Next Article in Journal
Update on the Phylodynamics of SADS-CoV
Next Article in Special Issue
Gut Hormones as Potential Therapeutic Targets or Biomarkers of Response in Depression: The Case of Motilin
Previous Article in Journal
Feasibility Study on the Use of Fly Maggots (Musca domestica) as Carriers to Inhibit Shrimp White Spot Syndrome
Previous Article in Special Issue
Zebrafish as an Animal Model for Testing Agents with Antidepressant Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Metabolic Dysfunction for the Treatment of Mood Disorders: Review of the Evidence

by
Brett D. M. Jones
1,2,
Salman Farooqui
3,
Stefan Kloiber
1,2,
Muhammad Omair Husain
1,2,
Benoit H. Mulsant
1,2 and
Muhammad Ishrat Husain
1,2,*
1
Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
2
Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada
3
Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
*
Author to whom correspondence should be addressed.
Submission received: 1 July 2021 / Revised: 23 July 2021 / Accepted: 26 July 2021 / Published: 11 August 2021
(This article belongs to the Special Issue Novel Pharmacological Approaches to the Treatment of Depression)

Abstract

:
Major depressive disorder (MDD) and bipolar disorder (BD) are often chronic with many patients not responding to available treatments. As these mood disorders are frequently associated with metabolic dysfunction, there has been increased interest in novel treatments that would target metabolic pathways. The objectives of this scoping review were to synthesize evidence on the impact on mood symptoms of lipid lowering agents and anti-diabetics drugs, while also reviewing current knowledge on the association between mood disorders and dyslipidemia or hyperglycemia. We propose that metabolic dysfunction is prevalent in both MDD and BD and it may contribute to the development of these disorders through a variety of pathophysiological processes including inflammation, brain structural changes, hormonal alterations, neurotransmitter disruptions, alteration on brain cholesterol, central insulin resistance, and changes in gut microbiota. Current evidence is conflicting on the use of statins, polyunsaturated fatty acids, thiazolidinediones, glucagon-like peptide agonists, metformin, or insulin for the treatment of MDD and BD. Given the paucity of high-quality randomized controlled trials, additional studies are needed before any of these medications can be repurposed in routine clinical practice. Future trials need to enrich patient recruitment, include evaluations of mechanism of action, and explore differential effects on specific symptom domains such as anhedonia, suicidality, and cognition.

1. Introduction

Major depressive disorder (MDD) and bipolar disorder (BD) are a leading cause of disability worldwide and result in substantial impairment in functioning for impacted individuals [1]. Current pharmacotherapies for mood disorders include antidepressants, mood-stabilizers, and antipsychotics. Although a beneficial option for many patients, a substantial portion of patients do not respond or reach remission [2,3]. Since many pharmacological approaches for mood disorders target similar biological mechanisms, such as monoamine neurotransmission, it is important to develop novel and repurposed pharmacotherapies that target differential pathological targets that may be implicated in mood disorders.
Given the high prevalence of treatment resistance in mood disorders, investigating the repurposing of medications targeting novel biological targets is necessary. The potential for the use of repurposed medication has been shown in previous work that has attempted to target putative pathological processes associated with mood disorders, such as an activated immune system [4]. Similarly, there has been increasing interest in exploring altered metabolic function in mood disorders as a potential treatment target. In recent years, several studies have evaluated metabolic modulating agents, such as lipid lowering agents and anti-diabetic drugs, for the treatment of mood disorders. The objective of this review is to summarize evidence for associations between mood disorders and metabolic dysfunction (i.e., dyslipidemia and hyperglycemia.) and to provide a scoping review on evidence from clinical trials of lipid lowering agents and anti-diabetics drugs for the treatment of mood disorders.

2. Materials and Methods

Relevant literature related to the topic of the current review was obtained by searching Medline using PubMed for reviews and primary studies of the associations between metabolic function and mood disorders (MDD or BD) using the following search terms (depress* or bipolar) and (metabolic function or metabolic dysfunction). Reference lists of related reviews were searched for additional studies. To identify clinical trials of metabolic modulating agents as monotherapy or augmentation treatments in MDD or BD, the following search terms were used: (antidiabet* OR lipid OR hyperglycemic OR statin OR insulin OR thiazolidinedione* OR glucagon-like peptide OR GLP OR metformin OR Poly Unsaturated Fatty Acid OR PUFA OR eicosapentaenoic acid EPA OR DHA OR Docosahexaenoic acid or omega-3 OR omega-6) and (depress* or bipolar) and (trial OR RCT)).
Studies selected were randomised controlled trials (RCTs), cluster RCTs, or cross-over trials.
When a significant number of RCTs were available for one specific class of metabolic-targeting medication, we reviewed recent meta-analyses from reputable peer-reviewed journals that followed standard (e.g., PRISMA) reporting guidelines. When no RCT was available, we reviewed open-label trials or cohort studies. Participants had to meet criteria for MDD or BD according to international classification of disease (ICD) ICD 10, diagnostic statistical manual of mental disorders (DSM)-IV or DSM-5 [5,6]. We only reviewed English language studies.

3. Results

3.1. Evidence for Associations between Metabolic Dysfunction and Mood Disorders

Individuals with MDD have a 4-fold higher rate of early death than healthy controls, in part due to a higher prevalence of metabolic disturbances such as dyslipidemia and hyperglycemia [7,8]. The association between metabolic disturbances and mood disorders appeared to be bidirectional [9]. In longitudinal studies, several metabolic dysregulations have been associated with sustained depressive states over time [10,11,12].One-year prior to diagnosis, patients with BD present with higher rates of hypertension, cardiovascular disease, gastrointestinal disease, respiratory diseases, metabolic disease, and musculoskeletal disease than the general population [13]. The following section will review specific abnormalities in lipid and glucose regulation.

3.1.1. Lipid Regulation

In a large cohort study (N = 766,427), when compared to controls, patients with MDD had both a higher prevalence of hyperlipidemia (14.4% vs. 7.9%, OR: 1.67; 95%CI: 1.53–1.82) and a higher incidence (3.6% vs. 2.6%, RR: 1.35; 95%CI: 1.24–1.47) [14]. In the same cohort, the prevalence of hyperlipidemia for BD was 13.5% (OR: 1.75, 95%CI: 1.52–2.02) and the incidence was 4.37% (RR: 1.66, 95%CI: 1.47–1.87) [15]. In a separate retrospective cohort study (N = 26,852), patients with newly diagnosed hyperlipidemia had a higher risk for MDD compared to healthy controls (HR: 1.64, 95%CI, 1.55–1.74) [16]. In a large cohort of men (N = 29,133) followed for 5–8 years, baseline low cholesterol was associated with a high relative risk of admission to hospital for MDD or death by suicide [17]. Lower levels of cholesterol have also frequently been associated with suicidality [18]. Alexithymia, which is an inability to recognize emotions frequently seen in MDD, has also been associated with lower high-density lipoprotein (HDL) and suicidality [19]. In a study in older adults, baseline low HDL was associated with a higher risk of MDD within 5 years [20]. In 1040 women, the OR of prevalence of MDD or high level of depressive symptoms was 1.5 for those with low HDL; in 752 men, those with low low-density lipoprotein (LDL) had a 2.0 OR of prevalence and subsequent depression [21]. In a population study, compared to patients with schizophrenia, those with BD had an increased OR of hyperlipidemia 1 year prior to receiving their respective psychiatric diagnosis (OR: 1.22 95%CI: 1.09–1.36) [13].
Alterations in polyunsaturated fatty acids (PUFA)- e.g., omega-3 PUFA, omega-6 PUFA, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA)- have also been reported in patients with MDD. The rates of MDD are lower in countries with a high fish consumption and those with lower PUFA consumption have higher rates of depressive symptoms [22,23]. Lower fish intake has been associated with higher rates of suicidality and higher prevalence of BD 1 or 2 [24,25]. Patients with MDD have lower omega-3 PUFA levels than healthy controls [26]. Furthermore, patients with MDD have altered ratio of omega-PUFA with higher amounts of omega-6 vs omega-3 PUFA [27]. In a meta-analysis of patients with BD 1 (n = 118) vs healthy controls (n = 147), patients with BD had significant lower levels of DHA and EPA [28]. There may also be a link between low EPA + DHA erythrocyte levels and risk of developing BD depression or mania in persons at high-risk of BD [29]. Taken together, meta-analyses have found, with high heterogeneity, that MDD is associated with increased total cholesterol, triglycerides, low LDL, low HDL, and low omega-3 PUFA [30,31,32,33].

3.1.2. Glucose Regulation

Clinically significant depressive symptoms are prevalent in up to 30% of patients with diabetes mellitus (DM) [34]. The prevalence of MDD in diabetic patients is twice that of non-diabetic patients (OR: 2.0, 95%CI: 1.8–2.2) and can be as high as 10% [35]. In a large meta-analysis, patients with MDD (N = 154,366) had a higher risk of type 2 DM than healthy persons (N = 2,098,063; RR: 1.49; 95%CI: 1.29–1.72) [36]. In a separate longitudinal study, depressive symptoms at baseline were associated with increased incident type 2 DM over a 3-year follow-up period [9]. In a recent study, hyperglycemia but not insulin resistance, irrespective of diagnosis of DM, was associated with an increased risk of developing depressive symptoms over 4 years [37]. In a cohort of 123,232 patients with DM and 1,933,218 controls, women with DM had 2.55 times higher risk of being diagnosed with MDD than women without DM (95%CI: 2.48–2.62) while the increased risk in men was not as marked (OR: 1.85, 95%CI: 1.80–1.91).
As with MDD, a strong association has been found between BD and DM. DM rates have been reported to be up three times as high in patients with BD as in healthy controls [38]. In a Taiwanese population study, prevalence of DM in patients with BD was higher than in the general population (10.77% vs. 5.57%, OR: 2.01; 99% CI: 1.64–2.48) [39]. Similarly, in a Swedish population study BD was more common in patients with DM (OR: 1.71, 95%CI: 1.54–1.91) than in non-diabetic persons [40]. Another study reported than in the 1 year prior to diagnosis of BD, the OR of presence of DM compared to the general population was 1.27 (95%CI: 1.12–1.45) and the 3-year pre diagnosis OR was 1.53 (95%CI: 1.31–1.79) [13].

3.2. Evidence on the Efficacy of Metabolic Modulating Agents for the Treatment of Mood Disorders

Based on our search, we screened 3923 articles and identified 1 open-label study, 19 RCTs, and 4 meta-analyses for this review (Table 1 and Table 2). Included medications were statins, PUFA, thiazolidinediones (TZD), glucagon-like peptide 1 (GLP-1) agonists, metformin (MET), and insulin.

3.2.1. Lipid Lowering Agents for Mood Disorders

Statins for the Treatment of MDD

Statins are 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors that are primarily used to treat dyslipidemia [41]. The first RCT of statins in mood disorders investigated the efficacy of lovastatin as an augmenting agent of fluoxetine (up to 40 mg/day) [42]. Participants (N = 68) had MDD and were randomized to either fluoxetine + lovastatin (30 mg/day) or fluoxetine + placebo for 6 weeks. Participants receiving lovastatin had a significantly larger reduction in depressive symptoms (mean (SD): −12.8 (6.3) vs. −8.2 (4.0), t = 3.4, df = 60, p < 0.001). A second RCT in patients with severe MDD (N = 60) compared the efficacy of citalopram (40 mg/day) + atorvastatin (20 mg/day) vs citalopram (40 mg/day) + placebo for 12 weeks [43]. The atorvastatin group had a significantly larger reduction in depressive symptoms than in the placebo group (F (3, 174) =8.93, p < 0.001). A third study in MDD (N = 48) investigated the efficacy of fluoxetine (40 mg/day) + simvastatin (20 mg/day) vs. fluoxetine (40 mg/day) + placebo for 6 weeks. Again, there was a significantly larger reduction in depressive symptoms in the simvastatin group than in the placebo group, (F (1.88, 78.94) = 3.78, p = 0.02) [44].
Two RCTs have investigated the efficacy of statins in patients with MDD with co-morbid medical conditions [45,46]. The first was an RCT in patients with MDD (N = 58) who had a coronary-artery bypass graft (CABG) within the past 6 months [45]. Patients were randomized to either simvastatin (20 mg) or atorvastatin (20 mg) monotherapy with no other antidepressant treatment. Both treatment groups had a significant reduction in depressive symptoms; however, there was a larger and faster improvement on depressive symptoms with simvastatin group than with atorvastatin (F (1.62, 71.06) = 3.41, p = 0.048). A second study was a retrospective secondary analysis of an RCT of escitalopram (N = 149) vs placebo (N = 151) for the treatment of MDD in patients who had an acute coronary syndrome 2–14 weeks prior [46]. Participants were prescribed statins as medically indicated by a cardiologist (N = 226). In a logistic regression predicting response rates, escitalopram use and not statins use significantly predicted rates of response (defined as 50% reduction in Hamilton depression rating scale (HDRS)) at 24 weeks. Conversely, statin use but not escitalopram uses significantly predicted response rates at 1-year follow-up. Some patients with MDD who did not participate in the escitalopram randomization received statin therapy (N = 97); depression response rates were significantly higher in those who took lipophilic statins (N = 52; 44.7%) than in those who took hydrophilic statins (N = 45; 17.1%) [46]. More recently, a study of youth and young adults with moderate to severe depression investigated the efficacy of rosuvastatin (N = 48), aspirin (N = 40), or placebo (N = 42) in addition to standard antidepressant treatment for patients with moderate to severe MDD over 12 weeks [47]. There was a significant reduction in depressive symptoms in all three groups and no significant differences in the primary or secondary outcomes between groups. In a recent meta-analysis that pooled the results from the aforementioned studies, statins were more efficacious than placebo in addition to antidepressants to reduce depressive symptoms at 8 weeks (N = 255, SMD = −0.48, 95%CI: −0.74 to −0.22) and 12 weeks (N = 134, SMD = −0.47, 95%CI: −0.89 to −0.05); an exploratory network meta-analysis suggested that simvastatin, which is more lipophilic, may be more efficacious than the rosuvastatin, with is less lipophilic [48].

Statins for the Treatment of BD

The data from an RCT of the efficacy of atorvastatin (N = 27) vs. placebo (N = 33) in treating lithium-induced diabetes insipidus in BD and MDD were used to explore the effect of atorvastatin on cognition and mood [49]. There were no differences between the two groups on global cognition, cognitive impairment, executive function, depression relapse, or mania relapse [49]. In a small RCT (N = 54), patients with BD in a manic phased received lovastatin in a addition to lithium compared to placebo and lithium [50]. Overall there was no significant difference in reduction in manic symptoms compared to placebo. In a subsequent small RCT (N = 27), patients with BD in a manic phase were randomized to addition of either placebo or lovastatin to standard of care [51]; there were no differences in mood symptoms between the two groups.

PUFAs for the Treatment of MDD

Converging evidence suggests that PUFA are anti-inflammatory, hypotriglyceridemic, can modulate lipid metabolism, regulate adipokines, promote adipogenesis, and alter epigenetic mechanisms [52]. In a recent meta-analysis of 26 trials (N = 2160) in MDD or depressive symptoms found that, PUFAs were more effective than placebo in reducing depressive symptoms (SMD= −0.28; p = 0.004) [53]. Pure EPA and a mixture of EPA and DHA (with >60% EPA) demonstrated clinical benefits compared to placebo (SMD = −0.50, p = 0.003, and SMD = −1.03, p = 0.03, respectively), while pure DHA did not exhibit benefits. In another meta-analysis s of RCTs including only participants with MDD and no medical comorbidity (N = 910), PUFAs were superior to placebo in reducing depressive symptoms (SMD: 1.24; 95%CI: 0.060–2.414) [54]. In a network meta-analysis of the same RCTs, high-dose omega-3 PUFA was more effective than low dose omega-3 PUFA [54].

PUFA for the Treatment of BD

RCTs have had mixed results for treating either manic or depressive phases of BD. In a meta-analysis of 5 RCTs (N = 291) for BD depression, omega-3 PUFA yielded a moderate effect size (0.34) [55]. However, PUFA were not better than placebo for the treatment of mania [55]. A recent study examined whether PUFA supplementation would prevent mood episodes in patients with BD over 52 weeks [56]. This small study (N = 80) found little benefit for prophylactic supplementation other than a small reduction in hypomanic symptoms in the omega-3 PUFA group. One small RCT (N = 31) explored the effects of omega-3 PUFA on cognition in BD [57], showing no improvement in cognition compared to placebo.

3.2.2. Antidiabetic Agents for the Treatment of Mood Disorders

Thiazolidinediones (TZD) for MDD

TZD function as agonists to the proliferator activated nuclear receptor (PPAR)-gamma, which primarily functions to enhance insulin sensitization and enhance glucose metabolism [58]. PPAR-gamma agonists had previously been assessed in the treatment of other neurological disorders, such as Alzheimer’s disease or multiple sclerosis [59,60]. Animal models of MDD and 2 open-label studies in adults with MDD or BD suggested potential benefit of these agents in the treatment of MDD [61,62,63,64,65,66]. A first RCT was completed in 40 patients with MDD and no history of DM or metabolic syndrome [67]. Participants received citalopram (30 mg/day) + pioglitazone (15 mg/day) or citalopram (30 mg/day) + placebo for 6 weeks. The pioglitazone group had significantly larger reduction in depressive symptoms than the placebo group (F (1, 38) =9.483, p = 0.004). A subsequent RCT compared the efficacy of pioglitazone and metformin, and assessed whether their antidepressant effects were due to insulin sensitization [68]. Patients had MDD and polycystic ovarian syndrome (PCOS) (N = 50) and were assigned to pioglitazone (15 mg/day) vs metformin (750 mg twice/day). Overall, pioglitazone was superior to metformin in reducing depressive symptoms (38.3% vs. 8.3% reduction respectively; F (1, 37) = 75.513, p < 0.001). At the end of the trial, there were no significant differences in biochemical profile or insulin resistance between the 2 groups, and change in insulin resistance was not associated with change in depressive symptoms [68]. A subsequent RCT in patients with MDD who did not achieve remission with standard antidepressants, randomized participants (N = 37) to either pioglitazone (15 mg/day) or placebo [69] and found no difference in reduction of depressive symptoms in the two groups [69]. However, in a post-hoc analysis splitting the participants based on insulin sensitivity, the insulin resistant pioglitazone group had a significantly larger reduction in depressive symptoms than the insulin resistant placebo group. Further, improvement in oral glucose tolerance test (OGTT) and fasting plasma glucose (FPG) was associated with a favourable reduction in depressive symptoms in the insulin resistant group only [69].

Thiazolidinediones (TZDs) for BD

In an open-label study in depressed patients with BD (N = 34) receiving pioglitazone (15–30 mg/day), higher baseline IL-6 was associated with a favourable antidepressant response and improvement in insulin resistance [70]. A subsequent 6-week RCT (N = 44) explored whether pioglitazone (30 mg/day) vs placebo would be an effective augmentation agent to lithium in patients with BD I depression. These patients did not have DM or evidence of metabolic dysfunction. The pioglitazone group had a significant larger reduction in depressive symptoms than the placebo group at week 2, 4 and 6 [71]. Another RCT in patients with BD depression (N = 37) evaluated pioglitazone (15–45 mg/day) vs placebo in addition to standard care72. It found no significant benefit of pioglitazone augmentation, with a near significant improvement favouring placebo [72]. Of note, participants recruited to this RCT were not enriched for evidence of metabolic dysfunction.

Glucagon-like Peptide 1 (GLP-1) Agonists (Liraglutide) for MDD and BD

GLP-1 is a hormone synthesized in the gastro-intestinal tract (GI) and central nervous system (CNS). Liraglutide, a GLP-1 agonist is primarily used in the treatment of type 2 DM through enhanced glucose utilization via increased insulin secretion and suppression of glucagon [73,74,75]. A small open-label study has investigated whether liraglutide, improves cognition in euthymic patients with MDD (N = 13) or BD (N = 6) [76]. Participants presented with executive dysfunction at baseline and received 1.8 mg/day of liraglutide in addition to standard pharmacotherapy for their mood disorder. A significant pre-post improvement in trail-making test-B (TMTB) scores was observed and it was larger in those with higher baseline BMI and insulin resistance.

Metformin for MDD

Metformin (MET), primarily used for treatment of type 2 DM, minimizes hepatic glucose output and improves insulin-mediated uptake of glucose [77]. In observational studies, patients with MDD and DM receiving an antidepressant and MET have improvement in both conditions [78]. In an RCT, 58 patients with MDD and type 2 DM were treated with either MET vs placebo for 24 weeks [79]. MET significantly enhanced cognitive functions across multiple domains (i.e., verbal memory, visual memory, attention, and delayed memory). Further, MET significantly decreased depressive symptoms compared to placebo. MET also decreased HBA1c, with a positive correlation between reduction in HbA1c and depressive symptoms in the MET (r = 0.618, n= 22, p < 0.01). A recent RCT explored whether MET would be effective as an adjunctive agent in patients with MDD without medical comorbidity. Patients with MDD (N = 80) were randomized to receive fluoxetine (20 mg/day) + MET (1000 mg/day) or fluoxetine (20 mg/day) + placebo [80]. The MET group had a significantly larger reduction in depression severity than the placebo group after 12 weeks of treatment (SMD: −3.454; 95%CI: −4.145–−2.76). The MET group also had a significantly larger reduction in TNF-alpha, IL-1B, IL-6, IGF-1, MDA, CRP, and significantly increased BDNF and serotonin than the placebo group.

Insulin for MDD

Insulin is an endogenous hormone secreted by pancreatic-B cells and is used in replacement therapy for both type 1 and 2 DM. In healthy adults, intranasal insulin has been associated with enhanced mood and well-being [81]. In a cross-over RCT, patients with MDD (N = 35) were randomized to either intranasal insulin (40 IU QID) or placebo [82]. Overall, there was no significant difference between groups in changes in cognitive function or in secondary measures such as quality of life.

Insulin for BD

In an RCT, adults with euthymic BD (N = 62) were randomized to intranasal insulin (40 IU QID) vs. placebo [83]. Treatment with insulin led to a significant improvement in executive function as measured by the trail making test-B.

4. Discussion

This review highlights converging evidence that mood disorders are associated with metabolic dysfunction of lipid, fatty acid, and glucose regulation. This is supported by animal models, large population-based cross-sectional studies, cohort studies, and observational studies. Small studies provide some evidence for the effectiveness of metabolic modulating agents for the treatment of MDD and BD. The best evidence exists for the use of statins and PUFAs in patients with MDD. Some evidence also supports the use of PUFAs in patients with BD depression, but there is only very limited evidence for the use of any metabolic modulating agent for manic phases. Overall, this field of studies is in its early stages and large RCTs are required to replicate and extend these findings before treatment recommendations can be made.
While it is clear that MDD and BD are associated with metabolic dysfunction, and treatments that target these pathways have shown promise in treating depressive symptoms, it is less clear how these disturbances contribute to the pathophysiology of mood symptoms. Several pathophysiological links and shared mechanisms between metabolic dysfunction and mood disorders have been proposed. Some of the proposed pathophysiological links through which metabolic dysfunction may contribute specifically to the development of mood disorders include and potential therapeutic benefit include; Brain Atrophy: Repeated periods of hypoglycemia/hyperglycemia, lacunar infarcts, and generalized microvascular dysfunction have been shown to impact brain function in regions that are associated with mood and cognition such as the pre-frontal cortex (PFC) and hippocampus (HC) [84]. Brain cholesterol metabolism: Brain cholesterol is responsible for regulation of several processes including membrane bound proteins, ion channels, synaptic transmission, synapse formation, dendrite formation, and axonal formation; disruptions of which have been associated with mood disorders [85]. Monoamine neurotransmission: Deficits in fatty acids have been associated with altered DA and serotonin neurotransmission in the frontal cortex, suggesting impacts on cognitive function and emotion evaluation and regulation [26,86,87,88,89,90]. Inflammation: Cholesterol, LDL, very-LDL (VLDL), and chylomicrons circulating in vessel walls can become oxidized, triggering an immunological cascade, reactive oxygen species (ROS), and immune cells such as macrophages, natural killers cells, mast cells, or dendritic cells [91]. Many of these proinflammatory molecules cross the blood brain barrier (BBB) leading to microglial activation and downstream processes that can lead to psychiatric symptoms and potentially mood disorders [4]. Medications that target metabolic variables, such as Omega-3 PUFA and statins, have also been shown to be anti-inflammatory [92]. Insulin Resistance: Insulin receptors are expressed in brain regions that are associated with mood disorders; the nucleus accumbens (NAc), the ventral tegmental area (VTA), the amygdala, and the raphe nuclei of which disrupted signaling may be contributing to depressive symptomatology [93]. Gut Microbiome: There is a rapidly increasing body of research that has shown that regulation of the gut microbiome has impact on psychiatric disorders [94]. The microbiome-brain axis has also been implicated in brain development, function, and metabolism [95]. Metabolic dysfunction such as obesity, glycemic control, and insulin resistance have been shown to be associated with a shift of microbiota [96].
A major limitation to most treatment studies thus far has been the lack of pre-treatment enrichment of samples. Some of the reviewed RCTs have attempted to stratify participants for evidence of metabolic dysfunction, for example patients with MDD after a CABG (who may benefit from lipid lowering agents) or patients with co-morbid DM/insulin resistance (who may benefit from anti-diabetic agents). Following this approach, a proof-of-concept study recruited participants with MDD and baseline inflammation and found that those with higher inflammation responded best to treatment with PUFAs [97]. If replicated, this finding could suggest that the mechanism of action of PUFAs may in part be anti-inflammatory and patients with baseline inflammation may be more likely to benefit from them. Similarly, many of the anti-diabetic agents reviewed were studied in patients with various baseline levels of insulin resistance or hyperglycemia, and changes in depressive symptoms was associated with changes in insulin resistance [69], glucose tolerance tests [69], inflammation [70], or HBA1C [79]. These findings, support the hypothesis that these agents may be most efficacious in patients with higher degree of metabolic dysfunction. Future studies should consider similar stratification approaches to improve our understanding of mechanism of action of these agents and achieve a higher likelihood of detecting significant treatment effect.
As discussed above, overwhelming evidence support a bi-directional association between metabolic dysfunction and mood disorders. However, a biomarker specific for metabolic dysfunction in individuals with, or at risk for, associated with mood disorders remains elusive. Numerous attempts over the past decades have failed to find a singular “biomarker” of pathology in mood disorders. A composite measure based on multiple biomarkers, similar to the validated measures that predict the risk of stroke, myocardial infarction, or death in patients with vascular risk factors, may be able to distinguish individuals with metabolic dysfunction at risk for mood disorders, or individuals with mood disorders at risk for metabolic disorders. A recent study using machine learning sought to determine the metabolic profile of MDD (N = 5283) compared to controls (N = 10,145) based on a panel of lipids, fatty acids, low molecular weight metabolites, and lipid/fatty acid ratios [98]. A profile of ten metabolomic molecules were associated with increased odds of MDD. In a separate analysis in a subset of these patients, patients who were currently depressed had differential metabolic profile compared to healthy controls [99]. These findings support the notion that in a subset of patients there is an association with lipid/metabolomic regulation and prevalence and incidence of MDD that may be best identified through an index combining multiple measures or a profile based on multiple markers. Future research should explore whether stratification of patients based upon such an index or profile would enhance treatment response to metabolic modulating agents and whether changes in metabolic parameters are associated with response.
Limitations: This scoping review may not have identified all the extant literature on the effect of pharmacotherapies that target metabolic dysfunction to ameliorate mood and related symptoms in patients with MDD and BD.

5. Conclusions

In summary, there is evidence of altered metabolic function either through lipid metabolism or hyperglycemia/insulin resistance in a subset of patients with MDD and BD. These metabolic disturbances may have both direct and indirect effects on the pathophysiology of MDD and BD. While there is promising evidence for the use of some metabolic agents for the treatment MDD and BD, larger and better designed studies are required before these agents can be repurposed and recommended for the treatment for mood disorders. These future studies also need to explore potential mechanisms of action and use innovative clinical trial design to address the heterogeneous clinical phenotypes of mood disorders.

Author Contributions

All authors contributed (B.D.M.J., S.F., S.K., M.O.H., B.H.M. and M.I.H.) to the conceptualization of the manuscript. B.D.M.J. and S.F. completed the search and screened articles to be included. All authors (B.D.M.J., S.F., S.K., M.O.H., B.H.M. and M.I.H.) contributed to the writing, editing, and finalization of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported in part by Academic Scholars Awards to M.I.H. and S.K. from the Department of Psychiatry, University of Toronto and by B.H.M.’s Labatt Family Chair in Biology of Depression in Late-Life Adults at the University of Toronto.

Conflicts of Interest

S.K. received honorarium from Empowerpharm for past consultation. B.H.M. currently holds and receives support from the Labatt Family Chair in Biology of Depression in Late-Life Adults at the University of Toronto. He currently receives research support from Brain Canada, the Canadian Institutes of Health Research, the CAMH Foundation, the Patient-Centered Outcomes Research Institute (PCORI), the US National Institute of Health (NIH), Capital Solution Design LLC (software used in a study founded by CAMH Foundation), and HAPPYneuron (software used in a study founded by Brain Canada). He has been an unpaid consultant to Myriad Neuroscience. M.I.H. is a PI for a trial sponsored by COMPASS Pathways Limited.

References

  1. The World Health Organization. WHO Global Burden of Disease (2008); 2004 Update; WHO: Geneva, Switzerland, 2008. [Google Scholar]
  2. Rush, A.J.; Trivedi, M.H.; Wisniewski, S.R.; Nierenberg, A.A.; Stewart, J.W.; Warden, D.; Niederehe, G.; Thase, M.E.; Lavori, P.W.; Lebowitz, B.; et al. Acute and Longer-Term Outcomes in Depressed Outpatients Requiring One or Several Treatment Steps: A STAR*D Report. Am. J. Psychiatry 2006, 163, 1905–1917. [Google Scholar] [CrossRef] [PubMed]
  3. Bowden, C.; Perlis, R.; Thase, M.; Ketter, T.; Ostacher, M.; Calabrese, J.; Reilly-Harrington, N.; Gonzalez, J.; Singh, V.; Nierenberg, A.; et al. Aims and Results of the NIMH Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD). CNS Neurosci. Ther. 2011, 18, 243–249. [Google Scholar] [CrossRef] [PubMed]
  4. Jones, B.D.M.; Daskalakis, Z.J.; Carvalho, A.F.; Strawbridge, R.; Young, A.H.; Mulsant, B.H.; Husain, M.I. Inflammation as a treatment target in mood disorders: Review. BJPsych. Open 2020, 6, e60. [Google Scholar] [CrossRef] [PubMed]
  5. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Association: Washington, DC, USA, 2013; Available online: https://0-doi-org.brum.beds.ac.uk/10.1176/appi.books.9780890425596 (accessed on 6 August 2021).
  6. World Health Organization. ICD-10: International Statistical Classification of Diseases and Related Health Problems: Tenth Revision, 2nd ed.; World Health Organization: Geneva, Switzerland, 2004; Available online: https://apps.who.int/iris/handle/10665/42980 (accessed on 6 August 2021).
  7. Vancampfort, D.; Vansteelandt, K.; Correll, C.U.; Mitchell, A.; De Herdt, A.; Sienaert, P.; Probst, M.; De Hert, M. Metabolic Syndrome and Metabolic Abnormalities in Bipolar Disorder: A Meta-Analysis of Prevalence Rates and Moderators. Am. J. Psychiatry 2013, 170, 265–274. [Google Scholar] [CrossRef]
  8. Wulsin, L.R.; Vaillant, G.E.; Wells, V.E. A Systematic Review of the Mortality of Depression. Psychosom. Med. 1999, 61, 6–17. [Google Scholar] [CrossRef]
  9. Golden, S.H.; Lazo, M.; Carnethon, M.; Bertoni, A.G.; Schreiner, P.J.; Diez Roux, A.V.; Lee, H.B.; Lykestos, C. Examining a bidirectional association between depressive symptoms and diabetes. JAMA J. Am. Med. Assoc. 2008, 299, 2751–2759. [Google Scholar] [CrossRef] [PubMed]
  10. Vogelzangs, N.; Beekman, A.T.F.; Boelhouwer, I.G.; Bandinelli, S.; Milaneschi, Y.; Ferrucci, L.; Penninx, B.W.J.H. Metabolic depression: A chronic depressive subtype? Findings from the InCHIANTI study of older persons. J. Clin. Psychiatry 2011, 72, 598–604. [Google Scholar] [CrossRef]
  11. Marijnissen, R.M.; Vogelzangs, N.; Mulder, M.E.; Brink, R.H.S.V.D.; Comijs, H.C.; Voshaar, R.C.O. Metabolic dysregulation and late-life depression: A prospective study. Psychol. Med. 2016, 47, 1041–1052. [Google Scholar] [CrossRef] [PubMed]
  12. Vogelzangs, N.; Beekman, A.T.; Dortland, A.K.V.R.; Schoevers, R.A.; Giltay, E.J.; de Jonge, P.; Penninx, B.W. Inflammatory and Metabolic Dysregulation and the 2-Year Course of Depressive Disorders in Antidepressant Users. Neuropsychopharmacology 2014, 39, 1624–1634. [Google Scholar] [CrossRef] [Green Version]
  13. Chen, P.H.; Tsai, S.Y.; Pan, C.H.; Chen, Y.L.; Chang, H.M.; Su, S.S.; Chen, C.C.; Kuo, C.J. High Risk and Trajectories of Physical Illnesses before the Diagnosis of Bipolar Disorder. J. Affect. Disord. 2021, 281, 99–108. [Google Scholar] [CrossRef]
  14. Chien, I.-C.; Lin, C.-H.; Chou, Y.-J.; Chou, P. Increased risk of hyperlipidemia in patients with major depressive disorder: A population-based study. J. Psychosom. Res. 2013, 75, 270–274. [Google Scholar] [CrossRef]
  15. Hsu, J.-H.; Chien, I.-C.; Lin, C.-H. Increased risk of hyperlipidemia in patients with bipolar disorder: A population-based study. Gen. Hosp. Psychiatry 2015, 37, 294–298. [Google Scholar] [CrossRef]
  16. Chuang, C.-S.; Yang, T.-Y.; Muo, C.-H.; Su, H.-L.; Sung, F.-C.; Kao, C.-H. Hyperlipidemia, statin use and the risk of developing depression: A nationwide retrospective cohort study. Gen. Hosp. Psychiatry 2014, 36, 497–501. [Google Scholar] [CrossRef] [PubMed]
  17. Partonen, T.; Haukka, J.; Virtamo, J.; Taylor, P.R.; Lönnqvist, J. Association of low serum total cholesterol with major depression and suicide. Br. J. Psychiatry 1999, 175, 259–262. [Google Scholar] [CrossRef] [PubMed]
  18. De Berardis, D.; Marini, S.; Piersanti, M.; Cavuto, M.; Perna, G.; Valchera, A.; Mazza, M.; Fornaro, M.; Iasevoli, F.; Martinotti, G.; et al. The Relationships between Cholesterol and Suicide: An Update. ISRN Psychiatry 2012, 2012, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. De Berardis, D.; Serroni, N.; Campanella, D.; Carano, A.; Gambi, F.; Valchera, A.; Conti, C.; Sepede, G.; Scali, M.; Fulcheri, M.; et al. Alexithymia and its relationships with C-reactive protein and serum lipid levels among drug naïve adult outpatients with major depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2008, 32, 1982–1986. [Google Scholar] [CrossRef]
  20. Almeida, O.; Yeap, B.; Hankey, G.; Golledge, J.; Flicker, L. HDL cholesterol and the risk of depression over 5 years. Mol. Psychiatry 2013, 19, 637–638. [Google Scholar] [CrossRef] [PubMed]
  21. Ancelin, M.-L.; Carriere, I.; Boulenger, J.-P.; Malafosse, A.; Stewart, R.; Cristol, J.-P.; Ritchie, K.; Chaudieu, I.; Dupuy, A.M. Gender and Genotype Modulation of the Association Between Lipid Levels and Depressive Symptomatology in Community-Dwelling Elderly (The ESPRIT Study). Biol. Psychiatry 2010, 68, 125–132. [Google Scholar] [CrossRef] [Green Version]
  22. Hibbeln, J.R.; Elsevier, B.V. Fish consumption and major depression. Lancet 1998, 351, 1213. [Google Scholar] [CrossRef]
  23. Tanskanen, A.; Hibbeln, J.R.; Tuomilehto, J.; Uutela, A.; Haukkala, A.; Viinamäki, H.; Lehtonen, J.; Vartiainen, E. Fish Consumption and Depressive Symptoms in the General Population in Finland. Psychiatr. Serv. 2001, 52, 529–531. [Google Scholar] [CrossRef] [PubMed]
  24. Noaghiul, S.; Hibbeln, J.R. Cross-National Comparisons of Seafood Consumption and Rates of Bipolar Disorders. Am. J. Psychiatry 2003, 160, 2222–2227. [Google Scholar] [CrossRef] [PubMed]
  25. Hirayama, T. Life-style and mortality: A large-scale census-based cohort study in Japan. Contrib. Epidemiol. Biostat. 1990. Available online: https://ci.nii.ac.jp/naid/10018977972 (accessed on 6 August 2021).
  26. Müller, C.P.; Reichel, M.; Mühle, C.; Rhein, C.; Gulbins, E.; Kornhuber, J. Brain membrane lipids in major depression and anxiety disorders. In Biochimica et Biophysica Acta—Molecular and Cell Biology of Lipids; Elsevier: Amsterdam, The Netherlands, 2015; Volume 1851, pp. 1052–1065. [Google Scholar]
  27. Shanmugam, N.; Reddy, M.A.; Guha, M.; Natarajan, R. High glucose-induced expression of proinflammatory cytokine and chemokine genes in monocytic cells. Diabetes 2003, 52, 1256–1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. McNamara, R.K.; Welge, J.A. Meta-analysis of erythrocyte polyunsaturated fatty acid biostatus in bipolar disorder. Bipolar Disord. 2016, 18, 300–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Messamore, E.; Almeida, D.M.; Jandacek, R.J.; McNamara, R.K. Polyunsaturated fatty acids and recurrent mood disorders: Phenomenology, mechanisms, and clinical application. Prog. Lipid Res. 2017, 66, 1–13. [Google Scholar] [CrossRef] [Green Version]
  30. Shin, J.Y.; Suls, J.; Martin, R. Are Cholesterol and Depression Inversely Related? A Meta-analysis of the Association between Two Cardiac Risk Factors. Ann. Behav. Med. 2008, 36, 33–43. [Google Scholar] [CrossRef] [PubMed]
  31. Pan, A.; Keum, N.; Okereke, O.I.; Sun, Q.; Kivimaki, M.; Rubin, R.R.; Hu, F.B. Bidirectional Association Between Depression and Metabolic Syndrome: A systematic review and meta-analysis of epidemiological studies. Diabetes Care 2012, 35, 1171–1180. [Google Scholar] [CrossRef] [Green Version]
  32. Lin, P.Y.; Huang, S.Y.; Su, K.P. A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression. Biol. Psychiatry 2010, 68, 140–147. [Google Scholar] [CrossRef]
  33. Persons, J.E.; Fiedorowicz, J.G. Depression and serum low-density lipoprotein: A systematic review and meta-analysis. J. Affect. Disord. 2016, 206, 55–67. [Google Scholar] [CrossRef]
  34. Ali, S.; Stone, M.A.; Peters, J.L.; Davies, M.J.; Khunti, K. The prevalence of co-morbid depression in adults with Type 2 diabetes: A systematic review and meta-analysis. Diabet. Med. 2001, 23, 1165–1173. [Google Scholar] [CrossRef]
  35. Anderson, R.J.; Freedland, K.E.; Clouse, R.E.; Lustman, P.J. The Prevalence of Comorbid Depression in Adults with Diabetes: A meta-analysis. Diabetes Care 2001, 24, 1069–1078. [Google Scholar] [CrossRef] [Green Version]
  36. Vancampfort, D.; Mitchell, A.J.; De Hert, M.; Sienaert, P.; Probst, M.; Buys, R.; Stubbs, B. Type 2 diabetes in patients with major depressive disorder: A meta-analysis of prevalence estimates and predictors. Depress. Anxiety 2015, 32, 763–773. [Google Scholar] [CrossRef]
  37. Geraets, A.F.J.; Köhler, S.; Muzambi, R.; Schalkwijk, C.G.; Oenema, A.; Eussen, S.J.P.M.; Dagnelie, P.C.; Stehouwer, C.D.A.; Schaper, N.C.; Henry, R.M.A.; et al. The association of hyperglycaemia and insulin resistance with incident depressive symptoms over 4 years of follow-up: The Maastricht Study. Diabetologia 2020, 63, 2315–2328. [Google Scholar] [CrossRef] [PubMed]
  38. Calkin, C.V.; Gardner, D.; Ransom, T.; Alda, M. The relationship between bipolar disorder and type 2 diabetes: More than just co-morbid disorders. Ann. Med. 2012, 45, 171–181. [Google Scholar] [CrossRef]
  39. Chien, I.-C.; Chang, K.-C.; Lin, C.-H.; Chou, Y.-J.; Chou, P. Prevalence of diabetes in patients with bipolar disorder in Taiwan: A population-based national health insurance study. Gen. Hosp. Psychiatry 2010, 32, 577–582. [Google Scholar] [CrossRef] [PubMed]
  40. Wändell, P.; Ljunggren, G.; Wahlström, L.; Carlsson, A.C. Diabetes and psychiatric illness in the total population of Stockholm. J. Psychosom. Res. 2014, 77, 169–173. [Google Scholar] [CrossRef]
  41. Lipid Modification: Cardiovascular Risk Assessment and the Modification of Blood Lipids for the Primary and Secondary Prevention of Cardiovascular Disease—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/25340243/ (accessed on 6 August 2021).
  42. Ghanizadeh, A.; Hedayati, A. Augmentation of fluoxetine with lovastatin for treating major depressive disorder, a randomized double-blind placebo controlled-clinical trial. Depress. Anxiety 2013, 30, 1084–1088. [Google Scholar] [CrossRef] [PubMed]
  43. Haghighi, M.; Khodakarami, S.; Jahangard, L.; Ahmadpanah, M.; Bajoghli, H.; Holsboer-Trachsler, E.; Brand, S. In a randomized, double-blind clinical trial, adjuvant atorvastatin improved symptoms of depression and blood lipid values in patients suffering from severe major depressive disorder. J. Psychiatr. Res. 2014, 58, 109–114. [Google Scholar] [CrossRef]
  44. Gougol, A.; Zareh-Mohammadi, N.; Raheb, S.; Farokhnia, M.; Salimi, S.; Iranpour, N.; Yekehtaz, H.; Akhondzadeh, S. Simvastatin as an adjuvant therapy to fluoxetine in patients with moderate to severe major depression: A double-blind placebo-controlled trial. J. Psychopharmacol. 2015, 29, 575–581. [Google Scholar] [CrossRef]
  45. Abbasi, S.H.; Mohammadinejad, P.; Shahmansouri, N.; Salehiomran, A.; Beglar, A.A.; Zeinoddini, A.; Forghani, S.; Akhondzadeh, S. Simvastatin versus atorvastatin for improving mild to moderate depression in post-coronary artery bypass graft patients: A double-blind, placebo-controlled, randomized trial. J. Affect. Disord. 2015, 183, 149–155. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, S.W.; Bae, K.Y.; Kim, J.M.; Shin, I.S.; Hong, Y.J.; Ahn, Y.; Jeong, M.H.; Berk, M.; Yoon, J.S. The use of statins for the treatment of depression in patients with acute coronary syndrome. Transl. Psychiatry 2015, 5, e620. [Google Scholar] [CrossRef] [Green Version]
  47. Berk, M.; Mohebbi, M.; Dean, O.M.; Cotton, S.M.; Chanen, A.M.; Dodd, S.; Ratheesh, A.; Amminger, G.P.; Phelan, M.; Weller, A.; et al. Youth Depression Alleviation with Anti-inflammatory Agents (YoDA-A): A randomised clinical trial of rosuvastatin and aspirin. BMC Med. 2020, 18, 16. [Google Scholar] [CrossRef] [PubMed]
  48. De Giorgi, R.; De Crescenzo, F.; Pesci, N.R.; Martens, M.; Howard, W.; Cowen, P.J.; Harmer, C.J. Statins for major depressive disorder: A systematic review and meta-analysis of randomized controlled trials. PLoS ONE 2021, 16, e0249409. [Google Scholar] [CrossRef] [PubMed]
  49. Soh, J.F.; Almadani, A.; Beaulieu, S.; Rajji, T.; Mulsant, B.H.; Su, C.-L.; Renaud, S.; Mucsi, I.; Torres-Platas, S.G.; Levinson, A.; et al. The effect of atorvastatin on cognition and mood in bipolar disorder and unipolar depression patients: A secondary analysis of a randomized controlled trial. J. Affect. Disord. 2020, 262, 149–154. [Google Scholar] [CrossRef]
  50. Ghanizadeh, A.; OmraniSigaroodi, M.; Javadpour, A.; Dabbaghmanesh, M.H.; Shafiee, S. Lovastatin as an Adjuvant to Lithium for Treating Manic Phase of Bipolar Disorder: A 4-Week, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Depression Res. Treat. 2014, 2014, 1–6. [Google Scholar] [CrossRef]
  51. Lotfi, M.; Shafiee, S.; Ghanizadeh, A.; Sigaroudi, M.O.; Razeghian, L. A Magnetic Resonance Spectroscopy Study of Lovastatin for Treating Bipolar Mood Disorder: A 4-Week Randomized Double-Blind, Placebo-Controlled Clinical Trial. Recent Pat. Inflamm. Allergy Drug Discov. 2017, 10, 133–141. [Google Scholar] [CrossRef] [PubMed]
  52. Albracht-Schulte, K.; Kalupahana, N.S.; Ramalingam, L.; Wang, S.; Rahman, S.M.; Robert-McComb, J.; Moustaid-Moussa, N. Omega-3 fatty acids in obesity and metabolic syndrome: A mechanistic update. J. Nutr. Biochem. 2018, 58, 1–16. [Google Scholar] [CrossRef]
  53. Liao, Y.; Xie, B.; Zhang, H.; He, Q.; Guo, L.; Subramaniapillai, M.; Fan, B.; Lu, C.; Mclntyer, R.S. Efficacy of omega-3 PUFAs in depression: A meta-analysis. Transl. Psychiatry 2019, 9, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Luo, X.D.; Feng, J.S.; Yang, Z.; Huang, Q.T.; Yang, B.; Su, K.P.; Pan, J.Y. High-dose omega-3 polyunsaturated fatty acid supplementation might be more superior than low-dose for major depressive disorder in early therapy period: A network meta-analysis. BMC Psychiatry 2020, 20, 248. [Google Scholar] [CrossRef] [PubMed]
  55. Saunders, E.F.H.; Ramsden, C.E.; Sherazy, M.S.; Gelenberg, A.J.; Davis, J.M.; Rapoport, S.I. Omega-3 and omega-6 polyunsaturated fatty acids in bipolar disorder: A review of biomarker and treatment studies. J. Clin. Psychiatry 2016, 77, e1301–e1308. [Google Scholar] [CrossRef]
  56. McPHILEMY, G.; Byrne, F.; Waldron, M.; Hibbeln, J.R.; Davis, J.; McDonald, C.; Hallahan, B. A 52-week prophylactic randomised control trial of omega-3 polyunsaturated fatty acids in bipolar disorder. Bipolar Disord. 2020. [Google Scholar] [CrossRef]
  57. Ciappolino, V.; DelVecchio, G.; Prunas, C.; Andreella, A.; Finos, L.; Caletti, E.; Siri, F.; Mazzocchi, A.; Botturi, A.; Turolo, S.; et al. The Effect of DHA Supplementation on Cognition in Patients with Bipolar Disorder: An Exploratory Randomized Control Trial. Nutrients 2020, 12, 708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Spiegelman, B.M. PPAR-γ: Adipogenic regulator and thiazolidinedione receptor. Diabetes 1998, 47, 507–514. [Google Scholar] [CrossRef]
  59. Kaiser, C.C.; Shukla, D.K.; Stebbins, G.T.; Skias, D.D.; Jeffery, D.R.; Stefoski, D.; Katsamakis, G.; Feinstein, D.L. A pilot test of pioglitazone as an add-on in patients with relapsing remitting multiple sclerosis. J. Neuroimmunol. 2009, 211, 124–130. [Google Scholar] [CrossRef]
  60. Miller, B.W.; Willett, K.C.; Desilets, A.R. Rosiglitazona y pioglitazona para el tratamiento de la enfermedad de Alzheimer. Ann. Pharmacother. 2011, 45, 1416–1424. [Google Scholar] [CrossRef]
  61. Rosa, A.O.; Kaster, M.; Binfaré, R.W.; Morales, S.; Martín-Aparicio, E.; Navarro-Rico, M.L.; Martinez, A.; Medina, M.; Garcia, A.G.; Lopez, M.G.; et al. Antidepressant-like effect of the novel thiadiazolidinone NP031115 in mice. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2008, 32, 1549–1556. [Google Scholar] [CrossRef]
  62. Eissa Ahmed, A.A.; Al-Rasheed, N.M.; Al-Rasheed, N.M. Antidepressant-like effects of rosiglitazone, a PPARγ agonist, in the rat forced swim and mouse tail suspension tests. Behav. Pharmacol. 2009, 20, 635–642. [Google Scholar] [CrossRef]
  63. Budni, J.; Lobato, K.R.; Binfaré, R.W.; Freitas, A.E.; Costa, A.P.; Martín-De-Saavedra, M.D.; Leal, R.B.; Lopez, M.G.; Rodrigues, A.L.S. Involvement of PI3K, GSK-3β and PPARγ in the antidepressant-like effect of folic acid in the forced swimming test in mice. J. Psychopharmacol. 2012, 26, 714–723. [Google Scholar] [CrossRef]
  64. Sadaghiani, M.S.; Paydar, M.J.; Gharedaghi, M.H.; Fard, Y.Y.; Dehpour, A.R. Antidepressant-like effect of pioglitazone in the forced swimming test in mice: The role of PPAR-gamma receptor and nitric oxide pathway. Behav. Brain Res. 2011, 224, 336–343. [Google Scholar] [CrossRef] [PubMed]
  65. Kemp, D.E.; Ismail-Beigi, F.; Ganocy, S.J.; Conroy, C.; Gao, K.; Obral, S.; Fein, E.; Findling, R.L.; Calabrese, J.R. Use of insulin sensitizers for the treatment of major depressive disorder: A pilot study of pioglitazone for major depression accompanied by abdominal obesity. J. Affect. Disord. 2012, 136, 1164–1173. [Google Scholar] [CrossRef] [Green Version]
  66. Rasgon, N.L.; Kenna, H.A.; Williams, K.E.; Powers, B.; Wroolie, T.; Schatzberg, A.F. Rosiglitazone Add-On in Treatment of Depressed Patients with Insulin Resistance: A Pilot Study. Sci. World J. 2010, 10, 321–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Sepanjnia, K.; Modabbernia, A.; Ashrafi, M.; Modabbernia, M.J.; Akhondzadeh, S. Pioglitazone adjunctive therapy for moderate-to-severe major depressive disorder: Randomized double-blind placebo-controlled trial. Neuropsychopharmacology 2012, 37, 2093–2100. [Google Scholar] [CrossRef] [Green Version]
  68. Kashani, L.; Omidvar, T.; Farazmand, B.; Modabbernia, A.; Ramzanzadeh, F.; Tehraninejad, E.S.; Ashrafi, M.; Tabrizi, M.; Akhondzadeh, S. Does pioglitazone improve depression through insulin-sensitization? Results of a randomized double-blind metformin-controlled trial in patients with polycystic ovarian syndrome and comorbid depression. Psychoneuroendocrinology 2013, 38, 767–776. [Google Scholar] [CrossRef] [PubMed]
  69. Lin, K.W.; Wroolie, T.E.; Robakis, T.K.; Rasgon, N.L. Adjuvant pioglitazone for unremitted depression: Clinical correlates of treatment response. Psychiatry Res. 2015, 230, 846–852. [Google Scholar] [CrossRef] [Green Version]
  70. Kemp, D.E.; Schinagle, M.; Gao, K.; Conroy, C.; Ganocy, S.J.; Ismail-Beigi, F.; Calabrese, J.R. PPAR-γ Agonism as a Modulator of Mood: Proof-of-Concept for Pioglitazone in Bipolar Depression. CNS Drugs 2014, 28, 571–581. [Google Scholar] [CrossRef] [Green Version]
  71. Zeinoddini, A.; Sorayani, M.; Hassanzadeh, E.; Arbabi, M.; Farokhnia, M.; Salimi, S.; Ghaleiha, A.; Akhondzadeh, S. Pioglitazone adjunctive therapy for depressive episode of bipolar disorder: A randomized, double-blind, placebo-controlled trial. Depress. Anxiety 2015, 32, 167–173. [Google Scholar] [CrossRef]
  72. Aftab, A.; Kemp, D.E.; Ganocy, S.J.; Schinagle, M.; Conroy, C.; Brownrigg, B.; D’Arcangelo, N.; Goto, T.; Woods, N.; Serrano, M.B.; et al. Double-blind, placebo-controlled trial of pioglitazone for bipolar depression. J. Affect. Disord. 2018, 245, 957–964. [Google Scholar] [CrossRef]
  73. Cabou, C.; Campistron, G.; Marsollier, N.; Leloup, C.; Cruciani-Guglielmacci, C.; Pénicaud, L.; Drucker, D.J.; Magnan, C.; Burcelin, R. Brain Glucagon-Like Peptide-1 Regulates Arterial Blood Flow, Heart Rate, and Insulin Sensitivity. Diabetes 2008, 57, 2577–2587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. During, M.J.; Cao, L.; Zuzga, D.S.; Francis, J.S.; Fitzsimons, H.L.; Jiao, X.; Bland, R.; Klugmann, M.; Banks, W.A.; Drucker, D.J.; et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat. Med. 2003, 9, 1173–1179. [Google Scholar] [CrossRef]
  75. McClean, P.L.; Gault, V.A.; Harriott, P.; Hölscher, C. Glucagon-like peptide-1 analogues enhance synaptic plasticity in the brain: A link between diabetes and Alzheimer’s disease. Eur. J. Pharmacol. 2010, 630, 158–162. [Google Scholar] [CrossRef]
  76. Mansur, R.B.; Ahmed, J.; Cha, D.S.; Woldeyohannes, H.O.; Subramaniapillai, M.; Lovshin, J.; Lee, J.G.; Lee, J.H.; Brietzke, E.; Reininghaus, E.Z.; et al. Liraglutide promotes improvements in objective measures of cognitive dysfunction in individuals with mood disorders: A pilot, open-label study. J. Affect. Disord. 2017, 207, 114–120. [Google Scholar] [CrossRef]
  77. Zhou, J.; Massey, S.; Story, D.; Li, L. Metformin: An old drug with new applications. Int. J. Mol. Sci. 2018, 19, 2863. [Google Scholar] [CrossRef] [Green Version]
  78. Abrahamian, H.; Hofmann, P.; Prager, R.; Toplak, H. Diabetes mellitus and co-morbid depression: Treatment with milnacipran results in significant improvement of both diseases (results from the Austrian MDDM study group). Neuropsychiatr. Dis. Treat. 2009, 5, 261–266. [Google Scholar] [CrossRef] [Green Version]
  79. Guo, M.; Mi, J.; Jiang, Q.M.; Xu, J.M.; Tang, Y.Y.; Tian, G.; Wang, B. Metformin may produce antidepressant effects through improvement of cognitive function among depressed patients with diabetes mellitus. Clin. Exp. Pharmacol. Physiol. 2014, 41, 650–656. [Google Scholar] [CrossRef]
  80. Abdallah, M.S.; Mosalam, E.M.; Zidan, A.-A.A.; Elattar, K.S.; Zaki, S.A.; Ramadan, A.N.; Ebeid, A.M. The Antidiabetic Metformin as an Adjunct to Antidepressants in Patients with Major Depressive Disorder: A Proof-of-Concept, Randomized, Double-Blind, Placebo-Controlled Trial. Neurotherapeutics 2020, 17, 1897–1906. [Google Scholar] [CrossRef] [PubMed]
  81. Benedict, C.; Hallschmid, M.; Hatke, A.; Schultes, B.; Fehm, H.L.; Born, J.; Kern, W. Intranasal insulin improves memory in humans. Psychoneuroendocrinology 2004, 29, 1326–1334. [Google Scholar] [CrossRef]
  82. Cha, D.S.; Best, M.W.; Bowie, C.R.; Gallaugher, L.A.; Woldeyohannes, H.O.; Soczynska, J.K.; Lewis, G.; MacQueen, G.; Sahakian, B.J.; Kennedy, S.H.; et al. A randomized, double-blind, placebo-controlled, crossover trial evaluating the effect of intranasal insulin on cognition and mood in individuals with treatment-resistant major depressive disorder. J. Affect. Disord. 2017, 210, 57–65. [Google Scholar] [CrossRef]
  83. McIntyre, R.S.; Soczynska, J.; Woldeyohannes, H.O.; Miranda, A.; Vaccarino, A.; MacQueen, G.; Lewis, G.F.; Kennedy, S. A randomized, double-blind, controlled trial evaluating the effect of intranasal insulin on neurocognitive function in euthymic patients with bipolar disorder. Bipolar Disord. 2012, 14, 697–706. [Google Scholar] [CrossRef]
  84. Holt, R.I.G.; de Groot, M.; Golden, S.H. Diabetes and depression. Curr. Diabetes Rep. 2014, 14, 491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Parekh, A.; Smeeth, D.; Milner, Y.; Thuret, S. The Role of Lipid Biomarkers in Major Depression. Healthcare 2017, 5, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kodas, E.; Page, G.; Zimmer, L.; Vancassel, S.; Guilloteau, D.; Durand, G.; Chalon, S. Neither the density nor function of striatal dopamine transporters were influenced by chronic n-3 polyunsaturated fatty acid deficiency in rodents. Neurosci. Lett. 2002, 321, 95–99. [Google Scholar] [CrossRef]
  87. Zimmer, L.; Hembert, S.; Durand, G.; Breton, P.; Guilloteau, D.; Besnard, J.-C.; Chalon, S. Chronic n-3 polyunsaturated fatty acid diet-deficiency acts on dopamine metabolism in the rat frontal cortex: A microdialysis study. Neurosci. Lett. 1998, 240, 177–181. [Google Scholar] [CrossRef]
  88. Zimmer, L.; Delpal, S.; Guilloteau, D.; Aїoun, J.; Durand, G.; Chalon, S. Chronic n-3 polyunsaturated fatty acid deficiency alters dopamine vesicle density in the rat frontal cortex. Neurosci. Lett. 2000, 284, 25–28. [Google Scholar] [CrossRef]
  89. McNamara, R.K.; Jandacek, R.; Rider, T.; Tso, P.; Cole-Strauss, A.; Lipton, J.W. Omega-3 fatty acid deficiency increases constitutive pro-inflammatory cytokine production in rats: Relationship with central serotonin turnover. Prostaglandins Leukot. Essent. Fat. Acids 2010, 83, 185–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. De La Presa Owens, S.; Innis, S.M. Docosahexaenoic and arachidonic acid prevent a decrease in dopaminergic and serotoninergic neurotransmitters in frontal cortex caused by a linoleic and α-linolenic acid deficient diet in formula-fed piglets. J. Nutr. 1999, 129, 2088–2093. [Google Scholar] [CrossRef] [Green Version]
  91. Linton, M.F.; Yancey, P.G.; Davies, S.S.; Jerome, W.G.; Linton, E.F.; Song, W.L.; Doran, A.C.; Vickers, K.C.; Feingold, K.R.; Anawalt, B.; et al. The Role of Lipids and Lipoproteins in Atherosclerosis. Science 1950, 111, 166–186. [Google Scholar]
  92. Ruscica, M.; Ferri, N.; Macchi, C.; Corsini, A.; Sirtori, C.R. Lipid lowering drugs and inflammatory changes: An impact on cardiovascular outcomes? Ann. Med. 2018, 50, 461–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lyra e Silva, N.D.M.; Lam, M.P.; Soares, C.N.; Munoz, D.P.; Milev, R.; de Felice, F.G. Insulin resistance as a shared pathogenic mechanism between depression and type 2 diabetes. Front. Psychiatry 2019, 10, 57. [Google Scholar] [CrossRef] [Green Version]
  94. Winter, G.; Hart, R.A.; Charlesworth, R.; Sharpley, C. Gut microbiome and depression: What we know and what we need to know. Rev. Neurosci. 2018, 29, 629–643. [Google Scholar] [CrossRef]
  95. Salvucci, E. The human-microbiome superorganism and its modulation to restore health. Int. J. Food Sci. Nutr. 2019, 70, 781–795. [Google Scholar] [CrossRef]
  96. Gérard, C.; Vidal, H. Impact of gut microbiota on host glycemic control. Front. Endocrinol. 2019, 10, 29. [Google Scholar] [CrossRef] [PubMed]
  97. Rapaport, M.H.; Nierenberg, A.A.; Schettler, P.J.; Kinkead, B.L.; Cardoos, A.; Walker, R.S.W.; Mischoulon, D. Inflammation as a predictive biomarker for response to omega-3 fatty acids in major depressive disorder: A proof-of-concept study. Mol. Psychiatry 2015, 21, 71–79. [Google Scholar] [CrossRef]
  98. Bot, M.; Milaneschi, Y.; Al-Shehri, T.; Amin, N.; Garmaeva, S.; Onderwater, G.L.; Pool, R.; Thesing, C.S.; Vijfhuizen, L.S.; Vogelzangs, N.; et al. Metabolomics Profile in Depression: A Pooled Analysis of 230 Metabolic Markers in 5283 Cases With Depression and 10,145 Controls. Biol. Psychiatry 2020, 87, 409–418. [Google Scholar] [CrossRef] [PubMed]
  99. De Kluiver, H.; Jansen, R.; Milaneschi, Y.; Bot, M.; Giltay, E.J.; Schoevers, R.; Penninx, B.W. Metabolomic profiles discriminating anxiety from depression. Acta Psychiatr. Scand. 2021. [Google Scholar] [CrossRef]
Table 1. Clinical trials of metabolic modulating agents in Major Depressive Disorder (MDD).
Table 1. Clinical trials of metabolic modulating agents in Major Depressive Disorder (MDD).
ReferenceParticipants (N, % Female)Age (SD) *Medication ComparatorDiagnosisEnrichmentOutcome Measure and Duration Result
Statin
Ghanizadeh et al. 2013N = 68, 63.2% 32.5 (10.2) and 31.7 (9.3)fluoxetine (40 mg) + lovastatin (30 mg)Fluoxetine (40 mg) + placebo DSM-IV MDDnone HDRS, 6 week RCTReduction of 12.8 (6.3) vs. 8.2 (4.0) p < 0.001
favouring lovastatin
Haghighi et al. 2014 N = 60, 56.6% 33.1 (8.9) and 31.4 (7.8)citalopram (40 mg) + atorvastatin (20 mg) citalopram (40 mg) + placeboDSM-5 MDD noneHDRS, 12 week RCTweek 12 HDRS
atorvastatin: 19.63 (3.16)
placebo: 22.03 (3.58)
Gougol et al. 2015 N= 48, 60.4% 36.4 (8.1) and 34.2 (10.8)fluoxetine (40 mg) + simvastatin (20 mg)fluoxetine (40 mg/day) + placebo DSM-IV MDDnoneHDRS, 6 week RCTgroup × time interaction favouring simvastatin (F (1.88, 78.94) = 3.78, p = 0.02)
(Cohen’s d at week 6: 0.61)
Abbasi et al. 2015 N = 46, 32.6% 56.9 (6.9) and 57.7 (7.3)simvastatin (20 mg/day) atorvastatin (20 mg/day)DSM-IV MDDCABG in the last 6 monthsHDRS, 6 week RCTsignificant effect for time × treatment interaction favouring simvastatin s (F (1.62, 71.06) = 3.41, p = 0.048)
Kim et al. 2015 N = 300, 39.6% 60.3 (10.8), 58.9 (12.9), 60.4 (10.8), 59.3 (10.0)escitalopram + statins Escitalopram only, statin only, placebo onlyDSM-IV MDDacute coronary syndrome (ACS)HDRS, 24-week RCT with 1-year follow-upstatin use associated with higher 1 year response rate 2.23 (1.11–4.51) p = 0.025
Berk et al. 2020 N = 130, 60%, 20.2 (2.6)Rosuvastatin (10 mg) Aspirin (100 mg) or placebo DSM-IV MDDNoneMADRS, 12-week RCT no difference between rosuvastatin and placebo (−4.2, 95%CI (−9.1, 0.6), p = 0.089)
Giorgi et al. 2021 Meta-analysis statins compared to placebo in 5 RCT at 8 weeks (N = 255, SMD = −0.48, 95%CI = −0.74 to −0.22) and 12 weeks (N = 134, SMD = −0.47, 95%CI = −0.89 to −0.05)
Poly Unsaturated Fatty Acid (PUFA)
Liao et al. 2019 Meta-analysisBeneficial effect of omega-3 polyunsaturated fatty acids on depression symptoms in 26 RCTs (SMD = −0.28, p = 0.004)
Luo et al. 2020 Meta-analysisn-3 PUFAs were superior to placebo in 10 RCTs (SMD: 1.243 ± 0.596; 95%CI: 0.060~2.414)
both the high (SMD: 0.908 ± 0.331; 95%CI: 0.262, 1.581) and the low-dose (SMD: 0.601 ± 0.286; 95%CI: 0.034, 1.18) n-3 PUFAs were superior to placebo, and the efficacy of high-dose n-3 PUFAs is superior to that of low-dose
Thiazolidinediones (TZD)
Sepanjnia et al. 2012 N = 40, 72.5% 31.4 (5.4) and 32.7 (5.4)citalopram (30 mg) + pioglitazone (15 mg BID)citalopram (30 mg) + placeboDSM-IV MDDNoneHDRS, 6-week RCTmean difference favouring pioglitazone (−3.4 (−5.6 to −1.2)).
Kashani et al. 2013N = 50, 100%, 21.2 (3.3) and 20.3 (4.6)pioglitazone (15 mg BID) metformin (750 mg BID)DSM-IV MDDPCOSHDRS, 6-week RCTpercentage reduction favouring pioglitazone [38.3% versus 8.3% reduction from baseline scores, F (1, 37) = 73.513, p < 0.001].
Lin et al. 2015 N = 37, 78.4% 49.4 (15.1) and 43.3 (11.8)pioglitazone (30 mg) +TAU placebo + treatment as usual DSM-IV MDDNoneHDRS, 12-week RCTno significant difference in mean decline of HDRS-21 scores between treatment groups (t [29] = −1.22, p = 0.23).
Glucagon-Like Peptide 1 (GLP-1) Agonists
Mansur et al. 2017 N = 19, 57.9% 38.2liraglutide (1.8 mg) + TAU DSM 5 MDD or BDmeasurable impairment in executive functionTMTB, 4-week open-labelsignificant improvement in TMTB
standard score with those with higher baseline BMI and IR having the best improvement (Cohen’s d = 0.64, p = 0.009).
Metformin (MET)
Guo et al. 2014 N= 58, 37.9%, 54.7 (7.3) and 53.3 (7.3)Metformin (1–2 g per day)placeboDSM-IV MDDDiabetes Mellitus type 2wechsler memory scale–revised and MADRS, 24-week RCTsignificant improvement in verbal memory index, visual memory index, general memory index, and delayed memory index compared to placebo.
metformin significantly decreased
MADRS (F
1112
= 26.43; p < 0.001
Metformin significantly decreased MADRS compared to placebo (F = 26.43; p < 0.001)
MET metformin significantly decreased
MADRS (F
1112
= 26.43; p < 0.00
Abdallah et al. 2020 N = 80, 47.5% 34.1 (8.4) and 35.1 (8.0)metformin (1 g) + fluoxetine (20 mg)placebo + fluoxetine 20 mgDSM-IV MDD noneHDRS, 12-week RCT12 week results favouring metformin LSMD −3.454, p = 0.000
Insulin
Cha et al. 2017 N = 35, 62.9% 47.1 (9.9)intranasal insulin (40 International Units (IU) QID)placeboDSM-IV MDDnonepositive and negative affect schedule (PANAS) and global index of neurocognition, 12-week cross over RCTno between group differences observed
HDRS: hamilton depression rating scale; RCT: Randomized Controlled Trial; SMD: Standard Mean Difference; TMTB: trail-making test- B; CABG: coronary artery bypass graft surgery; BMI: body mass index; IR: insulin resistance; MADRS: Montgomery–Åsberg Depression Rating Scale; TAU: treatment as usual; RCT: Randomized Controlled Trial. * age is reported for active medication group and then comparator group.
Table 2. Clinical trials of metabolic modulating agents in Bipolar Disorder (BD).
Table 2. Clinical trials of metabolic modulating agents in Bipolar Disorder (BD).
ReferenceParticipants (N, % Female, Age)Age (SD) *Medication ComparatorDiagnosisEnrichmentOutcome Measure and Trial DurationResult
Statin
Fotso Soh et al. 2020
(secondary analysis)
N = 60, 58.3% 47.8 (13.8) and 53.1 (11.8)atorvastatin (20 mg) + lithium placebo + lithium DSM-IV BD or MDD lithium induced diabetes insipidus global cognition Z-score, 12-week RCTNo between group differences
Lotfi et al. 2017 N = 27Not reportedLovastatin +TAUPlacebo + TAUDSM-IV BD (mania)noneYMRS, 4-week RCT No significant difference found between groups
Ghanizadeh et al. 2013
N = 54, 44.4% female 30.5 (8.1) and 29.5 (10.8)Lovastatin + lithium Placebo + lithium DSM-IV BD (mania) noneYMRS, 4 weeksNo significant difference found between groups
Poly Unsaturated Fatty Acid (PUFA)
Sarris et al. 2012 5 RCT (N = 291) for bipolar depression revealed a significant effect size (0.34) in favor of omega-3 (p = 0.029)
5 RCT (N = 291) for bipolar mania revealed a non-significant effect
McPhilemy et al. 2020 N = 80, 51.25%45 (13) and 48 (12)EPA (1 g) + TAUPlacebo + TAUDSM-IV BD (any mood state) 3 mood relapses in last 5 years and 2 in the last 3 yearsmood state relapse (mania or depression), 52-week RCTno significant differences in the number of mood episode relapses (U = 490.00, p = 0.14) and no significant difference for time to relapse between groups (Log Rank χ2 = 0.41, p = 0.52).
Ciappolino et al. 2020 N = 31, 71%, 36 (12) and 50.4 (11.3)DHA supplementation (1250 mg) + TAUPlacebo + TAU DSM-IV BD (euthymia) NoneBAC-A, 12-week RCTno significant effects on cognition between groups.
Thiazolidinediones (TZD)
Zeinoddini et al. 2015 N = 44, 34.1% 33.6 (3.5) and 31.2 (5.6)pioglitazone (30 mg) + lithium Placebo + lithium DSM-IV BD (depressed) None HDRS, YMRS, 6-week RCT significant effect for time × treatment interaction on the HDRS scores [F (2.78, 116.65) = 4.77, p = 0.005]
Aftab et al. 2019 N = 38, 64.9%, 46.7 (12.1) and 43.7 (13) pioglitazone (15–45 mg) + TAUPlacebo + TAUDSM-IV BD (depressed) NoneIDS-C30, 8-week RCTmean reduction from baseline to week 8 in IDS-C30 score was−6.59 for pioglitazone and −11.63 for placebo
Insulin
McIntyre et al. 2012 N = 62, 46.8%, 40.8 (10.2) and 39.3 (10.4)intranasal insulin (40 IU q.i.d). + TAUPlacebo + TAU DSM-IV BD (euthymia)NoneCVLT-II
and PDT, 8-week RCT
significant improvement versus placebo with intranasal insulin therapy on secondary measure of executive function (i.e., TMTB). No group differences on primary measure
CVLT-II: California Verbal Learning Test, second edition; PDT: Process Dissociation Task; TMTB: Trail-Making test-B; BAC-A: Brief Assessment of Cognition in Affective Disorder; TAU: Treatment as usual; YMRS: Young Mania Rating Scale; HDRS: Hamilton Depression Rating Scale; IDS-C30: Inventory of depressive symptomatology; DHA: docosahexaenoic acid; EPA; eicosapentaenoic acid. * age is reported for active medication group and then comparator group.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jones, B.D.M.; Farooqui, S.; Kloiber, S.; Husain, M.O.; Mulsant, B.H.; Husain, M.I. Targeting Metabolic Dysfunction for the Treatment of Mood Disorders: Review of the Evidence. Life 2021, 11, 819. https://0-doi-org.brum.beds.ac.uk/10.3390/life11080819

AMA Style

Jones BDM, Farooqui S, Kloiber S, Husain MO, Mulsant BH, Husain MI. Targeting Metabolic Dysfunction for the Treatment of Mood Disorders: Review of the Evidence. Life. 2021; 11(8):819. https://0-doi-org.brum.beds.ac.uk/10.3390/life11080819

Chicago/Turabian Style

Jones, Brett D. M., Salman Farooqui, Stefan Kloiber, Muhammad Omair Husain, Benoit H. Mulsant, and Muhammad Ishrat Husain. 2021. "Targeting Metabolic Dysfunction for the Treatment of Mood Disorders: Review of the Evidence" Life 11, no. 8: 819. https://0-doi-org.brum.beds.ac.uk/10.3390/life11080819

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop