell subsets that play regulatory roles. Nevertheless, macrophages are generally considered the principal effector cells responsible for reduced insulin signaling [95].
In lean individuals, adipose tissue macrophages constitute a minority of stromal-vascular cells and typically exhibit an M2-like polarization state [92,93]. However, in obesity, adipose tissue macrophages increase substantially, comprising a significant proportion of adipose tissue cells, primarily exhibiting a pro-inflammatory (M1-like) phenotype [96]. Importantly, visceral adiposity, primarily located around abdominal viscera, exhibits distinct metabolic characteristics compared to subcutaneous adipose tissue. Visceral obesity is more strongly associated with heightened lipotoxicity and impaired insulin sensitivity [97]. Furthermore, higher levels of inflammatory cells are found in visceral compared to subcutaneous fats [98]. Consequently, the intricate association between T2D and obesity is more dependent on the role of visceral adipose tissue on metabolic dysregulation.
Macrophages undergo notable changes during obesity, with an increased number of M1-polarized macrophages characterized by heightened pro-inflammatory phenotype and secretion of cytokines such as TNF-α. This increase in macrophage number and altered M1 to M2 ratio typifies adipose tissue inflammation in obesity and is associated with T2D development [99]. Additionally, upregulation of other pro-inflammatory cytokines and chemokines, such as IL-1β, IL-6, and monocyte chemoattractant protein-1 (MCP-1), in enlarged adipose tissue sites of individuals with obesity and those with T2D, further exacerbates inflammation and insulin resistance. It is important to note that, independent of body weight, a small number of studies have also shown a positive relationship between inflammation and T2D, with an increase in genes related to the recruitment of macrophages, including cluster of differentiation 68 (CD68), MCP-1, IL-6, and IL-8 [100].
Various lipid species elevated due to diet or obesity may also contribute to inflammation by promoting endoplasmic reticulum stress and activating intracellular inflammatory pathways [101]. For instance, nutrient overload (i.e., excess glucose and free fatty acids) can activate the inflammasome complex, which in turn triggers downstream inflammatory pathways, including IL-1β production [102].
In addition, hypoxia, particularly prevalent in obesogenic conditions due to impaired angiogenesis and reduced oxygen perfusion, serves as another initiator of inflammation within adipose tissue [103]. Hypoxia-inducible factor 1 alpha (HIF-α) induction, primarily driven by intra-adipocyte hypoxia, initiates an inflammatory response by upregulating chemokines, thereby recruiting and differentiating monocytes into pro-inflammatory M1-like adipose tissue macrophages [103]. Genetic deletion of adipocyte HIF-α has been shown to prevent obesity-induced inflammation and insulin resistance, further supporting the role of hypoxia in adipocyte tissue inflammation during obesity [103]. Furthermore, it has been evidenced that hypoxia can alter the secretome of murine 3T3-L1 adipocytes, leading to increased expression of prothymosin-alpha (ProT-α), an immunomodulatory protein [104]. Elevated levels of circulating ProT-α are associated with obesity and can be detected prior to the onset of cardiometabolic diseases. As such, ProT-α may serve as a biomarker for inflammation and insulin resistance in T2D [104].

4.4.2. Neuroinflammation and Insulin Resistance in T2D

In addition to peripheral metabolic dysfunction, obesity and associated inflammation have been implicated in brain function alterations, particularly in regions governing energy homeostasis and metabolism. The hypothalamus, crucial in regulating body weight through energy intake and expenditure, exhibits increased glial cells and/or astrocytes presence during obesity [105]. Studies have shown that elevated expression of inflammatory cytokines mediated by PRRs in obesogenic conditions is correlated with hypothalamic insulin resistance. Resistin emerges as a significant hormone linking obesity-induced hypothalamic inflammation and insulin resistance via TLR4 signaling pathways [106].
Furthermore, chronic exposure to glucocorticoids in humans is recognized to induce whole-body insulin resistance and obesity. Subtle forms of glucocorticoid excess are observed in chronic stress scenarios due to activation of the HPA axis, resulting in heightened adrenal cortisol production [107]. Moreover, obesity is intricately linked with abnormalities in the HPA axis, encompassing increased local production of glucocorticoids in adipose tissue, alterations in cortisol circadian rhythm, and heightened susceptibility to HPA axis activation. These factors collectively lead to prolonged exposure to glucocorticoids over time [108]. Glucocorticoids can induce insulin resistance by suppressing the transcription of IRS1 while enhancing the transcription of proteins which oppose insulin action, including protein tyrosine phosphatase type 1 B (PTP1B) and p38MAPK [109]. Moreover, glucocorticoids directly stimulate hepatic gluconeogenesis, which contributes to hyperglycemia and thus increases the risk of T2D [110]. This means that chronic exposure to glucocorticoids, whether through stress-induced activation of the HPA axis or exogenous administration, further exacerbates insulin resistance and obesity, highlighting the intricate interplay between neuroinflammation and metabolic dysregulation in the development of T2D.

4.4.3. Inflammation and the Gut Microbiome in T2D

Similarly to MDD, the gut microbiome composition also influences the inflammatory pathways involved in metabolic disorders. In mice with diet-induced obesity, alterations in intestinal immune cell populations, including decreased Treg cells and eosinophils and increased macrophages in the lamina propria, are observed [111]. These alterations, termed dysbiosis, affect body fat distribution, systemic inflammation, and insulin resistance [112]. Dysbiosis is implicated in systemic low-grade inflammation, facilitated by increased permeability of the gastrointestinal tract leading to leakage of bacterial products like LPS, as well as localized inflammation in the small bowel and colon [113,114]. Supporting this, microbiota transplantation from individuals with obesity into lean hosts exacerbates systemic insulin resistance [115].

5. Role of Inflammation in the Bidirectional Relationship between MDD and T2D

5.1. MDD Leading to T2D

5.1.1. Inflammation, Monoamine Neurotransmitters, and T2D in Individuals with MDD

MDD-associated alterations in monoamine metabolism (particularly 5-HT and DA), which, as discussed above, are partly related to inflammation, may play a role in the development of T2D via their involvement in appetite and food reward regulation [116,117]. More specifically, alterations in monoamine metabolism could stimulate greater consumption of energy-dense foods, potentially contributing to weight gain and excessive fat accumulation, thus increasing the risk of T2D [116,117]. In this regard, 5-HT depletion in the CNS has been shown to promote hyperphagia, whereas the administration of drugs enhancing 5-HT neurotransmission, 5-HT receptors agonists, and 5-HT itself all appear to reduce food and energy intake [116,118]. Similarly, DA antagonists are known to increase appetite, food intake, and body weight, while DA agonists reduce energy intake and lead to weight loss [117,119]. Decreased DA signaling in regions involved in food reward regulation has been shown to increase anticipatory food reward (the anticipated rewarding effect of food), while concomitantly decreasing consummatory food reward (the actual rewarding effect of food), thereby promoting cravings for highly palatable foods as well as increased consumption of high-fat, high-sugar, energy-dense foods [117,119].

5.1.2. Inflammation, the HPA Axis, and T2D in Individuals with MDD

MDD-associated alterations in the regulation of the HPA axis, which are partly related to inflammation, may play a role in the development of T2D via their impact on glucose homeostasis [120], food intake [121], and patterns of fat storage and distribution [122]. High cortisol levels and their resistance to feedback inhibition by the HPA axis enhance hepatic glucose release and disrupt insulin function by inhibiting its secretion from pancreatic β-cells and impairing insulin-mediated glucose absorption via the GLUT4 transporter, collectively leading to increased blood glucose levels [122]. Higher cortisol levels, as well as high cortisol reactivity, have been shown to influence food intake, particularly in individuals with obesity. A study conducted in women with obesity showed that cortisol excretion rate was positively correlated with weekly starchy food consumption as well as daily carbohydrate and lipid intakes [123]. Similarly, individuals with obesity displaying high cortisol reactivity had significantly higher food intakes following stress induction via a Trier Social Stress Test, which appeared to be partly mediated by decreased cognitive reappraisal [121]. Cortisol is also well-known to impact fat storage and distribution patterns, with hypercortisolemia promoting central adiposity, especially visceral fat depots, which are strongly associated with insulin resistance and other cardiometabolic disturbances, such as dyslipidemia [122]. In this regard, several studies have also shown that individuals with MDD having hypercortisolemia also had higher abdominal and visceral fat deposits than healthy controls as well as individuals with MDD without hypercortisolemia [124,125].

5.1.3. Inflammation, Neuroplasticity, and T2D in Individuals with MDD

While evidence is somewhat limited, MDD-associated alterations in neuroplasticity, which are partly related to inflammation, may play a role in the development of T2D via their impact on insulin resistance [126,127], blood glucose regulation [128], and food intake [129,130]. A study conducted in a mice model of high-fat diet-induced insulin resistance showed that insulin resistance was dependent on amygdalar neuroadaptation [126]. Along the same lines, high-fat diet-induced metabolic disturbances and depressive phenotypes in mice were underlined by an astrocyte-mediated disturbance in glutamatergic neurotransmission [131]. In humans, a study reported increased insulin resitance in normal weight individuals with lower serum BDNF levels [127]. Similarly, a meta-analysis of observational studies found that individuals with T2D had lower BDNF plasma/serum levels than healthy controls [132]. Along the same lines, in mice models of T2D, systemic and intracerebroventricular BDNF administration was shown to reduce blood glucose levels, possibly by increasing insulin secretion and inhibiting glucagon secretion by the pancreas [128,133]. BDNF also appears to play a role in the regulation of food intake, with central BDNF administration reducing appetite, food consumption, and body weight in rodents [129,130].

5.1.4. Inflammation, the Gut Microbiome, and T2D in Individuals with MDD

MDD-associated alterations in the composition of the gut microbiome may play a role in the development of T2D, partly via their involvement in the regulation of inflammatory processes and their potential subsequent impact on glycemic parameters [85,86,87,134,135,136]. Similar to what has been observed in individuals with MDD, individuals with T2D have a lower abundance of SCFA-producing bacterial species [137], particularly butyrate-producing species, as well as lower α and β-diversity [138]. Lower abundance of butyrate-producing bacteria has been associated with poorer glycemic parameters, including higher A1c levels, higher fasting blood glucose levels, higher insulin resistance, as well as lower levels of insulin and C-peptide, an indicator of insulin synthesis [137]. As previously described, these alterations in glycemic parameters could be partly mediated by the effects of SCFAs on circulating levels of pro-inflammatory and anti-inflammatory cytokines [86], as well as by the pro-inflammatory effects of macrophage infiltration and active LPS translocation into the systemic circulation due to increased intestinal permeability or “leaky gut” [85,134,135]. Similarly, lower α and β-diversity were both associated with higher insulin resistance [137], which could be mediated by the impact of bacterial diversity on the severity of systemic inflammation [87].

5.2. T2D Leading to MDD

5.2.1. Inflammation and MDD in Individuals with T2D

The precise pathophysiological mechanisms leading to MDD in individuals with T2D remain incompletely understood. However, a prominent mechanism implicated in comorbidity is the inflammation hypothesis. This theory suggests that the elevation of pro-inflammatory markers seen in obesity-induced T2D fosters a systemic pro-inflammatory milieu across various tissues, potentially contributing to the onset of MDD [139]. Notably, studies have shown that adults and elderly individuals with the MDD and T2D comorbidity exhibit higher levels of CRP and IL-6 in their plasma than those with only T2D [140]. Furthermore, hyperglycemia and hyperinsulinemia in T2D may further exacerbate this pro-inflammatory state, potentially facilitating access of pro-inflammatory mediators to the CNS and thereby activating pathways associated with depressive symptoms. In diabetic animal models, both central and peripheral anti-inflammatory responses are compromised, correlating with an increase in depressive-like behaviors [141]. Another study found that combinatorial treatment with Metformin and Telmisartan (hypertension medication) alleviated depressive behaviors in rats while reducing levels of inflammatory mediators, including nitric oxide, IL-6, and IL-1β [142].
Furthermore, dysfunction of the BBB emerges as a hallmark linking T2D to neuropathology [143]. Studies in diabetic mice have shown a significant increase in BBB permeability, a phenomenon also observed in MDD [144]. However, the specific role of BBB dysfunction in precipitating depressive symptoms in T2D requires further investigation. Nonetheless, the downstream consequences of BBB disruption, including inflammation, oxidative stress, and glial activation, have been extensively associated with MDD and may thus represent a shared mechanism underlying the comorbidity [145,146].
Additionally, elevated inflammatory cytokines downregulate BNDF expression, crucial for neurogenesis, in the hippocampus and prefrontal cortex, thus inducing depressive phenotypes in individuals with T2D [147]. Studies on insulin resistance-induced mice corroborate this, demonstrating increased immobility time, anhedonia, and anxiety-like behaviors alongside diminished plasmatic and amygdalar BDNF levels [148]. This suggests that insulin resistance may influence this behavioral phenotype in the modulation of neurotrophic factor expression.

5.2.2. Inflammation, Oxidative Stress, and MDD in Individuals with T2D

Another proposed mechanism involves oxidative stress stemming from T2D. In individuals with T2D, hyperglycemia hampers the activity of antioxidant enzymes in the brain, leading to an accumulation of reactive oxygen species, which instigates apoptotic and necrotic cell death, culminating in cerebral injury and inhibition of neurogenesis [149]. Reactive oxygen species trigger the activation of NF-κB (regulator of brain inflammation), thereby upregulating the expression of pro-inflammatory cytokines like TNF-α and IL-1β [150], further contributing to MDD pathogenesis. Although oxidative stress may have independent effects on the comorbidity, it is still relevant to the context of this review as there is a bidirectional relationship between oxidative stress and inflammation, whereby oxidative stress can lead to chronic inflammation, while chronic inflammation increases oxidative stress by promoting reactive oxygen species production and decreasing antioxidant reserves [151]. This suggests that oxidative stress may exacerbate the negative effects of inflammation, which may, in turn, exacerbate the detrimental effects of oxidative stress, ultimately increasing the risk of developing MDD in individuals with T2D.

5.2.3. Inflammation, the HPA Axis, and MDD in Individuals with T2D

Emerging evidence suggests that disturbances in the HPA axis link stress exposure to mood disorders [152]. Diabetic conditions hyperactivate the brain renin-angiotensin system, a system involved in brain homeostasis, by acting on angiotensin receptor subtypes, including the angiotensin II receptor type 1. The activation of this receptor promotes inflammation and oxidative stress [153]. Further, this subsequently hyperactivates the HPA axis and elevates cortisol and proinflammatory cytokine levels, fostering an environment favorable to MDD onset. Notably, hyperinsulinemia exacerbates HPA axis hyperactivation, leading to elevated plasma cortisol levels and subsequent hippocampal atrophy, which, in turn, disrupts HPA axis feedback modulation [154]. Furthermore, neuroinflammation attributed to microglial and astrocytic activation, alongside increased hippocampal microglial and astrocyte numbers, was observed in hyperglycemic animal models of metabolic syndrome [155]. In diabetic rats, impaired adaptation of the HPA response to various stress forms was noted, partially due to defects in glucocorticoid receptors [156]. Nonetheless, neuroinflammation appears linked to MDD in T2D, suggesting immunomodulatory strategies targeting inflammatory mediators to mitigate neuroinflammation and disturbed redox homeostasis may hold promise.

5.2.4. Inflammation, Neuroplasticity, and MDD in Individuals with T2D

The activation of innate immunity and increased cytokine levels seen in T2D can lead to microglial activation and subsequent reductions in neurogenesis [157]. Studies in T2D rodent models have revealed decreased dendritic branching and plasticity of neurons, along with cognitive deficits and depression-like behaviors correlated with reduced hippocampal neurogenesis [157,158]. It is worth noting that hyperglycemia and hyperinsulinemia, other characteristics of T2D, may also lead to decreased neurogenesis [159]. Thus, further investigations must be performed to pinpoint the independent impact of T2D-associated inflammation on neurogenesis and the associated increased vulnerability to MDD.

6. Targeting Inflammation through Lifestyle Interventions in the Management of MDD and T2D

6.1. Diet Interventions

Diet plays a fundamental role in the management of T2D and is increasingly recognized for its potential role in the management of depressive symptoms in individuals with MDD. Healthy dietary patterns and dietary improvements have the potential to elicit a variety of biological changes, including changes in inflammation, which might be relevant for the prevention and management of MDD and T2D.
Numerous systematic reviews and meta-analyses of observational studies have shown that healthy dietary patterns, characterized by high intakes of fruit, vegetables, nuts, whole grain, legumes, low-fat dairy products, fish, and lean cuts of meat, decrease the risk of developing MDD in generally healthy adults [160,161,162]. Similarly, interventions promoting the adherence to healthy dietary patterns, such as the Mediterranean diet, have the potential to significantly decrease depressive symptoms among adults with MDD or high depressive symptoms [163,164]. Along the same lines, consumption of healthy dietary patterns has been associated with a lower risk of T2D in observational studies [165,166,167] and improvements in glycemic control in intervention studies conducted in adults with T2D [168,169].
In line with this review’s topic, diet is known to play a pivotal role in influencing the body’s inflammatory response, with certain dietary patterns, foods, and food components capable of either promoting or alleviating inflammation. Healthy dietary patterns, such as the Mediterranean diet, the Dietary Approach to Stop Hypertensions (DASH) diet, and vegetarian diets, have been demonstrated to reduce inflammation in both healthy individuals and individuals with chronic diseases such as T2D [170]. Similarly, specific foods, beverages, nutrients, and other food components, including tea, omega-3 fatty acids, and flavonoids, have been found to have beneficial impacts on inflammation [171,172,173]. Conversely, unhealthy dietary patterns [174] as well as high intakes of some nutrients, such as saturated fatty acids [175], can lead to increased inflammation. Particularly, saturated fatty acids have been shown to promote a pro-inflammatory response predominantly through the activation of TLR4 signaling [176]. TLR4 binding to diet-derived saturated fatty acids also contributes to adipose tissue inflammation and dysfunction [177].
Diet has also been associated with other pathways linking inflammation to the development of MDD and T2D, including HPA axis dysregulation, neuroplasticity, and gut microbiome modulations. For example, in a sample of healthy men, fish oil supplementation over a period of 3 weeks has been shown to reduce basal cortisol levels and to prevent adrenal activation elicited by mental stress [178]. Similarly, high doses of vitamin C as well as flavonoid-rich foods (e.g., dark chocolate, pomegranate juice) were shown to decrease cortisol levels in healthy individuals as well as individuals with overweight or obesity [179,180]. In contrast, high glycemic index diets, even when consumed over short periods of time (e.g., ≤3 weeks), were shown to increase cortisol levels in healthy adults [181,182]. In rodents, diets high in fats and refined sugars have been shown to decrease neurogenesis, potentially through a decrease in hippocampal BDNF [183]. Conversely, dietary compounds such as omega-3 fatty acids, fiber, vitamins, and flavonoids appear to have a beneficial impact on hippocampal neurogenesis [184,185]. The diversity, composition, and metabolic function of the gut microbiota are also well recognized to be influenced by both short-term nutrient intake and long-term dietary patterns [186,187]. Notably, the abundance of SCFA-producing bacterial species as well as bacterial species involved in nutrient metabolisms has been positively correlated with the consumption of plant-based and Mediterranean-like diets, while the abundance of pro-inflammatory bacterial species has been positively associated with the consumption of processed and animal-derived foods [188,189].
While dietary improvements have beneficial effects on the risk and management of both MDD and T2D, a limited number of studies have assessed the role of diet in the prevention and management of the MDD and T2D comorbidity. It would be relevant to study the association between diet and inflammation in individuals with this comorbidity.

6.2. Physical Activity Interventions

Physical activity has been demonstrated to have beneficial effects on both MDD and T2D management. The extensive array of biological changes observed in exercise studies highlights its potential to elicit beneficial effects through various pathways. Regarding MDD, it is widely acknowledged that exercise effectively treats mild to moderate MDD, with response rates comparable to pharmacotherapies and cognitive behavioral therapy. Evidence suggests that physical activity may alleviate depressive phenotypes through several biological mechanisms. For example, exercise is linked with an increase in BDNF, which is known to play a role in alleviating symptoms by promoting neuron health and synaptic plasticity [190]. Moreover, as reviewed in this article, MDD is also associated with an upregulation of inflammatory pathways in the brain. At the same time, physical activity is linked with a reduction in systemic inflammatory signaling, suggesting a potential for mitigating these pathways [191]. More specifically, a study conducted on rodents found that the number and activation of hippocampal microglia, along with neuroinflammation in the hippocampus, may contribute to the pathogenesis of MDD [192]. A running exercise regimen was shown to reverse these changes, suggesting that the antidepressant effects of exercise may be, in part, due to alternations in hippocampal microglia and neuroinflammation [192]. Additionally, endurance exercise training has been shown to increase the release of transcriptional factors that enhance the expression of the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) gene. The PGC1α gene is heavily involved in preserving and protecting against neuronal loss [193] and is associated with a reduction in inflammatory markers, notably IL-6 and TNF-α [193,194].
In the context of T2D, both aerobic and resistance exercises have been established to enhance glycemic control, thereby aiding in the management of T2D [195]. Evidence has also linked exercise to lower BMI, with a reduction in pro-inflammatory cytokine secretion and an increase in anti-inflammatory cytokines [196]. Reducing the body’s inflammatory state through physical activity holds significant importance in mitigating the development of T2D. Although findings in the literature are mixed, long-term exercise has demonstrated a capacity to decrease the levels of inflammatory markers such as CRP, TNF-a, and IL-6, potentially alleviating chronic low-grade inflammation [197]. Hyperglycemia in individuals with T2D upregulates the expression of inflammation markers, and exercise has been shown to diminish their expression, thereby improving insulin sensitivity and reducing blood glucose levels [198]. Furthermore, physical inactivity is strongly correlated with obesity, particularly the accumulation of visceral fat, which exacerbates systemic inflammation [199]. Conversely, long-term physical exercise serves as a protective mechanism against abdominal fat accumulation and subsequent chronic inflammation [200]. This is partly mediated by myokines, in particular IL-15, which is implicated in the control of visceral adiposity. Indeed, a study demonstrated that elevated IL-15 levels protected mice from obesity, particularly by limiting visceral fat deposition. Interestingly, a 12-week endurance training study conducted in men has shown an increase in the expression of muscular IL-15 following endurance training [201], potentially contributing to decreasing visceral fats and subsequent inflammation. Additionally, exercise has been demonstrated in both men and women following a 12-week training program to lessen oxidative stress by enhancing the expression of antioxidant enzyme genes, increasing nitrogen oxide availability, and decreasing the production of reactive oxygen species [202]. In turn, this indirectly reduces the levels of inflammatory markers.
In the context of T2D, both aerobic and resistance exercises have been established to enhance glycemic control, thereby aiding in the management of T2D [195,203]. Various studies have also linked exercise to lower BMI, with a reduction in pro-inflammatory cytokine secretion and an increase in anti-inflammatory cytokines [196,204]. Reducing the body’s inflammatory state through physical activity holds significant importance in mitigating the development of T2D. Although findings in the literature are mixed, long-term exercise has demonstrated a capacity to decrease the levels of inflammatory markers such as CRP, TNF-a, and IL-6, potentially alleviating chronic low-grade inflammation [197]. Hyperglycemia in individuals with T2D upregulates the expression of inflammation markers, and exercise has been shown to diminish their expression, thereby improving insulin sensitivity and reducing blood glucose levels [198]. Furthermore, physical inactivity is strongly correlated with obesity, particularly the accumulation of visceral fat, which exacerbates systemic inflammation [199]. Conversely, long-term physical exercise serves as a protective mechanism against abdominal fat accumulation and subsequent chronic inflammation [200]. This is partly mediated by myokines, particularly IL-15, which is implicated in the control of visceral adiposity. Indeed, a study demonstrated that elevated IL-15 levels protected mice from obesity, particularly by limiting visceral fat deposition. Interestingly, a 12-week endurance training study conducted in men has shown an increase in the expression of muscular IL-15 following endurance training [201], potentially contributing to decreasing visceral fats and subsequent inflammation. Additionally, exercise has been demonstrated in both men and women following a 12-week training program to lessen oxidative stress by enhancing the expression of antioxidant enzyme genes, increasing nitrogen oxide availability, and decreasing the production of reactive oxygen species [202]. In turn, this indirectly reduces the levels of inflammatory markers.

7. Conclusions

There is a bidirectional relationship between the risk of MDD and the risk of T2D. Additionally, MDD exacerbates the complications of T2D by promoting unhealthy behaviors, inadequate self-care, and difficulties in adhering to T2D management strategies, making it a crucial comorbidity to further investigate. Although the precise mechanisms linking T2D and MDD are still not fully understood, shared inflammatory mechanisms likely contribute to the heightened risk of developing this comorbidity. As described in this review, inflammation serves as a common denominator exacerbating the adverse impacts of both MDD and T2D, complicating the management of the comorbidity. Indeed, chronic low-grade inflammation characterizes both conditions, exerting profound effects on the HPA axis, neuroplastic processes, gut microbiome, insulin resistance, and adipose tissue dysfunction. Deepening our understanding of the mechanisms underlying the augmented inflammatory responses observed in individuals with this comorbidity is essential for tailoring appropriate therapeutic strategies. Interventions targeting inflammation through dietary modifications and physical activity hold significant promise as adjunctive strategies in the management of the MDD and T2D comorbidity. However, it is important to note that most studies included in this review involved Caucasian North American populations, which may limit the generalizability of these findings. To better understand the relationship between MDD and T2D across different demographic groups and to ascertain whether inflammatory pathways vary among these populations, it is crucial to broaden the scope of research to include more diverse populations.
Lastly, policymakers should prioritize recommendations for preventing both disorders, recognizing their significance as public health concerns. The effective management of MDD leads to improved glycemic control, thereby enhancing overall health outcomes. A collaborative interprofessional approach involving the individual with MDD and T2D, mental health professionals, primary care providers, and other care partners is therefore essential to mitigate the impact of this comorbid psychopathology.

Author Contributions

Conceptualization, A.M.B. and M.R.; writing—original draft preparation, A.M.B. and M.R.; writing—review and editing, A.M.B., M.R., I.G. and C.A.; visualization, A.M.B. and M.R.; supervision, I.G. and C.A.; project administration, I.G. and C.A.; funding acquisition, I.G. and C.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by an Interstrategic Direction Grant (2016) from the Institut du Savoir Montfort (https://savoirmontfort.ca (accessed on 22 April 2024)) to I.G. and C.A. The Institut du Savoir Montfort is an organization dedicated to education, research and community engagement, and often collaborates with Universities. A.B. was funded by a Nutrition and Mental Health Doctoral Scholarship from the School of Nutrition Sciences of the University of Ottawa. M.R. was funded by a Canadian Institutes of Health Research (CIHR) Master’s Scholarship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Incidence, Prevalence, and Years Lived with Disability for 354 Diseases and Injuries for 195 Countries and Territories, 1990-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [PubMed]
  2. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, J.C.; Mbanya, J.C.; et al. IDF Diabetes Atlas: Global, Regional and Country-Level Diabetes Prevalence Estimates for 2021 and Projections for 2045. Diabetes Res. Clin. Pract. 2022, 183, 109119. [Google Scholar] [CrossRef] [PubMed]
  3. Berk, M.; Köhler-Forsberg, O.; Turner, M.; Penninx, B.W.J.H.; Wrobel, A.; Firth, J.; Loughman, A.; Reavley, N.J.; McGrath, J.J.; Momen, N.C.; et al. Comorbidity between Major Depressive Disorder and Physical Diseases: A Comprehensive Review of Epidemiology, Mechanisms and Management. World Psychiatry 2023, 22, 366–387. [Google Scholar] [CrossRef] [PubMed]
  4. Rotella, F.; Mannucci, E. Depression as a Risk Factor for Diabetes: A Meta-Analysis of Longitudinal Studies. J. Clin. Psychiatry 2013, 74, 4231. [Google Scholar] [CrossRef] [PubMed]
  5. Hasan, S.S.; Clavarino, A.M.; Mamun, A.A.; Kairuz, T. Incidence and Risk of Diabetes Mellitus Associated with Depressive Symptoms in Adults: Evidence from Longitudinal Studies. Diabetes Metab. Syndr. Clin. Res. Rev. 2014, 8, 82–87. [Google Scholar] [CrossRef]
  6. Chireh, B.; Li, M.; D’Arcy, C. Diabetes Increases the Risk of Depression: A Systematic Review, Meta-Analysis and Estimates of Population Attributable Fractions Based on Prospective Studies. Prev. Med. Rep. 2019, 14, 100822. [Google Scholar] [CrossRef]
  7. Schmitt, A.; Reimer, A.; Hermanns, N.; Kulzer, B.; Ehrmann, D.; Krichbaum, M.; Huber, J.; Haak, T. Depression Is Linked to Hyperglycaemia via Suboptimal Diabetes Self-Management: A Cross-Sectional Mediation Analysis. J. Psychosom. Res. 2017, 94, 17–23. [Google Scholar] [CrossRef]
  8. Ismail, K.; Moulton, C.D.; Winkley, K.; Pickup, J.C.; Thomas, S.M.; Sherwood, R.A.; Stahl, D.; Amiel, S.A. The Association of Depressive Symptoms and Diabetes Distress with Glycaemic Control and Diabetes Complications over 2 Years in Newly Diagnosed Type 2 Diabetes: A Prospective Cohort Study. Diabetologia 2017, 60, 2092–2102. [Google Scholar] [CrossRef]
  9. Beran, M.; Muzambi, R.; Geraets, A.; Albertorio-Diaz, J.R.; Adriaanse, M.C.; Iversen, M.M.; Kokoszka, A.; Nefs, G.; Nouwen, A.; Pouwer, F.; et al. The Bidirectional Longitudinal Association between Depressive Symptoms and HbA1c: A Systematic Review and Meta-Analysis. Diabet. Med. 2022, 39, e14671. [Google Scholar] [CrossRef]
  10. Jing, X.; Chen, J.; Dong, Y.; Han, D.; Zhao, H.; Wang, X.; Gao, F.; Li, C.; Cui, Z.; Liu, Y.; et al. Related Factors of Quality of Life of Type 2 Diabetes Patients: A Systematic Review and Meta-Analysis. Health Qual. Life Outcomes 2018, 16, 189. [Google Scholar] [CrossRef]
  11. Teli, M.; Thato, R.; Rias, Y.A. Predicting Factors of Health-Related Quality of Life Among Adults with Type 2 Diabetes: A Systematic Review. SAGE Open Nurs. 2023, 9, 23779608231185921. [Google Scholar] [CrossRef] [PubMed]
  12. Prigge, R.; Wild, S.H.; Jackson, C.A. Depression, Diabetes, Comorbid Depression and Diabetes and Risk of All-Cause and Cause-Specific Mortality: A Prospective Cohort Study. Diabetologia 2022, 65, 1450–1460. [Google Scholar] [CrossRef] [PubMed]
  13. Cai, J.; Zhang, S.; Wu, R.; Huang, J. Association between Depression and Diabetes Mellitus and the Impact of Their Comorbidity on Mortality: Evidence from a Nationally Representative Study. J. Affect. Disord. 2024, 354, 11–18. [Google Scholar] [CrossRef] [PubMed]
  14. König, H.; König, H.H.; Konnopka, A. The Excess Costs of Depression: A Systematic Review and Meta-Analysis. Epidemiol. Psychiatr. Sci. 2020, 29, e30. [Google Scholar] [CrossRef] [PubMed]
  15. Soley-Bori, M.; Ashworth, M.; Bisquera, A.; Dodhia, H.; Lynch, R.; Wang, Y.; Fox-Rushby, J. Impact of Multimorbidity on Healthcare Costs and Utilisation: A Systematic Review of the UK Literature. Br. J. General. Pract. 2021, 71, e39–e46. [Google Scholar] [CrossRef]
  16. Mehdi, S.; Wani, S.U.D.; Krishna, K.L.; Kinattingal, N.; Roohi, T.F. A Review on Linking Stress, Depression, and Insulin Resistance via Low-Grade Chronic Inflammation. Biochem. Biophys. Rep. 2023, 36, 101571. [Google Scholar] [CrossRef]
  17. Uher, R.; Payne, J.L.; Pavlova, B.; Perlis, R.H. Major Depressive Disorder in DSM-5: Implications for Clinical Practice and Research of Changes from DSM-IV. Depress. Anxiety 2014, 31, 459–471. [Google Scholar] [CrossRef]
  18. Dunn, E.C.; Brown, R.C.; Dai, Y.; Rosand, J.; Nugent, N.R.; Amstadter, A.B.; Smoller, J.W. Genetic Determinants of Depression: Recent Findings and Future Directions. Harv. Rev. Psychiatry 2015, 23, 1–18. [Google Scholar] [CrossRef]
  19. Marx, W.; Penninx, B.W.J.H.; Solmi, M.; Furukawa, T.A.; Firth, J.; Carvalho, A.F.; Berk, M. Major Depressive Disorder. Nat. Rev. Dis. Primers 2023, 9, 44. [Google Scholar] [CrossRef]
  20. Köhler, C.A.; Freitas, T.H.; Maes, M.; de Andrade, N.Q.; Liu, C.S.; Fernandes, B.S.; Stubbs, B.; Solmi, M.; Veronese, N.; Herrmann, N.; et al. Peripheral Cytokine and Chemokine Alterations in Depression: A Meta-Analysis of 82 Studies. Acta Psychiatr. Scand. 2017, 135, 373–387. [Google Scholar] [CrossRef]
  21. Leighton, S.P.; Nerurkar, L.; Krishnadas, R.; Johnman, C.; Graham, G.J.; Cavanagh, J. Chemokines in Depression in Health and in Inflammatory Illness: A Systematic Review and Meta-Analysis. Mol. Psychiatry 2017, 23, 48–58. [Google Scholar] [CrossRef] [PubMed]
  22. Enache, D.; Pariante, C.M.; Mondelli, V. Markers of Central Inflammation in Major Depressive Disorder: A Systematic Review and Meta-Analysis of Studies Examining Cerebrospinal Fluid, Positron Emission Tomography and Post-Mortem Brain Tissue. Brain Behav. Immun. 2019, 81, 24–40. [Google Scholar] [CrossRef] [PubMed]
  23. Bush, G.; Luu, P.; Posner, M.I. Cognitive and Emotional Influences in Anterior Cingulate Cortex. Trends Cogn. Sci. 2000, 4, 215–222. [Google Scholar] [CrossRef] [PubMed]
  24. Conway, B.R. The Organization and Operation of Inferior Temporal Cortex. Annu. Rev. Vis. Sci. 2018, 4, 381–402. [Google Scholar] [CrossRef]
  25. Benros, M.E.; Waltoft, B.L.; Nordentoft, M.; Ostergaard, S.D.; Eaton, W.W.; Krogh, J.; Mortensen, P.B. Autoimmune Diseases and Severe Infections as Risk Factors for Mood Disorders: A Nationwide Study. JAMA Psychiatry 2013, 70, 812–820. [Google Scholar] [CrossRef]
  26. Patki, G.; Solanki, N.; Atrooz, F.; Allam, F.; Salim, S. Depression, Anxiety-like Behavior and Memory Impairment Are Associated with Increased Oxidative Stress and Inflammation in a Rat Model of Social Stress. Brain Res. 2013, 1539, 73–86. [Google Scholar] [CrossRef]
  27. Yin, R.; Zhang, K.; Li, Y.; Tang, Z.; Zheng, R.; Ma, Y.; Chen, Z.; Lei, N.; Xiong, L.; Guo, P.; et al. Lipopolysaccharide-Induced Depression-like Model in Mice: Meta-Analysis and Systematic Evaluation. Front. Immunol. 2023, 14, 1181973. [Google Scholar] [CrossRef]
  28. Ohgi, Y.; Futamura, T.; Kikuchi, T.; Hashimoto, K. Effects of Antidepressants on Alternations in Serum Cytokines and Depressive-like Behavior in Mice after Lipopolysaccharide Administration. Pharmacol. Biochem. Behav. 2013, 103, 853–859. [Google Scholar] [CrossRef]
  29. Ramirez, K.; Sheridan, J.F. Antidepressant Imipramine Diminishes Stress-Induced Inflammation in the Periphery and Central Nervous System and Related Anxiety- and Depressive- like Behaviors. Brain Behav. Immun. 2016, 57, 293–303. [Google Scholar] [CrossRef]
  30. Udina, M.; Castellví, P.; Moreno-España, J.; Navinés, R.; Valdés, M.; Forns, X.; Langohr, K.; Solà, R.; Vieta, E.; Martín-Santos, R. Interferon-Induced Depression in Chronic Hepatitis C: A Systematic Review and Meta-Analysis. J. Clin. Psychiatry 2012, 73, 1128–1138. [Google Scholar] [CrossRef]
  31. Udina, M.; Hidalgo, D.; Navinés, R.; Forns, X.; Solà, R.; Farré, M.; Capuron, L.; Vieta, E.; Martín-Santos, R. Prophylactic Antidepressant Treatment of Interferon-Induced Depression in Chronic Hepatitis C: A Systematic Review and Meta-Analysis. J. Clin. Psychiatry 2014, 75, 15762. [Google Scholar] [CrossRef] [PubMed]
  32. Więdłocha, M.; Marcinowicz, P.; Krupa, R.; Janoska-Jaździk, M.; Janus, M.; Dębowska, W.; Mosiołek, A.; Waszkiewicz, N.; Szulc, A. Effect of Antidepressant Treatment on Peripheral Inflammation Markers—A Meta-Analysis. Prog. Neuropsychopharmacol. Biol. Psychiatry 2018, 80, 217–226. [Google Scholar] [CrossRef] [PubMed]
  33. Kappelmann, N.; Lewis, G.; Dantzer, R.; Jones, P.B.; Khandaker, G.M. Antidepressant Activity of Anti-Cytokine Treatment: A Systematic Review and Meta-Analysis of Clinical Trials of Chronic Inflammatory Conditions. Mol. Psychiatry 2018, 23, 335–343. [Google Scholar] [CrossRef] [PubMed]
  34. Na, K.S.; Lee, K.J.; Lee, J.S.; Cho, Y.S.; Jung, H.Y. Efficacy of Adjunctive Celecoxib Treatment for Patients with Major Depressive Disorder: A Meta-Analysis. Prog. Neuropsychopharmacol. Biol. Psychiatry 2014, 48, 79–85. [Google Scholar] [CrossRef] [PubMed]
  35. Van Der Veen, F.M.; Evers, E.A.T.; Deutz, N.E.P.; Schmitt, J.A.J. Effects of Acute Tryptophan Depletion on Mood and Facial Emotion Perception Related Brain Activation and Performance in Healthy Women with and without a Family History of Depression. Neuropsychopharmacology 2007, 32, 216–224. [Google Scholar] [CrossRef]
  36. Bell, C.; Abrams, J.; Nutt, D. Tryptophan Depletion and Its Implications for Psychiatry. Br. J. Psychiatry 2001, 178, 399–405. [Google Scholar] [CrossRef] [PubMed]
  37. Ogawa, S.; Tsuchimine, S.; Kunugi, H. Cerebrospinal Fluid Monoamine Metabolite Concentrations in Depressive Disorder: A Meta-Analysis of Historic Evidence. J. Psychiatr. Res. 2018, 105, 137–146. [Google Scholar] [CrossRef]
  38. Meyer, J.H.; Ginovart, N.; Boovariwala, A.; Sagrati, S.; Hussey, D.; Garcia, A.; Young, T.; Praschak-Rieder, N.; Wilson, A.A.; Houle, S. Elevated Monoamine Oxidase A Levels in the Brain: An Explanation for the Monoamine Imbalance of Major Depression. Arch. Gen. Psychiatry 2006, 63, 1209–1216. [Google Scholar] [CrossRef]
  39. Mousten, I.V.; Sørensen, N.V.; Christensen, R.H.B.; Benros, M.E. Cerebrospinal Fluid Biomarkers in Patients with Unipolar Depression Compared with Healthy Control Individuals: A Systematic Review and Meta-Analysis. JAMA Psychiatry 2022, 79, 571–581. [Google Scholar] [CrossRef]
  40. Kennis, M.; Gerritsen, L.; van Dalen, M.; Williams, A.; Cuijpers, P.; Bockting, C. Prospective Biomarkers of Major Depressive Disorder: A Systematic Review and Meta-Analysis. Mol. Psychiatry 2019, 25, 321–338. [Google Scholar] [CrossRef]
  41. Perrin, A.J.; Horowitz, M.A.; Roelofs, J.; Zunszain, P.A.; Pariante, C.M. Glucocorticoid Resistance: Is It a Requisite for Increased Cytokine Production in Depression? A Systematic Review and Meta-Analysis. Front. Psychiatry 2019, 10, 443676. [Google Scholar] [CrossRef] [PubMed]
  42. Lino de Oliveira, C.; Bolzan, J.A.; Surget, A.; Belzung, C. Do Antidepressants Promote Neurogenesis in Adult Hippocampus? A Systematic Review and Meta-Analysis on Naive Rodents. Pharmacol. Ther. 2020, 210, 107515. [Google Scholar] [CrossRef] [PubMed]
  43. Molendijk, M.L.; Spinhoven, P.; Polak, M.; Bus, B.A.A.; Penninx, B.W.J.H.; Elzinga, B.M. Serum BDNF Concentrations as Peripheral Manifestations of Depression: Evidence from a Systematic Review and Meta-Analyses on 179 Associations (N = 9484). Mol. Psychiatry 2013, 19, 791–800. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, C.; Zhong, J.; Zou, B.; Fang, L.; Chen, J.; Deng, X.; Zhang, L.; Zhao, X.; Qu, Z.; Lei, Y.; et al. Meta-Analyses of Comparative Efficacy of Antidepressant Medications on Peripheral BDNF Concentration in Patients with Depression. PLoS ONE 2017, 12, e0172270. [Google Scholar] [CrossRef]
  45. Kempton, M.J.; Salvador, Z.; Munafò, M.R.; Geddes, J.R.; Simmons, A.; Frangou, S.; Williams, S.C.R. Structural Neuroimaging Studies in Major Depressive Disorder: Meta-Analysis and Comparison with Bipolar Disorder. Arch. Gen. Psychiatry 2011, 68, 675–690. [Google Scholar] [CrossRef]
  46. Amirkhanzadeh Barandouzi, Z.; Starkweather, A.R.; Henderson, W.A.; Gyamfi, A.; Cong, X.S. Altered Composition of Gut Microbiota in Depression: A Systematic Review. Front. Psychiatry 2020, 11, 541. [Google Scholar] [CrossRef]
  47. Simpson, C.A.; Diaz-Arteche, C.; Eliby, D.; Schwartz, O.S.; Simmons, J.G.; Cowan, C.S.M. The Gut Microbiota in Anxiety and Depression—A Systematic Review. Clin. Psychol. Rev. 2021, 83, 101943. [Google Scholar] [CrossRef]
  48. Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X.; et al. Gut Microbiome Remodeling Induces Depressive-like Behaviors through a Pathway Mediated by the Host’s Metabolism. Mol. Psychiatry 2016, 21, 786–796. [Google Scholar] [CrossRef]
  49. GBD 2019 Mental Disorders Collaborators. Global, Regional, and National Burden of 12 Mental Disorders in 204 Countries and Territories, 1990–2019: A Systematic Analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry 2022, 9, 137–150. [Google Scholar] [CrossRef]
  50. Amiri, S.; Behnezhad, S.; Nadinlui, K.B. Body Mass Index (BMI) and Risk of Depression in Adults: A Systematic Review and Meta-Analysis of Longitudinal Studies. Obes. Med. 2018, 12, 1–12. [Google Scholar] [CrossRef]
  51. Graham, E.A.; Deschênes, S.S.; Khalil, M.N.; Danna, S.; Filion, K.B.; Schmitz, N. Measures of Depression and Risk of Type 2 Diabetes: A Systematic Review and Meta-Analysis. J. Affect. Disord. 2020, 265, 224–232. [Google Scholar] [CrossRef] [PubMed]
  52. Saha, S.; Lim, C.C.W.; Cannon, D.L.; Burton, L.; Bremner, M.; Cosgrove, P.; Huo, Y.; McGrath, J.J. Co-Morbidity between Mood and Anxiety Disorders: A Systematic Review and Meta-Analysis. Depress. Anxiety 2021, 38, 286–306. [Google Scholar] [CrossRef] [PubMed]
  53. Chesney, E.; Goodwin, G.M.; Fazel, S. Risks of All-Cause and Suicide Mortality in Mental Disorders: A Meta-Review. World Psychiatry 2014, 13, 153–160. [Google Scholar] [CrossRef]
  54. Chatterjee, S.; Khunti, K.; Davies, M.J. Type 2 Diabetes. Lancet 2017, 389, 2239–2251. [Google Scholar] [CrossRef] [PubMed]
  55. Punthakee, Z.; Goldenberg, R.; Katz, P. 2018 Clinical Practice Guidelines Definition, Classification and Diagnosis of Diabetes, Prediabetes and Metabolic Syndrome Diabetes Canada Clinical Practice Guidelines Expert Committee. Can. J. Diabetes 2018, 42, S10–S15. [Google Scholar] [CrossRef] [PubMed]
  56. Stumvoll, M.; Goldstein, B.J.; Van Haeften, T.W. Type 2 Diabetes: Principles of Pathogenesis and Therapy. Lancet 2005, 365, 1333–1346. [Google Scholar] [CrossRef]
  57. Sesti, G. Pathophysiology of Insulin Resistance. Best Pract. Res. Clin. Endocrinol. Metab. 2006, 20, 665–679. [Google Scholar] [CrossRef]
  58. Schwartz, S.S.; Epstein, S.; Corkey, B.E.; Grant, S.F.A.; Gavin, J.R.; Aguilar, R.B. The Time Is Right for a New Classification System for Diabetes: Rationale and Implications of the β-Cell–Centric Classification Schema. Diabetes Care 2016, 39, 179–186. [Google Scholar] [CrossRef]
  59. Iglay, K.; Hannachi, H.; Howie, P.J.; Xu, J.; Li, X.; Engel, S.S.; Moore, L.M.; Rajpathak, S. Prevalence and Co-Prevalence of Comorbidities among Patients with Type 2 Diabetes Mellitus. Curr. Med. Res. Opin. 2016, 32, 1243–1252. [Google Scholar] [CrossRef]
  60. Hannoodee, S.; Nasuruddin, D. Acute Inflammatory Response. StatPearls 2020, 206, 20. [Google Scholar] [CrossRef]
  61. Pahwa, R.; Goyal, A.; Jialal, I. Chronic Inflammation. In Pathobiology of Human Disease: A Dynamic Encyclopedia of Disease Mechanisms; Elsevier: Amsterdam, The Netherlands, 2023; pp. 300–314. [Google Scholar] [CrossRef]
  62. Huang, X.; Hussain, B.; Chang, J. Peripheral Inflammation and Blood–Brain Barrier Disruption: Effects and Mechanisms. CNS Neurosci. Ther. 2021, 27, 36–47. [Google Scholar] [CrossRef] [PubMed]
  63. Sun, Y.; Koyama, Y.; Shimada, S. Inflammation from Peripheral Organs to the Brain: How Does Systemic Inflammation Cause Neuroinflammation? Front. Aging Neurosci. 2022, 14, 903455. [Google Scholar] [CrossRef] [PubMed]
  64. Galea, I. The Blood–Brain Barrier in Systemic Infection and Inflammation. Cell Mol. Immunol. 2021, 18, 2489–2501. [Google Scholar] [CrossRef] [PubMed]
  65. Mondelli, V.; Vernon, A.C.; Turkheimer, F.; Dazzan, P.; Pariante, C.M. Brain Microglia in Psychiatric Disorders. Lancet Psychiatry 2017, 4, 563–572. [Google Scholar] [CrossRef]
  66. Cipriani, A.; Furukawa, T.A.; Salanti, G.; Chaimani, A.; Atkinson, L.Z.; Ogawa, Y.; Leucht, S.; Ruhe, H.G.; Turner, E.H.; Higgins, J.P.T.; et al. Comparative Efficacy and Acceptability of 21 Antidepressant Drugs for the Acute Treatment of Adults with Major Depressive Disorder: A Systematic Review and Network Meta-Analysis. Focus 2018, 16, 420–429. [Google Scholar] [CrossRef]
  67. Munkholm, K.; Winkelbeiner, S.; Homan, P. Individual Response to Antidepressants for Depression in Adults-a Meta-Analysis and Simulation Study. PLoS ONE 2020, 15, e0237950. [Google Scholar] [CrossRef]
  68. Zhu, C.-B.; Lindler, K.M.; Owens, A.W.; Daws, L.C.; Blakely, R.D.; Hewlett, W.A. Interleukin-1 Receptor Activation by Systemic Lipopolysaccharide Induces Behavioral Despair Linked to MAPK Regulation of CNS Serotonin Transporters. Neuropsychopharmacology 2010, 35, 2510–2520. [Google Scholar] [CrossRef]
  69. Bonaccorso, S.; Puzella, A.; Marino, V.; Pasquini, M.; Biondi, M.; Artini, M.; Almerighi, C.; Levrero, M.; Egyed, B.; Bosmans, E.; et al. Immunotherapy with Interferon-Alpha in Patients Affected by Chronic Hepatitis C Induces an Intercorrelated Stimulation of the Cytokine Network and an Increase in Depressive and Anxiety Symptoms. Psychiatry Res. 2001, 105, 45–55. [Google Scholar] [CrossRef]
  70. Capuron, L.; Neurauter, G.; Musselman, D.L.; Lawson, D.H.; Nemeroff, C.B.; Fuchs, D.; Miller, A.H. Interferon-Alpha–Induced Changes in Tryptophan Metabolism: Relationship to Depression and Paroxetine Treatment. Biol. Psychiatry 2003, 54, 906–914. [Google Scholar] [CrossRef]
  71. Raison, C.L.; Dantzer, R.; Kelley, K.W.; Lawson, M.A.; Woolwine, B.J.; Vogt, G.; Spivey, J.R.; Saito, K.; Miller, A.H. CSF Concentrations of Brain Tryptophan and Kynurenines during Immune Stimulation with IFN-α: Relationship to CNS Immune Responses and Depression. Mol. Psychiatry 2010, 15, 393–403. [Google Scholar] [CrossRef]
  72. Neurauter, G.; Schrocksnadel, K.; Scholl-Burgi, S.; Sperner-Unterweger, B.; Schubert, C.; Ledochowski, M.; Fuchs, D. Chronic Immune Stimulation Correlates with Reduced Phenylalanine Turnover. Curr. Drug Metab. 2008, 9, 622–627. [Google Scholar] [CrossRef] [PubMed]
  73. Pariante, C.M.; Lightman, S.L. The HPA Axis in Major Depression: Classical Theories and New Developments. Trends Neurosci. 2008, 31, 464–468. [Google Scholar] [CrossRef] [PubMed]
  74. Nandam, L.S.; Brazel, M.; Zhou, M.; Jhaveri, D.J. Cortisol and Major Depressive Disorder—Translating Findings from Humans to Animal Models and Back. Front. Psychiatry 2020, 10, 476719. [Google Scholar] [CrossRef]
  75. Pariante, C.M. Why Are Depressed Patients Inflamed? A Reflection on 20 Years of Research on Depression, Glucocorticoid Resistance and Inflammation. Eur. Neuropsychopharmacol. 2017, 27, 554–559. [Google Scholar] [CrossRef] [PubMed]
  76. Matsubara, T.; Funato, H.; Kobayashi, A.; Nobumoto, M.; Watanabe, Y. Reduced Glucocorticoid Receptor α Expression in Mood Disorder Patients and First-Degree Relatives. Biol. Psychiatry 2006, 59, 689–695. [Google Scholar] [CrossRef]
  77. Pace, T.W.W.; Hu, F.; Miller, A.H. Cytokine-Effects on Glucocorticoid Receptor Function: Relevance to Glucocorticoid Resistance and the Pathophysiology and Treatment of Major Depression. Brain Behav. Immun. 2007, 21, 9–19. [Google Scholar] [CrossRef]
  78. Kossintseva, I.; Wong, S.; Johnstone, E.; Guilbert, L.; Olson, D.M.; Mitchell, B.F. Proinflammatory Cytokines Inhibit Human Placental 11β-Hydroxysteroid Dehydrogenase Type 2 Activity through Ca2+ and CAMP Pathways. Am. J. Physiol. Endocrinol. Metab. 2006, 290, 282–288. [Google Scholar] [CrossRef]
  79. Pariante, C.M. The Role of Multi-Drug Resistance p-Glycoprotein in Glucocorticoid Function: Studies in Animals and Relevance in Humans. Eur. J. Pharmacol. 2008, 583, 263–271. [Google Scholar] [CrossRef]
  80. Cramer, S.C.; Sur, M.; Dobkin, B.H.; O’Brien, C.; Sanger, T.D.; Trojanowski, J.Q.; Rumsey, J.M.; Hicks, R.; Cameron, J.; Chen, D.; et al. Harnessing Neuroplasticity for Clinical Applications. Brain 2011, 134, 1591–1609. [Google Scholar] [CrossRef]
  81. Bezzi, P.; Domercq, M.; Brambilla, L.; Galli, R.; Schols, D.; De Clercq, E.; Vescovi, A.; Bagetta, G.; Kollias, G.; Meldolesi, J.; et al. CXCR4-Activated Astrocyte Glutamate Release via TNFα: Amplification by Microglia Triggers Neurotoxicity. Nat. Neurosci. 2001, 4, 702–710. [Google Scholar] [CrossRef]
  82. Haroon, E.; Miller, A.H. Inflammation Effects on Brain Glutamate in Depression: Mechanistic Considerations and Treatment Implications. Curr. Top. Behav. Neurosci. 2017, 31, 173–198. [Google Scholar] [CrossRef] [PubMed]
  83. Guillemin, G.J. Quinolinic Acid: Neurotoxicity. FEBS J. 2012, 279, 1355. [Google Scholar] [CrossRef] [PubMed]
  84. Hardingham, G.E.; Bading, H. Synaptic versus Extrasynaptic NMDA Receptor Signalling: Implications for Neurodegenerative Disorders. Nat. Rev. Neurosci. 2010, 11, 682–696. [Google Scholar] [CrossRef] [PubMed]
  85. Ghosh, S.S.; Wang, J.; Yannie, P.J.; Ghosh, S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J. Endocr. Soc. 2020, 4, bvz039. [Google Scholar] [CrossRef]
  86. Sun, M.; Wu, W.; Liu, Z.; Cong, Y. Microbiota Metabolite Short Chain Fatty Acids, GPCR, and Inflammatory Bowel Diseases. J. Gastroenterol. 2017, 52, 1–8. [Google Scholar] [CrossRef]
  87. Appiah, S.A.; Foxx, C.L.; Langgartner, D.; Palmer, A.; Zambrano, C.A.; Braumüller, S.; Schaefer, E.J.; Wachter, U.; Elam, B.L.; Radermacher, P.; et al. Evaluation of the Gut Microbiome in Association with Biological Signatures of Inflammation in Murine Polytrauma and Shock. Sci. Rep. 2021, 11, 6665. [Google Scholar] [CrossRef]
  88. Christensen, A.A.; Gannon, M. The Beta Cell in Type 2 Diabetes. Curr. Diab. Rep. 2019, 19, 81. [Google Scholar] [CrossRef]
  89. Rohm, T.V.; Meier, D.T.; Olefsky, J.M.; Donath, M.Y. Inflammation in Obesity, Diabetes, and Related Disorders. Immunity 2022, 55, 31–55. [Google Scholar] [CrossRef]
  90. Wharton, S.; Pedersen, S.D.; Lau, D.C.W.; Sharma, A.M. Weight Management in Diabetes. Can. J. Diabetes 2018, 42, S124–S129. [Google Scholar] [CrossRef]
  91. Lasselin, J.; Capuron, L. Chronic Low-Grade Inflammation in Metabolic Disorders: Relevance for Behavioral Symptoms. Neuroimmunomodulation 2014, 21, 95–101. [Google Scholar] [CrossRef]
  92. Xu, H.; Barnes, G.T.; Yang, Q.; Tan, G.; Yang, D.; Chou, C.J.; Sole, J.; Nichols, A.; Ross, J.S.; Tartaglia, L.A.; et al. Chronic Inflammation in Fat Plays a Crucial Role in the Development of Obesity-Related Insulin Resistance. J. Clin. Investig. 2003, 112, 1821–1830. [Google Scholar] [CrossRef] [PubMed]
  93. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W. Obesity Is Associated with Macrophage Accumulation in Adipose Tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef] [PubMed]
  94. Lee, Y.S.; Olefsky, J. Chronic Tissue Inflammation and Metabolic Disease. Genes. Dev. 2021, 35, 307–328. [Google Scholar] [CrossRef] [PubMed]
  95. McLaughlin, T.; Ackerman, S.E.; Shen, L.; Engleman, E. Role of Innate and Adaptive Immunity in Obesity-Associated Metabolic Disease. J. Clin. Investig. 2017, 127, 5–13. [Google Scholar] [CrossRef]
  96. Fuchs, A.; Samovski, D.; Smith, G.I.; Cifarelli, V.; Farabi, S.S.; Yoshino, J.; Pietka, T.; Chang, S.W.; Ghosh, S.; Myckatyn, T.M.; et al. Associations Among Adipose Tissue Immunology, Inflammation, Exosomes and Insulin Sensitivity in People with Obesity and Nonalcoholic Fatty Liver Disease. Gastroenterology 2021, 161, 968–981. [Google Scholar] [CrossRef]
  97. Samaras, K.; Botelho, N.K.; Chisholm, D.J.; Lord, R.V. Subcutaneous and Visceral Adipose Tissue Gene Expression of Serum Adipokines That Predict Type 2 Diabetes. Obesity 2010, 18, 884–889. [Google Scholar] [CrossRef]
  98. Bruun, J.M.; Lihn, A.S.; Pedersen, S.B.; Richelsen, B. Monocyte Chemoattractant Protein-1 Release Is Higher in Visceral than Subcutaneous Human Adipose Tissue (AT): Implication of Macrophages Resident in the AT. J. Clin. Endocrinol. Metab. 2005, 90, 2282–2289. [Google Scholar] [CrossRef]
  99. Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity Induces a Phenotypic Switch in Adipose Tissue Macrophage Polarization. J. Clin. Investig. 2007, 117, 175–184. [Google Scholar] [CrossRef]
  100. McLaughlin, T.; Deng, A.; Gonzales, O.; Aillaud, M.; Yee, G.; Lamendola, C.; Abbasi, F.; Connolly, A.J.; Sherman, A.; Cushman, S.W.; et al. Insulin Resistance Is Associated with a Modest Increase in Inflammation in Subcutaneous Adipose Tissue of Moderately Obese Women. Diabetologia 2008, 51, 2303–2308. [Google Scholar] [CrossRef]
  101. Hotamisligil, G.S.; Erbay, E. Nutrient Sensing and Inflammation in Metabolic Diseases. Nat. Rev. Immunol. 2008, 8, 923–934. [Google Scholar] [CrossRef]
  102. Böni-Schnetzler, M.; Boller, S.; Debray, S.; Bouzakri, K.; Meier, D.T.; Prazak, R.; Kerr-Conte, J.; Pattou, F.; Ehses, J.A.; Schuit, F.C.; et al. Free Fatty Acids Induce a Proinflammatory Response in Islets via the Abundantly Expressed Interleukin-1 Receptor I. Endocrinology 2009, 150, 5218–5229. [Google Scholar] [CrossRef] [PubMed]
  103. Lee, Y.S.; Kim, J.-W.; Osborne, O.; Oh, D.Y.; Sasik, R.; Schenk, S.; Chen, A.; Chung, H.; Murphy, A.; Watkins, S.M.; et al. Increased Adipocyte O2 Consumption Triggers HIF-1a, Causing Inflammation and Insulin Resistance in Obesity. Cell 2014, 157, 1339–1352. [Google Scholar] [CrossRef] [PubMed]
  104. Greco, M.; Mirabelli, M.; Tocci, V.; Mamula, Y.; Salatino, A.; Brunetti, F.S.; Dragone, F.; Sicilia, L.; Tripolino, O.; Chiefari, E.; et al. Prothymosin-Alpha, a Novel and Sensitive Biomarker of the Inflammatory and Insulin-Resistant Statuses of Obese Individuals: A Pilot Study Involving Humans. Endocrines 2023, 4, 427–436. [Google Scholar] [CrossRef]
  105. Jais, A.; Brüning, J.C. Hypothalamic Inflammation in Obesity and Metabolic Disease. J. Clin. Investig. 2017, 127, 24–32. [Google Scholar] [CrossRef]
  106. Benomar, Y.; Taouis, M. Molecular Mechanisms Underlying Obesity-Induced Hypothalamic Inflammation and Insulin Resistance: Pivotal Role of Resistin/Tlr4 Pathways. Front. Endocrinol. 2019, 10, 417170. [Google Scholar] [CrossRef]
  107. Whirledge, S.; Cidlowski, J.A. Glucocorticoids, Stress, and Fertility. Minerva Endocrinol. 2010, 35, 109–125. [Google Scholar]
  108. Geer, E.B.; Islam, J.; Buettner, C. Mechanisms of Glucocorticoid-Induced Insulin Resistance: Focus on Adipose Tissue Function and Lipid Metabolism. Endocrinol. Metab. Clin. 2014, 43, 75–102. [Google Scholar] [CrossRef]
  109. Almon, R.R.; Dubois, D.C.; Jin, J.Y.; Jusko, W.J. Temporal Profiling of the Transcriptional Basis for the Development of Corticosteroid-Induced Insulin Resistance in Rat Muscle. J. Endocrinol. 2005, 184, 219–232. [Google Scholar] [CrossRef]
  110. Mazziotti, G.; Gazzaruso, C.; Giustina, A. Diabetes in Cushing Syndrome: Basic and Clinical Aspects. Trends Endocrinol. Metab. 2011, 22, 499–506. [Google Scholar] [CrossRef]
  111. Winer, D.A.; Winer, S.; Dranse, H.J.; Lam, T.K.T. Immunologic Impact of the Intestine in Metabolic Disease. J. Clin. Investig. 2017, 127, 33–42. [Google Scholar] [CrossRef]
  112. Membrez, M.; Blancher, F.; Jaquet, M.; Bibiloni, R.; Cani, P.D.; Burcelin, R.G.; Corthesy, I.; Macé, K.; Chou, C.J. Gut Microbiota Modulation with Norfloxacin and Ampicillin Enhances Glucose Tolerance in Mice. FASEB J. 2008, 22, 2416–2426. [Google Scholar] [CrossRef] [PubMed]
  113. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef]
  114. De La Serre, C.B.; Ellis, C.L.; Lee, J.; Hartman, A.L.; Rutledge, J.C.; Raybould, H.E. Propensity to High-Fat Diet-Induced Obesity in Rats Is Associated with Changes in the Gut Microbiota and Gut Inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, 440–448. [Google Scholar] [CrossRef] [PubMed]
  115. Marotz, C.A.; Zarrinpar, A. Treating Obesity and Metabolic Syndrome with Fecal Microbiota Transplantation. Yale J. Biol. Med. 2016, 89, 383–388. [Google Scholar] [PubMed]
  116. Voigt, J.P.; Fink, H. Serotonin Controlling Feeding and Satiety. Behav. Brain Res. 2015, 277, 14–31. [Google Scholar] [CrossRef] [PubMed]
  117. Volkow, N.D.; Wang, G.J.; Baler, R.D. Reward, Dopamine and the Control of Food Intake: Implications for Obesity. Trends Cogn. Sci. 2011, 15, 37–46. [Google Scholar] [CrossRef]
  118. Lam, D.D.; Heisler, L.K. Serotonin and Energy Balance: Molecular Mechanisms and Implications for Type 2 Diabetes. Expert Rev. Mol. Med. 2007, 9, 1–24. [Google Scholar] [CrossRef]
  119. Stice, E.; Spoor, S.; Ng, J.; Zald, D.H. Relation of Obesity to Consummatory and Anticipatory Food Reward. Physiol. Behav. 2009, 97, 551. [Google Scholar] [CrossRef]
  120. Hoogendoorn, C.J.; Roy, J.F.; Gonzalez, J.S. Shared Dysregulation of Homeostatic Brain-Body Pathways in Depression and Type 2 Diabetes. Curr. Diab Rep. 2017, 17, 90. [Google Scholar] [CrossRef]
  121. Herhaus, B.; Ullmann, E.; Chrousos, G.; Petrowski, K. High/Low Cortisol Reactivity and Food Intake in People with Obesity and Healthy Weight. Transl. Psychiatry 2020, 10, 40. [Google Scholar] [CrossRef]
  122. Incollingo Rodriguez, A.C.; Epel, E.S.; White, M.L.; Standen, E.C.; Seckl, J.R.; Tomiyama, A.J. Hypothalamic-Pituitary-Adrenal Axis Dysregulation and Cortisol Activity in Obesity: A Systematic Review. Psychoneuroendocrinology 2015, 62, 301–318. [Google Scholar] [CrossRef] [PubMed]
  123. Vicennati, V.; Pasqui, F.; Cavazza, C.; Garelli, S.; Casadio, E.; di Dalmazi, G.; Pagotto, U.; Pasquali, R. Cortisol, Energy Intake, and Food Frequency in Overweight/Obese Women. Nutrition 2011, 27, 677–680. [Google Scholar] [CrossRef] [PubMed]
  124. Everson-Rose, S.A.; Lewis, T.T.; Karavolos, K.; Dugan, S.A.; Wesley, D.; Powell, L.H. Depressive Symptoms and Increased Visceral Fat in Middle-Aged Women. Psychosom. Med. 2009, 71, 410–416. [Google Scholar] [CrossRef] [PubMed]
  125. Kahl, K.G.; Schweiger, U.; Pars, K.; Kunikowska, A.; Deuschle, M.; Gutberlet, M.; Lichtinghagen, R.; Bleich, S.; Hüper, K.; Hartung, D. Adrenal Gland Volume, Intra-Abdominal and Pericardial Adipose Tissue in Major Depressive Disorder. Psychoneuroendocrinology 2015, 58, 1–8. [Google Scholar] [CrossRef] [PubMed]
  126. Tsai, S.F.; Wu, H.T.; Chen, P.C.; Chen, Y.W.; Yu, M.; Tzeng, S.F.; Wu, P.H.; Chen, P.S.; Kuo, Y.M. Stress Aggravates High-Fat-Diet-Induced Insulin Resistance via a Mechanism That Involves the Amygdala and Is Associated with Changes in Neuroplasticity. Neuroendocrinology 2018, 107, 147–157. [Google Scholar] [CrossRef]
  127. Karczewska-Kupczewska, M.; Straczkowski, M.; Adamska, A.; Nikołajuk, A.; Otziomek, E.; Górska, M.; Kowalska, I. Decreased Serum Brain-Derived Neurotrophic Factor Concentration in Young Nonobese Subjects with Low Insulin Sensitivity. Clin. Biochem. 2011, 44, 817–820. [Google Scholar] [CrossRef]
  128. Yamanaka, M.; Itakura, Y.; Ono-Kishino, M.; Tsuchida, A.; Nakagawa, T.; Taiji, M. Intermittent Administration of Brain-Derived Neurotrophic Factor (BDNF) Ameliorates Glucose Metabolism and Prevents Pancreatic Exhaustion in Diabetic Mice. J. Biosci. Bioeng. 2008, 105, 395–402. [Google Scholar] [CrossRef]
  129. Pelleymounter, M.A.; Cullen, M.J.; Wellman, C.L. Characteristics of BDNF-Induced Weight Loss. Exp. Neurol. 1995, 131, 229–238. [Google Scholar] [CrossRef]
  130. Nonomura, T.; Tsuchida, A.; Ono-Kishino, M.; Nakagawa, T.; Taiji, M.; Noguchi, H. Brain-Derived Neurotrophic Factor Regulates Energy Expenditure Through the Central Nervous in Obese Diabetic Mice. J. Diabetes Res. 2001, 2, 201–209. [Google Scholar] [CrossRef]
  131. Tsai, S.F.; Hsu, P.L.; Chen, Y.W.; Hossain, M.S.; Chen, P.C.; Tzeng, S.F.; Chen, P.S.; Kuo, Y.M. High-Fat Diet Induces Depression-like Phenotype via Astrocyte-Mediated Hyperactivation of Ventral Hippocampal Glutamatergic Afferents to the Nucleus Accumbens. Mol. Psychiatry 2022, 27, 4372–4384. [Google Scholar] [CrossRef]
  132. Davarpanah, M.; Shokri-mashhadi, N.; Ziaei, R.; Saneei, P. A Systematic Review and Meta-Analysis of Association between Brain-Derived Neurotrophic Factor and Type 2 Diabetes and Glycemic Profile. Sci. Rep. 2021, 11, 13773. [Google Scholar] [CrossRef]
  133. Tonra, J.R.; Ono, M.; Liu, X.; Garcia, K.; Jackson, C.; Yancopoulos, G.D.; Wiegand, S.J.; Wong, V. Brain-Derived Neurotrophic Factor Improves Blood Glucose Control and Alleviates Fasting Hyperglycemia in C57BLKS-Lepr(Db)/Lepr(Db) Mice. Diabetes 1999, 48, 588–594. [Google Scholar] [CrossRef] [PubMed]
  134. Ukena, S.N.; Singh, A.; Dringenberg, U.; Engelhardt, R.; Seidler, U.; Hansen, W.; Bleich, A.; Bruder, D.; Franzke, A.; Rogler, G.; et al. Probiotic Escherichia Coli Nissle 1917 Inhibits Leaky Gut by Enhancing Mucosal Integrity. PLoS ONE 2007, 2, e1308. [Google Scholar] [CrossRef] [PubMed]
  135. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet–Induced Obesity and Diabetes in Mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  136. Cao, S.; Zhang, Q.; Wang, C.C.; Wu, H.; Jiao, L.; Hong, Q.; Hu, C. LPS Challenge Increased Intestinal Permeability, Disrupted Mitochondrial Function and Triggered Mitophagy of Piglets. Innate Immun. 2018, 24, 221–230. [Google Scholar] [CrossRef]
  137. Umirah, F.; Neoh, C.F.; Ramasamy, K.; Lim, S.M. Differential Gut Microbiota Composition between Type 2 Diabetes Mellitus Patients and Healthy Controls: A Systematic Review. Diabetes Res. Clin. Pract. 2021, 173, 108689. [Google Scholar] [CrossRef]
  138. Chen, Z.; Radjabzadeh, D.; Chen, L.; Kurilshikov, A.; Kavousi, M.; Ahmadizar, F.; Ikram, M.A.; Uitterlinden, A.G.; Zhernakova, A.; Fu, J.; et al. Association of Insulin Resistance and Type 2 Diabetes with Gut Microbial Diversity: A Microbiome-Wide Analysis from Population Studies. JAMA Netw. Open 2021, 4, e2118811. [Google Scholar] [CrossRef]
  139. Stuart, M.J.; Baune, B.T. Depression and Type 2 Diabetes: Inflammatory Mechanisms of a Psychoneuroendocrine Co-Morbidity. Neurosci. Biobehav. Rev. 2012, 36, 658–676. [Google Scholar] [CrossRef]
  140. Nguyen, M.M.; Perlman, G.; Kim, N.; Wu, C.Y.; Daher, V.; Zhou, A.; Mathers, E.H.; Anita, N.Z.; Lanctôt, K.L.; Herrmann, N.; et al. Depression in Type 2 Diabetes: A Systematic Review and Meta-Analysis of Blood Inflammatory Markers. Psychoneuroendocrinology 2021, 134, 105448. [Google Scholar] [CrossRef]
  141. O’Connor, J.C.; Satpathy, A.; Hartman, M.E.; Horvath, E.M.; Kelley, K.W.; Dantzer, R.; Johnson, R.W.; Freund, G.G. IL-1β-Mediated Innate Immunity Is Amplified in the Db/Db Mouse Model of Type 2 Diabetes. J. Immunol. 2005, 174, 4991–4997. [Google Scholar] [CrossRef]
  142. Aswar, U.; Chepurwar, S.; Shintre, S.; Aswar, M. Telmisartan Attenuates Diabetes Induced Depression in Rats. Pharmacol. Rep. 2017, 69, 358–364. [Google Scholar] [CrossRef] [PubMed]
  143. Bogush, M.; Heldt, N.A.; Persidsky, Y. Blood Brain Barrier Injury in Diabetes: Unrecognized Effects on Brain and Cognition. J. Neuroimmune Pharmacol. 2017, 12, 593–601. [Google Scholar] [CrossRef] [PubMed]
  144. Qiao, J.; Lawson, C.M.; Rentrup, K.F.G.; Kulkarni, P.; Ferris, C.F. Evaluating Blood-Brain Barrier Permeability in a Rat Model of Type 2 Diabetes. J. Transl. Med. 2020, 18, 256. [Google Scholar] [CrossRef]
  145. Moylan, S.; Berk, M.; Dean, O.M.; Samuni, Y.; Williams, L.J.; O’Neil, A.; Hayley, A.C.; Pasco, J.A.; Anderson, G.; Jacka, F.N.; et al. Oxidative & Nitrosative Stress in Depression: Why so Much Stress? Neurosci. Biobehav. Rev. 2014, 45, 46–62. [Google Scholar] [CrossRef] [PubMed]
  146. Haroon, E.; Miller, A.H.; Sanacora, G. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology 2017, 42, 193–215. [Google Scholar] [CrossRef]
  147. Autry, A.E.; Monteggia, L.M. Brain-Derived Neurotrophic Factor and Neuropsychiatric Disorders. Pharmacol. Rev. 2012, 64, 238–258. [Google Scholar] [CrossRef]
  148. Grillo, C.A.; Piroli, G.G.; Kaigler, K.F.; Wilson, S.P.; Wilson, M.A.; Reagan, L.P. Downregulation of Hypothalamic Insulin Receptor Expression Elicits Depressive-like Behaviors in Rats. Behav. Brain Res. 2011, 222, 230–235. [Google Scholar] [CrossRef]
  149. Cui, X.; Zuo, P.; Zhang, Q.; Li, X.; Hu, Y.; Long, J.; Packer, L.; Liu, J. Chronic Systemic D-Galactose Exposure Induces Memory Loss, Neurodegeneration, and Oxidative Damage in Mice: Protective Effects of R-α-Lipoic Acid. J. Neurosci. Res. 2006, 83, 1584–1590. [Google Scholar] [CrossRef]
  150. Shih, R.H.; Wang, C.Y.; Yang, C.M. NF-KappaB Signaling Pathways in Neurological Inflammation: A Mini Review. Front. Mol. Neurosci. 2015, 8, 165218. [Google Scholar] [CrossRef]
  151. Khansari, N.; Shakiba, Y.; Mahmoudi, M. Chronic Inflammation and Oxidative Stress as a Major Cause of Age-Related Diseases and Cancer. Recent. Pat. Inflamm. Allergy Drug Discov. 2009, 3, 73–80. [Google Scholar] [CrossRef]
  152. Bartolomucci, A.; Leopardi, R. Stress and Depression: Preclinical Research and Clinical Implications. PLoS ONE 2009, 4, e4265. [Google Scholar] [CrossRef] [PubMed]
  153. Cosarderelioglu, C.; Nidadavolu, L.S.; George, C.J.; Oh, E.S.; Bennett, D.A.; Walston, J.D.; Abadir, P.M. Brain Renin–Angiotensin System at the Intersect of Physical and Cognitive Frailty. Front. Neurosci. 2020, 14, 586314. [Google Scholar] [CrossRef] [PubMed]
  154. Jacobson, L.; Sapolsky, R. The Role of the Hippocampus in Feedback Regulation of the Hypothalamic-Pituitary-Adrenocortical Axis. Endocr. Rev. 1991, 12, 118–134. [Google Scholar] [CrossRef] [PubMed]
  155. Wanrooy, B.J.; Kumar, K.P.; Wen, S.W.; Qin, C.X.; Ritchie, R.H.; Wong, C.H.Y. Distinct Contributions of Hyperglycemia and High-Fat Feeding in Metabolic Syndrome-Induced Neuroinflammation. J. Neuroinflamm. 2018, 15, 293. [Google Scholar] [CrossRef]
  156. Chan, O.; Inouye, K.; Akirav, E.; Park, E.; Riddell, M.C.; Vranic, M.; Matthews, S.G. Insulin Alone Increases Hypothalamo-Pituitary-Adrenal Activity, and Diabetes Lowers Peak Stress Responses. Endocrinology 2005, 146, 1382–1390. [Google Scholar] [CrossRef]
  157. Chesnokova, V.; Pechnick, R.N.; Wawrowsky, K. Chronic Peripheral Inflammation, Hippocampal Neurogenesis, and Behavior. Brain Behav. Immun. 2016, 58, 1–8. [Google Scholar] [CrossRef]
  158. Bliss, T.V.P.; Collingridge, G.L. A Synaptic Model of Memory: Long-Term Potentiation in the Hippocampus. Nature 1993, 361, 31–39. [Google Scholar] [CrossRef]
  159. Rafalski, V.A.; Brunet, A. Energy Metabolism in Adult Neural Stem Cell Fate. Prog. Neurobiol. 2011, 93, 182–203. [Google Scholar] [CrossRef]
  160. Shafiei, F.; Salari-Moghaddam, A.; Larijani, B.; Esmaillzadeh, A. Adherence to the Mediterranean Diet and Risk of Depression: A Systematic Review and Updated Meta-Analysis of Observational Studies. Nutr. Rev. 2019, 77, 230–239. [Google Scholar] [CrossRef]
  161. Tolkien, K.; Bradburn, S.; Murgatroyd, C. An Anti-Inflammatory Diet as a Potential Intervention for Depressive Disorders: A Systematic Review and Meta-Analysis. Clin. Nutr. 2019, 38, 2045–2052. [Google Scholar] [CrossRef]
  162. Iguacel, I.; Huybrechts, I.; Moreno, L.A.; Michels, N. Vegetarianism and Veganism Compared with Mental Health and Cognitive Outcomes: A Systematic Review and Meta-Analysis. Nutr. Rev. 2021, 79, 361–381. [Google Scholar] [CrossRef] [PubMed]
  163. Jacka, F.N.; O’Neil, A.; Opie, R.; Itsiopoulos, C.; Cotton, S.; Mohebbi, M.; Castle, D.; Dash, S.; Mihalopoulos, C.; Chatterton, M.; et al. A Randomised Controlled Trial of Dietary Improvement for Adults with Major Depression (the “SMILES” Trial). BMC Med. 2017, 15, 23. [Google Scholar] [CrossRef] [PubMed]
  164. Bayes, J.; Schloss, J.; Sibbritt, D. The Effect of a Mediterranean Diet on the Symptoms of Depression in Young Males (the “AMMEND: A Mediterranean Diet in MEN with Depression” Study): A Randomized Controlled Trial. Am. J. Clin. Nutr. 2022, 116, 572–580. [Google Scholar] [CrossRef] [PubMed]
  165. Toi, P.L.; Anothaisintawee, T.; Chaikledkaew, U.; Briones, J.R.; Reutrakul, S.; Thakkinstian, A. Preventive Role of Diet Interventions and Dietary Factors in Type 2 Diabetes Mellitus: An Umbrella Review. Nutrients 2020, 12, 2722. [Google Scholar] [CrossRef] [PubMed]
  166. Zeraattalab-Motlagh, S.; Jayedi, A.; Shab-Bidar, S. Mediterranean Dietary Pattern and the Risk of Type 2 Diabetes: A Systematic Review and Dose–Response Meta-Analysis of Prospective Cohort Studies. Eur. J. Nutr. 2022, 61, 1735–1748. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, Y.; Liu, B.; Han, H.; Hu, Y.; Zhu, L.; Rimm, E.B.; Hu, F.B.; Sun, Q. Associations between Plant-Based Dietary Patterns and Risks of Type 2 Diabetes, Cardiovascular Disease, Cancer, and Mortality—A Systematic Review and Meta-Analysis. Nutr. J. 2023, 22, 46. [Google Scholar] [CrossRef]
  168. de Carvalho, G.B.; Dias-Vasconcelos, N.L.; Santos, R.K.F.; Brandão-Lima, P.N.; da Silva, D.G.; Pires, L.V. Effect of Different Dietary Patterns on Glycemic Control in Individuals with Type 2 Diabetes Mellitus: A Systematic Review. Crit. Rev. Food Sci. Nutr. 2020, 60, 1999–2010. [Google Scholar] [CrossRef]
  169. Chiavaroli, L.; Lee, D.; Ahmed, A.; Cheung, A.; Khan, T.A.; Blanco, S.; Mejia; Mirrahimi, A.; Jenkins, D.J.A.; Livesey, G.; et al. Effect of Low Glycaemic Index or Load Dietary Patterns on Glycaemic Control and Cardiometabolic Risk Factors in Diabetes: Systematic Review and Meta-Analysis of Randomised Controlled Trials. BMJ 2021, 374, 16. [Google Scholar] [CrossRef]
  170. Koelman, L.; Egea Rodrigues, C.; Aleksandrova, K. Effects of Dietary Patterns on Biomarkers of Inflammation and Immune Responses: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2022, 13, 101–115. [Google Scholar] [CrossRef]
  171. Haghighatdoost, F.; Hariri, M. The Effect of Green Tea on Inflammatory Mediators: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Phytother. Res. 2019, 33, 2274–2287. [Google Scholar] [CrossRef]
  172. Fallah, A.A.; Sarmast, E.; Fatehi, P.; Jafari, T. Impact of Dietary Anthocyanins on Systemic and Vascular Inflammation: Systematic Review and Meta-Analysis on Randomised Clinical Trials. Food Chem. Toxicol. 2020, 135, 110922. [Google Scholar] [CrossRef]
  173. Tosatti, J.A.G.; Alves, M.T.; Cândido, A.L.; Reis, F.M.; Araújo, V.E.; Gomes, K.B. Influence of N-3 Fatty Acid Supplementation on Inflammatory and Oxidative Stress Markers in Patients with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. Br. J. Nutr. 2021, 125, 657–668. [Google Scholar] [CrossRef] [PubMed]
  174. Norde, M.M.; Collese, T.S.; Giovannucci, E.; Rogero, M.M. A Posteriori Dietary Patterns and Their Association with Systemic Low-Grade Inflammation in Adults: A Systematic Review and Meta-Analysis. Nutr. Rev. 2021, 79, 331–350. [Google Scholar] [CrossRef] [PubMed]
  175. Ruiz-Núñez, B.; Dijck-Brouwer, D.A.J.; Muskiet, F.A.J. The Relation of Saturated Fatty Acids with Low-Grade Inflammation and Cardiovascular Disease. J. Nutr. Biochem. 2016, 36, 1–20. [Google Scholar] [CrossRef] [PubMed]
  176. Rocha, D.M.; Caldas, A.P.; Oliveira, L.L.; Bressan, J.; Hermsdorff, H.H. Saturated Fatty Acids Trigger TLR4-Mediated Inflammatory Response. Atherosclerosis 2016, 244, 211–215. [Google Scholar] [CrossRef] [PubMed]
  177. Rogero, M.M.; Calder, P.C. Obesity, Inflammation, Toll-Like Receptor 4 and Fatty Acids. Nutrients 2018, 10, 432. [Google Scholar] [CrossRef]
  178. Delarue, J.; Matzinger, O.; Binnert, C.; Schneiter, P.; Chioléro, R.; Tappy, L. Fish Oil Prevents the Adrenal Activation Elicited by Mental Stress in Healthy Men. Diabetes Metab. 2003, 29, 289–295. [Google Scholar] [CrossRef]
  179. Tsang, C.; Hodgson, L.; Bussu, A.; Farhat, G.; Al-Dujaili, E. Effect of Polyphenol-Rich Dark Chocolate on Salivary Cortisol and Mood in Adults. Antioxidants 2019, 8, 149. [Google Scholar] [CrossRef]
  180. Al-Dujaili, E.A.S.; Good, G.; Tsang, C.; Al-Dujaili, E.; Tsang, C. Consumption of Pomegranate Juice Attenuates Exercise-Induced Oxidative Stress, Blood Pressure and Urinary Cortisol/Cortisone Ratio in Human Adults. EC Nutr. 2016, 4, 982–995. [Google Scholar]
  181. Whittaker, J.; Harris, M. Low-Carbohydrate Diets and Men’s Cortisol and Testosterone: Systematic Review and Meta-Analysis. Nutr. Health 2022, 28, 543–554. [Google Scholar] [CrossRef]
  182. Al-Dujaili, E.A.S.; Ashmore, S.; Tsang, C. A Short Study Exploring the Effect of the Glycaemic Index of the Diet on Energy Intake and Salivary Steroid Hormones. Nutrients 2019, 11, 260. [Google Scholar] [CrossRef] [PubMed]
  183. Park, H.R.; Park, M.; Choi, J.; Park, K.Y.; Chung, H.Y.; Lee, J. A High-Fat Diet Impairs Neurogenesis: Involvement of Lipid Peroxidation and Brain-Derived Neurotrophic Factor. Neurosci. Lett. 2010, 482, 235–239. [Google Scholar] [CrossRef] [PubMed]
  184. Rendeiro, C.; Vauzour, D.; Rattray, M.; Waffo-Téguo, P.; Mérillon, J.M.; Butler, L.T.; Williams, C.M.; Spencer, J.P.E. Dietary Levels of Pure Flavonoids Improve Spatial Memory Performance and Increase Hippocampal Brain-Derived Neurotrophic Factor. PLoS ONE 2013, 8, e63535. [Google Scholar] [CrossRef] [PubMed]
  185. Ziaei, S.; Mohammadi, S.; Hasani, M.; Morvaridi, M.; Belančić, A.; Daneshzad, E.; Saleh, S.A.K.; Adly, H.M.; Heshmati, J. A Systematic Review and Meta-Analysis of the Omega-3 Fatty Acids Effects on Brain-Derived Neurotrophic Factor (BDNF). Nutr. Neurosci. 2023, 27, 715–725. [Google Scholar] [CrossRef]
  186. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [PubMed]
  187. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet Rapidly and Reproducibly Alters the Human Gut Microbiome. Nature 2013, 505, 559–563. [Google Scholar] [CrossRef]
  188. Bolte, L.A.; Vich Vila, A.; Imhann, F.; Collij, V.; Gacesa, R.; Peters, V.; Wijmenga, C.; Kurilshikov, A.; E Campmans-Kuijpers, M.J.; Fu, J.; et al. Gut Microbiota Long-Term Dietary Patterns Are Associated with pro-Inflammatory and Anti-Inflammatory Features of the Gut Microbiome. Gut 2021, 70, 1287–1298. [Google Scholar] [CrossRef]
  189. Turpin, W.; Dong, M.; Sasson, G.; Raygoza Garay, J.A.; Espin-Garcia, O.; Lee, S.H.; Neustaeter, A.; Smith, M.I.; Leibovitzh, H.; Guttman, D.S.; et al. Mediterranean-Like Dietary Pattern Associations with Gut Microbiome Composition and Subclinical Gastrointestinal Inflammation. Gastroenterology 2022, 163, 685–698. [Google Scholar] [CrossRef]
  190. Russo-Neustadt, A.A.; Beard, R.C.; Huang, Y.M.; Cotman, C.W. Physical Activity and Antidepressant Treatment Potentiate the Expression of Specific Brain-Derived Neurotrophic Factor Transcripts in the Rat Hippocampus. Neuroscience 2000, 101, 305–312. [Google Scholar] [CrossRef]
  191. Mathur, N.; Pedersen, B.K. Exercise as a Mean to Control Low-Grade Systemic Inflammation. Mediat. Inflamm. 2008, 2008, 109502. [Google Scholar] [CrossRef]
  192. Xiao, K.; Luo, Y.; Liang, X.; Tang, J.; Wang, J.; Xiao, Q.; Qi, Y.; Li, Y.; Zhu, P.; Yang, H.; et al. Beneficial Effects of Running Exercise on Hippocampal Microglia and Neuroinflammation in Chronic Unpredictable Stress-Induced Depression Model Rats. Transl. Psychiatry 2021, 11, 461. [Google Scholar] [CrossRef] [PubMed]
  193. Jodeiri Farshbaf, M.; Ghaedi, K.; Megraw, T.L.; Curtiss, J.; Shirani Faradonbeh, M.; Vaziri, P.; Nasr-Esfahani, M.H. Does PGC1α/FNDC5/BDNF Elicit the Beneficial Effects of Exercise on Neurodegenerative Disorders? Neuromol. Med. 2015, 18, 1–15. [Google Scholar] [CrossRef] [PubMed]
  194. Handschin, C.; Cheol, S.C.; Chin, S.; Kim, S.; Kawamori, D.; Kurpad, A.J.; Neubauer, N.; Hu, J.; Mootha, V.K.; Kim, Y.B.; et al. Abnormal Glucose Homeostasis in Skeletal Muscle–Specific PGC-1α Knockout Mice Reveals Skeletal Muscle–Pancreatic β Cell Crosstalk. J. Clin. Investig. 2007, 117, 3463–3474. [Google Scholar] [CrossRef] [PubMed]
  195. Lumb, A. Diabetes and Exercise. Clin. Med. 2014, 14, 673. [Google Scholar] [CrossRef] [PubMed]
  196. Lincoln, K.D. Social Stress, Obesity, and Depression among Women: Clarifying the Role of Physical Activity. Ethn. Health 2019, 24, 662–678. [Google Scholar] [CrossRef] [PubMed]
  197. Chen, X.; Sun, X.; Wang, C.; He, H. Effects of Exercise on Inflammatory Cytokines in Patients with Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Oxid. Med. Cell Longev. 2020, 2020, 660557. [Google Scholar] [CrossRef]
  198. Little, J.P.; Thyfault, J.P. Modification of Insulin Sensitivity and Glycemic Control by Activity and Exercise. Med. Sci. Sports Exerc. 2013, 45, 1868–1877. [Google Scholar] [CrossRef]
  199. Pedersen, B.K. The Diseasome of Physical Inactivity—And the Role of Myokines in Muscle–Fat Cross Talk. J. Physiol. 2009, 587, 5559–5568. [Google Scholar] [CrossRef] [PubMed]
  200. Rosenkilde, M.; Nordby, P.; Stallknecht, B. Maintenance of Improvements in Fitness and Fatness 1 Year after a 3-Month Lifestyle Intervention in Overweight Men. Eur. J. Clin. Nutr. 2016, 70, 1212–1214. [Google Scholar] [CrossRef]
  201. Rinnov, A.; Yfanti, C.; Nielsen, S.; Åkerström, T.C.A.; Peijs, L.; Zankari, A.; Fischer, C.P.; Pedersen, B.K. Endurance Training Enhances Skeletal Muscle Interleukin-15 in Human Male Subjects. Endocrine 2014, 45, 271–278. [Google Scholar] [CrossRef]
  202. De Oliveira, V.N.; Bessa, A.; Jorge, M.L.M.P.; da Silva Oliveira, R.J.; de Mello, M.T.; de Agostini, G.G.; Jorge, P.T.; Espindola, F.S. The Effect of Different Training Programs on Antioxidant Status, Oxidative Stress, and Metabolic Control in Type 2 Diabetes. Appl. Physiol. Nutr. Metab. 2012, 37, 334–344. [Google Scholar] [CrossRef] [PubMed]
  203. Yang, Z.; Scott, C.A.; Mao, C.; Tang, J.; Farmer, A.J. Resistance Exercise versus Aerobic Exercise for Type 2 Diabetes: A Systematic Review and Meta-Analysis. Sports Med. 2014, 44, 487–499. [Google Scholar] [CrossRef] [PubMed]
  204. Barazzoni, R.; Gortan Cappellari, G.; Ragni, M.; Nisoli, E. Insulin Resistance in Obesity: An Overview of Fundamental Alterations. Eat. Weight Disord. 2018, 23, 149–157. [Google Scholar] [CrossRef] [PubMed]
Table 1. Characteristics of acute versus chronic inflammation.
Table 1. Characteristics of acute versus chronic inflammation.
Acute InflammationChronic Inflammation
OnsetImmediateDelayed
DurationDays to weeksMonths to years
Cause
-
Pathogens
-
Tissue damage
-
Persistent foreign bodies
-
Persistent acute inflammation
-
Autoimmune reactions
Primary cells
-
Neutrophils
-
Monocytes
-
Macrophages
-
Lymphocytes
-
Fibroblasts
Primary meditators
-
Vasoactive amines
-
Eicosanoids
-
Cytokines
-
Growth factors
-
Hydrolytic enzymes
-
Reactive oxygen species
Tissue damageMinimal
(promotes tissue repair)
Prolonged
(promotes tissue damage)
Outcomes
-
Resolution
-
Accesses formation
-
Chronic inflammation
-
Tissue fibrosis and destruction
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bodnaruc, A.M.; Roberge, M.; Giroux, I.; Aguer, C. The Bidirectional Link between Major Depressive Disorder and Type 2 Diabetes: The Role of Inflammation. Endocrines 2024, 5, 478-500. https://doi.org/10.3390/endocrines5040035

AMA Style

Bodnaruc AM, Roberge M, Giroux I, Aguer C. The Bidirectional Link between Major Depressive Disorder and Type 2 Diabetes: The Role of Inflammation. Endocrines. 2024; 5(4):478-500. https://doi.org/10.3390/endocrines5040035

Chicago/Turabian Style

Bodnaruc, Alexandra M., Mathilde Roberge, Isabelle Giroux, and Céline Aguer. 2024. "The Bidirectional Link between Major Depressive Disorder and Type 2 Diabetes: The Role of Inflammation" Endocrines 5, no. 4: 478-500. https://doi.org/10.3390/endocrines5040035

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop