Next Article in Journal
CRIF1 Deficiency Increased Homocysteine Production by Disrupting Dihydrofolate Reductase Expression in Vascular Endothelial Cells
Next Article in Special Issue
Dietary Phytoestrogens and Their Metabolites as Epigenetic Modulators with Impact on Human Health
Previous Article in Journal
Current Status and Future Perspectives on Therapeutic Potential of Apigenin: Focus on Metabolic-Syndrome-Dependent Organ Dysfunction
Previous Article in Special Issue
Comprehensive Biological and Chemical Evaluation of Two Seseli Species (S. gummiferum and S. transcaucasicum)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Exploring the Impact of Flavonoids on Symptoms of Depression: A Systematic Review and Meta-Analysis

1
Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
2
Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
*
Author to whom correspondence should be addressed.
Submission received: 13 September 2021 / Revised: 13 October 2021 / Accepted: 15 October 2021 / Published: 20 October 2021

Abstract

:
Recent evidence suggests that diet modifies key biological factors associated with the development of depression. It has been suggested that this could be due to the high flavonoid content commonly found in many plant foods, beverages and dietary supplements. Our aim was to conduct a systematic review to evaluate the effects of dietary flavonoids on the symptoms of depression. A total of 46 studies met the eligibility criteria. Of these, 36 were intervention trials and 10 were observational studies. A meta-analysis of 36 clinical trials involving a total of 2788 participants was performed. The results showed a statistically significant effect of flavonoids on depressive symptoms (mean difference = −1.65; 95% C.I., −2.54, −0.77; p < 0.01). Five of the 10 observational studies included in the systematic review reported significant results, suggesting that a higher flavonoid intake may improve symptoms of depression. Further studies are urgently required to elucidate whether causal and mechanistic links exist, along with substantiation of functional brain changes associated with flavonoid consumption.

1. Introduction

Depressive disorders are recognized as some of the main causes of disability globally. The World Health Organization (WHO) estimates that 322 million people around the world demonstrate symptoms of depression, nearly 4.4% of the global burden of all diseases [1]. Depressive disorders include two main sub-categories: major depressive disorder (MDD)/depressive episode and dysthymia (i.e., chronic form of mild depression). In both cases, the manifestation of symptoms severely limits psychosocial functioning, diminishes the quality of life and increases the risk of suicidal behaviors [2,3]. Despite intense ongoing research efforts, the exact etiology of depressive disorders remains elusive. However, the complex biological pattern underlying depressive symptoms is thought to be associated with biochemical, genetic and environmental factors, including exposure to physical, social and psychological stressors during early and adult life [4].
The brain appears to be more susceptible than other organs to free radical attacks. Overproduction of free radical species may impair neuronal membrane phospholipids and consequently affect serotonergic and catecholaminergic receptor functions [5]. An altered redox status has been confirmed in the central nervous system (CNS) of depressed patients and oxidative disturbances have been associated with lowered concentrations of several endogenous antioxidant enzymes [6]. Pharmacological treatments, although proven to be effective in treating from moderate to severe symptoms in MDD, have only modest effect sizes and approximately 30% of depressed patients do not respond sufficiently to the existing drug therapies. Moreover, antidepressants are associated with a plethora of side effects and a propensity for abuse [7].
Nutritional psychiatry is an emerging discipline that focuses on the role of diet and nutrients in diverse aspects of mental health [8]. Numerous mechanisms were identified through which diet and its components may affect depression. These include the modulation of the pathways involved in inflammation, oxidative stress, epigenetics, mitochondrial dysfunction, the gut microbiota, tryptophan–kynurenine metabolism, the hypothalamic–pituitary–adrenal (HPA) axis and neurogenesis [9]. Although Molendijk et al. highlighted various issues regarding study recruitment and blinding, the SMILES trial showed that a 12-week healthy diet intervention improved ratings of depression in adults with a mean age of 40 years [10,11]. Further randomized clinical trials (RCTs) confirmed the benefits of a Mediterranean-style diet on mental health in depression, namely the HELFIMED and PREDIMED trials [12,13]. A meta-analysis of 16 RCTs that involved some type of diet improvement showed that diet interventions overall reduced depressive symptoms [14]. Currently, there are also human studies showing that the antioxidant/anti-inflammatory effects of dietary components may improve cognitive and depression outcomes [15,16].
Flavonoids belong to a family of polyphenolic plant compounds widely present in fruits, vegetables and certain beverages. Based on their molecular structure, flavonoids are classified into six main subclasses that include flavones, flavonols, flavanones, flavan-3-ols, anthocyanins and isoflavones [17]. Experimental studies have repeatedly demonstrated that dietary flavonoids and their representative subclasses exert modulatory effects on key cellular mechanisms associated with endogenous antioxidant systems [18], inhibition of immune-inflammatory pathways [19], antidepressant-like effects and enhancement of mitochondrial functions and neurotransmission [20,21]. Another advantage is that some flavonoids may have some clinical efficacy in individuals with depressive disorders without causing the side effects usually associated with conventional antidepressants [22]. However, to date, there are no meta-analyses synthesizing the relationship between dietary flavonoids and depressive disorders. Hence, the aim of the current paper is to conduct a systematic review and meta-analysis of human studies to critically evaluate whether flavonoids have some clinical efficacy in patients with depressive symptoms.

2. Methods

The search strategy, screening and selection criteria were developed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) Statement [23]. The registration number in PROSPERO is CRD42020173649.

2.1. Eligibility Criteria and Search Strategies

A literature search was conducted in the following databases: Scopus, PubMed and Web of Science. Observational and clinical studies, which clearly assessed the effect of flavonoids on the symptoms of depression, published up to 30 July 2021, were included in the review. We included articles that determined the flavonoid content of foods or the dose of the flavonoid-containing supplements. Articles were excluded from the review for the following reasons: studies not published in English; articles that used secondary data, such as reviews, meta-analysis, conference papers and book chapters; studies on animal models or in vitro experiments; studies that did not use a depression rating scale. In order to conduct a comprehensive systematic literature search, we used both controlled vocabulary and free text terms. The search was conducted using Boolean operators “AND” and “OR” to combine the following terms: “flavonoid” OR “anthocyanin” OR “anthocyanidin” OR “cyanidin” OR “delphinidin” OR “malvidin” OR “pelargonidin” OR “peonidin” OR “petunidin” OR “flavan-3-ol” OR “catechin” OR “epicatechin” OR “epigallocatechin” OR “gallocatechin” OR “proanthocyanidin” OR “theaflavin” OR “thearubigin” OR “flavonol” OR “isorhamnetin” OR “kaempferol” OR “myricetin” OR “quercetin” OR “flavone” OR “apigenin” OR “luteolin” OR “baicalein” OR “chrysin” OR “flavanone” OR “eriodictyol” OR “hesperetin” OR “naringenin” OR “isoflavone” OR “daidzein” OR “genistein” OR “glycitein” OR “biochanin” OR “formononetin” AND “depression” OR “depressive disorder” OR “depressive symptom” OR “antidepressant” OR “antidepressive”. At the same time, similar queries were respectively used for controlled vocabulary search: “flavonoids” [Mesh] AND “depression” [Mesh], INDEXTERMS “flavonoids” AND “depression”.

2.2. Study Selection and Data Extraction

Two authors (S.D. and S.A.) independently reviewed the titles and abstracts obtained from all the databases. After the removal of duplicate records with a reference management software (EndNote X8; Clarivate Analytics, Philadelphia, PA, USA) the titles and abstracts were reviewed. The full texts were screened by two authors (S.D. and G.C.) and all articles that did not meet the inclusion criteria were excluded. In the case of disagreement about the eligibility of a study, a third author (G.S.) decided which articles were included. Data were extracted independently by two authors (S.D. and S.A.) and confirmed where necessary by the principal investigator (G.S.). A data extraction spreadsheet was developed and the information from the studies included in the review was extracted and tabulated. The following information was recorded: author’s name, publication year, study country, study design, participant characteristics (sample size, gender and age), health status, duration of follow-up, outcome assessment for depression, intervention (type of compounds and dose) and results.

2.3. Study Quality Assessment

The study quality and risk of bias of each included study were assessed using the Cochrane risk of bias tool for RCTs [24] and the Newcastle–Ottawa Scale (NOS) for observational studies [25]. The Cochrane risk of bias tool is made up of 7 components: (1) sequence generation, (2) allocation sequence concealment, (3) blinding of participants and personnel, (4) blinding of outcome assessment, (5) incomplete outcome data and (6) selective outcome reporting. The NOS, which was identified as one of the most useful tools for assessing methodological quality of observational studies, evaluates three quality domains (selection, comparability and outcome) divided across eight specific items. The NOS assigns a maximum score of 9 points. Studies with NOS scores of 0–3, 4–6 and 7–9 were considered low, moderate and high quality, respectively.

2.4. Statistical Analysis

The meta-analysis was carried out using the R Software, version 4.0.3 (R Foundation for Statistical Computing, Vienna, Austria) and the interface R-Studio version 1.4.1717 (R studio, PBC, Boston, MA, USA). A p value < 0.05 was considered to be statistically significant. Continuous data from the studies included were expressed as mean difference with a 95% confidence interval. Participants who consumed the flavonoid intervention were recorded as the experimental group, while those who consumed the placebo intervention were recorded as the control group. Since measurement time periods differed among studies, final measurement time points of each study were used. The summary statistics required for each outcome were the number of participants in the intervention and control groups at baseline and post-test, the mean change from baseline and the SD of the mean change. If change-from-baseline scores were not provided, they were calculated using baseline and post-test means and SDs. The mean difference was used to express the results across studies. Statistical heterogeneity was calculated using the Higgins I2 statistic, which describes the percentage of variability in the effect estimate due to heterogeneity rather than sampling error. Inconsistency was examined using I2 and the following grades were applied: <25% (very low), from 25 to <50% (low), from 50 to <75% (moderate) and ≥75% (large) [26]. A random effects model was chosen for the meta-analyses because this method of analysis is favored when there is evidence of heterogeneity among studies. In order to assess whether the pooled estimate was biased by the effect of any particular study, we also carried out a sensitivity analysis by removing outlier studies and recalculating the pooled estimate. Subgroup analyses were also performed to evaluate the potential effect of different factors on the depressive outcomes. These factors were flavonoid subclasses, type of clinical trials (controlled and uncontrolled) and level of blinding. The visual inspection of funnel plots and Egger’s linear regression were used to assess the publication bias [27,28].

3. Results

3.1. Study Selection

As presented in Figure 1, the combined search resulted in 3928 published articles from the three databases, among which 1414 were duplicates. During the screening phase, 2514 records were discarded because they clearly did not meet the inclusion criteria, as determined from the title and abstract. The remaining 111 articles were examined for eligibility assessment through full-text reading. Of these, 65 records did not meet the eligibility criteria, or the full text was unavailable. Thus, a total of 46 studies were included in the final analyses.

3.2. Study Characteristics

The 46 included studies were conducted between 1999 and 2021. Thirty-six out of the 46 studies selected were clinical trials [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64] and 10 studies were observational cohort studies [65,66,67,68,69,70,71,72,73,74]. Among the 36 included clinical trials, 31 were RCTs with an average number of 67.4 randomized participants. The clinical trials varied in time duration from as few as 5 days to 2 years with the most common time frame being 8 weeks. The observational studies included both longitudinal cohort and cross-sectional designs and had a mean of 9426 participants. Most of the studies assessed both genders (n = 25), 20 assessed only females and only 1 study assessed only men. The age of all participants varied widely from 18 to 92 years. However, the majority of studies included subjects aged ≥40 years.
Most of the studies measured depression symptomatology in disease states, including osteopenia [52], osteoarthritis [45], fibromyalgia [55], multiple sclerosis [47,63], neurodegeneration or age-associated cognitive decline [32,38,39,54], obesity/overweight [34,49], schizophrenia and bipolar disorder [43,56] and chronic hepatitis C infection [44]. Sixteen of the studies were in perimenopausal, menopausal, or postmenopausal women [31,33,35,36,37,42,48,50,51,52,53,61,62,64,70,71]. A summary of the characteristics of all the included studies are presented in Table 1 and Table 2.

3.3. Flavonoids

A wide variety of flavonoids was assessed in the research reports included in this review. The majority of clinical studies assessed the effect of flavonoids consumed via tablets or dry extract capsules [29,30,31,32,33,34,35,36,39,40,41,42,43,44,45,47,48,49,50,51,52,53,54,56,57,58,59,60,61,62,64], liquids [38,46,55,63] and powder [37]. The observational studies looked at flavonoids consumed in their biological whole food form. The exposure was measured using the diet history questionnaire (DHQ) [65,68,69,74] and food frequency questionnaire (FFQ) [66,67,70,72,73]. Only 1 observational study measured urinary concentrations of isoflavones using liquid chromatography coupled with tandem mass spectrometry [71]. The most commonly tested groups of flavonoids in the intervention trials were isoflavones [31,32,33,35,36,37,42,48,50,51,52,55,61,62,64]. Although several clinical studies assessed total flavonoid intake or considered the combined effect of mixed flavonoid subclasses, other classes of flavonoids tested included flavan-3-ols, flavonols, flavones, anthocyanins and proanthocyanidins. Two of the observational studies estimated habitual dietary consumption of total polyphenols and then determined total flavonoid intake [67,72]. In addition to the intake of total flavonoids, two observational studies also estimated flavonoid subclasses, such as flavonols, flavones, flavanones, anthocyanins, flavan-3-ols and proanthocyanidins [66,67]. However, the main measures of exposure in the observational studies were isoflavones [65,68,69,70,71,73,74].

3.4. Depression Scales

As shown in Table 1 and Table 2, a large number of depression scales has been used in the studies included in this review. The most common depression scales used in both clinical and observational studies were the Geriatric Depression Scale (GDS), Center for Epidemiological Studies Depression Scale (CES-D), Beck Depression Inventory (BDI) and Hamilton Depression Scale (HAM-D). The GDS was used in 6 clinical trials [29,32,37,38,50,54] and 3 observational studies [65,66,73] of the 46 included studies. The GDS is a commonly used depression scale in geriatric populations. This scale has several versions and it can be applied to normal, medically ill or cognitively impaired older adults [75]. Another popular depression scale used was the CES-D, which was used in three clinical trials [46,55,64] and five observational studies [66,67,68,69,70]. The CES-D, designed for large-scale studies, is a 20-item self-report inventory that can be completed in approximately 5 min [76]. The BDI was used in seven intervention trials [34,44,45,47,48,49,61]. The BDI is a 21-item, multiple-choice inventory designed to assess the level of key depressive symptoms (e.g., guilt, low self-worth and suicidal ideation) in adults [77]. Another popular scale was the HAM-D, which was used in five of the studies [30,31,43,56,62]. This scale consists of 17 items that are rated by the observer, rather than the patient [78].

3.5. Flavonoids and Depressive Symptoms

Overall, dietary flavonoids were associated with an improvement in depressive symptoms. The majority of trials (n = 22) found significant effects of consuming flavonoids on the symptoms of depression [29,30,31,33,34,35,36,41,42,44,46,47,48,49,51,52,56,58,60,61,62,64]. Zarghami et al. observed a significant decrease in the depression score after flavonoid intervention but the antidepressant drug fluoxetine had a greater anti-depressant activity than flavonoids [57]. Thirteen clinical studies showed no significant effect after the intervention with flavonoids [32,37,38,39,40,43,45,50,53,54,55,59,63]. Significant results on depressive symptoms associated with perimenopause, menopause, or postmenopause were reported in 11 trials of the 14 studies investigating the effect of flavonoids during the menopause transition [31,33,35,36,42,48,51,52,61,62,64]. One clinical study also noted a statistically significant effect on depressive symptoms in postmenopausal women with osteopenia [52]. Two out of the three trials conducted on anxious individuals found a significant improvement in depressive symptoms after treatment with flavonoids [30,41]. There were also significant effects of flavonoids on overweight or obese adults [34,49] and subjects affected by multiple sclerosis [47], chronic hepatitis C infection [44] and bipolar disorder [56]. No effects were reported in Alzheimer’s disease or individuals affected by age-associated memory impairment. Five out of the 10 observational studies reported that higher flavonoid intakes are associated with lower depression symptoms [66,67,68,69,74]. No significant associations were found between flavonoid intake and depressive symptoms in elderly subjects, menopausal women and subjects affected by mental health problems.

3.6. Meta-Analysis of the Effect of Flavonoids on Depressive Symptoms

Overall, we meta-analyzed 36 trials involving a total of 2788 participants. Observational studies were not included in the meta-analysis because sufficient data were not available. The meta-analysis showed a statistically significant reduction of depressive symptoms after consumption of dietary flavonoids (mean difference = −1.65; 95% CI, −2.54, −0.77; p < 0.01). Figure 2A depicts a forest plot of the effect of flavonoids on the reduction in depressive symptoms compared with controls. There was evidence of significant heterogeneity among studies (I2 = 92%). We carried out a sensitivity analysis by omitting outlier studies and recalculating the effect size. Using a random effect model, the sensitivity analysis did not affect the overall findings. By eliminating 10 of the studies from the primary analysis, the positive effects of flavonoids on depression symptoms remained significant (mean difference = −0.69; 95% CI, −1.13, −0.25; p < 0.01) (Figure 2B). After excluding outlier studies, the heterogeneity was reduced to I2 = 35%. The subgroup analyses were conducted based on the type of flavonoid subclass, type of clinical trial (controlled and uncontrolled trials) and level of blinding. As presented in Figure 3A, isoflavone (mean difference = −1.47; 95% CI; −2.83, −0.11; p = 0.03), flavonols (mean difference = −3.14; 95% CI, −5.79, −0.49; p = 0.02) and flavan-3-ol (mean difference = −1.80; 95% CI, −3.07, −0.52; p < 0.01) (Figure 3A) subclasses showed statistically significant effects. In the subgroup analysis based on the presence or absence of control group, pooled results from 31 controlled studies involving 2358 participants were significant (mean difference = −1.09; 95% CI, −1.89, −0.30; p < 0.01). Likewise, the effect size of five studies conducted with no control was also significant (mean difference = −5.59; 95% CI, −9.78, −1.41; p < 0.01) (Figure 3B). The subgroup analysis based on the level of blinding revealed a significant effect of flavonoids in 25 double-blind studies (mean difference = −0.92; 95% CI, −1.85, −0.01; p = 0.05) and 7 unblind intervention trials (mean difference = −3.11; 95% CI, −5.69, −0.54; p = 0.02). (Figure 3C).

3.7. Publication Bias

The publication bias was assessed using funnel plots and Egger’s test. The visual inspection of the funnel plot revealed a symmetrical distribution of studies (Figure 4A,B). Additionally, Egger’s test (p = 0.54) did not show significant publication bias. Thus, the likelihood of publication bias was low. Results for the risk of bias are shown in Supplementary Figure S1 and Supplementary Table S1. Overall, results from the critical appraisal tool show good methodology in the intervention trials. Most included clinical studies (25 of 36) were double-blind. However, although these trials claimed to be double blinded, many failed to provide details of how the investigators and those delivering the interventions were blinded in the method sections. The majority of trials were randomized, but most of the studies did not indicate the technique for sequence generation. A common weakness observed in the clinical trials was also the lack of information regarding the methods for evaluating or verifying participant compliance. Almost all manuscripts reported the number of dropouts during the interventions. The study qualities of the selected observational studies were diverse. Based on the NOS quality assessment results, eight studies achieved NOS scores between 4 and 6 (moderate quality) [65,67,68,69,70,71,72,73], one study was rated as “low” quality (3 points) [74] and one study was rated as “high” quality (8 points) [66]. This variation in quality of observational studies could possibly be explained by the following reasons: (a) no selection of the unexposed cohort; (b) no reliability of outcome assessment; (c) 20% or over losses to follow-up after baseline.

4. Discussion

A recent study highlighted the importance of a systematic method for summarizing the evidence on dietary interventions in depression [79]. Currently, systematic reviews and meta-analyses have shown that unhealthy dietary patterns are associated with an increased risk of depression, while high-quality diets are related to a lower risk of depressive symptoms [15,80]. In addition, several studies have indicated that nutritional deficiencies, such as vitamins (B vitamins), minerals (magnesium, selenium, zinc and iron) and amino acids, are common in depressed patients [81,82,83]. Although hotly debated, recent findings have also suggest that omega-3 polyunsaturated fatty acids, vitamin D and minerals may have a protective effect against the development of depressive symptoms [84,85,86].
Dietary flavonoids possess a large number of neuroprotective actions in various pathophysiological conditions, including depression. Although the antidepressant effects of flavonoids have been confirmed in various animal models of depression [22,87], the results of human studies remain controversial. The purpose of this manuscript is to provide a comprehensive review of the literature regarding the effects of flavonoids on depressive symptoms. A considerable number of the included studies observed significant results, suggesting that dietary flavonoids may improve depressive symptoms.
Our meta-analysis pooled results from 36 clinical trials to estimate the effects of the consumption of flavonoids on outcomes associated with depressive symptoms. The results show a significant antidepressant effect of flavonoids on subjects affected by depressive symptoms. However, the 36 clinical trials included in our meta-analysis were associated with a considerable heterogeneity. The sensitivity analysis restricted to 26 clinical studies also found a significant improvement in depressive symptoms with a low level of heterogeneity.
In the subgroup analyses by flavonoid subclasses, we only found a significant effect of isoflavones, flavonols and flavan3-ols on depressive symptoms. Likewise, separate subgroup analyses confined to double-blind and unblind trials showed a significant effect of flavonoids against depressive symptoms. Additional subgroup analyses of controlled and uncontrolled clinical studies also revealed a statistically significant effect of flavonoids.
Most of the trials examined here used different treatment durations and involved a large variety of dosages and sources of flavonoids. Depressive symptoms were frequently accompanied by other conditions and, in many cases, the effect of flavonoids on depressive symptoms was not always the primary outcome of the study. Moreover, many different types of depression scales were used. There were no large-scale randomized trials and most of the intervention studies were underpowered. It should be also noticed that the capture of the dietary background through an initial dietary assessment was not performed in the included clinical studies. This is crucial to evaluate flavonoid content in the habitual diet (i.e., listing the food sources of flavonoids), avoid underestimation of flavonoid exposure and know whether the planned flavonoid-based intervention contains a flavonoid intake level sufficiently above the expected background intake. Indeed, when there was evidence of effectiveness of flavonoids, it was not clear that the flavonoids themselves (rather than other bioactive components consumed as part of the diet) are solely or partially responsible for the observed effects. Therefore, the results from these clinical studies do not provide a clear picture on optimal dosage, treatment duration and estimated flavonoid intakes. However, although our findings should be interpreted cautiously, this meta-analysis provides preliminary evidence that treatments with flavonoids may potentially have some clinical efficacy in the treatment of depressive symptoms.
The observational studies reported the flavonoid intake of individuals in a real-life setting and estimated the prevalence of depressive symptoms among consumers of flavonoids. Although observational studies were not included in the meta-analyses as they did not present statistically homogeneous categories, five studies found significant results [66,67,68,69,74], suggesting that a higher flavonoid intake may improve symptoms of depression. Flavonoid intake was measured with different FFQ and DHQ. However, there are some issues with these tools, such as under- or over-reporting consumption and measurement error [88]. Additionally, errors of exposure measurement may be due in large part to the lack of analytic values for specific flavonoid classes. Except for one study [71], none of these observational studies took advantage of the more recent flavonoid analytic techniques and biomarkers of intake were not used. Moreover, most of the studies focused on intake of a specific flavonoid-rich food (typically, soy) or adherence to dietary patterns [65,68,69,73,74] and they did not perform a comprehensive evaluation of flavonoid subclasses. Therefore, all these factors must be considered when interpreting the results.
Both clinical trials and observational studies measured depressive symptoms rather than MDD and did not fully assess the severity of depressive symptoms. Therefore, the results obtained here cannot be generalized to treatment of MDD. Although the specific links between flavonoids and depression are not fully clarified, several mechanisms have been suggested [89]. Extensive evidence indicates that depression is associated with activated immune-inflammatory, neuro-oxidative and neuro-nitrosative pathways [90]. Animal studies have demonstrated that flavonoids possess an antidepressant-like property via interactions with oxidative pathways and antioxidant systems [21]. Flavonoids may also exert beneficial effects on depression through their anti-oxidants and anti-inflammatory properties and inhibition of proinflammatory mediators [91,92,93]. Another possible mechanism is that flavonoids may modulate signaling pathways, which are responsible for maintaining neuron survival and synaptic plasticity [94,95,96]. It is important to note that the beneficial effects of flavonoids may depend on their bioavailability, which differs greatly by subclasses. Bioavailability is influenced by absorption, metabolism and disposition in tissues and cells. The structure of flavonoids influences the rate and extent of intestinal absorption which, in turn, affects the metabolites circulating in the plasma. Flavonoids are metabolized by colonic microflora and several studies suggest that the metabolites of flavonoids may be one of the characteristics responsible for their therapeutic effects [97]. The differences in bioavailability and absorption rates of various flavonoids are lacking in the included studies and this aspect should be considered when interpreting the results. Moreover, flavonoid content between foods and supplements is highly variable and the type of source (i.e., foods or supplements) may affect numerous factors associated with bioavailability and health outcomes [98,99]. A common theme in this review is the use of isoflavones for peri- and postmenopausal women and during pregnancy. Isoflavones are found mainly in soy-based foods and they exhibit estrogenic activity by binding estrogen receptors. It has been suggested that isoflavones may alleviate the symptoms of depression by acting as phytoestrogens; therefore, they may only be appropriate for use in specific population groups [100,101].
Our manuscript has several strengths. First, to our knowledge, this is the first paper which systematically reviewed available human studies on the effects of flavonoids on depressive symptoms. The literature search was conducted applying specific inclusion and exclusion criteria and using controlled vocabulary queries to identify relevant studies. The publication bias was evaluated with the visual inspection of the funnel plot and Egger’s test. The visual inspection of the funnel plot of studies included in the sensitivity analysis showed no evidence of publication bias. Furthermore, a quality assessment of the observational studies and clinical trials was performed and most of the included studies had a relatively good quality.
Recent research efforts are beginning to highlight differences in which men and women respond to diet and express symptoms of depression. The interactions between flavonoid intake, gender and depressive symptoms was not tested in any of the studies. No clinical study investigated the association between flavonoid consumption and neurochemical factors, which are substantial contributors to the pathogenesis of depression. Moreover, social and cultural factors were not considered as contributory factors to the development of depressive symptoms.

5. Conclusions

Based on our findings, we may provide some directions for future research delineating the effects of flavonoids on depressive phenomenology and MDD. First, given that flavonoids and their subclasses are commonly consumed as part of a normal diet, more carefully designed studies should be performed to improve dietary intake assessment and determine the specific effects on symptoms of depression. Second, well-powered and long-term trials are required to explore the optimal dosage and treatment duration. Third, studies investigating biomarkers of intake are also required to clarify interactions of flavonoids with potentially causal mechanisms. Fourth, prospective studies are needed to confirm the association between flavonoid consumption and depressive symptoms or MDD and between immune and nitro-oxidative biomarkers of depression in order to decipher whether the clinical efficacy of flavonoids is associated with an improvement of those biomarkers. If confirmed, the findings may have important implications for depression prevention.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/antiox10111644/s1, Figure S1: Risk of bias assessment for the included randomized controlled trials, Table S1: Quality assessment of included observational studies using the New-Castle Ottawa Scale.

Author Contributions

Conceptualization, S.D.; methodology, S.D., G.C. and S.A.; software, S.D., G.C. and S.A.; validation, G.C. and S.A.; formal analysis, G.C. and S.A.; investigation, S.D., G.C. and S.A.; resources, G.S.; data curation, S.D., G.C. and S.A.; writing—original draft preparation, S.D.; writing—review and editing, S.D., M.M and G.S; visualization, S.D., M.M. and G.S.; supervision, M.M. and G.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BDI: Beck Depression Inventory; CES-D, Center for Epidemiological Studies Depression Scale; CNS, central nervous system; DHQ, Diet History Questionnaire; FFQ, Food Frequency Questionnaire; GDS, Geriatric Depression Scale; HAM-D, Hamilton Depression Scale; HPA, hypothalamic–pituitary–adrenal; MDD, major depressive disorder; NOS, Newcastle–Ottawa Scale; PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analysis; RCT, randomized controlled trial; WHO, World Health Organization.

References

  1. Friedrich, M.J. Depression Is the Leading Cause of Disability Around the World. JAMA 2017, 317, 1517. [Google Scholar] [CrossRef]
  2. Klein, D.N.; Santiago, N.J. Dysthymia and chronic depression: Introduction, classification, risk factors, and course. J. Clin. Psychol. 2003, 59, 807–816. [Google Scholar] [CrossRef]
  3. Rotenstein, L.S.; Ramos, M.A.; Torre, M.; Segal, J.B.; Peluso, M.J.; Guille, C.; Sen, S.; Mata, D.A. Prevalence of Depression, Depressive Symptoms, and Suicidal Ideation Among Medical Students: A Systematic Review and Meta-Analysis. JAMA 2016, 316, 2214–2236. [Google Scholar] [CrossRef] [Green Version]
  4. van den Bosch, M.; Meyer-Lindenberg, A. Environmental Exposures and Depression: Biological Mechanisms and Epidemiological Evidence. Annu. Rev. Public Health 2019, 40, 239–259. [Google Scholar] [CrossRef] [Green Version]
  5. Bhatt, S.; Nagappa, A.N.; Patil, C.R. Role of oxidative stress in depression. Drug Discov. Today 2020, 25, 1270–1276. [Google Scholar] [CrossRef]
  6. Liu, T.; Zhong, S.; Liao, X.; Chen, J.; He, T.; Lai, S.; Jia, Y. A Meta-Analysis of Oxidative Stress Markers in Depression. PLoS ONE 2015, 10, e0138904. [Google Scholar] [CrossRef]
  7. 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. Lancet 2018, 391, 1357–1366. [Google Scholar] [CrossRef] [Green Version]
  8. Marx, W.; Moseley, G.; Berk, M.; Jacka, F. Nutritional psychiatry: The present state of the evidence. Proc. Nutr. Soc. 2017, 76, 427–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Marx, W.; Lane, M.; Hockey, M.; Aslam, H.; Berk, M.; Walder, K.; Borsini, A.; Firth, J.; Pariante, C.M.; Berding, K.; et al. Diet and depression: Exploring the biological mechanisms of action. Mol. Psychiatry 2021, 26, 134–150. [Google Scholar] [CrossRef] [PubMed]
  10. Jacka, F.N.; O’Neil, A.; Opie, R.; Itsiopoulos, C.; Cotton, S.; Mohebbi, M.; Castle, D.; Dash, S.; Mihalopoulos, C.; Chatterton, M.L.; 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] [Green Version]
  11. Molendijk, M.L.; Fried, E.I.; Van Der Does, W. The SMILES trial: Do undisclosed recruitment practices explain the remarkably large effect? BMC Med. 2018, 16, 243. [Google Scholar] [CrossRef]
  12. Parletta, N.; Zarnowiecki, D.; Cho, J.; Wilson, A.; Bogomolova, S.; Villani, A.; Itsiopoulos, C.; Niyonsenga, T.; Blunden, S.; Meyer, B.; et al. A Mediterranean-style dietary intervention supplemented with fish oil improves diet quality and mental health in people with depression: A randomized controlled trial (HELFIMED). Nutr. Neurosci. 2019, 22, 474–487. [Google Scholar] [CrossRef] [Green Version]
  13. Sánchez-Villegas, A.; Martínez-González, M.A.; Estruch, R.; Salas-Salvadó, J.; Corella, D.; Covas, M.I.; Arós, F.; Romaguera, D.; Gómez-Gracia, E.; Lapetra, J.; et al. Mediterranean dietary pattern and depression: The PREDIMED randomized trial. BMC Med. 2013, 11, 208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Firth, J.; Marx, W.; Dash, S.; Carney, R.; Teasdale, S.B.; Solmi, M.; Stubbs, B.; Schuch, F.B.; Carvalho, A.F.; Jacka, F.; et al. The Effects of Dietary Improvement on Symptoms of Depression and Anxiety: A Meta-Analysis of Randomized Controlled Trials. Psychosom. Med. 2019, 81, 265–280. [Google Scholar] [CrossRef] [PubMed]
  15. Li, Y.; Lv, M.-R.; Wei, Y.-J.; Sun, L.; Zhang, J.-X.; Zhang, H.-G.; Li, B. Dietary patterns and depression risk: A meta-analysis. Psychiatry Res. 2017, 253, 373–382. [Google Scholar] [CrossRef] [PubMed]
  16. Davinelli, S.; Ali, S.; Solfrizzi, V.; Scapagnini, G.; Corbi, G. Carotenoids and cognitive outcomes: A meta-analysis of randomized intervention trials. Antioxidants 2021, 10, 223. [Google Scholar] [CrossRef]
  17. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Ali, T.; Kim, T.; Rehman, S.U.; Khan, M.S.; Amin, F.U.; Khan, M.; Ikram, M.; Kim, M.O. Natural Dietary Supplementation of Anthocyanins via PI3K/Akt/Nrf2/HO-1 Pathways Mitigate Oxidative Stress, Neurodegeneration, and Memory Impairment in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 6076–6093. [Google Scholar] [CrossRef]
  19. Chen, L.; Teng, H.; Jia, Z.; Battino, M.; Miron, A.; Yu, Z.; Cao, H.; Xiao, J. Intracellular signaling pathways of inflammation modulated by dietary flavonoids: The most recent evidence. Crit. Rev. Food Sci. Nutr. 2018, 58, 2908–2924. [Google Scholar] [CrossRef]
  20. Yi, L.-T.; Liu, B.-B.; Li, J.; Luo, L.; Liu, Q.; Geng, D.; Tang, Y.; Xia, Y.; Wu, D. BDNF signaling is necessary for the antidepressant-like effect of naringenin. Prog. Neuropsychopharmacol. Biol. Psychiatry 2014, 48, 135–141. [Google Scholar] [CrossRef]
  21. Yi, L.-T.; Li, C.-F.; Zhan, X.; Cui, C.-C.; Xiao, F.; Zhou, L.-P.; Xie, Y. Involvement of monoaminergic system in the antidepressant-like effect of the flavonoid naringenin in mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 2010, 34, 1223–1228. [Google Scholar] [CrossRef]
  22. Khan, H.; Perviz, S.; Sureda, A.; Nabavi, S.M.; Tejada, S. Current standing of plant derived flavonoids as an antidepressant. Food Chem. Toxicol. 2018, 119, 176–188. [Google Scholar] [CrossRef]
  23. Moher, D.; Shamseer, L.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; Group, P.-P. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst. Rev. 2015, 4, 1. [Google Scholar] [CrossRef] [Green Version]
  24. Higgins, J.P.T.; Altman, D.G.; Gøtzsche, P.C.; Jüni, P.; Moher, D.; Oxman, A.D.; Savovic, J.; Schulz, K.F.; Weeks, L.; Sterne, J.A.C.; et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 2011, 343, d5928. [Google Scholar] [CrossRef] [Green Version]
  25. Stang, A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur. J. Epidemiol. 2010, 25, 603–605. [Google Scholar] [CrossRef] [Green Version]
  26. Higgins, J.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions. Res. Syn. Meth. 2011, 2, 126–130. [Google Scholar]
  27. Begg, C.B.; Mazumdar, M. Operating Characteristics of a Rank Correlation Test for Publication Bias. Biometrics 1994, 50, 1088. [Google Scholar] [CrossRef] [PubMed]
  28. Egger, M.; Smith, G.D.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test. Br. Med. J. 1997, 315, 629–634. [Google Scholar] [CrossRef] [Green Version]
  29. Abdelhamid, Z.S.A.; Serry, Z.M.; Elnahas, N.M.G.; Ammar, N.M. Serum serotonin response to aerobic exercise verus phoenix. Int. J. PharmTech Res. 2016, 9, 108–114. [Google Scholar]
  30. Amsterdam, J.D.; Shults, J.; Soeller, I.; Mao, J.J.; Rockwell, K.; Newberg, A.B. Chamomile (Matricaria recutita) may provide antidepressant activity in anxious, depressed humans: An exploratory study. Altern. Ther. Health Med. 2012, 18, 44–49. [Google Scholar] [PubMed]
  31. Estrella, R.E.N.; Landa, A.I.; Lafuente, J.V.; Gargiulo, P.A. Effects of antidepressants and soybean association in depressive menopausal women. Acta Pol. Pharm. 2014, 71, 323–327. [Google Scholar]
  32. Gleason, C.E.; Fischer, B.L.; Dowling, N.M.; Setchell, K.D.R.; Atwood, C.S.; Carlsson, C.M.; Asthana, S. Cognitive Effects of Soy Isoflavones in Patients with Alzheimer’s Disease. J. Alzheimer’s Dis. JAD 2015, 47, 1009–1019. [Google Scholar] [CrossRef] [Green Version]
  33. Hirose, A.; Terauchi, M.; Akiyoshi, M.; Owa, Y.; Kato, K.; Kubota, T. Low-dose isoflavone aglycone alleviates psychological symptoms of menopause in Japanese women: A randomized, double-blind, placebo-controlled study. Arch. Gynecol. Obstet. 2016, 293, 609–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ibero-Baraibar, I.; Perez-Cornago, A.; Ramirez, M.J.; Martínez, J.A.; Zulet, M.A. An Increase in Plasma Homovanillic Acid with Cocoa Extract Consumption Is Associated with the Alleviation of Depressive Symptoms in Overweight or Obese Adults on an Energy Restricted Diet in a Randomized Controlled Trial. J. Nutr. 2015, 146, 897S–904S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ishiwata, N.; Melby, M.K.; Mizuno, S.; Watanabe, S. New equol supplement for relieving menopausal symptoms: Randomized, placebo-controlled trial of Japanese women. Menopause 2009, 16, 141–148. [Google Scholar] [CrossRef]
  36. Jou, H.J.; Ling, P.Y.; Wu, S.C. Comparison of 70 mg and 35 mg isoflavone soya supplement for menopause symptoms. Int. J. Gynecol. Obstet. 2005, 90, 159–160. [Google Scholar] [CrossRef] [PubMed]
  37. Kok, L.; Kreijkamp-Kaspers, S.; Grobbee, D.E.; Lampe, J.W.; van der Schouw, Y.T. A randomized, placebo-controlled trial on the effects of soy protein containing isoflavones on quality of life in postmenopausal women. Menopause 2005, 12, 56–62. [Google Scholar] [CrossRef] [Green Version]
  38. Krikorian, R.; Shidler, M.D.; Nash, T.A.; Kalt, W.; Vinqvist-Tymchuk, M.R.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplementation improves memory in older adults. J. Agric. Food Chem. 2010, 58, 3996–4000. [Google Scholar] [CrossRef] [Green Version]
  39. Lau, H.; Shahar, S.; Mohamad, M.; Rajab, N.F.; Yahya, H.M.; Din, N.C.; Hamid, H.A. The effects of six months Persicaria minor extract supplement among older adults with mild cognitive impairment: A double-blinded, randomized, and placebo-controlled trial. BMC Complement. Med. Ther. 2020, 20, 315. [Google Scholar] [CrossRef]
  40. Lingaerde, O.; Føreland, A.R.; Magnusson, A. Can winter depression be prevented by Ginkgo biloba extract? A placebo-controlled trial. Acta Psychiatr. Scand. 1999, 100, 62–66. [Google Scholar] [CrossRef]
  41. Amsterdam, J.D.; Li, Q.S.; Xie, S.X.; Mao, J.J. Putative Antidepressant Effect of Chamomile (Matricaria chamomilla L.) Oral Extract in Subjects with Comorbid Generalized Anxiety Disorder and Depression. J. Altern. Complement. Med. 2020, 26, 813–819. [Google Scholar] [CrossRef]
  42. Lipovac, M.; Chedraui, P.; Gruenhut, C.; Gocan, A.; Stammler, M.; Imhof, M. Improvement of postmenopausal depressive and anxiety symptoms after treatment with isoflavones derived from red clover extracts. Maturitas 2010, 65, 258–261. [Google Scholar] [CrossRef]
  43. Loftis, J.M.; Wilhelm, C.J.; Huckans, M. Effect of epigallocatechin gallate supplementation in schizophrenia and bipolar disorder: An 8-week, randomized, double-blind, placebo-controlled study. Ther. Adv. Psychopharmacol. 2013, 3, 21–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Malaguarnera, G.; Bertino, G.; Chisari, G.; Motta, M.; Vecchio, M.; Vacante, M.; Caraci, F.; Greco, C.; Drago, F.; Nunnari, G.; et al. Silybin supplementation during HCV therapy with pegylated interferon-α plus ribavirin reduces depression and anxiety and increases work ability. BMC Psychiatry 2016, 16, 398. [Google Scholar] [CrossRef] [Green Version]
  45. Morgan, S.L.; Baggott, J.E.; Moreland, L.; Desmond, R.; Kendrach, A.C. The safety of flavocoxid, a medical food, in the dietary management of knee osteoarthritis. J. Med. Food 2009, 12, 1143–1148. [Google Scholar] [CrossRef] [PubMed]
  46. Park, M.; Choi, J.; Lee, H.J. Flavonoid-rich orange juice intake and altered gut microbiome in young adults with depressive symptom: A randomized controlled study. Nutrients 2020, 12, 1815. [Google Scholar] [CrossRef] [PubMed]
  47. Platero, J.L.; Cuerda-Ballester, M.; Sancho-Cantus, D.; Benlloch, M.; Ceron, J.J.; Rubio, C.P.; García-Pardo, M.P.; López-Rodríguez, M.M.; de la Rubia Ortí, J.E. The impact of epigallocatechin gallate and coconut oil treatment on cortisol activity and depression in multiple sclerosis patients. Life 2021, 11, 353. [Google Scholar] [CrossRef] [PubMed]
  48. Quattrocchi, T.; Micali, E.; Gentile, A.; La Ferrera, E.G.; Barbaro, L.; Ciarcià, S.; Corrado, F.; Di Costa, M.; Fazio, R.; Licenziato, R.; et al. Effects of a phyto complex on well-being of climacteric women. J. Obstet. Gynaecol. Res. 2015, 41, 1093–1098. [Google Scholar] [CrossRef]
  49. Rondanelli, M.; Opizzi, A.; Solerte, S.B.; Trotti, R.; Klersy, C.; Cazzola, R. Administration of a dietary supplement (N-oleyl-phosphatidylethanolamine and epigallocatechin-3-gallate formula) enhances compliance with diet in healthy overweight subjects: A randomized controlled trial. Br. J. Nutr. 2009, 101, 457–464. [Google Scholar] [CrossRef] [Green Version]
  50. Santos-Galduróz, R.F.; Galduróz, J.C.F.; Facco, R.L.; Hachul, H.; Tufik, S. Effects of isoflavone on the learning and memory of women in menopause: A double-blind placebo-controlled study. Braz. J. Med. Biol. Res. 2010, 43, 1123–1126. [Google Scholar] [CrossRef] [Green Version]
  51. Singhal, S.; Shullai, W. Comparative study of gabapentin and isoflavone in menopausal vasomotor symptoms. J. MidLife Health 2016, 7, 132–139. [Google Scholar] [CrossRef]
  52. Atteritano, M.; Mazzaferro, S.; Bitto, A.; Cannata, M.L.; D’Anna, R.; Squadrito, F.; Macrì, I.; Frisina, A.; Frisina, N.; Bagnato, G. Genistein effects on quality of life and depression symptoms in osteopenic postmenopausal women: A 2-year randomized, double-blind, controlled study. Osteoporos. Int. J. Establ. Result Coop. Eur. Found. Osteoporos. Natl. Osteoporos. Found. USA 2014, 25, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  53. Terauchi, M.; Horiguchi, N.; Kajiyama, A.; Akiyoshi, M.; Owa, Y.; Kato, K.; Kubota, T. Effects of grape seed proanthocyanidin extract on menopausal symptoms, body composition, and cardiovascular parameters in middle-aged women: A randomized, double-blind, placebo-controlled pilot study. Menopause 2014, 21, 990–996. [Google Scholar] [CrossRef] [PubMed]
  54. van Dongen, M.C.J.M.; van Rossum, E.; Kessels, A.G.H.; Sielhorst, H.J.G.; Knipschild, P.G. The efficacy of ginkgo for elderly people with dementia and age-associated memory impairment: New results of a randomized clinical trial. J. Am. Geriatr. Soc. 2000, 48, 1183–1194. [Google Scholar] [CrossRef] [PubMed]
  55. Wahner-Roedler, D.L.; Thompson, J.M.; Luedtke, C.A.; King, S.M.; Cha, S.S.; Elkin, P.L.; Bruce, B.K.; Townsend, C.O.; Bergeson, J.R.; Eickhoff, A.L.; et al. Dietary soy supplement on fibromyalgia symptoms: A randomized, double-blind, placebo-controlled, early phase trial. Evid. Based Complement. Altern. Med. 2011, 2011, 350697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Xiao, H.; Wignall, N.; Brown, E.S. An open-label pilot study of icariin for co-morbid bipolar and alcohol use disorder. Am. J. Drug Alcohol Abuse 2016, 42, 162–167. [Google Scholar] [CrossRef] [PubMed]
  57. Zarghami, M.; Chabra, A.; Azadbakht, M.; Khalilian, A.; Asghar Hoseini, A. Antidepressant Effect of Asperugo procumbens L. in Comparison with Fluoxetine: A Randomized Double Blind Clinical Trial. Res. J. Pharmacogn. 2018, 5, 15–20. [Google Scholar] [CrossRef]
  58. Zhang, Q.; Yang, H.; Wang, J.; Li, A.; Zhang, W.; Cui, X.; Wang, K. Effect of green tea on reward learning in healthy individuals: A randomized, double-blind, placebo-controlled pilot study. Nutr. J. 2013, 12, 84. [Google Scholar] [CrossRef] [Green Version]
  59. Brock, C.; Whitehouse, J.; Tewfik, I.; Towell, T. American Skullcap (Scutellaria lateriflora): A randomised, double-blind placebo-controlled crossover study of its effects on mood in healthy volunteers. Phyther. Res. PTR 2014, 28, 692–698. [Google Scholar] [CrossRef]
  60. Brown, E.S.; Bice, C.; Putnam, W.C.; Leff, R.; Kulikova, A.; Nakamura, A.; Ivleva, E.I.; Van Enkevort, E.; Holmes, T.; Miingi, N. Human safety and pharmacokinetics study of orally administered icariin: Randomized, double-blind, placebo-controlled trial. Nat. Prod. Commun. 2019, 14. [Google Scholar] [CrossRef]
  61. Casini, M.L.; Marelli, G.; Papaleo, E.; Ferrari, A.; D’Ambrosio, F.; Unfer, V. Psychological assessment of the effects of treatment with phytoestrogens on postmenopausal women: A randomized, double-blind, crossover, placebo-controlled study. Fertil. Steril. 2006, 85, 972–978. [Google Scholar] [CrossRef]
  62. Chedraui, P.; San Miguel, G.; Schwager, G. The effect of soy-derived isoflavones over hot flushes, menopausal symptoms and mood in climacteric women with increased body mass index. Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol. 2011, 27, 307–313. [Google Scholar] [CrossRef] [PubMed]
  63. Coe, S.; Cossington, J.; Collett, J.; Soundy, A.; Izadi, H.; Ovington, M.; Durkin, L.; Kirsten, M.; Clegg, M.; Cavey, A.; et al. A randomised double-blind placebo-controlled feasibility trial of flavonoid-rich cocoa for fatigue in people with relapsing and remitting multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2019, 90, 507–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. de Sousa-Muñoz, R.L.; Filizola, R.G. Efficacy of soy isoflavones for depressive symptoms of the climacteric syndrome. Maturitas 2009, 63, 89–93. [Google Scholar] [CrossRef] [PubMed]
  65. Hakim, B.N.; Suzana, S.; Haron, H. Association between isoflavones consumption and cognitive function and comorbidities among older adults residing in the State of Johor, Malaysia. Malays. J. Nutr. 2016, 22, 29–39. [Google Scholar]
  66. Chang, S.-C.; Cassidy, A.; Willett, W.C.; Rimm, E.B.; O’Reilly, E.J.; Okereke, O.I. Dietary flavonoid intake and risk of incident depression in midlife and older women. Am. J. Clin. Nutr. 2016, 104, 704–714. [Google Scholar] [CrossRef] [Green Version]
  67. Godos, J.; Castellano, S.; Ray, S.; Grosso, G.; Galvano, F. Dietary polyphenol intake and depression: Results from the mediterranean healthy eating, lifestyle and aging (MEAL) study. Molecules 2018, 23, 999. [Google Scholar] [CrossRef] [Green Version]
  68. Miyake, Y.; Tanaka, K.; Okubo, H.; Sasaki, S.; Furukawa, S.; Arakawa, M. Soy isoflavone intake and prevalence of depressive symptoms during pregnancy in Japan: Baseline data from the Kyushu Okinawa Maternal and Child Health Study. Eur. J. Nutr. 2018, 57, 441–450. [Google Scholar] [CrossRef]
  69. Miyake, Y.; Tanaka, K.; Okubo, H.; Sasaki, S.; Furukawa, S.; Arakawa, M. Dietary patterns and depressive symptoms during pregnancy in Japan: Baseline data from the Kyushu Okinawa Maternal and Child Health Study. J. Affect. Disord. 2018, 225, 552–558. [Google Scholar] [CrossRef]
  70. Nagata, C.; Shimizu, H.; Takami, R.; Hayashi, M.; Takeda, N.; Yasuda, K. Hot flushes and other menopausal symptoms in relation to soy product intake in Japanese women. Climacteric J. Int. Menopause Soc. 1999, 2, 6–12. [Google Scholar] [CrossRef]
  71. Richard, A.; Rohrmann, S.; Mohler-Kuo, M.; Rodgers, S.; Moffat, R.; Güth, U.; Eichholzer, M. Urinary phytoestrogens and depression in perimenopausal US women: NHANES 2005–2008. J. Affect. Disord. 2014, 156, 200–205. [Google Scholar] [CrossRef] [PubMed]
  72. Rosli, H.; Shahar, S.; Din, N.C.; Haron, H.; Rajab, N.F. Prevalence of Poor Mental Health and Cognitive Status among Middle-Aged Adults and Its Predictors in Relation to Polyphenols Intake. Malays. J. Med. Sci. MJMS 2019, 26, 72–89. [Google Scholar] [CrossRef] [PubMed]
  73. Woo, J.; Lynn, H.; Lau, W.Y.; Leung, J.; Lau, E.; Wong, S.Y.S.; Kwok, T. Nutrient intake and psychological health in an elderly Chinese population. Int. J. Geriatr. Psychiatry 2006, 21, 1036–1043. [Google Scholar] [CrossRef]
  74. Cui, Y.; Huang, C.; Momma, H.; Niu, K.; Nagatomi, R. Daily dietary isoflavone intake in relation to lowered risk of depressive symptoms among men. J. Affect. Disord. 2020, 261, 121–125. [Google Scholar] [CrossRef] [PubMed]
  75. Krishnamoorthy, Y.; Rajaa, S.; Rehman, T. Diagnostic accuracy of various forms of geriatric depression scale for screening of depression among older adults: Systematic review and meta-analysis. Arch. Gerontol. Geriatr. 2020, 87, 104002. [Google Scholar] [CrossRef] [PubMed]
  76. Lewinsohn, P.M.; Seeley, J.R.; Roberts, R.E.; Allen, N.B. Center for epidemiologic studies depression scale (CES-D) as a screening instrument for depression among community-residing older adults. Psychol. Aging 1997, 12, 277–287. [Google Scholar] [CrossRef]
  77. Richter, P.; Werner, J.; Heerlein, A.; Kraus, A.; Sauer, H. On the validity of the Beck Depression Inventory. A review. Psychopathology 1998, 31, 160–168. [Google Scholar] [CrossRef]
  78. Dunlop, B.W.; Parikh, S.V.; Rothschild, A.J.; Thase, M.E.; Debattista, C.; Conway, C.R.; Forester, B.P.; Mondimore, F.M.; Shelton, R.C.; Macaluso, M.; et al. Comparing sensitivity to change using the 6-item versus the 17-item Hamilton depression rating scale in the GUIDED randomized controlled trial. BMC Psychiatry 2019, 19, 420. [Google Scholar] [CrossRef] [Green Version]
  79. Thomas-Odenthal, F.; Molero, P.; van der Does, W.; Molendijk, M. Impact of review method on the conclusions of clinical reviews: A systematic review on dietary interventions in depression as a case in point. PLoS ONE 2020, 15, e0238131. [Google Scholar] [CrossRef]
  80. Molendijk, M.; Molero, P.; Ortuño Sánchez-Pedreño, F.; Van der Does, W.; Angel Martínez-González, M. Diet quality and depression risk: A systematic review and dose-response meta-analysis of prospective studies. J. Affect. Disord. 2018, 226, 346–354. [Google Scholar] [CrossRef] [Green Version]
  81. Kimball, S.M.; Mirhosseini, N.; Rucklidge, J. Database analysis of depression and anxiety in a community sample—response to a micronutrient intervention. Nutrients 2018, 10, 152. [Google Scholar] [CrossRef] [Green Version]
  82. Edman, J.S.; Morrow, F.D.; Marby, D.W.; Mirages, S.; Perrone, G.; Kayne, H.L.; Cole, J.O. B Complex Vitamin Patterns in Geriatric and Young Adult Inpatients with Major Depression. J. Am. Geriatr. Soc. 1991, 39, 252–257. [Google Scholar] [CrossRef]
  83. Eby, G.A.; Eby, K.L. Rapid recovery from major depression using magnesium treatment. Med. Hypotheses 2006, 67, 362–370. [Google Scholar] [CrossRef]
  84. Deacon, G.; Kettle, C.; Hayes, D.; Dennis, C.; Tucci, J. Omega 3 polyunsaturated fatty acids and the treatment of depression. Crit. Rev. Food Sci. Nutr. 2017, 57, 212–223. [Google Scholar] [CrossRef]
  85. Spedding, S. Vitamin D and depression: A systematic review and meta-analysis comparing studies with and without biological flaws. Nutrients 2014, 6, 1501–1518. [Google Scholar] [CrossRef]
  86. Li, Z.; Wang, W.; Xin, X.; Song, X.; Zhang, D. Association of total zinc, iron, copper and selenium intakes with depression in the US adults. J. Affect. Disord. 2018, 228, 68–74. [Google Scholar] [CrossRef]
  87. Guan, L.P.; Liu, B.Y. Antidepressant-like effects and mechanisms of flavonoids and related analogues. Eur. J. Med. Chem. 2016, 121, 47–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Brown, D. Do Food Frequency Questionnaires Have Too Many Limitations? J. Am. Diet. Assoc. 2006, 106, 1541–1542. [Google Scholar] [CrossRef] [PubMed]
  89. Rendeiro, C.; Rhodes, J.S.; Spencer, J.P.E. The mechanisms of action of flavonoids in the brain: Direct versus indirect effects. Neurochem. Int. 2015, 89, 126–139. [Google Scholar] [CrossRef] [PubMed]
  90. Maes, M.; Fišar, Z.; Medina, M.; Scapagnini, G.; Nowak, G.; Berk, M. New drug targets in depression: Inflammatory, cell-mediated immune, oxidative and nitrosative stress, mitochondrial, antioxidant, and neuroprogressive pathways. And new drug candidates—Nrf2 activators and GSK-3 inhibitors. Inflammopharmacology 2012, 20, 127–150. [Google Scholar] [CrossRef] [PubMed]
  91. Felger, J.C.; Lotrich, F.E. Inflammatory cytokines in depression: Neurobiological mechanisms and therapeutic implications. Neuroscience 2013, 246, 199–229. [Google Scholar] [CrossRef] [Green Version]
  92. Scapagnini, G.; Davinelli, S.; Drago, F.; De Lorenzo, A.; Oriani, G. Antioxidants as antidepressants: Fact or fiction? CNS Drugs 2012, 26, 477–490. [Google Scholar] [CrossRef]
  93. Davinelli, S.; Maes, M.; Corbi, G.; Zarrelli, A.; Willcox, D.C.; Scapagnini, G. Dietary phytochemicals and neuro-inflammaging: From mechanistic insights to translational challenges. Immun. Ageing 2016, 13, 16. [Google Scholar] [CrossRef] [Green Version]
  94. Williams, C.M.; El Mohsen, M.A.; Vauzour, D.; Rendeiro, C.; Butler, L.T.; Ellis, J.A.; Whiteman, M.; Spencer, J.P.E. Blueberry-induced changes in spatial working memory correlate with changes in hippocampal CREB phosphorylation and brain-derived neurotrophic factor (BDNF) levels. Free Radic. Biol. Med. 2008, 45, 295–305. [Google Scholar] [CrossRef]
  95. Spencer, J.P.E.; Rice-Evans, C.; Williams, R.J. Modulation of pro-survival Akt/protein kinase B and ERK1/2 signaling cascades by quercetin and its in vivo metabolites underlie their action on neuronal viability. J. Biol. Chem. 2003, 278, 34783–34793. [Google Scholar] [CrossRef] [Green Version]
  96. Spencer, J.P.E. Flavonoids and brain health: Multiple effects underpinned by common mechanisms. Genes Nutr. 2009, 4, 243–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Pei, R.; Liu, X.; Bolling, B. Flavonoids and gut health. Curr. Opin. Biotechnol. 2020, 61, 153–159. [Google Scholar] [CrossRef] [PubMed]
  98. Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Rémésy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81, 230S–242S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Williamson, G.; Manach, C. Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am. J. Clin. Nutr. 2005, 81, 243S–255S. [Google Scholar] [CrossRef]
  100. Messina, M.; Gleason, C. Evaluation of the potential antidepressant effects of soybean isoflavones. Menopause 2016, 23, 1348–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Davinelli, S.; Scapagnini, G.; Marzatico, F.; Nobile, V.; Ferrara, N.; Corbi, G. Influence of equol and resveratrol supplementation on health-related quality of life in menopausal women: A randomized, placebo-controlled study. Maturitas 2017, 96, 77–83. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA flow diagram for systematic review and meta-analysis.
Figure 1. PRISMA flow diagram for systematic review and meta-analysis.
Antioxidants 10 01644 g001
Figure 2. Forest plots showing the effects of flavonoid consumption on depressive symptoms. (A) Forest plot of the effect of dietary flavonoids on depressive symptoms in 36 clinical trials. (B) Sensitivity analysis summarizing the effect of flavonoids on depressive symptoms.
Figure 2. Forest plots showing the effects of flavonoid consumption on depressive symptoms. (A) Forest plot of the effect of dietary flavonoids on depressive symptoms in 36 clinical trials. (B) Sensitivity analysis summarizing the effect of flavonoids on depressive symptoms.
Antioxidants 10 01644 g002
Figure 3. Forest plots for subgroup analyses. (A) Forest plot based on the type of flavonoid subclass; (B) type of clinical trial (controlled and uncontrolled) and (C) level of blinding.
Figure 3. Forest plots for subgroup analyses. (A) Forest plot based on the type of flavonoid subclass; (B) type of clinical trial (controlled and uncontrolled) and (C) level of blinding.
Antioxidants 10 01644 g003
Figure 4. Funnel plots for publication bias. (A) Primary analysis with 36 studies and (B) sensitivity analysis with 26 studies.
Figure 4. Funnel plots for publication bias. (A) Primary analysis with 36 studies and (B) sensitivity analysis with 26 studies.
Antioxidants 10 01644 g004
Table 1. Data summary of clinical intervention trials assessing the effects of flavonoids on depressive symptoms.
Table 1. Data summary of clinical intervention trials assessing the effects of flavonoids on depressive symptoms.
Study
(Author, Year,
Country, Ref.)
Study DesignSubjectsConditionInterventionDepression
Scale
Results
Abdelhamid et al.,
2016; Egypt [29]
Randomized clinical
trial
Duration: 24 weeks
100 subjects (60–70 y)
(n = 50 women; n = 50 men)
Mild depressionFlavonoid-rich extract
from date palm fruit
(rutin 11.2 mg/100 mg)
GDSSignificant effect
compared with baseline
(p = 0.0001)
Amsterdam et al.,
2012; US [30]
Randomized double-blind
placebo-controlled trial
Duration: 8 weeks
41 subjects (mean age 42.9 y)
(n = 26 women; n = 15 men)
Anxiety with or
without depression
Chamomile capsules
(220 mg/d;
1.2% apigenin)
HAM-DSignificant effect
(treatment vs. placebo
p < 0.05)
Amsterdam et al.,
2019; US [41]
Randomized open-label
trial
Duration: 8 weeks
179 subjects (mean age 45.7 y)
(n = 119 women; n = 60 men)
Anxiety with or
without depression
Chamomile capsules
(1500 mg/d; 18 mg of
flavonoids)
HRSD Significant effect in
subjects with depression (p < 0.02)
Atteritano et al.,
2014; Italy [52]
Randomized double-blind
controlled trial
Duration: 2 years
262 women
(49–67 y)
Osteopenia and
Postmenopause
Genistein tablets
(54 mg/d)
ZSDSSignificant effect
(treatment vs. placebo
p < 0.01)
Brock et al.,
2014; UK [59]
Randomized double-blind
placebo-controlled
crossover trial
Duration: 2 weeks
31 subjects (mean age 34 y)
(n = 25 women; n = 6 men)
Anxious and
non-anxious
population
Scutellaria lateriflora
capsules (350 mg/d;
11.7 mg/g baicalein;
7.7 mg/g wogonin)
POMSNo significant effect
Brown et al.,
2019; US [60]
Randomized double-blind
placebo-controlled trial
Duration: 5 days
24 subjects (18–50 y)
(men and women)
Healthy
subjects
Icariin capsules
(from 100 to 1.680 mg/d)
QIDS-CSignificant effect
(treatment vs. placebo
p = 0.02)
Casini et al.,
2006; Italy [61]
Randomized double-blind
crossover
placebo-controlled study
Duration: 6 months
77 women
(49–50 y)
PostmenopausePhytoestrogens tablets
(soy isoflavones 60 mg/d;
45% genisytein
45% daidzein
10% glycitein)
BDI and
POMS
Significant effect in BDI
and POMS scores
(treatment vs. placebo
BDI, p < 0.01;
POMS, p < 0.001)
Chedraui et al.,
2011; Ecuador [62]
Single blinded pilot clinical
trial
Duration: 3 months
45 women
(age 40–59 y)
Menopausal
symptoms
Soy isoflavone capsules
(100 mg/d; 40%
isoflavones)
HAM-DSignificant effect
compared with baseline
(p < 0.05)
Coe et al.,
2019; UK [63]
Randomized, double-blind
placebo-
controlled trial
Duration: 6 weeks
41 subjects (mean age 44 y)
(n = 30 women; n = 10 men)
Relapsing and
remitting
multiple sclerosis
Flavonoid-rich cocoa
drink
(intervention group
200 mg/day)
HADSNo significant effect
de Sousa-Muñoz,
and Filizola,
2009; Brazil [64]
Placebo-controlled
double-blind randomized
trial
Duration:16 weeks
79 women
(mean age 53.3 y)
Depressive
symptoms in
climacteric
syndrome
Soy isoflavone capsules
(120 mg/d; 60 mg
isoflavones; 20 mg
daidzein; 14 mg
genistein)
CES-DSignificant effect
compared with baseline
(p = 0.001)
Estrella et al.,
2014; Chile [31]
Randomized clinical trial
Duration: 3 months
20 women
(45–55 y)
Depression in
menopause
Soybean
concentrate (100 mg/d;
50 mg isoflavones)
HAM-D
and ZSDS
Significant effect in
depression scores
compared with baseline
(p < 0.001)
Gleason et al.,
2015; US [32]
Randomized double-blind
parallel group clinical trial
Duration:6 months
59 subjects (>60 y)
(men and women)
Alzheimer’s
disease
Soy isoflavone capsules
(100 mg/d; 85% daidzein,
and genistein)
GDS and
POMS
No significant effect
Hirose et al.,
2016; Japan [33]
Randomized double-blind
placebo-controlled trial
Duration 8 weeks
58 women
(40–60 y)
MenopauseSoy isoflavone tablets
(12.5 mg/d; 25 mg/d;
genistein 51.8%, daidzein
43.3%, glycitein 4.9%)
HADSSignificant effect in
compared with placebo
(p = 0.033)
Ibero-Baraibar et al.,
2015; Spain [34]
Randomized double-blind
placebo-controlled
parallel trial
Duration: 4 weeks
37 subjects (mean age 57 y)
(n = 25 women; n = 22 men)
Overweight or
obese adults
Cocoa extract
(1.4 g/d; 414 mg
flavan-3-ols; 153 mg
epicatechin; 15 mg
catechin; 246 mg
procyanidins),
BDISignificant effect
compared with baseline
(p < 0.01)
Ishiwata et al.,
2009; Japan [35]
Randomized double-blind
placebo-controlled trial
Duration: 12 weeks
37 women
(mean age 46.6 y)
MenopauseEquol capsules
(10 or 30 mg/d)
POMSSignificant effect
compared with placebo
(p < 0.05)
Jou et al.,
2005; Taiwan [36]
Single-centre prospective
randomized trial
Duration: 6 weeks
20 women
(age N/A)
MenopauseSoy isoflavone capsules
(35 or 70 mg/d isoflavones)
GCSSignificant effect in 35 mg
groups compared with
baseline (p < 0.05)
Kok et al.,
2005; Netherlands [37]
Randomized double-blind
placebo-controlled trial
Duration: 12 months
202 women
(60–75 y)
PostmenopauseSoy powder (25.6 g/d;
52 mg/g genistein;
41 mg/g daidzein;
6 mg/g glycitein)
GDSNo significant effect
Krikorian et al.,
2010; Canada [38]
Single-blind
placebo-controlled trial
Duration:12 weeks
9 subjects (mean age 76.2)
(men and women)
Memory declineBlueberry juice
(three doses; 444 mL/d, 0.42 g
anthocyanins; 532 mL/d,
0.51 g anthocyanins;
621 mL/d, 0.59 g
anthocyanins)
GDSNo significant effect
Lau et al.,
2020; Malaysia [39]
Multi-center randomized double-blind
placebo-controlled trial
Duration: 6 months
30 subjects (mean age 66.4 y)
(n = 23 women; n = 7 men)
Mild cognitive
impairment
Persicaria minor extract
(500 mg/d; 0.45%
quercetin-3-glucuronide;
0.15% quercitrin)
POMSNo significant effect
Lingaerde et al.,
1999; Norway [40]
Randomized double-blind
placebo-controlled trial
Duration: 10 weeks
27 subjects (26–58 y)
(n = 21 women; n = 6 men)
Seasonal affective
disorder
Ginkgo biloba tablets
(120–160 mg/d; 24
mg flavones)
MADRSNo significant effect
Lipovac et al.,
2010; Austria [42]
Randomized double-blind
placebo-controlled trial
Duration: 3 months
109 women
(mean age 53.5 y)
Postmenopausal
symptoms
Isoflavone capsules
(80 mg/d;
extract in form of
HADS and
ZSDS
Significant effect in
HADS and ZSDS
scores compared
Loftis et al.,
2013; US [43]
Randomized double-blind
placebo-controlled trial
Duration: 8 weeks
25 subjects (≥18 years)
(men and women)
Schizophrenia and
bipolar disorder
biochanin A,
formononetin,
genistein and daidzein)
EGCG capsules
(N/A mg/d;
capsules enriched with
150 mg of theaflavin)
HAM-Dwith control
(p < 0.001)
No significant effect
Malaguarnera et al.;
2016; Italy [44]
Randomized double-blind
placebo-controlled trial
Duration: 1 year
62 subjects (mean age 46 y)
(n = 26 women; n = 36 men)
Hepatitis C virus
infection
Silybin pills (active group:
1.5 mg/kg/week
Peg–IFN plus RBV +
silybin 94 mg +
vitamin E 30 mg +
phospholipids 194 mg)
BDISignificant effect
in intervention group
compared with placebo
(p < 0.05)
Morgan et al.,
2009; US [45]
Randomized double-blind
placebo-controlled trial
Duration: 12 weeks
19 subjects (40–75 y)
(men and women)
Moderate
osteoarthritis
Scutellaria baicalensis
and Acacia catechu
pills
(500 mg/d; N/A mg/g of
flavonoids)
BDINo significant effect
Park et al.,
2020; South Korea [46]
Randomized single-blind
controlled trial
Duration: 8 weeks
40 subjects (mean age 21.8 y)
(n = 24 women; n = 16 men)
Depressed and
non-depressed
population
Flavonoid-rich orange juice
(380 mL/d; 600 mg
flavonoids)
CES-DSignificant effect
compared with baseline
(p < 0.0001)
Platero et al.,
2021; Spain [47]
Randomized
placebo-controlled trial
Duration: 4 months
46 subjects (mean age 47.1 y)
(n = 32 women; n = 14 men)
Multiple
sclerosis
EGCG capsules
(800 mg/d)
with
coconut oil (60 mL/d)
BDISignificant effect
compared with baseline
(p = 0.007)
Quattrocchi et al.,
2015; Italy [48]
Intervention trial
Duration: 6 months
131 women
(42–67 y)
Menopausal
symptoms
Phyto complex (200 mg/d;
20% isoflavones, 1.5%
vitexin)
BDISignificant effect
compared with baseline
(p < 0.05)
Rondanelli et al.,
2009; Italy [49]
Randomized double-blind
placebo-controlled
parallel trial
Duration: 2 months
116 subjects (18–50 y)
(n = 89 women; n = 27 men)
OverweightEGCG capsules
(50 mg/d)
BDISignificant effect
compared with placebo
(p < 0·005)
Santos- Galduróz et al.,
2010; Brazil [50]
Randomized double-blind
placebo-controlled trial
Duration: 4 months
38 women
(50–65 y)
MenopauseIsoflavone tablets
(80 mg/d;
60.8 mg genistein; 16 mg
daidzein; 3.2 mg
glycitein)
GDSNo significant effect
Singhal et al.,
2016; India [51]
Comparative clinical
Trial
Duration: 3 months weeks
100 women
(46–55 y)
Menopausal
Vasomotor
symptoms
Isoflavone tablets
(60 mg/d)
ZSDSSignificant effect
compared with control
(p = 0.02)
Terauchi et al.,
2014; Japan [53]
Randomized double-blind
placebo-controlled pilot
trial
Duration: 8 weeks
91 women
(40–60 y)
Menopausal
symptoms
Grape seed
Proanthocyanidin
tablets (100 or 200 mg/d;
85% proanthocyanidins;
15% flavan-3-ols)
HADSNo significant effect
van Dongen et al.,
2000; Netherlands [54]
Randomized double-blind
placebo-controlled
parallel group
multicenter trial
Duration: 24 weeks
123 subjects (mean age 82.8)
(men and women)
Age-associated
Memory
impairment
Ginko biloba tablets
(160 or 240 mg/d;
24% ginkgo flavonols)
GDSNo significant effect
Wahner-Roedler et al., 2011; US [55]Randomized double-blind
placebo-controlled
early phase trial.
Duration: 6 weeks
28 women
(mean age 53.9 y)
Fibromyalgia
symptoms
Soy shake
(N/A mL/d;
160 mg soy
isoflavones)
CES-DNo significant effect
Xiao et al.,
2016; US [56]
Open-label
Trial
Duration: 8 weeks
10 subjects (18–70 y)
(men and women)
Alcohol abuse and
bipolar disorder
Icariin capsules
(300 mg/d)
HAM-DSignificant effect
compared with baseline
(p = 0.01)
Zarghami et al.,
2018; Iran [57]
Randomized double-blind
clinical trial
Duration: 6 weeks
25 subjects (18–70 y)
(n = 19 women; n = 6 men)
Mild depressionAsperugo procumbens
capsules (1.2 g/d;
6 mg total flavonoids)
HRSDSignificant lower effect
compared with antidepressant
control
(p = 0.03)
Zhang et al.,
2013; China [58]
Randomized double-blind
placebo-controlled
pilot trial
Duration: 5 weeks
46 subjects (mean age: 25.7 y)
(n = 23 women; n = 23 men)
Reward function
in healthy subject
Green tea
(400 mg/d;
EGCG 45.6%;
epigallocatechin 16.7%;
epicatechin-3-gallate
11.4%; epicatechin
6.8%)
MADRS
And
HRSD
Significant effect
compared with control
(MADRS p < 0.01;
HRSD p < 0.001)
GDS, Geriatric Depression Scale; HRSD, Hamilton Rating Scale for Depression; HAM-D, Hamilton Depression Rating; ZSDS, Zung Self-Rating Depression Scale; POMS, Profile of Mood States; QIDS-C, Quick Inventory of Depressive Symptomatology Clinician-Rated; HADS, Hospital Anxiety and Depression Scale; CES-D, Center of Epidemiologic Studies of Depression; EGCG, Epigallocatechin-gallate; BDI, Beck Depression Inventory; N/A, Not Available; GCS, Greene Climacteric Scale; MADRS, Montgomery–Asberg Depression Rating Scale; Peg–IFN, Pegylated Interferon.
Table 2. Data summary of observational studies assessing the effects of flavonoids on depressive symptoms.
Table 2. Data summary of observational studies assessing the effects of flavonoids on depressive symptoms.
Study
(Author, Year,
Country, Ref.)
Study DesignSubjects
and Sample Size
Main
Variable
Exposure
Measure
Depression
Scale
Results
Chang et al.,
2016; US [66]
Longitudinal cohort study
Duration: 1976–2001
82,648 women who
participated in the NHS
(mean age of 67 y at baseline)
and NHSII
(mean age of 47 y at baseline)
Dietary
Flavonoid
intake
FFQ
(total flavonoid
intake ranged from
127.6 to 779.4 mg/d)
CES-D and
GDS
Participants in the highest
flavonoid consumption group
had a 7–10% reduction in
depression risk
compared with the
lowest intake group
(p-trend = 0.0004–0.08).
Flavones and
proanthocyanidins showed
the strongest associations
Cui et al.,
2020; Japan [74]
Cross-sectional study
Duration: 2008–2011
1335 men
(19–83 y)
Dietary
Isoflavone
intake
DHQ
(isoflavone intake
ranged from ≤10.61
to ≥25.79 mg/d)
SDSA high level of dietary
isoflavone intake was
associated with a lower
prevalence of depressive
symptoms in all the adjusted
models
(p-trend = 0.002–0.029)
Godos et al.,
2018; Italy [67]
Cross-sectional study
Duration: 2014–2015
1572 participants who
participated in the MHELAS
(18–92 y)
(n = 912 women; n = 660 men)
Dietary
Polyphenol
intake
FFQ
(total flavonoid
intake ranged from
157.0 to 543.7 mg/d)
CES-DHigher dietary intake of
flavanones and anthocyanins
was inversely associated with
depressive symptoms
(p-trend = 0.001)
Hakim et al.,
2016; Malaysia [65]
Longitudinal cohort study
Duration: 18 months
400 elderly participants
(≥60 y)
(n = 231 women; n = 169 men)
Dietary
Isoflavone
intake
DHQ
(mean intake:
isoflavones 19.1 mg/d,
daidzein 11.7 mg/d,
genistein 7.6 mg/d)
GDSNo association
between
isoflavone intake
and depression
Miyake et al.,
2018; Japan [68]
Cross-sectional study1745 pregnant women (mean
age 31 y) who participated
in the KOMCHS
(an ongoing prospective
prebirth cohort study)
Soy
Isoflavone
intake
DHQ
(isoflavone intake
ranged from
10.5 to 49.4 mg/d)
CES-DIsoflavone intake was
associated with a lower
prevalence of depressive
symptoms during pregnancy
in all the quartiles
(p-trend = 0.002)
Miyake et al.,
2018; Japan [69]
Cross-sectional study1744 pregnant women (mean
age 31.2 y) who participated
in the KOMCHS
Dietary
patterns
DHQ
(maximum isoflavone intake:
Healthy pattern 43.5 mg/d
Japanese pattern 32.7 mg/d
Western pattern 31.2 mg/d)
CES-DThe healthy and Japanese
patterns were
inversely associated with
depressive symptoms during
pregnancy
(Healthy pattern p-trend< 0.0001;
Japanese pattern p-trend = 0.008)
Nagata et al.,
1999; Japan [70]
Cross-sectional study
Duration: September 1996–
August 1997
284 menopausal women
(mean age 47.1 y)
Soy product
intake
FFQ (total isoflavone
intake from soy product:
38.6 mg/d;
isoflavone intake from
fermented soy product
12.6 mg/d)
CES-DNo association
between
soy products and depression
Richard et al.,
2014; Switzerland [71]
Cross-sectional study
Duration: 2005–2008
193 perimenopausal women
who participated in the
NHANES
(mean age 49 y)
Urinary
phyto-
estrogens
HPLC-APPI-MS/MS
(mean value depressed vs.
non-depressed women:
isoflavones 128.3 and 97.4
μg/g; daidzein 57.4 and 48.3
μg/g; genistein 27.6 and 21.6 μg/g)
PHQ-9No association between urinary
isoflavone
concentrations and depression
Rosli et al.,
2019; Malaysia [72]
Cross-sectional study349 participants
with mild mental
health problem
(45–59 y)
(n = 228 women; n = 121 men)
Dietary
Polyphenols
intake
FFQ
(total flavonoid
intake 265 mg/d)
GHQ-28No association
between flavonoid
intake and depression
Woo et al.,
2006; Hong Kong [73]
Cross-sectional study3999 participants
(≥65 y) (women and men)
Nutrient
intake
FFQ
(total isoflavone
intake ranged from
4 to ≥19 mg/d)
GDSNo association
between isoflavone
intake and depression
DHQ, Diet History Questionnaire; GDS, Geriatric Depression Scale; NHS, Nurses’ Health Study; FFQ, Food-Frequency Questionnaire; SDS, Self-rating Depression Scale; MHELAS, Mediterranean Healthy Eating, Lifestyle and Aging Study; CES-D, Center of Epidemiologic Studies of Depression; SDS, KOMCHS, Kyushu Okinawa Maternal and Child Health Study; NHANES, National Health and Nutrition Survey; Patient Health Questionnaire-9; HPLC-APPI-MS/MS, High Performance Liquid Chromatography/Atmospheric Pressure Photoionization//Tandem mass spectrometry; GHQ-28, General Health Questionnaire-28.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ali, S.; Corbi, G.; Maes, M.; Scapagnini, G.; Davinelli, S. Exploring the Impact of Flavonoids on Symptoms of Depression: A Systematic Review and Meta-Analysis. Antioxidants 2021, 10, 1644. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10111644

AMA Style

Ali S, Corbi G, Maes M, Scapagnini G, Davinelli S. Exploring the Impact of Flavonoids on Symptoms of Depression: A Systematic Review and Meta-Analysis. Antioxidants. 2021; 10(11):1644. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10111644

Chicago/Turabian Style

Ali, Sawan, Graziamaria Corbi, Michael Maes, Giovanni Scapagnini, and Sergio Davinelli. 2021. "Exploring the Impact of Flavonoids on Symptoms of Depression: A Systematic Review and Meta-Analysis" Antioxidants 10, no. 11: 1644. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10111644

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