Next Article in Journal
The Attachment-Caregiving Questionnaire as a Personality Inventory Sensitive to Psychological Vulnerabilities: A Pilot Study
Previous Article in Journal
Examining Intentions for Impact: Understanding What Influences the Planning of High-Level Team Sport Coaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Integrative Interventions for Improving Outcomes in Depression: A Narrative Review

by
Matthew Halma
1,*,
Christof Plothe
1 and
Paul E. Marik
2,*
1
EbMC Squared CIC, Bath BA2 4BL, UK
2
Frontline COVID-19 Critical Care Alliance, Washington, DC 20036, USA
*
Authors to whom correspondence should be addressed.
Psychol. Int. 2024, 6(2), 550-577; https://doi.org/10.3390/psycholint6020033
Submission received: 18 February 2024 / Revised: 28 March 2024 / Accepted: 17 April 2024 / Published: 29 April 2024
(This article belongs to the Section Neuropsychology, Mental Health and Brain Disorders)

Abstract

:
Antidepressants are among the most used medications in the US, with significant deleterious effects on people’s well-being. At any given time, depression impacts approximately 1 in 10 Americans, causing wide and broad societal costs. Interest is developing for non-pharmacological treatments and preventative measures. We summarize the literature on non-invasive dietary and lifestyle approaches for treating depression. This review aims to inform future research and treatment programs for depression by providing an evidentiary summary of integrative therapeutic approaches for depression.

1. Introduction

Worldwide, the prevalence of depression among adults is estimated to be 28.4% [1]. Among adolescents, the one-year prevalence of depression is 8% and the lifetime prevalence of depression is 19% [2]. Among adolescents, the prevalence of elevated depressive symptoms rose by nearly 50% between the decade of the 2000s and the decade of the 2010s [2]. Depression also affects healthcare workers and is more common among medical professionals than in the general population [3,4].
Beyond the immediate effects of depression, those with depression are at higher risk for many other conditions, including heart attacks [5], diabetes [6], and suicide [7]. The debilitating nature of the condition makes it difficult for one to enjoy a fulfilling life with social connections [8]. Depression has many multifarious impacts on one’s career prospects and one’s ability to experience joy from goal pursuit or enacting a hobby [9].
Depression can also affect others besides the depressed person; depressed people often withdraw from social relationships [10], and those they do still meet with can be influenced through mental contagion [11,12].
In the Netherlands, about 1 in every 13 adults is currently using an antidepressant [13]. In recent years, the trend has been towards the increasing duration of antidepressant use [14], and in the US two-thirds of patients continue antidepressants for at least two years [15,16].

2. Epidemiology

While depression is seen as a condition affecting solely the mind, still epidemiological factors underlie depression at the population level. Depression is more common in women [17], and peaks in the 45 to 59 years age group [18]. Rural residents also have lower rates of depression compared to urban dwellers [19]. Those involved in some spiritual practice reduce the risk of developing depression by half [20].
Characteristics of one’s family of origin can predispose one to depression. While the mechanism of causation is unclear, the children of parents with major depressive disorder (MDD) are three times more likely to develop MDD than the children of parents without MDD [21]. A history of childhood abuse is associated with a greater risk of depression [22,23], as is a lack of parental affection [24]. Social contagion of moods is possible, and being around other depressed people may contribute to depression [11].
Other factors associated with depression are low education, recent negative life events, loneliness, alcohol consumption, low physical activity [25,26], and smoking [27]. The personality traits of low agreeableness, low extraversion, low openness, low mastery, low conscientiousness, and high neuroticism are also associated with depression [27]. Internet addiction is also associated with a greater depression risk [28]. Significant life disruptions can also precipitate the onset of depression, including heart attacks [29] and even childbirth [30]. Due to the mental nature of depression, a discussion about its causes traverses many questions about how one is living one’s life, including work [31,32], relationships, and self-care.
Nutritional associations have also been investigated. Coffee consumption was associated with a lower risk of depression in women [33]. Vegetable and fruit consumption was also associated with a lower risk of depression [34], and dietary magnesium and calcium significantly lowered the depression risk [35]. A recent umbrella review of the dietary associations with depression prevention and treatment demonstrated a significant protective benefit from healthy diet patterns [36]. Unhealthy beverage consumption habits, as parametrized by the Healthy Beverage Index (HBI) score [37], were also associated with an increased depression risk [38].
Specific factors showing strong evidence for decreased depression risk included fish consumption, coffee or tea consumption, dietary zinc, and light to moderate alcohol (<40 g/day) [36]. Consumption of sugar-sweetened beverages also raised the risk of depression [36]. Moderate-quality evidence exists for the association of consumption of probiotics, omega-3 polyunsaturated fatty acid, and acetyl-L-carnitine with decreased depression risk [36].
Other dietary factors studied for their anti-depression effect, revealing equivocal evidence for efficacy, include cocoa-rich foods [39], red or processed meat [40], vitamin D [41], folic acid [42], and B vitamins [43]. Genetic factors can contribute to or protect against the depression phenotype [44,45,46,47], and heritability of depression is estimated at 37% from twin studies [48].
Exposure to nature is associated with better affective states and lower risk of depression [49,50,51,52], an effect which can be mediated by the quality of the urban built environment [53,54]. Sociality can also protect against depression, as evidenced by an interventional study, where women with chronic depression in London made new friends, and observed a significant improvement in their present state examination (PSE) scores [55], an assessment of effect [56]. Additionally, having hobbies is associated with a lower risk of depression [57,58].
High-stress environments and jobs can also contribute to depression, though this relationship is mediated by other factors [59], including (perceived) level of support [60] and psychosocial safety in the workplace [61].

3. Aims and Methods

This narrative review aims to include non-pharmacological, evidence-based treatments for the treatment of depression. We divide these into two broad categories of interventions: dietary/nutraceutical and lifestyle therapies.
The methodology of this article begins with first performing a manual search for (1) dietary factors impacting depression, including specific supplements and herbal treatments, and (2) lifestyle factors influencing depression. The search strategy searches for reviews summarizing the interventions in each category. When reviews are found, the interventions summarized in the review are included in Supplementary Table S1 for dietary interventions with clinical evidence. Agents with only preclinical evidence are included in Supplementary Table S2. Lifestyle interventions are included in Supplementary Table S3. The dietary interventions with human clinical evidence for depression treatment are included in Section 4. The lifestyle interventions with human clinical evidence are included in Section 5.

4. Nutritional Support for Depression Treatment

As mentioned, the nutritional factors behind depression have been elucidated in meta-analyses. Vegetarian diets are associated with a higher rate of depression in people [62], whereas Mediterranean diets are associated with a lower risk of depression [63,64]. Other specific nutrient deficiencies and their impact on depression are outlined in Table 1.
Nutraceuticals in the context of depression have been reviewed in [42,65,66,67,68,69,70,71,72,73]. Several nutritional deficiencies may exist in the depressed patient [74], which, if addressed, may positively influence the prognosis of depression.
We searched for reviews on nutritional supplements in depression and found several reviews providing an evidentiary overview of different nutraceutical and nutritional supplementary protocols for depression (Supplementary Table S1) [42,65,66,67,68,69,70,75,76,77,78,79,80,81,82]. The specific interventions are included in Table 1.
Table 1. A summary of dietary agents and their impacts on depression.
Table 1. A summary of dietary agents and their impacts on depression.
FactorImpactOptimal Serum LevelsDaily Intake in Depression Treating ContextSources
ZincZinc supplementation significantly lowered depressive symptom scores (Beck’s Depression Inventory, BDI) WMD = −4.15; [−6.56, −1.75] [83]70–120 micrograms per deciliter (mcg/dL) for adults25 mg zinc sulfate or 30 mg zinc gluconate [83]Meat, shellfish, dairy, legumes, nuts
MagnesiumConsumption associated with lower risk of depression RR = 0.81 [0.70, 0.92] [35]0.75–0.95 millimoles per liter (mmol/L) for adults248 mg [84]Leafy greens, nuts and seeds, legumes
CaffeineAssociated with reduced depression risk RR = 0.72 [0.52, 1.00] highest vs. lowest consumption [85]N/ABetween 68 mg/day and 509 mg/day [85]Coffee, tea
CocoaDecrease in depressive symptoms g = −0.42 [−0.67, −0.17] [39]N/A50–100 g/day cocoaCocoa
FishLowers depression risk RR = 0.89 [0.80, 0.99] highest vs. lowest consumption [86]
RR = 0.83 [0.74, 0.93] highest vs. lowest consumption [87]
N/A>1 serving per week [86]Fish
Omega 3 polyunsaturated fatty acidsLowered depression risk RR = 0.87 [0.74, 1.04] highest vs. lowest consumption [86]
EPA + DHA consumption associated with lower depression risk [88]
N/A500 mg/day [86]Fatty Fish
SeleniumIntake associated with lower risk of postpartum depression OR = 0.97 [0.95, 0.99] and reduction in depressive symptoms WMD = −0.37 [−0.56, −0.18] [89]Average level 124 ng/mL [90]100 to 200 μg [89]Wheat products, meat [91]
B-vitaminsNon-significant reduction in depressive symptoms (SMD = 0.15 [−0.01, 0.32]) [43]N/AN/ALiver, fish, leafy greens, eggs, seeds
BiotinAssociated with lower odds of depression (OR = 0.71 [0.55, 0.91]) [92]>400 ng/L [93]30 μg [94]Organ meat, egg yolk, some vegetables, milk [95]
Folic acidAssociated with lower odds of depression OR = 0.78 [0.61, 0.99] [92]Deficiency is defined as serum folate < 10 nmol/L and RBC folate < 340 nmol/L [96]240 μg [94]Legumes, leafy greens, citrus, vegetables, liver, dairy products [97]
Vitamin B12No significant effect on depressive symptoms [98]Deficiency is defined as plasma vitamin B12 < 150 pmol/L [96]2.4 μg [94]Liver, fish, leafy greens, eggs, seeds
Vitamin DIn cases of deficiency, vitamin D supplementation may help depressive symptoms [99]
Inverse correlation between serum vitamin D levels and depression [100]
Serum 25-Hydroxyvitamin D: 50–100
nmol/L [101]
>1000 IU [99]Sunlight [102], oily fish, fortified foods [103]
ProbioticsSmall but significant effects for trials lasting at least one month (SMD = −0.28, [−0.44, −0.13]) [104]
Significant difference in depression score (SMD = −0.47 [−0.67, −0.27]) [105]
Other meta-analyses reveal no significant difference, though very close to statistical threshold of p = 0.05 (SMD = −0.128, [−0.261, 0.005]) [106]
Biomarkers are multifactorial [107]10 billion CFU [108]Yogurts, kefir [109], kombucha [110], fermented meat and fish products, sauerkraut, kimchi, natto, miso, sourdough bread [111,112]
Acetyl-L-CarnitineSignificant reduction in depressive symptoms (SMD = −1.10, [−1.65, −0.56]) [113]10–15 μmol/L [114]2 g [115]Meat [116]
CreatineReduction in depression associated with level of dietary creatine consumption
AOR = 0.68 [0.52, 0.88] [117]
N/A2–10 g [118]Meat [119]
Amino acids (a.a.)Reduction in depressive symptoms greater than placebo SMD = −1.21 [0.57, 1.95] [120]Varies by specific a.a.
For tryptophan: 40–120 µmol/L [121]
Recommended Daily Allowance (RDA) doses of 8 essential and 2 semi-essential amino acids (arginine and histidine) [122]Protein rich foods, supplements
MethylfolateImprovement in depression profile SMD = −0.38 [−0.59, −0.17] [123]
SMD = −0.61 [−0.97, −0.24] [124]
Serum 5-methyltetrahydrofolate
24–51
nmol/L [125]
15 mg [124]Leafy greens,
legumes,
fortified cereals, liver
5-HTPSignificant improvements in depression symptoms (g = 1.11 [0.53, 1.69] [126]N/A150–300 mg [126]Turkey, chicken, fish, dairy products, supplements
St. John’s WortSimilar response to SSRI treatment. RR = 0.96 [0.83, 1.10] relative to second generation antidepressants [127]N/A500 mg [128]Hypericum perforatum
SaffronSignificantly better than placebo improvement in depressive symptoms g = 0.891 [0.369, –1.412] [129]N/A100 mg [129]Saffron spice derived from the Crocus sativus flower
CurcuminSignificant clinical efficacy in depression (HAM-D SMD = −0.34 [−0.56 to −0.13]) [130]
Effective as adjunctive therapy [131]
N/A1 g [130]Turmeric spice, commonly used in curry dishes and various recipes
Methylene BlueReduction in manic depressive attacks [132]
Marked improvement in depressive symptoms SMD = −0.99 [−1.82, −0.16] [133]
N/A15 mg/daySupplements
Chinese Herbal MedicinePositive effect [134,135]
CHM better than placebo (HAMD-17, MD = −4.53, [−5.69, −3.37]) [134]
N/AN/ADepends on formulation
Nigella SativaDecreased depression score SMD = −1.4 [−1.94, −0.86] [136]N/A1000 mg oil extractBlack cumin seed
S-adenosyl methionineLow-quality evidence for efficacy [137]N/A1600 mg orally [138]Supplements
Bacopa MonnieriNonsignificant improvement
SMD = −0.32 [−0.86, 0.22]
[139]
N/A300 mg extract [139]Bacopa Monnieri
SHR-5 (Rhodiola metabolite)Improves depressive symptoms SMD = −1.66 [−2.17, −1.16] [140]N/A340−680 mg Rhodiola [140]Rhodiola rosea L.
Kava kavaImprovement in symptoms in human subjects
SMD = 2.24 (p < 0.0001) [141]
N/A3.2 g [141]Piper methysticum
InositolEquivocal evidence [68]7 μg/mL [142]12 g [143]Fruits, beans, grains, and nuts [144]
ChromiumRCT shows effectiveness compared to placebo nonsignificant
SMD = −0.538 [−1.72, 0.65] [145]
<0.60 μg/L [146]600 μg chromium picolinate [145]Meats, grain products, fruits, vegetables, nuts, spices, brewer’s yeast, beer, and wine [147]
Co-enzyme Q10SMD = 0.97 [0.01, 1.93] p < 0.00001 [69]Males: 0.9 μmol/L
Females: 0.8 μmol/L [148]
300 mg [69]Meat, fish, nuts, and some oils [149]
CrocinSMD = 6.04 [3.43, 8.65] p = 0.01 [69]N/A30 mg [69]Saffron
Antioxidant supplementsSignificant improvement (SMD = 0.40, 95% CI = 0.28–0.51, p < 0.00001) [69]N/AN/ASupplements
Extra Virgin Olive OilAntidepressant activity in severely depressed patients SMD = −0.75 [−1.23, −0.27] [150]N/A25 mL extra virgin olive oil [150]Extra virgin olive oil
LavenderPositive impact of lavender with imipramine (antidepressant) compared to imipramine monotherapy SMD = 2.45 [1.67, 3.23] [78]N/A60 drops lavandula tincture [151]Lavandula angustifolia
Dan zhi xiao yaoDecrease in Self-Rating Depression Scale scores [WMD = 0.89, 95% CI (−6.33, 8.11); p = 0.81] [152]N/A24 g [152]Mixture of Bupleurum chinense, Scutellaria baicalensis, Paeonia lactiflora, Glycyrrhiza uralensis, Mentha haplocalyx, Zingiber officinale, and Ziziphus jujuba
Alpha Lipoic AcidEquivocal evidence [68]N/A600–1800 mg [153]Muscle meats, heart, kidney, and liver [154]
N-acetyl Cysteine (NAC)Positive evidence from trials [155,156]N/A1 g [156]Supplements
GinsengImprovements in QOL in patients complaining of stressor fatigue [157]N/A17.4 mg Panax Ginseng extract with a blend of multivitamins [157]Ginseng
Other agents with preclinical data are shown in Supplementary Table S2 [141,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218]. Preclinical studies typically focus on several behavioral tests in mice.

4.1. The Gut Microbiome and Depression: The Importance of the Gut–Brain Axis

Depression is a complex mental health disorder that affects millions worldwide. While the exact causes of depression are not fully understood, emerging research suggests that the gut microbiome may play a significant role in its development and progression. The gut–brain axis, a bidirectional communication system between the enteric microbiota and the central nervous system, is thought to be a key mediator in this relationship and seems to have significant implications for depression.

4.1.1. The Gut–Brain Axis

The gut–brain axis refers to the intricate interactions between the enteric microbiota, the central nervous system (CNS), and the enteric nervous system (ENS) [219], creating a paradigm change in neuroscience [220]. The enteric microbiota, consisting of trillions of microorganisms residing in the gastrointestinal tract, influences various physiological processes, including the immune function, metabolism, and neurotransmitter production [221]. These microorganisms produce neurotransmitters, such as serotonin and gamma-aminobutyric acid (GABA), which are known to regulate mood and emotions [222] and even modify epigenetic processes of the gut–brain axis [223].

4.1.2. The Role of the Gut Microbiome in Depression

Studies have found alterations in the composition and diversity of the gut microbiome in individuals with depression [224]. Researchers [225] highlighted the bidirectional communication between the gut microbiota and the CNS, emphasizing the gut microbiome’s influence on neurological and psychiatric disorders [226]. Additionally, others [227] discussed the impact of the gut–brain axis on mental health, emphasizing the potential therapeutic benefits of modulating the gut microbiota.

4.1.3. Mechanisms

Several mechanisms have been proposed to explain how the gut microbiome may contribute to depression [222]. Short-chain fatty acids (SCFAs) [228], produced by the gut microbiota through the fermentation of dietary fibers, have been shown to modulate brain function and behavior [229]. Scientists [230] discussed the role of SCFAs in microbiota–gut–brain communication, highlighting their potential as therapeutic targets. Moreover, a dysregulated microbiota–gut–brain axis has been observed in patients with bipolar depression [231,232] and associated with depressive-like behaviors in animal models [224,226]. Emerging evidence suggests that alterations in gut permeability [233] and the subsequent inflammatory response may play a crucial role in the relationship between the gut microbiome and depression [234]. Research demonstrated [221] how the gut microbiome influences the production and metabolism of neurotransmitters such as serotonin [235], dopamine [236], and gamma-aminobutyric acid (GABA) [237], and how alterations in these neurotransmitter systems may contribute to depressive symptoms.

4.1.4. Clinical Implications and Treatment Approaches

Understanding the gut–brain axis and its association with depression opens up possibilities for novel therapeutic interventions. In studies, a predominance of some potentially harmful bacterial groups or a reduction in beneficial bacteria [232] has been found in depressive patients. Dietary interventions have been the subject of research and studies examining their potential impact on symptoms of depression. There is emerging evidence that suggests a link between diet and mental well-being, indicating that dietary improvements may positively affect symptoms of depression [71,238]. Probiotics, which are live microorganisms that confer health benefits when consumed, have shown promise in modulating the gut microbiota and improving depressive symptoms. Research [239] reviewed the mechanisms of action of probiotics as potential therapeutic targets for depression and anxiety disorders. For example, Lactobacillus rhamnosus directly regulates the GABAergic system in a vagus nerve-dependent way and mitigates depression- and anxiety-like behaviors in mice [240]. Bifidobacterium breve, proven to have an antidepressant-like effect, could stimulate the production of intestinal 5-hydroxytryptophan in mice and then regulate the host’s serotonin metabolism [241,242]. Pediococcus acidilactici could mitigate anxiety symptoms in mice by producing lactic acid and inhibiting the over-proliferated gut pathogenic bacteria under stress [243]. Fecal Microbiota Transplantation (FMT) is a procedure in which a healthy donor’s fecal matter is transplanted into a recipient’s gastrointestinal tract to restore a healthy balance of gut bacteria. There is growing interest in the potential therapeutic effects of FMT on various conditions, including depression [244,245].
Overall, the gut microbiome and the gut–brain axis are emerging areas of research in the field of depression. The bidirectional communication between the gut microbiota and the CNS highlights the potential for microbiome-based interventions in treating depression. While promising, more research is required to elucidate the underlying mechanisms and develop targeted therapies to modulate the gut–brain axis to alleviate depressive symptoms effectively.

4.2. The Link between Depression and Inflammation

Depression, a prevalent mental health disorder, has long been associated with alterations in the immune system and chronic inflammation. The findings highlight potential therapeutic targets and the importance of a holistic approach to managing depression. The etiology of depression remains multifactorial and complex; emerging evidence suggests a strong connection between depression and inflammation [246]. Inflammation, traditionally associated with the immune response to infection or injury, has been implicated in the pathophysiology of various psychiatric disorders. Numerous studies have demonstrated elevated levels of pro-inflammatory cytokines [247], such as interleukin-6 (IL-6) [248,249] and tumor necrosis factor-alpha (TNF-α) [250], in individuals with depression. Conversely, chronic inflammation, often triggered by external factors such as stress, trauma, or medical conditions, has been shown to contribute to developing or exacerbating depressive symptoms. The dysregulation of the immune system, particularly the imbalance in pro-inflammatory and anti-inflammatory cytokines, plays a crucial role in altering neurotransmitter metabolism, neuroplasticity, and the neuroendocrine function, ultimately affecting mood regulation [251].
The bidirectional relationship between depression and inflammation suggests a complex interplay between the immune and central nervous systems. Inflammation-induced activation of the kynurenine pathway [252], dysregulation of the hypothalamic–pituitary–adrenal (HPA) axis [253], and disruption of the blood–brain barrier [254] are among the proposed mechanisms linking inflammation to depressive symptoms. Moreover, chronic inflammation may impair the efficacy of conventional antidepressant treatments, emphasizing the need for personalized approaches that target both the neurochemical imbalances and the underlying inflammatory processes. It is worth noting that studies indicate that EMF exposure can increase the secretion of pro-inflammatory cytokines [255], including IL-6, TNF-alpha, and IL-1. The increasing intensity of EMF via mobile phones, Wi-Fi, etc., should initiate more research into the potential association between depression and EMF devices. This pro-inflammatory effect has been shown to be inhibited by curcumin [256].
Curcumin, a compound found in turmeric, has been the subject of scientific research exploring its potential use in depression [130,257,258,259]. Curcumin has been found to possess anti-inflammatory properties, which, as we can see, may be relevant to depression. Inflammation has been implicated in the development and progression of depression, and curcumin’s anti-inflammatory effects may help alleviate depressive symptoms [260]. Curcumin has also shown neuroprotective properties in preclinical studies, including antioxidant and anti-apoptotic effects. These effects may help protect against neuronal damage and promote neuroplasticity, essential factors in depression [131]. It has been found to modulate various neurotransmitters, including serotonin, dopamine, and glutamate, which are involved in mood regulation. By influencing these neurotransmitter systems, curcumin may impact depressive symptoms [260]. BDNF is a protein that plays a crucial role in the growth and maintenance of neurons. Reduced levels of BDNF have been associated with depression. Curcumin has been shown to increase BDNF levels, possibly contributing to its potential antidepressant effects [261]. Some studies have explored the combination of curcumin with other antidepressant medications, suggesting possible synergistic effects. Combining curcumin with standard antidepressant treatment may enhance the therapeutic response [258,259].

4.3. The Complex Relationship between Thyroid Dysfunction and Depression

Thyroid dysfunction refers to the abnormal functioning of the thyroid gland, which can result in either hyperthyroidism (overactive thyroid) or hypothyroidism (underactive thyroid). Depression, on the other hand, is a mood disorder characterized by persistent feelings of sadness, loss of interest, and a lack of motivation. While the connection between thyroid dysfunction and depression has been the subject of scientific inquiry [262,263,264], the relationship between these two conditions remains complex and multifaceted [265]. Research has shown a bidirectional relationship between thyroid dysfunction and depression, with each condition potentially influencing the other [266]. Several studies have found that individuals with thyroid dysfunction are at a higher risk of developing depression. For instance, a meta-analysis found a significant association between hypothyroidism and depression [267], suggesting that individuals with an underactive thyroid may be more prone to depressive symptoms. Moreover, thyroid hormones play a crucial role in regulating neurotransmitters [268] such as serotonin [235], dopamine [269], and norepinephrine [270], which are involved in mood regulation. Imbalances in these neurotransmitters have been linked to the development of depression. Therefore, disruptions in thyroid hormone levels can impact the functioning of these neurotransmitters, potentially contributing to the development of depressive symptoms [271]. Conversely, depression may also affect thyroid function. Chronic stress, a common contributor to depression, can lead to dysregulation of the hypothalamic–pituitary–thyroid (HPT) axis [272] which controls thyroid hormone production. This dysregulation can result in alterations in thyroid hormone levels [273,274], potentially leading to thyroid dysfunction. Autoimmune thyroiditis is also associated with an increased risk of depression [275]. Elevated thyroid-stimulating hormone (TSH), antithyroglobulin (TgAb), and thyroid peroxidase antibodies (TPOAb) levels have all been linked to depression and an increased risk of suicide [266]. Moreover, hypothyroidism is known to be one of the leading causes of treatment-resistant depression. Furthermore, chronic inflammation, often observed in individuals with depression, can also impact thyroid function. The complex relationship between thyroid dysfunction and depression necessitates comprehensive treatment approaches that address both conditions. For individuals with thyroid dysfunction, appropriate thyroid hormone replacement therapy can help restore hormonal balance and alleviate depressive symptoms. It is crucial to closely monitor thyroid hormone levels and micronutrients, such as iodine, zinc [276], iron [277], and selenium [278], and adjust medication dosages as necessary. If an individual with depression also exhibits symptoms of thyroid dysfunction, it is important to assess thyroid function and consider appropriate interventions to optimize treatment outcomes [279]. Thyroid dysfunction and depression share a complex and bidirectional relationship. While individuals with thyroid dysfunction may be at a higher risk of developing depression [280], depression can also impact thyroid function [273]. Addressing both conditions simultaneously is crucial for effective treatment outcomes. Further research is needed to unravel the precise mechanisms underlying this relationship and develop targeted interventions that can improve the lives of individuals affected by both thyroid dysfunction and depression.

5. Lifestyle Changes for Treatment of Depression

There are several changes that one can make in one’s life to recover from depression. These useful strategies have an evidence base documenting their efficacy. We performed a literature search on lifestyle treatment for depression and found several reviews (Supplementary Table S3) [281,282,283,284,285,286,287,288,289]. These findings are summarized below in Table 2.

5.1. Exercise

There is growing recognition that lifestyle behaviors, such as physical activity and exercise, can be useful strategies for treating depression, reducing depressive symptoms, improving quality of life, and improving physical health outcomes. Cross-sectional studies have shown that people with higher levels of physical activity present decreased depressive symptoms, and these results are consistent across different countries and cultures. For example, recent evidence using data from the Brazilian National Health Survey, accounting for 59,399 individuals, demonstrated that a lack of physical activity for leisure was associated with depression in young males, and middle-aged and older adults [301]. A study across 36 countries demonstrated that lower levels of physical activity (defined as less than 150 min of moderate–vigorous physical activity per week) were consistently associated with elevated depression (OR, 1.42; 95%CI, 1.24–1.63) [302]. However, mental health benefits have been noted from being physically active, even at levels below the public health recommendations [303]. In The Irish Longitudinal Study on Ageing, participants performing 400 to less than 600 MET-min/wk had a 16% lower rate of depressive symptoms (adjusted incidence rate ratio [AIRR], 0.84; 95% CI, 0.81–0.86) and 43% lower odds of depression compared with 0 MET-min/wk [304]. These findings are consistent with recent meta-analytic data suggesting that salutary mental health benefits among adults can be achieved with physical activity below public health recommendations; specifically, an activity volume equivalent to 2.5 h per week of brisk walking was associated with a 25% lower risk of depression, and half that activity volume was associated with an 18% lower risk compared with no activity [303]. The findings of The Irish Longitudinal Study on Ageing suggest that accumulating as little as 100 min per week or 20 min per day for 5 days per week of moderate-intensity activity (e.g., brisk walking; 4 METs) may be sufficient to significantly lower the risk of depressive symptoms and odds of major depression over time among older adults.
A large body of trials has been performed over the last 40 years evaluating the role of exercise as a therapy for depression. These results have been summarized in several meta-analyses. In a Cochrane analysis of 35 trials (1356 participants) comparing exercise with no treatment or a control intervention, the pooled outcome for the primary outcome of depression at the end of treatment was a standardized mean difference (SMD) of −0.62; 95% CI −0.81 to −0.42, indicating a moderate clinical effect. Schuch et al. performed a meta-analysis which included 25 RCTs comparing exercise versus control comparison groups. [305] Overall, exercise had a large and significant effect on depression. Similarly, Krogh et al. performed a meta-analysis which included 35 trials enrolling 2498 participants [306]. The effect of exercise versus control on depression severity was −0.66 SMD [95% CI −0.86 to −0.46; p < 0.001).
Exercise can improve depressive symptoms in people with depression. However, like other treatments, exercise is not a panacea and may not work equally for all. A seminal study by Dunn et al. named “The Depression Outcomes Study of Exercise” found a response rate of about 40% in depressed people free from other treatments [307]. However, it is likely that when combined with other interventions (i.e., vitamin D, L-methyl-folate, etc.) the response rate and degree of response will be much greater. In essence, exercise has multiple benefits to several domains of physical and mental health and should be promoted to everyone. To ensure compliance, adapting exercise prescriptions for people with depression should account for personal preferences and previous experiences in terms of making it the most enjoyable experience possible. Acute exercise should be used as a symptom management tool to improve mood in depression, with even light exercise an effective recommendation [308]. These data suggest that physical activity is beneficial for the depressed patient regardless of the intensity of the exercise.
The neurobiological mechanisms underpinning the antidepressant effects of exercise are largely unclear. However, some hypotheses involving inflammation, oxidative stress, and neuronal regeneration are speculated. Exercise training can promote increases in anti-inflammatory and antioxidant enzymes, referred to as a hormesis response, and subsequently decrease IL-6 levels. This effect was demonstrated in the REGASSA trial, where decreases in IL-6 serum levels were associated with reductions in depressive symptoms [309].

5.2. Time in Nature

Time in nature is associated with increases in positive mood and lowered feelings of depression [310,311].

Animal-Assisted Therapy

Time spent with animals can be an effective way of reducing depression [295].

5.3. Mindfulness

Several mindfulness-based therapies can potently treat depression. The most studied treatments are cognitive behavioral therapy (CBT), mindfulness-based stress reduction (MBSR), and mindfulness-based cognitive therapy (MBCT), which have important distinctions. Mindfulness-based therapies demonstrate significant reductions in depression [292].

5.4. Connection with Others

In the middle of depression, some of the things that fall by the wayside are plans and social interactions. Existing in large cities, one lives a largely anonymized existence, where one does not experience connection with others, including seeing others and being seen by others.

Purpose and Goals

Positive, goal-directed activity is associated with a decrease in depressive symptoms and has the added benefit of providing structure and a reason to positively interact and create with others. Progress in any aspect increases positive self-regard, confidence, and a sense of self-efficacy, as well as one’s social status. These factors are associated with a decrease in depressive symptoms [312,313].
Another benefit is that learning positively uses neural pathways and grows new neurons and is also associated with a sense of optimism. Furthermore, a challenging task necessarily takes much of the mental bandwidth, leaving less space for ruminations characteristic of depression. During periods of intense stress including the London Blitz during World War II, there was a paradoxical decrease in psychiatric presentation to hospitals, owing to the dire need of hospital beds [314]. The efforts of every man and woman were needed, and this sense of purpose is protective against depression.
Depression is often a reason for introspection into the aspects of one’s life that are not working. Often a major life area, such as one’s career or one’s close relationships (or lack thereof), is brought into focus. In these cases where dissatisfaction with one’s current life is the proximal cause of one’s depression, working with a life coach or otherwise reflecting on one’s ideal life (and how to achieve it) is a powerful practice for inspiring hope and action which follows that. A significant proportion of depression is a lack of meaning and purpose.
Interestingly, regular Argentinian tango was comparable to mindfulness meditation in terms of the impact on depression [290], suggesting a value in novel pursuits and hobbies.
Indigenous communities living traditional existences do not suffer from psychiatric issues. The differences in the sense of purpose between modern and tribal cultures can be attributed to the tight-knit tribal communities where everybody feels a sense of importance in the eyes of the community. Furthermore, practices such as initiation into manhood and womanhood, most notably the vision quest, provide the individual with a clear role to play in the community.
Over millennia, these practices have corrected wayward youth and integrated at-risk youth into constructive roles within the community. While this review mostly focuses on the individual treatment of depression, it should be noted that initiatives like Upward Bound, which provide a similar experience for youth on the cusp of adulthood, increase the likelihood of post-secondary education [315].
These programs involve people getting out in nature, which in itself has positive benefits for mood disorders [316], and additionally provides the benefits of physical activity [317]. The program positively impacts self-concept [318].

5.5. Gratitude

An outlook of gratitude has been valued by all the major monotheistic religions [319]. Furthermore, in modernity, when gratitude is operationalized as an explicit practice, it is associated with positive mental health, including alleviating depression [298,320,321,322,323]. Gratitude journaling, which simply involves recording 3–5 things that one is grateful for daily, is one of the most accessible ways that one can practice journaling [324,325].

5.6. Deep Brain Stimulation

Noninvasive brain stimulation methods have been studied for their favorable modulation of a wide variety of neural states. For the treatment of depression, some promising data exist for these therapies, showing a small but significant effect [298,321].
Non-invasive brain stimulation (NIBS) using transcranial direct current stimulation or transcranial magnetic stimulation has been demonstrated to be highly effective in the treatment of depression [326,327,328,329,330].

5.7. Whole-Body Hyperthermia

Historically, hyperthermia interventions have been utilized to address depressive symptoms, with evidence dating back to ancient times, such as the practices of Galen of Pergamon (129–198 C.E.), who treated melancholia by immersing patients in hot tubs and providing skin massages [331]. Contemporary research has highlighted the positive effects of regular sauna bathing, including reductions in all-cause and cardiovascular mortality, increased lifespan, improved exercise performance, and the activation of autophagy through the expression of heat-shock proteins [332,333,334,335]. Heat therapy also enhances cell stress pathways, possesses antioxidant and anti-inflammatory properties, and enhances mitochondrial function. Sauna bathing exhibits physiological similarities to aerobic exercise, increasing heart rate, stroke volume, and cardiac output [336,337]. Furthermore, whole-body hyperthermia (WBH) selectively raises IL-6 levels [338] and shows promise in conditions like chronic fatigue syndrome [339,340].
Animal studies have indicated that WBH activates portions of the dorsal raphe nucleus associated with mood regulation and produces antidepressant-like responses [341]. Clinical studies have shown that a single session of WBH can significantly reduce depressive symptoms when assessed five days post-treatment [342]. Additionally, a randomized, double-blind study comparing WBH with a sham condition in depressed patients revealed significant reductions in Depression Rating Scale scores over a six-week post-intervention period in the active WBH group [343]. Hanusch et al. conducted a meta-analysis on the effect of WBH on depression indices, encompassing seven studies with a total of 148 subjects. Six out of seven studies reported statistically significant reductions in depressive symptoms between one and six weeks post-intervention. The treatment effect appeared to be independent of the total number of WBH sessions, with target temperatures between 38 °C and 39 °C and a slower increase in core body temperature during the intervention resulting in larger treatment effects. This suggests potential benefits of a near-infrared (NIR) sauna over a regular sauna, as NIR sauna sessions can be more controlled, shorter in duration (5–10 min initially, increasing to 20 min), and performed two to three times a week for maximal cardiovascular benefit [331].

5.8. Photobiomodulation

Photobiomodulation (PBM) is referred to in the literature as low-level light therapy, red-light therapy, and near-infrared (NIR)-light therapy. Depression is associated with brain hypometabolism and cerebral as well as systemic mitochondrial dysfunction [344,345,346,347,348,349,350,351,352,353,354,355,356]. In a rat model of depression, vital steps in the production of adenosine triphosphate (ATP) were inhibited in the cerebral cortex and cerebellum [347].
Peripheral blood mononuclear cells of depressed patients were shown to have significantly impaired mitochondrial function [348,349], and greater mitochondrial dysfunction correlated with the severity of neuro-vegetative symptoms, including fatigue and poor concentration [348]. Muscle biopsy samples from depressed patients with physical symptoms had a decreased rate of ATP production and more frequent mitochondrial DNA deletions than controls [346].
The most well-studied mechanism of action of PBM centers around enhancing the activity of cytochrome C oxidase (CCO), which is unit four of the mitochondrial respiratory chain, responsible for the final reduction of oxygen to water [350]. The theory is that CCO enzyme activity may be inhibited by nitric oxide (NO). This inhibitory NO can be dissociated by photons of light that are absorbed by CCO. These absorption peaks are mainly in the red (600–700 nm) and near-infrared (760–940 nm) spectral regions. When NO is dissociated, the mitochondrial membrane potential is increased, more oxygen is consumed, more glucose is metabolized and more ATP is produced by the mitochondria [351].
PBM has been found to specifically increase CCO activity and expression [350,352,353]. Studies have also shown increases in complex II, III, and IV activity, as well as upregulation of gene coding for subunits of complex I, complex IV, and ATP synthase. [350] Low-level laser therapy has been shown to increase levels of ATP, the rate of oxygen consumption, and cerebral oxygenation [350]. Though t-PBM with red and NIR light can include wavelengths from 600 to 1070 nm, specific wavelengths have been directly linked to mitochondrial activity. Near Infrared activates CCO, increases mitochondrial oxygen consumption, and leads to higher levels of ATP [354,355,356].
There is some evidence that PBM applied peripherally, not just transcranially, may have an effect in attenuating depressive symptoms [350]. There is no clear mechanism proposed explaining this effect. In a recent study, five outpatients with lower back pain and concurrent self-reported depression were treated over five weeks with physical therapy (PT) (5 sessions) and concurrent PBM (3 sessions) and matched to five control patients treated with PT alone (5 sessions) [357]. Participants receiving s-PBM reported a larger decrease in their depression scores. Oron and co-workers have shown that delivering NIR light to mouse tibia resulted in improvement in a transgenic mouse model of Alzheimer’s disease [358].

6. Conclusions

More integrative approaches, including diet and lifestyle, may improve the quality of life of people with depression and enable them to live a fulfilling life. Currently, practitioner understanding is a barrier, as well as the limited time that primary care physicians spend with patients. Additionally, other systemic issues remain regarding the cost of therapy, while simultaneously therapists themselves are overburdened and under-compensated. Expectations of one’s self can be a barrier to those experiencing depression even in acknowledging it, let alone seeking help [359]. Much work remains to be carried out on the public health understanding of depression, and an integrative approach, combining dietary and lifestyle change with other therapies and embracing a trauma-aware perspective, can help greatly. Additionally, the resiliency of the person experiencing depression must be acknowledged, and he or she must be captain of the process. Practitioners can help by educating and coaching the person recovering from depression, and by pointing them to resources and therapeutics for their specific case.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/psycholint6020033/s1, Table S1: Dietary, nutraceutical and herbal interventions for treating depression in human subjects found in reviews of dietary interventions for depression. Table S2: Dietary, nutraceutical and herbal interventions for treating depression in preclinical models found in reviews of dietary interventions for depression. Table S3: Lifestyle interventions for treating depression in humans found in reviews of lifestyle interventions for depression.

Author Contributions

Conceptualization, P.E.M.; methodology, M.H. and P.E.M.; investigation, M.H., C.P. and P.E.M.; data curation, M.H.; writing—original draft preparation, M.H., C.P. and P.E.M.; writing—review and editing, M.H., C.P. and P.E.M.; supervision, P.E.M.; funding acquisition, P.E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Frontline COVID-19 Critical Care Alliance and The APC was funded by the Frontline COVID-19 Critical Care Alliance.

Acknowledgments

We thank Mazhar Hussain for assisting with reference formatting.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hu, T.; Zhao, X.; Wu, M.; Li, Z.; Luo, L.; Yang, C.; Yang, F. Prevalence of depression in older adults: A systematic review and meta-analysis. Psychiatry Res. 2022, 311, 114511. [Google Scholar] [CrossRef] [PubMed]
  2. Shorey, S.; Ng, E.D.; Wong, C.H.J. Global prevalence of depression and elevated depressive symptoms among adolescents: A systematic review and meta-analysis. Br. J. Clin. Psychol. 2022, 61, 287–305. [Google Scholar] [CrossRef] [PubMed]
  3. Bailey, E.; Robinson, J.; McGorry, P. Depression and suicide among medical practitioners in Australia. Intern. Med. J. 2018, 48, 254–258. [Google Scholar] [CrossRef] [PubMed]
  4. Outhoff, K. Depression in doctors: A bitter pill to swallow. S. Afr. Fam. Pract. 2019, 61 (Suppl. 1), S11–S14. [Google Scholar] [CrossRef]
  5. Glassman, A.H. Depression and cardiovascular comorbidity. Dialogues Clin. Neurosci. 2007, 9, 9–17. [Google Scholar] [CrossRef] [PubMed]
  6. Holt, R.I.G.; de Groot, M.; Golden, S.H. Diabetes and Depression. Curr. Diabetes Rep. 2014, 14, 491. [Google Scholar] [CrossRef] [PubMed]
  7. Li, X.; Mu, F.; Liu, D.; Zhu, J.; Yue, S.; Liu, M.; Liu, Y.; Wang, J. Predictors of suicidal ideation, suicide attempt and suicide death among people with major depressive disorder: A systematic review and meta-analysis of cohort studies. J. Affect. Disord. 2022, 302, 332–351. [Google Scholar] [CrossRef] [PubMed]
  8. Achterbergh, L.; Pitman, A.; Birken, M.; Pearce, E.; Sno, H.; Johnson, S. The experience of loneliness among young people with depression: A qualitative meta-synthesis of the literature. BMC Psychiatry 2020, 20, 415. [Google Scholar] [CrossRef] [PubMed]
  9. Street, H. Exploring Relationships Between Goal Setting, Goal Pursuit and Depression: A Review. Aust. Psychol. 2002, 37, 95–103. [Google Scholar] [CrossRef]
  10. Bosc, M. Assessment of social functioning in depression. Compr. Psychiatry 2000, 41, 63–69. [Google Scholar] [CrossRef]
  11. Eisenberg, D.; Golberstein, E.; Whitlock, J.L.; Downs, M.F. Social contagion of mental health: Evidence from college roommates. Health Econ. 2013, 22, 965–986. [Google Scholar] [CrossRef] [PubMed]
  12. Neumann, R.; Strack, F. “Mood contagion”: The automatic transfer of mood between persons. J. Personal. Soc. Psychol. 2000, 79, 211–223. [Google Scholar] [CrossRef] [PubMed]
  13. Huijbregts, K.M.; Hoogendoorn, A.; Slottje, P.; van Balkom, A.J.L.M.; Batelaan, N.M. Long-Term and Short-Term Antidepressant Use in General Practice: Data from a Large Cohort in the Netherlands. Psychother. Psychosom. 2017, 86, 362–369. [Google Scholar] [CrossRef]
  14. Moore, M.; Yuen, H.M.; Dunn, N.; Mullee, M.A.; Maskell, J.; Kendrick, T. Explaining the rise in antidepressant prescribing: A descriptive study using the general practice research database. BMJ 2009, 339, b3999. [Google Scholar] [CrossRef]
  15. Olfson, M.; Marcus, S.C. National patterns in antidepressant medication treatment. Arch. Gen. Psychiatry 2009, 66, 848–856. [Google Scholar] [CrossRef]
  16. Mojtabai, R.; Olfson, M. National trends in long-term use of antidepressant medications: Results from the U. S. National Health and Nutrition Examination Survey. J. Clin. Psychiatry 2014, 75, 169–177. [Google Scholar] [PubMed]
  17. Noble, R.E. Depression in women. Metabolism 2005, 54 (Suppl. 5), 49–52. [Google Scholar] [CrossRef]
  18. de la Torre, J.A.; Vilagut, G.; Ronaldson, A.; Dregan, A.; Ricci-Cabello, I.; Hatch, S.L.; Serrano-Blanco, A.; Valderas, J.M.; Hotopf, M.; Alonso, J. Prevalence and age patterns of depression in the United Kingdom. A population-based study. J. Affect. Disord. 2021, 279, 164–172. [Google Scholar] [CrossRef]
  19. Romans, S.; Cohen, M.; Forte, T. Rates of depression and anxiety in urban and rural Canada. Soc. Psychiatry Psychiatr. Epidemiol. 2011, 46, 567–575. [Google Scholar] [CrossRef]
  20. Portnoff, L.; McClintock, C.; Lau, E.; Choi, S.; Miller, L. Spirituality cuts in half the relative risk for depression: Findings from the United States, China, and India. Spiritual. Clin. Pract. 2017, 4, 22–31. [Google Scholar] [CrossRef]
  21. Weissman, M.M.; Wickramaratne, P.; Nomura, Y.; Warner, V.; Verdeli, H.; Pilowsky, D.J.; Grillon, C.; Bruder, G. Families at High and Low Risk for Depression: A 3-Generation Study. Arch. Gen. Psychiatry 2005, 62, 29–36. [Google Scholar] [CrossRef] [PubMed]
  22. Sneed, J.R.; Kasen, S.; Cohen, P. Early-life risk factors for late-onset depression. Int. J. Geriatr. Psychiatry 2007, 22, 663–667. [Google Scholar] [CrossRef] [PubMed]
  23. Comijs, H.C.; van Exel, E.; van der Mast, R.C.; Paauw, A.; Oude Voshaar, R.; Stek, M.L. Childhood abuse in late-life depression. J. Affect. Disord. 2013, 147, 241–246. [Google Scholar] [CrossRef] [PubMed]
  24. Parker, G. Parental ‘Affectionless Control’ as an Antecedent to Adult Depression: A Risk Factor Delineated. Arch. Gen. Psychiatry 1983, 40, 956–960. [Google Scholar] [CrossRef] [PubMed]
  25. Zhai, L.; Zhang, Y.; Zhang, D. Sedentary behaviour and the risk of depression: A meta-analysis. Br. J. Sports Med. 2015, 49, 705–709. [Google Scholar] [CrossRef] [PubMed]
  26. Mekary, R.A.; Lucas, M.; Pan, A.; Okereke, O.I.; Willett, W.C.; Hu, F.B.; Ding, E.L. Isotemporal Substitution Analysis for Physical Activity, Television Watching, and Risk of Depression. Am. J. Epidemiol. 2013, 178, 474–483. [Google Scholar] [CrossRef] [PubMed]
  27. Schaakxs, R.; Comijs, H.C.; van der Mast, R.C.; Schoevers, R.A.; Beekman, A.T.F.; Penninx, B.W.J.H. Risk Factors for Depression: Differential Across Age? Am. J. Geriatr. Psychiatry 2017, 25, 966–977. [Google Scholar] [CrossRef] [PubMed]
  28. Kim, Y.-J.; Jang, H.M.; Lee, Y.; Lee, D.; Kim, D.-J. Effects of Internet and Smartphone Addictions on Depression and Anxiety Based on Propensity Score Matching Analysis. Int. J. Environ. Res. Public Health 2018, 15, 859. [Google Scholar] [CrossRef]
  29. Ladwig, K.H.; Ladwig, K.H.; Roll, G.; Breithard, G.; Budde, T.; Borggrefe, M. Post-infarction depression and incomplete recovery 6 months after acute myocardial infarction. Lancet 1994, 343, 20–23. [Google Scholar] [CrossRef]
  30. Miller, L.J. Postpartum Depression. JAMA 2002, 287, 762–765. [Google Scholar] [CrossRef]
  31. Bonde, J.P.E. Psychosocial factors at work and risk of depression: A systematic review of the epidemiological evidence. Occup. Environ. Med. 2008, 65, 438–445. [Google Scholar] [CrossRef] [PubMed]
  32. Netterstrøm, B.; Conrad, N.; Bech, P.; Fink, P.; Olsen, O.; Rugulies, R.; Stansfeld, S. The Relation between Work-related Psychosocial Factors and the Development of Depression. Epidemiol. Rev. 2008, 30, 118–132. [Google Scholar] [CrossRef]
  33. Lucas, M.; Mirzaei, F.; Pan, A.; Okereke, O.I.; Willett, W.C.; O’reilly, J.; Koenen, K.; Ascherio, A. Coffee, Caffeine, and Risk of Depression Among Women. Arch. Intern. Med. 2023, 171, 1571–1578. [Google Scholar] [CrossRef]
  34. Liu, X.; Yan, Y.; Li, F.; Zhang, D. Fruit and vegetable consumption and the risk of depression: A meta-analysis. Nutrition 2016, 32, 296–302. [Google Scholar] [CrossRef] [PubMed]
  35. Li, B.; Lv, J.; Wang, W.; Zhang, D. Dietary magnesium and calcium intake and risk of depression in the general population: A meta-analysis. Aust. N. Z. J. Psychiatry 2016, 51, 219–229. [Google Scholar] [CrossRef]
  36. Xu, Y.; Zeng, L.; Zou, K.; Shan, S.; Wang, X.; Xiong, J.; Zhao, L.; Zhang, L.; Cheng, G. Role of dietary factors in the prevention and treatment for depression: An umbrella review of meta-analyses of prospective studies. Transl. Psychiatry 2021, 11, 478. [Google Scholar] [CrossRef]
  37. Duffey, K.J.; Davy, B.M. The Healthy Beverage Index Is Associated with Reduced Cardiometabolic Risk in US Adults: A Preliminary Analysis. J. Acad. Nutr. Diet. 2015, 115, 1682–1689.e2. [Google Scholar] [CrossRef] [PubMed]
  38. Rasaei, N.; Ghaffarian-Ensaf, R.; Shiraseb, F.; Abaj, F.; Gholami, F.; Clark, C.C.T.; Mirzaei, K. The association between Healthy Beverage Index and psychological disorders among overweight and obese women: A cross-sectional study. BMC Women’s Health 2022, 22, 295. [Google Scholar] [CrossRef]
  39. Fusar-Poli, L.; Gabbiadini, A.; Ciancio, A.; Vozza, L.; Signorelli, M.S.; Aguglia, E. The effect of cocoa-rich products on depression, anxiety, and mood: A systematic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 2022, 62, 7905–7916. [Google Scholar] [CrossRef]
  40. Nucci, D.; Fatigoni, C.; Amerio, A.; Odone, A.; Gianfredi, V. Red and Processed Meat Consumption and Risk of Depression: A Systematic Review and Meta-Analysis. Int. J. Environ. Res. Public Health 2020, 17, 6686. [Google Scholar] [CrossRef]
  41. Lázaro Tomé, A.; Reig Cebriá, M.J.; González-Teruel, A.; Carbonell-Asíns, J.A.; Cañete Nicolás, C.; Hernández-Viadel, M. Efficacy of vitamin D in the treatment of depression: A systematic review and meta-analysis. Actas Esp. Psiquiatr. 2021, 49, 12–23. [Google Scholar] [PubMed]
  42. Sarris, J.; Murphy, J.; Mischoulon, D.; Papakostas, G.I.; Fava, M.; Berk, M.; Ng, C.H. Adjunctive Nutraceuticals for Depression: A Systematic Review and Meta-Analyses. Am. J. Psychiatry 2016, 173, 575–587. [Google Scholar] [CrossRef] [PubMed]
  43. Young, L.M.; Pipingas, A.; White, D.J.; Gauci, S.; Scholey, A. A Systematic Review and Meta-Analysis of B Vitamin Supplementation on Depressive Symptoms, Anxiety, and Stress: Effects on Healthy and ‘At-Risk’ Individuals. Nutrients 2019, 11, 2232. [Google Scholar] [CrossRef] [PubMed]
  44. Levey, D.F.; Stein, M.B.; Wendt, F.R.; Pathak, G.A.; Zhou, H.; Aslan, M.; Quaden, R.; Harrington, K.M.; Nuñez, Y.Z.; Overstreet, C.; et al. Bi-ancestral depression GWAS in the Million Veteran Program and meta-analysis in >1.2 million individuals highlight new therapeutic directions. Nat. Neurosci. 2021, 24, 954–963. [Google Scholar] [CrossRef] [PubMed]
  45. Mullins, N.; Bigdeli, T.B.; Børglum, A.D.; Coleman, J.R.; Demontis, D.; Mehta, D.; Power, R.A.; Ripke, S.; Stahl, E.A.; Starnawska, A.; et al. GWAS of Suicide Attempt in Psychiatric Disorders and Association With Major Depression Polygenic Risk Scores. Am. J. Psychiatry 2019, 176, 651–660. [Google Scholar] [CrossRef] [PubMed]
  46. Ni, H.; Xu, M.; Zhan, G.-L.; Fan, Y.; Zhou, H.; Jiang, H.-Y.; Lu, W.-H.; Tan, L.; Zhang, D.-F.; Yao, Y.-G.; et al. The GWAS Risk Genes for Depression May Be Actively Involved in Alzheimer’s Disease. J. Alzheimers Dis. 2018, 64, 1149–1161. [Google Scholar] [CrossRef] [PubMed]
  47. Xie, T.; Stathopoulou, M.G.; de Andrés, F.; Siest, G.; Murray, H.; Martin, M.; Cobaleda, J.; Delgado, A.; Lamont, J.; Peñas-Liedó, E.; et al. VEGF-related polymorphisms identified by GWAS and risk for major depression. Transl. Psychiatry 2017, 7, e1055. [Google Scholar] [CrossRef] [PubMed]
  48. Sullivan, P.F.; Neale, M.C.; Kendler, K.S. Genetic Epidemiology of Major Depression: Review and Meta-Analysis. Am. J. Psychiatry 2000, 157, 1552–1562. [Google Scholar] [CrossRef] [PubMed]
  49. Beute, F.; de Kort, Y.A.W. The natural context of wellbeing: Ecological momentary assessment of the influence of nature and daylight on affect and stress for individuals with depression levels varying from none to clinical. Health Place. 2018, 49, 7–18. [Google Scholar] [CrossRef]
  50. Jakstis, K.; Fischer, L.K. Urban Nature and Public Health: How Nature Exposure and Sociocultural Background Relate to Depression Risk. Int. J. Environ. Res. Public Health 2021, 18, 9689. [Google Scholar] [CrossRef]
  51. Bezold, C.P.; Banay, R.F.; Coull, B.A.; Hart, J.E.; James, P.; Kubzansky, L.D.; Missmer, S.A.; Laden, F. The Association between Natural Environments and Depressive Symptoms in Adolescents Living in the United States. J. Adolesc. Health 2018, 62, 488–495. [Google Scholar] [CrossRef] [PubMed]
  52. Dempsey, S.; Devine, M.T.; Gillespie, T.; Lyons, S.; Nolan, A. Coastal blue space and depression in older adults. Health Place. 2018, 54, 110–117. [Google Scholar] [CrossRef] [PubMed]
  53. Galea, S.; Ahern, J.; Rudenstine, S.; Wallace, Z.; Vlahov, D. Urban built environment and depression: A multilevel analysis. J. Epidemiol. Community Health 2005, 59, 822–827. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, H.; Cui, X.; Dijst, M.; Tian, S.; Chen, J.; Huang, J. Association Between Natural/Built Campus Environment and Depression Among Chinese Undergraduates: Multiscale Evidence for the Moderating Role of Socioeconomic Factors After Controlling for Residential Self-Selection. Front. Public Health 2022, 10, 844541. [Google Scholar] [CrossRef] [PubMed]
  55. Harris, T.; Brown, G.W.; Robinson, R. Befriending as an intervention for chronic depression among women in an inner city: 1: Randomised controlled trial. Br. J. Psychiatry 1999, 174, 219–224. [Google Scholar] [CrossRef] [PubMed]
  56. Wing, J.K.; Birley, J.; Graham, P.; Isaacs, A. Present state examination. Br. J. Psychiatry 1974. [Google Scholar] [CrossRef]
  57. Fancourt, D.; Opher, S.; de Oliveira, C. Fixed-Effects Analyses of Time-Varying Associations between Hobbies and Depression in a Longitudinal Cohort Study: Support for Social Prescribing? Psychother. Psychosom. 2019, 89, 111–113. [Google Scholar] [CrossRef]
  58. Li, Z.; Dai, J.; Wu, N.; Jia, Y.; Gao, J.; Fu, H. Effect of Long Working Hours on Depression and Mental Well-Being among Employees in Shanghai: The Role of Having Leisure Hobbies. Int. J. Environ. Res. Public Health 2019, 16, 4980. [Google Scholar] [CrossRef] [PubMed]
  59. Hammen, C. Stress and Depression. Annu. Rev. Clin. Psychol. 2005, 1, 293–319. [Google Scholar] [CrossRef]
  60. Santa Maria, A.; Wörfel, F.; Wolter, C.; Gusy, B.; Rotter, M.; Stark, S.; Kleiber, D.; Renneberg, B. The Role of Job Demands and Job Resources in the Development of Emotional Exhaustion, Depression, and Anxiety Among Police Officers. Police Q. 2018, 21, 109–134. [Google Scholar] [CrossRef]
  61. Hall, G.B.; Dollard, M.F.; Winefield, A.H.; Dormann, C.; Bakker, A.B. Psychosocial safety climate buffers effects of job demands on depression and positive organizational behaviors. Anxiety Stress Coping 2013, 26, 355–377. [Google Scholar] [CrossRef] [PubMed]
  62. Ocklenburg, S.; Borawski, J. Vegetarian diet and depression scores: A meta-analysis. J. Affect. Disord. 2021, 294, 813–815. [Google Scholar] [CrossRef] [PubMed]
  63. Psaltopoulou, T.; Sergentanis, T.N.; Panagiotakos, D.B.; Sergentanis, I.N.; Kosti, R.; Scarmeas, N. Mediterranean diet, stroke, cognitive impairment, and depression: A meta-analysis. Ann. Neurol. 2013, 74, 580–591. [Google Scholar] [CrossRef]
  64. 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]
  65. Nabavi, S.M.; Daglia, M.; Braidy, N.; Nabavi, S.F. Natural products, micronutrients, and nutraceuticals for the treatment of depression: A short review. Nutr. Neurosci. 2017, 20, 180–194. [Google Scholar] [CrossRef] [PubMed]
  66. Alvarez-Mon, M.A.; Ortega, M.A.; García-Montero, C.; Fraile-Martinez, O.; Monserrat, J.; Lahera, G.; Mora, F.; Rodriguez-Quiroga, A.; Fernandez-Rojo, S.; Quintero, J.; et al. Exploring the Role of Nutraceuticals in Major Depressive Disorder (MDD): Rationale, State of the Art and Future Prospects. Pharmaceuticals 2021, 14, 821. [Google Scholar] [CrossRef]
  67. Bonokwane, M.B.; Lekhooa, M.; Struwig, M.; Aremu, A.O. Antidepressant Effects of South African Plants: An Appraisal of Ethnobotanical Surveys, Ethnopharmacological and Phytochemical Studies. Front. Pharmacol. 2022, 13, 895286. [Google Scholar] [CrossRef]
  68. Mischoulon, D.; Iovieno, N. Supplements and Natural Remedies for Depression. In The Massachusetts General Hospital Guide to Depression: New Treatment Insights and Options; Shapero, B.G., Mischoulon, D., Cusin, C., Eds.; Springer International Publishing: Cham, Switerland, 2019; pp. 195–209. [Google Scholar]
  69. Wang, H.; Jin, M.; Xie, M.; Yang, Y.; Xue, F.; Li, W.; Zhang, M.; Li, Z.; Li, X.; Jia, N.; et al. Protective role of antioxidant supplementation for depression and anxiety: A meta-analysis of randomized clinical trials. J. Affect. Disord. 2023, 323, 264–279. [Google Scholar] [CrossRef] [PubMed]
  70. Hoffmann, K.; Emons, B.; Brunnhuber, S.; Karaca, S.; Juckel, G. The Role of Dietary Supplements in Depression and Anxiety—A Narrative Review. Pharmacopsychiatry 2019, 52, 261–279. [Google Scholar] [CrossRef]
  71. 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]
  72. Subermaniam, K.; Teoh, S.L.; Yow, Y.-Y.; Tang, Y.-Q.; Lim, L.W.; Wong, K.-H. Marine algae as emerging therapeutic alternatives for depression: A review. Iran. J. Basic Med. Sci. 2021, 24, 997–1013. [Google Scholar] [PubMed]
  73. Varteresian, T.C.; Merrill, D.A.; Lavretsky, H. The Use of Natural Products and Supplements in Late-Life Mood and Cognitive Disorders. Focus 2013, 11, 15–21. [Google Scholar] [CrossRef]
  74. Petridou, E.T.; Kousoulis, A.A.; Michelakos, T.; Papathoma, P.; Dessypris, N.; Papadopoulos, F.C.; Stefanadis, C. Folate and B12 serum levels in association with depression in the aged: A systematic review and meta-analysis. Aging Ment. Health 2016, 20, 965–973. [Google Scholar] [CrossRef] [PubMed]
  75. Mischoulon, D. Popular herbal and natural remedies used in psychiatry. Focus. Am. Psychiatr. Publ. 2018, 16, 2–11. [Google Scholar] [CrossRef]
  76. Varteresian, T.; Lavretsky, H. Natural products and supplements for geriatric depression and cognitive disorders: An evaluation of the research. Curr. Psychiatry Rep. 2014, 16, 456. [Google Scholar] [CrossRef]
  77. Schefft, C.; Kilarski, L.L.; Bschor, T.; Koehler, S. Efficacy of adding nutritional supplements in unipolar depression: A systematic review and meta-analysis. Eur. Neuropsychopharmacol. 2017, 27, 1090–1109. [Google Scholar] [CrossRef]
  78. Dwyer, A.V.; Whitten, D.L.; Hawrelak, J.A. Herbal medicines, other than St. John’s Wort, in the treatment of depression: A systematic review. Altern. Med. Rev. 2011, 16, 40–49. [Google Scholar] [PubMed]
  79. Szafrański, T. Herbal remedies in depression–state of the art. Psychiatr. Pol. 2014, 48, 59–73. [Google Scholar] [CrossRef]
  80. Ernst, E. Herbal remedies for depression and anxiety. Adv. Psychiatr. Treat. 2007, 13, 312–316. [Google Scholar] [CrossRef]
  81. Sarris, J.; Panossian, A.; Schweitzer, I.; Stough, C.; Scholey, A. Herbal medicine for depression, anxiety and insomnia: A review of psychopharmacology and clinical evidence. Eur. Neuropsychopharmacol. 2011, 21, 841–860. [Google Scholar] [CrossRef]
  82. Sarris, J. Herbal medicines in the treatment of psychiatric disorders: A systematic review. Phytother. Res. 2007, 21, 703–716. [Google Scholar] [CrossRef] [PubMed]
  83. Yosaee, S.; Clark, C.C.T.; Keshtkaran, Z.; Ashourpour, M.; Keshani, P.; Soltani, S. Zinc in depression: From development to treatment: A comparative/ dose response meta-analysis of observational studies and randomized controlled trials. Gen. Hosp. Psychiatry 2022, 74, 110–117. [Google Scholar] [CrossRef] [PubMed]
  84. Tarleton, E.K.; Littenberg, B.; MacLean, C.D.; Kennedy, A.G.; Daley, C. Role of magnesium supplementation in the treatment of depression: A randomized clinical trial. PLoS ONE 2017, 12, e0180067. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, L.; Shen, X.; Wu, Y.; Zhang, D. Coffee and caffeine consumption and depression: A meta-analysis of observational studies. Aust. N. Z. J. Psychiatry 2016, 50, 228–242. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, Y.; Kim, Y.; Je, Y. Fish consumption and risk of depression: Epidemiological evidence from prospective studies. Asia-Pac. Psychiatry 2018, 10, e12335. [Google Scholar] [CrossRef] [PubMed]
  87. Li, F.; Liu, X.; Zhang, D. Fish consumption and risk of depression: A meta-analysis. J. Epidemiol. Community Health 2016, 70, 299–304. [Google Scholar] [CrossRef] [PubMed]
  88. Hoffmire, C.A.; Block, R.C.; Thevenet-Morrison, K.; van Wijngaarden, E. Associations between omega-3 poly-unsaturated fatty acids from fish consumption and severity of depressive symptoms: An analysis of the 2005–2008 National Health and Nutrition Examination Survey. Prostaglandins Leukot. Essent. Fat. Acids 2012, 86, 155–160. [Google Scholar] [CrossRef] [PubMed]
  89. Sajjadi, S.S.; Foshati, S.; Haddadian-Khouzani, S.; Rouhani, M.H. The role of selenium in depression: A systematic review and meta-analysis of human observational and interventional studies. Sci. Rep. 2022, 12, 1045. [Google Scholar] [CrossRef] [PubMed]
  90. Bleys, J.; Navas-Acien, A.; Guallar, E. Serum Selenium Levels and All-Cause, Cancer, and Cardiovascular Mortality Among US Adults. Arch. Intern. Med. 2008, 168, 404–410. [Google Scholar] [CrossRef]
  91. Finley, J.W. Bioavailability of Selenium from Foods. Nutr. Rev. 2006, 64, 146–151. [Google Scholar] [CrossRef]
  92. Mahdavifar, B.; Hosseinzadeh, M.; Salehi-Abargouei, A.; Mirzaei, M.; Vafa, M. Dietary intake of B vitamins and their association with depression, anxiety, and stress symptoms: A cross-sectional, population-based survey. J. Affect. Disord. 2021, 288, 92–98. [Google Scholar] [CrossRef] [PubMed]
  93. Trüeb, R. Serum Biotin Levels in Women Complaining of Hair Loss. Int. J. Trichology 2016, 8, 73. [Google Scholar] [CrossRef] [PubMed]
  94. Institute of Medicine Staff, & Food and Nutrition Board Staff. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin and Choline; National Academies Press: Washington, DC, USA, 2000. [Google Scholar]
  95. Said, H.M. Biotin: Biochemical, Physiological and Clinical Aspects. In Water Soluble Vitamins: Clinical Research and Future Application; Stanger, O., Ed.; Springer: Dordrecht, The Netherlands, 2012; pp. 1–19. [Google Scholar]
  96. de Benoist, B. Conclusions of a WHO Technical Consultation on folate and vitamin B12 deficiencies. Food Nutr. Bull. 2008, 29 (Suppl. 2), S238–S244. [Google Scholar] [CrossRef] [PubMed]
  97. Iyer, R.; Tomar, S.K. Folate: A Functional Food Constituent. J. Food Sci. 2009, 74, R114–R122. [Google Scholar] [CrossRef] [PubMed]
  98. Markun, S.; Gravestock, I.; Jäger, L.; Rosemann, T.; Pichierri, G.; Burgstaller, J.M. Effects of Vitamin B12 Supplementation on Cognitive Function, Depressive Symptoms, and Fatigue: A Systematic Review, Meta-Analysis, and Meta-Regression. Nutrients 2021, 13, 923. [Google Scholar] [CrossRef] [PubMed]
  99. Parker, G.B.; Brotchie, H.; Graham, R.K. Vitamin D and depression. J. Affect. Disord. 2017, 208, 56–61. [Google Scholar] [CrossRef] [PubMed]
  100. Menon, V.; Kar, S.K.; Suthar, N.; Nebhinani, N. Vitamin D and depression: A critical appraisal of the evidence and future directions. Indian. J. Psychol. Med. 2020, 42, 11–21. [Google Scholar] [CrossRef]
  101. Standing Committee on the Scientific Evaluation of Dietary Reference Intakes. Dietary Reference Intakes for Calcium, Phosphorus, Magnesium, Vitamin D, and Fluoride; National Academies Press: Washington, DC, USA, 1997. [Google Scholar]
  102. Baggerly, C.A.; Cuomo, R.E.; French, C.B.; Garland, C.F.; Gorham, E.D.; Grant, W.B.; Heaney, R.P.; Holick, M.F.; Hollis, B.W.; McDonnell, S.L.; et al. Sunlight and Vitamin D: Necessary for Public Health. J. Am. Coll. Nutr. 2015, 34, 359–365. [Google Scholar] [CrossRef]
  103. Moore, C.; Murphy, M.M.; Keast, D.R.; Holick, M.F. Vitamin D intake in the United States. J. Am. Diet. Assoc. 2004, 104, 980–983. [Google Scholar] [CrossRef]
  104. Liu, R.T.; Walsh, R.F.L.; Sheehan, A.E. Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci. Biobehav. Rev. 2019, 102, 13–23. [Google Scholar] [CrossRef]
  105. Chao, L.; Liu, C.; Sutthawongwadee, S.; Li, Y.; Lv, W.; Chen, W.; Yu, L.; Zhou, J.; Guo, A.; Li, Z.; et al. Effects of Probiotics on Depressive or Anxiety Variables in Healthy Participants Under Stress Conditions or With a Depressive or Anxiety Diagnosis: A Meta-Analysis of Randomized Controlled Trials. Front. Neurol. 2020, 11, 421. [Google Scholar] [CrossRef] [PubMed]
  106. Ng, Q.X.; Peters, C.; Ho, C.Y.X.; Lim, D.Y.; Yeo, W.-S. A meta-analysis of the use of probiotics to alleviate depressive symptoms. J. Affect. Disord. 2018, 228, 13–19. [Google Scholar] [CrossRef] [PubMed]
  107. Metwaly, A.; Reitmeier, S.; Haller, D. Microbiome risk profiles as biomarkers for inflammatory and metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 383–397. [Google Scholar] [CrossRef] [PubMed]
  108. Pinto-Sanchez, M.I.; Hall, G.B.; Ghajar, K.; Nardelli, A.; Bolino, C.; Lau, J.T.; Martin, F.-P.; Cominetti, O.; Welsh, C.; Rieder, A.; et al. Probiotic Bifidobacterium longum NCC3001 Reduces Depression Scores and Alters Brain Activity: A Pilot Study in Patients With Irritable Bowel Syndrome. Gastroenterology 2017, 153, 448–459.e8. [Google Scholar] [CrossRef] [PubMed]
  109. Granato, D.; Branco, G.F.; Nazzaro, F.; Cruz, A.G.; Faria, J.A.F. Functional Foods and Nondairy Probiotic Food Development: Trends, Concepts, and Products. Compr. Rev. Food Sci. Food Saf. 2010, 9, 292–302. [Google Scholar] [CrossRef] [PubMed]
  110. Kapp, J.M.; Sumner, W. Kombucha: A systematic review of the empirical evidence of human health benefit. Ann. Epidemiol. 2019, 30, 66–70. [Google Scholar] [CrossRef]
  111. Şanlier, N.; Gökcen, B.B.; Sezgin, A.C. Health benefits of fermented foods. Crit. Rev. Food Sci. Nutr. 2019, 59, 506–527. [Google Scholar] [CrossRef] [PubMed]
  112. Cuamatzin-García, L.; Rodríguez-Rugarcía, P.; El-Kassis, E.G.; Galicia, G.; Meza-Jiménez, M.d.L.; Baños-Lara, M.d.R.; Zaragoza-Maldonado, D.S.; Pérez-Armendáriz, B. Traditional Fermented Foods and Beverages from around the World and Their Health Benefits. Microorganisms 2022, 10, 1151. [Google Scholar] [CrossRef] [PubMed]
  113. Veronese, N.; Stubbs, B.; Solmi, M.; Ajnakina, O.; Carvalho, A.F.; Maggi, S. Acetyl-l-Carnitine Supplementation and the Treatment of Depressive Symptoms: A Systematic Review and Meta-Analysis. Psychosom. Med. 2018, 80, 154–159. [Google Scholar] [CrossRef]
  114. Nasca, C.; Bigio, B.; Lee, F.S.; Young, S.P.; Kautz, M.M.; Albright, A.; Beasley, J.; Millington, D.S.; Mathé, A.A.; Kocsis, J.H.; et al. Acetyl-l-carnitine deficiency in patients with major depressive disorder. Proc. Natl. Acad. Sci. USA 2018, 115, 8627–8632. [Google Scholar] [CrossRef]
  115. Malaguarnera, M.; Bella, R.; Vacante, M.; Giordano, M.; Malaguarnera, G.; Gargante, M.P.; Motta, M.; Mistretta, A.; Rampello, L.; Pennisi, G. Acetyl-l-carnitine reduces depression and improves quality of life in patients with minimal hepatic encephalopathy. Scand. J. Gastroenterol. 2011, 46, 750–759. [Google Scholar] [CrossRef] [PubMed]
  116. Roseiro, L.C.; Santos, C. Chapter 2.5—Carnitines (Including l-Carnitine, Acetyl-Carnitine, and Proprionyl-Carnitine). In Nonvitamin and Nonmineral Nutritional Supplements; Nabavi, S.M., Silva, A.S., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 45–52. [Google Scholar]
  117. Bakian, A.V.; Huber, R.S.; Scholl, L.; Renshaw, P.F.; Kondo, D. Dietary creatine intake and depression risk among U.S. Adults. Transl. Psychiatry 2020, 10, 52. [Google Scholar] [CrossRef] [PubMed]
  118. Kondo, D.G.; Forrest, L.N.; Shi, X.; Sung, Y.-H.; Hellem, T.L.; Huber, R.S.; Renshaw, P.F. Creatine target engagement with brain bioenergetics: A dose-ranging phosphorus-31 magnetic resonance spectroscopy study of adolescent females with SSRI-resistant depression. Amino Acids 2016, 48, 1941–1954. [Google Scholar] [CrossRef]
  119. Brosnan, M.E.; Brosnan, J.T. The role of dietary creatine. Amino Acids 2016, 48, 1785–1791. [Google Scholar] [CrossRef]
  120. Ille, R.; Spona, J.; Zickl, M.; Hofmann, P.; Lahousen, T.; Dittrich, N.; Bertha, G.; Hasiba, K.; Mahnert, F.A.; Kapfhammer, H.-P. “Add-On”-therapy with an individualized preparation consisting of free amino acids for patients with a major depression. Eur. Arch. Psychiatry Clin. Neurosci. 2007, 257, 222–229. [Google Scholar] [CrossRef]
  121. Pangborn, J. Nutritionally Correct Amino Acid Ranges: Urine and Plasma. Technical Memorandum 1, Biostatistics. 1986.
  122. Bralley, J.A.; Lord, R.S. Treatment of chronic fatigue syndrome with specific amino acid supplementation. J. Appl. Nutr. 1994, 46, 74–78. [Google Scholar]
  123. Maruf, A.A.; Poweleit, E.A.; Brown, L.C.; Strawn, J.R.; Bousman, C.A. Systematic Review and Meta-Analysis of L-Methylfolate Augmentation in Depressive Disorders. Pharmacopsychiatry 2021, 55, 139–147. [Google Scholar] [CrossRef] [PubMed]
  124. Altaf, R.; Gonzalez, I.; Rubino, K.; Nemec, E.C. Folate as adjunct therapy to SSRI/SNRI for major depressive disorder: Systematic review & meta-analysis. Complement. Ther. Med. 2021, 61, 102770. [Google Scholar]
  125. Liu, M.; Zhang, Z.; Zhou, C.; Li, Q.; He, P.; Zhang, Y.; Li, H.; Liu, C.; Liang, M.; Wang, X.; et al. Relationship of several serum folate forms with the risk of mortality: A prospective cohort study. Clin. Nutr. 2021, 40, 4255–4262. [Google Scholar] [CrossRef]
  126. Javelle, F.; Lampit, A.; Bloch, W.; Häussermann, P.; Johnson, S.L.; Zimmer, P. Effects of 5-hydroxytryptophan on distinct types of depression: A systematic review and meta-analysis. Nutr. Rev. 2019, 78, 77–88. [Google Scholar] [CrossRef]
  127. Asher, G.N.; Gartlehner, G.; Gaynes, B.N.; Amick, H.R.; Forneris, C.; Morgan, L.C.; Coker-Schwimmer, E.; Boland, E.; Lux, L.J.; Gaylord, S.; et al. Comparative Benefits and Harms of Complementary and Alternative Medicine Therapies for Initial Treatment of Major Depressive Disorder: Systematic Review and Meta-Analysis. J. Altern. Complement. Med. 2017, 23, 907–919. [Google Scholar] [CrossRef] [PubMed]
  128. Brattström, A. Long-term effects of St. John’s wort (Hypericum perforatum) treatment: A 1-year safety study in mild to moderate depression. Phytomedicine 2009, 16, 277–283. [Google Scholar] [CrossRef] [PubMed]
  129. Mazidi, M.; Shemshian, M.; Mousavi, S.H.; Norouzy, A.; Kermani, T.; Moghiman, T.; Sadeghi, A.; Mokhber, N.; Ghayour-Mobarhan, M.; Ferns, G.A.A. A double-blind, randomized and placebo-controlled trial of Saffron (Crocus sativus L.) in the treatment of anxiety and depression. J. Complement. Integr. Med. 2016, 13, 195–199. [Google Scholar] [CrossRef] [PubMed]
  130. Ng, Q.X.; Koh, S.S.H.; Chan, H.W.; Ho, C.Y.X. Clinical Use of Curcumin in Depression: A Meta-Analysis. J. Am. Med. Dir. Assoc. 2017, 18, 503–508. [Google Scholar] [CrossRef] [PubMed]
  131. Fusar-Poli, L.; Vozza, L.; Gabbiadini, A.; Vanella, A.; Concas, I.; Tinacci, S.; Petralia, A.; Signorelli, M.S.; Aguglia, E. Curcumin for depression: A meta-analysis. Crit. Rev. Food Sci. Nutr. 2020, 60, 2643–2653. [Google Scholar] [CrossRef] [PubMed]
  132. Naylor, G.J.; Martin, B.; Hopwood, S.E.; Watson, Y. A two-year double-blind crossover trial of the prophylactic effect of methylene blue in manic-depressive psychosis. Biol. Psychiatry 1986, 21, 915–920. [Google Scholar] [CrossRef] [PubMed]
  133. Naylor, G.J.; Smith, A.H.; Connelly, P. A controlled trial of Methylene Blue in severe depressive illness. Biol. Psychiatry 1987, 22, 657–659. [Google Scholar] [CrossRef]
  134. Wang, Y.; Shi, Y.-H.; Xu, Z.; Fu, H.; Zeng, H.; Zheng, G.-Q. Efficacy and safety of Chinese herbal medicine for depression: A systematic review and meta-analysis of randomized controlled trials. J. Psychiatr. Res. 2019, 117, 74–91. [Google Scholar] [CrossRef]
  135. Yeung, W.-F.; Chung, K.-F.; Ng, K.-Y.; Yu, Y.-M.; Ziea, E.T.-C.; Ng, B.F.-L. A systematic review on the efficacy, safety and types of Chinese herbal medicine for depression. J. Psychiatr. Res. 2014, 57, 165–175. [Google Scholar] [CrossRef]
  136. Zadeh, A.R.; Eghbal, A.F.; Mirghazanfari, S.M.; Ghasemzadeh, M.R.; Nassireslami, E.; Donyavi, V. Nigella sativa extract in the treatment of depression and serum Brain-Derived Neurotrophic Factor (BDNF) levels. J. Res. Med. Sci. 2022, 27, 28. [Google Scholar]
  137. Galizia, I.; Oldani, L.; Macritchie, K.; Amari, E.; Dougall, D.; Jones, T.N.; Lam, R.W.; Massei, G.J.; Yatham, L.N.; Young, A.H. S-adenosyl methionine (SAMe) for depression in adults. Cochrane Database Syst. Rev. 2016, 2016, CD011286. [Google Scholar] [CrossRef]
  138. Delle Chiaie, R.; Pancheri, P.; Scapicchio, P. Efficacy and tolerability of oral and intramuscular S-adenosyl-L-methionine 1, 4-butanedisulfonate (SAMe) in the treatment of major depression: Comparison with imipramine in 2 multicenter studies. Am. J. Clin. Nutr. 2002, 76, 1172S–1176S. [Google Scholar] [CrossRef]
  139. Calabrese, C.; Gregory, W.L.; Leo, M.; Kraemer, D.; Bone, K.; Oken, B. Effects of a standardized Bacopa monnieri extract on cognitive performance, anxiety, and depression in the elderly: A randomized, double-blind, placebo-controlled trial. J. Altern. Complement. Med. 2008, 14, 707–713. [Google Scholar] [CrossRef]
  140. Darbinyan, V.; Aslanyan, G.; Amroyan, E.; Gabrielyan, E.; Malmström, C.; Panossian, A. Clinical trial of Rhodiola rosea L. extract SHR-5 in the treatment of mild to moderate depression. Nord. J. Psychiatry 2007, 61, 343–348. [Google Scholar] [CrossRef]
  141. Sarris, J.; Kavanagh, D.J.; Byrne, G.; Bone, K.M.; Adams, J.; Deed, G. The Kava Anxiety Depression Spectrum Study (KADSS): A randomized, placebo-controlled crossover trial using an aqueous extract of Piper methysticum. Psychopharmacology 2009, 205, 399–407. [Google Scholar] [CrossRef]
  142. Lewin, L.M.; Melmed, S.; Bank, H. Rapid screening test for detection of elevated MYO-Inositol levels in human blood serum. Clin. Chim. Acta 1974, 54, 377–379. [Google Scholar] [CrossRef]
  143. Iovieno, N.; Dalton, E.D.; Fava, M.; Mischoulon, D. Second-tier natural antidepressants: Review and critique. J. Affect. Disord. 2011, 130, 343–357. [Google Scholar] [CrossRef]
  144. Clements, R.S.; Jr Darnell, B. Myo-inositol content of common foods: Development of a high-myo-inositol diet. Am. J. Clin. Nutr. 1980, 33, 1954–1967. [Google Scholar] [CrossRef]
  145. Davidson, J.R.; Abraham, K.; Connor, K.M.; McLeod, M.N. Effectiveness of chromium in atypical depression: A placebo-controlled trial. Biol. Psychiatry 2003, 53, 261–264. [Google Scholar] [CrossRef]
  146. Molin Christensen, J.; Holst, E.; Peter Bonde, J.; Knudsen, L. Determination of chromium in blood and serum: Evaluation of quality control procedures and estimation of reference values in Danish subjects. Sci. Total Environ. 1993, 132, 11–25. [Google Scholar] [CrossRef]
  147. Swaroop, A.; Bagchi, M.; Preuss, H.; Zafra-Stone, S.; Ahmad, T.; Bagchi, D. Benefits of chromium(III) complexes in animal and human health. In The Nutritional Biochemistry of Chromium (III); Elsevier: Amsterdam, The Netherlands, 2019; pp. 251–278. [Google Scholar]
  148. Niklowitz, P.; Onur, S.; Fischer, A.; Laudes, M.; Palussen, M.; Menke, T.; Döring, F. Coenzyme Q10 serum concentration and redox status in European adults: Influence of age, sex, and lipoprotein concentration. J. Clin. Biochem. Nutr. 2016, 58, 240–245. [Google Scholar] [CrossRef]
  149. Pravst, I.; Žmitek, K.; Žmitek, J. Coenzyme Q10 Contents in Foods and Fortification Strategies. Crit. Rev. Food Sci. Nutr. 2010, 50, 269–280. [Google Scholar] [CrossRef]
  150. Foshati, S.; Ghanizadeh, A.; Akhlaghi, M. Extra-Virgin Olive Oil Improves Depression Symptoms Without Affecting Salivary Cortisol and Brain-Derived Neurotrophic Factor in Patients with Major Depression: A Double-Blind Randomized Controlled Trial. J. Acad. Nutr. Diet. 2022, 122, 284–297.e1. [Google Scholar] [CrossRef]
  151. Akhondzadeh, S.; Kashani, L.; Fotouhi, A.; Jarvandi, S.; Mobaseri, M.; Moin, M.; Khani, M.; Jamshidi, A.H.; Baghalian, K.; Taghizadeh, M. Comparison of Lavandula angustifolia Mill. tincture and imipramine in the treatment of mild to moderate depression: A double-blind, randomized trial. Prog. Neuropsychopharmacol. Biol. Psychiatry 2003, 27, 123–127. [Google Scholar] [CrossRef]
  152. Wang, X.-L.; Feng, S.-T.; Wang, Y.-T.; Zhang, N.-N.; Wang, Z.-Z.; Zhang, Y. Canonical Chinese medicine formula Danzhi-Xiaoyao-San for treating depression: A systematic review and meta-analysis. J. Ethnopharmacol. 2022, 287, 114960. [Google Scholar] [CrossRef]
  153. Brennan, B.P.; Jensen, J.E.; Hudson, J.I.; Coit, C.E.; Beaulieu, A.; Pope, H.G., Jr.; Renshaw, P.F.; Cohen, B.M. A Placebo-Controlled Trial of Acetyl-L-Carnitine and α-Lipoic Acid in the Treatment of Bipolar Depression. J. Clin. Psychopharmacol. 2013, 33, 627–635. [Google Scholar] [CrossRef]
  154. Shay, K.P.; Moreau, R.F.; Smith, E.J.; Smith, A.R.; Hagen, T.M. Alpha-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2009, 1790, 1149–1160. [Google Scholar] [CrossRef]
  155. Berk, M.; Copolov, D.L.; Dean, O.; Lu, K.; Jeavons, S.; Schapkaitz, I.; Anderson-Hunt, M.; Bush, A.I. N-acetyl cysteine for depressive symptoms in bipolar disorder--a double-blind randomized placebo-controlled trial. Biol. Psychiatry 2008, 64, 468–475. [Google Scholar] [CrossRef]
  156. Berk, M.; Dean, O.; Cotton, S.M.; Gama, C.S.; Kapczinski, F.; Fernandes, B.S.; Kohlmann, K.; Jeavons, S.; Hewitt, K.; Allwang, C.; et al. The efficacy of N-acetylcysteine as an adjunctive treatment in bipolar depression: An open label trial. J. Affect. Disord. 2011, 135, 389–394. [Google Scholar] [CrossRef]
  157. Caso Marasco, A.; Vargas Ruiz, R.; Salas Villagomez, A.; Begoña Infante, C. Double-blind study of a multivitamin complex supplemented with ginseng extract. Drugs Exp. Clin. Res. 1996, 22, 323–329. [Google Scholar]
  158. Ahmadpoor, J.; Chahardahcheric, S.V.; Setorki, M. The Protective effect of hydroalcoholic extract of the southern maidenhair fern (adiantum capillus-veneris) on the depression and anxiety caused by chronic stress in adult male mice: An experimental randomized study. Iran. Red. Crescent Med. J. 2019, 21, e86750. [Google Scholar] [CrossRef]
  159. Rabiei, Z.; Setorki, M. Effect of ethanol Adiantum capillus-veneris extract in experimental models of anxiety and depression. Braz. J. Pharm. Sci. 2019, 55, e18099. [Google Scholar] [CrossRef]
  160. Pedersen, M.E.; Szewczyk, B.; Stachowicz, K.; Wieronska, J.; Andersen, J.; Stafford, G.I.; van Staden, J.; Pilc, A.; Jäger, A.K. Effects of South African traditional medicine in animal models for depression. J. Ethnopharmacol. 2008, 119, 542–548. [Google Scholar] [CrossRef]
  161. Aderibigbe, A. Antidepressant activity of ethanol extract of Albizia adianthifolia (Schumach) WF Wight leaf in mice. Afr. J. Med. Med. Sci. 2018, 47, 133–140. [Google Scholar]
  162. Beppe, G.J.; Dongmo, A.B.; Foyet, H.S.; Dimo, T.; Mihasan, M.; Hritcu, L. The aqueous extract of Albizia adianthifolia leaves attenuates 6-hydroxydopamine-induced anxiety, depression and oxidative stress in rat amygdala. BMC Complement. Altern. Med. 2015, 15, 374. [Google Scholar] [CrossRef]
  163. Jahani, R.; Khaledyan, D.; Jahani, A.; Jamshidi, E.; Kamalinejad, M.; Khoramjouy, M.; Faizi, M. Evaluation and comparison of the antidepressant-like activity of Artemisia dracunculus and Stachys lavandulifolia ethanolic extracts: An in vivo study. Res. Pharm. Sci. 2019, 14, 544. [Google Scholar]
  164. Ilkhanizadeh, A.; Asghari, A.; Hassanpour, S.; Safi, S. Anti-depressant effect of Artemisia dracunculus extract is mediated via GABAergic and serotoninergic systems in ovariectomized mice. J. Basic Clin. Pathophysiol. 2021, 9, 32–41. [Google Scholar]
  165. Zanelati, T.; Biojone, C.; Moreira, F.; Guimarães, F.S.; Joca, S.R.L. Antidepressant-like effects of cannabidiol in mice: Possible involvement of 5-HT1A receptors. Br. J. Pharmacol. 2010, 159, 122–128. [Google Scholar] [CrossRef]
  166. Sales, A.J.; Crestani, C.C.; Guimarães, F.S.; Joca, S.R. Antidepressant-like effect induced by Cannabidiol is dependent on brain serotonin levels. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 86, 255–261. [Google Scholar] [CrossRef]
  167. El-Alfy, A.T.; Ivey, K.; Robinson, K.; Ahmed, S.; Radwan, M.; Slade, D.; Khan, I.; ElSohly, M.; Ross, S. Antidepressant-like effect of Δ9-tetrahydrocannabinol other cannabinoids isolated from Cannabis sativa L. Pharmacol. Biochem. Behav. 2010, 95, 434–442. [Google Scholar] [CrossRef]
  168. Selvi, P.T.; Kumar, M.S.; Rajesh, R.; Kathiravan, T. Antidepressant activity of ethanolic extract of leaves of Centella asiatica. Linn by in vivo methods. Asian J. Res. Pharm. Sci. 2012, 2, 76–79. [Google Scholar]
  169. Rabadia, J.; Satish, S.; Ramanjaneyulu, J.; Narayanaswamy, V. An investigation of anti-depressant activity of Cinnamomum camphora oil in experimental mice. Asian J. Biomed. Pharm. Sci. 2013, 3, 44. [Google Scholar]
  170. Citó, M.; Silva, M.; Santos, L.; Fernandes, M.; Melo, F.; Aguiar, J.; Lopes, I.; Sousa, P.; Vasconcelos, S.; Macêdo, D.; et al. Antidepressant-like effect of Hoodia gordonii in a forced swimming test in mice: Evidence for involvement of the monoaminergic system. Braz. J. Med. Biol. Res. 2014, 48, 57–64. [Google Scholar] [CrossRef] [PubMed]
  171. Tian, J.; Zhang, F.; Cheng, J.; Guo, S.; Liu, P.; Wang, H. Antidepressant-like activity of adhyperforin, a novel constituent of Hypericum perforatum L. Sci. Rep. 2014, 4, 5632. [Google Scholar] [CrossRef] [PubMed]
  172. Fiebich, B.L.; Knörle, R.; Appel, K.; Kammler, T.; Weiss, G. Pharmacological studies in an herbal drug combination of St. John’s Wort (Hypericum perforatum) and passion flower (Passiflora incarnata): In vitro and in vivo evidence of synergy between Hypericum and Passiflora in antidepressant pharmacological models. Fitoterapia 2011, 82, 474–480. [Google Scholar] [CrossRef] [PubMed]
  173. Ejigu, A.; Engidawork, E. Screening of the antidepressant-like activity of two hypericum species found in Ethiopia. Ethiop Pharm. J. 2014, 30, 21–32. [Google Scholar] [CrossRef]
  174. Benneh, C.K.; Biney, R.P.; Adongo, D.W.; Mante, P.K.; Ampadu, F.A.; Tandoh, A.; Jato, J.; Woode, E. Anxiolytic antidepressant effects of Maerua angolensis DC. Stem bark extract in mice. Depress. Res. Treat. 2018, 2018, 1537371. [Google Scholar] [PubMed]
  175. Ishaq, H. Anxiolytic and antidepressant activity of different methanolic extracts of Melia azedarach Linn. Pak. J. Pharm. Sci. 2016, 29, 1649–1655. [Google Scholar]
  176. Jedi-Behnia, B.; Abbasi Maleki, S.; Mousavi, E. The antidepressant-like effect of Mentha spicata essential oil in animal models of depression in male mice. J. Adv. Biomed. Sci. 2017, 7, 141–149. [Google Scholar]
  177. Badr, A.M.; Attia, H.A.; Al-Rasheed, N. Oleuropein reverses repeated corticosterone-induced depressive-like behavior in mice: Evidence of modulating effect on biogenic amines. Sci. Rep. 2020, 10, 3336. [Google Scholar] [CrossRef]
  178. Perveen, T.; Hashmi, B.M.; Haider, S.; Tabassum, S.; Saleem, S.; Siddiqui, M.A. Role of monoaminergic system in the etiology of olive oil induced antidepressant and anxiolytic effects in rats. Int. Sch. Res. Not. 2013, 2013, 615685. [Google Scholar] [CrossRef]
  179. Tariq, U.; Butt, M.S.; Pasha, I.; Faisal, M.N. Neuroprotective effects of Olea europaea L. fruit extract against cigarette smoke-induced depressive-like behaviors in Sprague–Dawley rats. J. Food Biochem. 2021, 45, e14014. [Google Scholar] [CrossRef]
  180. Machado, D.G.; Bettio, L.E.; Cunha, M.P.; Capra, J.C.; Dalmarco, J.B.; Pizzolatti, M.G.; Rodrigues, A.L.S. Antidepressant-like effect of the extract of Rosmarinus officinalis in mice: Involvement of the monoaminergic system. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2009, 33, 642–650. [Google Scholar] [CrossRef]
  181. Sasaki, K.; El Omri, A.; Kondo, S.; Han, J.; Isoda, H. Rosmarinus officinalis polyphenols produce anti-depressant like effect through monoaminergic and cholinergic functions modulation. Behav. Brain Res. 2013, 238, 86–94. [Google Scholar] [CrossRef]
  182. Sasaki, K.; Ferdousi, F.; Fukumitsu, S.; Kuwata, H.; Isoda, H. Antidepressant-and anxiolytic-like activities of Rosmarinus officinalis extract in rodent models: Involvement of oxytocinergic system. Biomed. Pharmacother. 2021, 144, 112291. [Google Scholar] [CrossRef]
  183. Abdelhalim, A.; Karim, N.; Chebib, M.; Aburjai, T.; Khan, I.; Johnston, G.A.; Hanrahan, J. Antidepressant, anxiolytic and antinociceptive activities of constituents from Rosmarinus officinalis. J. Pharm. Pharm. Sci. 2015, 18, 448–459. [Google Scholar] [CrossRef] [PubMed]
  184. Adebiyi, R.; Elsa, A.; Agaie, B.; Etuk, E. Antinociceptive and antidepressant like effects of Securidaca longepedunculata root extract in mice. J. Ethnopharmacol. 2006, 107, 234–239. [Google Scholar] [CrossRef] [PubMed]
  185. Loria, M.J.; Ali, Z.; Abe, N.; Sufka, K.J.; Khan, I.A. Effects of Sceletium tortuosum in rats. J. Ethnopharmacol. 2014, 155, 731–735. [Google Scholar] [CrossRef] [PubMed]
  186. Machado, D.G.; Kaster, M.P.; Binfaré, R.W.; Dias, M.; Santos, A.R.; Pizzolatti, M.G.; Brighente, I.M.; Rodrigues, A.L.S. Antidepressant-like effect of the extract from leaves of Schinus molle L. in mice: Evidence for the involvement of the monoaminergic system. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2007, 31, 421–428. [Google Scholar] [CrossRef]
  187. Wado, E.K.; Kubicki, M.; Ngatanko, A.H.H.; Blondelle, K.D.L.; Linda, D.J.; Roland, R.N.; Balbine, K.; Lamshoeft, M.; Assongalem, A.E.; Foyet, H.S. Anxiolytic and antidepressant effects of Ziziphus mucronata hydromethanolic extract in male rats exposed to unpredictable chronic mild stress: Possible mechanisms of actions. J. Ethnopharmacol. 2020, 260, 112987. [Google Scholar] [CrossRef]
  188. Li, H.; Xiao, Y.; Han, L.; Jia, Y.; Luo, S.; Zhang, D.; Zhang, L.; Wu, P.; Xiao, C.; Kan, W.; et al. Ganoderma lucidum polysaccharides ameliorated depression-like behaviors in the chronic social defeat stress depression model via modulation of Dectin-1 and the innate immune system. Brain Res. Bull. 2021, 171, 16–24. [Google Scholar] [CrossRef] [PubMed]
  189. Mi, X.; Zeng, G.-R.; Liu, J.-Q.; Luo, Z.-S.; Zhang, L.; Dai, X.-M.; Fang, W.-T.; Zhang, J.; Chen, X.-C. Ganoderma lucidum triterpenoids improve maternal separation-induced anxiety-and depression-like behaviors in mice by mitigating inflammation in the periphery and brain. Nutrients 2022, 14, 2268. [Google Scholar] [CrossRef]
  190. Nagano, M.; Shimizu, K.; Kondo, R.; Hayashi, C.; Sato, D.; Kitagawa, K.; Ohnuki, K. Reduction of depression and anxiety by 4 weeks Hericium erinaceus intake. Biomed. Res. 2010, 31, 231–237. [Google Scholar] [CrossRef] [PubMed]
  191. Zhou, Y.; Ma, C.; Li, B.-M.; Sun, C. Polygala japonica Houtt. reverses depression-like behavior and restores reduced hippocampal neurogenesis in chronic stress mice. Biomed. Pharmacother. 2018, 99, 986–996. [Google Scholar] [CrossRef] [PubMed]
  192. Kim, N.-H.; Jeong, H.-J.; Lee, J.-Y.; Go, H.; Ko, S.-G.; Hong, S.-H.; Kim, H.-M.; Um, J.-Y. The effect of hydrolyzed Spirulina by malted barley on forced swimming test in ICR mice. Int. J. Neurosci. 2008, 118, 1523–1533. [Google Scholar] [CrossRef] [PubMed]
  193. Suresh, D.; Madhu, M.; Saritha, C.; Shankaraiah, P. Antidepressant activity of spirulina platensis in experimentally induced dipression in mice. Int. J. Res. Dev. Pharm. Life Sci. 2014, 3, 1026–1035. [Google Scholar]
  194. Soetantyo, G.I.; Sarto, M. The antidepressant effect of Chlorella vulgaris on female Wistar rats (Rattus norvegicus Berkenhout, 1769) with chronic unpredictable mild stress treatment. J. Trop. Biodivers. Biotechnol. 2019, 4, 72–81. [Google Scholar] [CrossRef]
  195. Miyake, Y.; Tanaka, K.; Okubo, H.; Sasaki, S.; Arakawa, M. Seaweed consumption and prevalence of depressive symptoms during pregnancy in Japan: Baseline data from the Kyushu Okinawa Maternal and Child Health Study. BMC Pregnancy Childbirth 2014, 14, 301. [Google Scholar] [CrossRef] [PubMed]
  196. Allaert, F.-A.; Demais, H.; Collén, P.N. A randomized controlled double-blind clinical trial comparing versus placebo the effect of an edible algal extract (Ulva Lactuca) on the component of depression in healthy volunteers with anhedonia. BMC Psychiatry 2018, 18, 215. [Google Scholar] [CrossRef]
  197. Guo, F.; Huang, C.; Cui, Y.; Momma, H.; Niu, K.; Nagatomi, R. Dietary seaweed intake and depressive symptoms in Japanese adults: A prospective cohort study. Nutr. J. 2019, 18, 58. [Google Scholar] [CrossRef]
  198. Sasaki, K.; Othman, M.B.; Demura, M.; Watanabe, M.; Isoda, H. Modulation of neurogenesis through the promotion of energy production activity is behind the antidepressant-like effect of colonial green alga, Botryococcus braunii. Front. Physiol. 2017, 8, 900. [Google Scholar] [CrossRef] [PubMed]
  199. Panahi, Y.; Badeli, R.; Karami, G.-R.; Badeli, Z.; Sahebkar, A. A randomized controlled trial of 6-week Chlorella vulgaris supplementation in patients with major depressive disorder. Complement. Ther. Med. 2015, 23, 598–602. [Google Scholar] [CrossRef]
  200. Talbott, S.; Hantla, D.; Capelli, B.; Ding, L.; Li, Y.; Artaria, C. Astaxanthin supplementation reduces depression and fatigue in healthy subjects. EC Nutr. 2019, 14, 239–246. [Google Scholar]
  201. Siddiqui, P.J.A.; Khan, A.; Uddin, N.; Khaliq, S.; Rasheed, M.; Nawaz, S.; Hanif, M.; Dar, A. Antidepressant-like deliverables from the sea: Evidence on the efficacy of three different brown seaweeds via involvement of monoaminergic system. Biosci. Biotechnol. Biochem. 2017, 81, 1369–1378. [Google Scholar] [CrossRef]
  202. Abreu, T.M.; Monteiro, V.S.; Martins, A.B.S.; Teles, F.B.; Rivanor, R.L.d.C.; Mota, F.; Macedo, D.S.; de Vasconcelos, S.M.M.; Júnior, J.E.R.H.; Benevides, N.M.B. Involvement of the dopaminergic system in the antidepressant-like effect of the lectin isolated from the red marine alga Solieria filiformis in mice. Int. J. Biol. Macromol. 2018, 111, 534–541. [Google Scholar] [CrossRef]
  203. Violle, N.; Rozan, P.; Demais, H.; Nyvall Collen, P.; Bisson, J.-F. Evaluation of the antidepressant-and anxiolytic-like effects of a hydrophilic extract from the green seaweed Ulva sp. in rats. Nutr. Neurosci. 2018, 21, 248–256. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, X.; Xiu, Z.; Du, Y.; Li, Y.; Yang, J.; Gao, Y.; Li, F.; Yin, X.; Shi, H. Brazilin treatment produces antidepressant-and anxiolytic-like effects in mice. Biol. Pharm. Bull. 2019, 42, 1268–1274. [Google Scholar] [CrossRef] [PubMed]
  205. Xu, Y.; Wang, Z.; You, W.; Zhang, X.; Li, S.; Barish, P.A.; Vernon, M.M.; Du, X.; Li, G.; Pan, J. Antidepressant-like effect of trans-resveratrol: Involvement of serotonin and noradrenaline system. Eur. Neuropsychopharmacol. 2010, 20, 405–413. [Google Scholar] [CrossRef]
  206. Shewale, P.B.; Patil, R.A.; Hiray, Y.A. Antidepressant-like activity of anthocyanidins from Hibiscus rosa-sinensis flowers in tail suspension test and forced swim test. Indian. J. Pharmacol. 2012, 44, 454. [Google Scholar] [CrossRef]
  207. Ghazizadeh, J.; Sadigh-Eteghad, S.; Marx, W.; Fakhari, A.; Hamedeyazdan, S.; Torbati, M.; Taheri-Tarighi, S.; Araj-khodaei, M.; Mirghafourvand, M. The effects of lemon balm (Melissa officinalis L.) on depression and anxiety in clinical trials: A systematic review and meta-analysis. Phytother. Res. 2021, 35, 6690–6705. [Google Scholar] [CrossRef]
  208. Guzmán-Gutiérrez, S.; Gómez-Cansino, R.; García-Zebadúa, J.; Jiménez-Pérez, N.; Reyes-Chilpa, R. Antidepressant activity of Litsea glaucescens essential oil: Identification of β-pinene and linalool as active principles. J. Ethnopharmacol. 2012, 143, 673–679. [Google Scholar] [CrossRef] [PubMed]
  209. Kim, M.; Nam, E.S.; Lee, Y.; Kang, H.-J. Effects of lavender on anxiety, depression, and physiological parameters: Systematic review and meta-analysis. Asian Nurs. Res. 2021, 15, 279–290. [Google Scholar] [CrossRef] [PubMed]
  210. Fajemiroye, J.O.; Martins, J.L.R.; Ghedini, P.C.; Galdino, P.M.; Paula, J.A.M.d.; Realino de Paula, J.; Da Rocha, F.F.; Costa, E.A. Antidepressive-like property of dichloromethane fraction of Pimenta pseudocaryophyllus and relevance of monoamine metabolic enzymes. Evid.-Based Complement. Altern. Med. 2013, 2013, 659391. [Google Scholar] [CrossRef]
  211. Molina, M.; Contreras, C.; Tellez-Alcantara, P. Mimosa pudica may possess antidepressant actions in the rat. Phytomedicine 1999, 6, 319–323. [Google Scholar] [CrossRef] [PubMed]
  212. Martínez-Vázquez, M.; Estrada-Reyes, R.; Escalona, A.A.; Velázquez, I.L.; Martínez-Mota, L.; Moreno, J.; Heinze, G. Antidepressant-like effects of an alkaloid extract of the aerial parts of Annona cherimolia in mice. J. Ethnopharmacol. 2012, 139, 164–170. [Google Scholar] [CrossRef]
  213. Gabriela, G.-C.; Javier, A.-A.F.; Elisa, V.-A.; Gonzalo, V.-P.; Herlinda, B.-J. Antidepressant-like effect of Tagetes lucida Cav. extract in rats: Involvement of the serotonergic system. Am. J. Chin. Med. 2012, 40, 753–768. [Google Scholar] [CrossRef]
  214. Ali, S.M.; Shamim, S.; Younus, I.; Anwer, L.; Khaliq, S.A. Anxiolytic, antidepressant and inhibitory effect on MAO isoenzymes by Bougainvillea glabra flower extract in rats. Pak. J. Pharm. Sci. 2021, 34, 1963–1968. [Google Scholar] [PubMed]
  215. Kazemian, A.; Parvin, N.; Raisi Dehkordi, Z.; Rafieian-Kopaei, M. The effect of valerian on the anxiety and depression symptoms of the menopause in women referred to shahrekord medical centers. J. Med. Plants 2017, 16, 94–101. [Google Scholar]
  216. Tammadon, M.R.; Nobahar, M.; Hydarinia-Naieni, Z.; Ebrahimian, A.; Ghorbani, R.; Vafaei, A.A. The effects of valerian on sleep quality, depression, and state anxiety in hemodialysis patients: A randomized, double-blind, crossover clinical trial. Oman Med. J. 2021, 36, e255. [Google Scholar] [CrossRef]
  217. Doron, R.; Lotan, D.; Einat, N.; Yaffe, R.; Winer, A.; Marom, I.; Meron, G.; Kately, N.; Rehavi, M. A novel herbal treatment reduces depressive-like behaviors and increases BDNF levels in the brain of stressed mice. Life Sci. 2014, 94, 151–157. [Google Scholar] [CrossRef]
  218. Li, J.-M.; Yang, C.; Zhang, W.-Y.; Kong, L.-D. The effects of banxia houpu decoction on a chronic mild stress model of depression. Zhongguo Zhong Yao Za Zhi = Zhongguo Zhongyao Zazhi = China J. Chin. Mater. Medica 2003, 28, 55–59. [Google Scholar]
  219. Mayer, E.A. Gut feelings: The emerging biology of gut–brain communication. Nat. Rev. Neurosci. 2011, 12, 453–466. [Google Scholar] [CrossRef] [PubMed]
  220. Mayer, E.A.; Knight, R.; Mazmanian, S.K.; Cryan, J.F.; Tillisch, K. Gut microbes and the brain: Paradigm shift in neuroscience. J. Neurosci. 2014, 34, 15490–15496. [Google Scholar] [CrossRef] [PubMed]
  221. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef] [PubMed]
  222. Foster, J.A.; Neufeld, K.-A.M. Gut–brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef] [PubMed]
  223. Stilling, R.M.; Dinan, T.G.; Cryan, J.F. Microbial genes, brain & behaviour—Epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014, 13, 69–86. [Google Scholar] [PubMed]
  224. Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G.; et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109–118. [Google Scholar] [CrossRef] [PubMed]
  225. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  226. Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; Chen, J.; Fan, S.; Du, X.; et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatry 2016, 21, 786–796. [Google Scholar] [CrossRef]
  227. Clapp, M.; Aurora, N.; Herrera, L.; Bhatia, M.; Wilen, E.; Wakefield, S. Gut microbiota’s effect on mental health: The gut-brain axis. Clin. Pract. 2017, 7, 987. [Google Scholar] [CrossRef]
  228. Van de Wouw, M.; Boehme, M.; Lyte, J.M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain–gut axis alterations. J. Physiol. 2018, 596, 4923–4944. [Google Scholar] [CrossRef] [PubMed]
  229. Slyepchenko, A.; Maes, M.; Jacka, F.N.; Köhler, C.A.; Barichello, T.; McIntyre, R.S.; Berk, M.; Grande, I.; Foster, J.A.; Vieta, E.; et al. Gut microbiota, bacterial translocation, and interactions with diet: Pathophysiological links between major depressive disorder and non-communicable medical comorbidities. Psychother. Psychosom. 2016, 86, 31–46. [Google Scholar] [CrossRef] [PubMed]
  230. Dalile, B. Short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
  231. Li, Z.; Lai, J.; Zhang, P.; Ding, J.; Jiang, J.; Liu, C.; Huang, H.; Zhen, H.; Xi, C.; Sun, Y.; et al. Multi-omics analyses of serum metabolome, gut microbiome and brain function reveal dysregulated microbiota-gut-brain axis in bipolar depression. Mol. Psychiatry 2022, 27, 4123–4135. [Google Scholar] [CrossRef] [PubMed]
  232. Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef]
  233. Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef] [PubMed]
  234. Maes, M.; Kubera, M.; Leunis, J.-C.; Berk, M. Increased IgA and IgM responses against gut commensals in chronic depression: Further evidence for increased bacterial translocation or leaky gut. J. Affect. Disord. 2012, 141, 55–62. [Google Scholar] [CrossRef] [PubMed]
  235. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed]
  236. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  237. Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2019, 4, 396–403. [Google Scholar] [CrossRef]
  238. Bear, T.L.; Dalziel, J.E.; Coad, J.; Roy, N.C.; Butts, C.A.; Gopal, P.K. The role of the gut microbiota in dietary interventions for depression and anxiety. Adv. Nutr. 2020, 11, 890–907. [Google Scholar] [CrossRef] [PubMed]
  239. Slyepchenko, A.; FCarvalho, A.; SCha, D.; Kasper, S.; SMcIntyre, R. Gut emotions-mechanisms of action of probiotics as novel therapeutic targets for depression and anxiety disorders. CNS Neurol. Disord.-Drug Targets (Former. Curr. Drug Targets-CNS Neurol. Disord.) 2014, 13, 1770–1786. [Google Scholar] [CrossRef]
  240. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed]
  241. Tian, P.; O’Riordan, K.J.; Lee, Y.-K.; Wang, G.; Zhao, J.; Zhang, H.; Cryan, J.F.; Chen, W. Towards a psychobiotic therapy for depression: Bifidobacterium breve CCFM1025 reverses chronic stress-induced depressive symptoms and gut microbial abnormalities in mice. Neurobiol. Stress. 2020, 12, 100216. [Google Scholar] [CrossRef] [PubMed]
  242. Tian, P.; Chen, Y.; Zhu, H.; Wang, L.; Qian, X.; Zou, R.; Zhao, J.; Zhang, H.; Qian, L.; Wang, Q.; et al. Bifidobacterium breve CCFM1025 attenuates major depression disorder via regulating gut microbiome and tryptophan metabolism: A randomized clinical trial. Brain Behav. Immun. 2022, 100, 233–241. [Google Scholar] [CrossRef] [PubMed]
  243. Tian, P.; Chen, Y.; Qian, X.; Zou, R.; Zhu, H.; Zhao, J.; Zhang, H.; Wang, G.; Chen, W. Pediococcus acidilactici CCFM6432 mitigates chronic stress-induced anxiety and gut microbial abnormalities. Food Funct. 2021, 12, 11241–11249. [Google Scholar] [CrossRef] [PubMed]
  244. Knudsen, J.K.; Michaelsen, T.Y.; Bundgaard-Nielsen, C.; Nielsen, R.E.; Hjerrild, S.; Leutscher, P.; Wegener, G.; Sørensen, S. Faecal microbiota transplantation from patients with depression or healthy individuals into rats modulates mood-related behaviour. Sci. Rep. 2021, 11, 21869. [Google Scholar] [CrossRef] [PubMed]
  245. Doll, J.P.K.; Vázquez-Castellanos, J.F.; Schaub, A.-C.; Schweinfurth, N.; Kettelhack, C.; Schneider, E.; Yamanbaeva, G.; Mählmann, L.; Brand, S.; Beglinger, C.; et al. Fecal Microbiota Transplantation (FMT) as an Adjunctive Therapy for Depression-Case Report. Front. Psychiatry 2022, 13, 815422. [Google Scholar] [CrossRef] [PubMed]
  246. Miller, A.H.; Raison, C.L. The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2016, 16, 22–34. [Google Scholar] [CrossRef]
  247. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctôt, K.L. A meta-analysis of cytokines in major depression. Biol. Psychiatry 2010, 67, 446–457. [Google Scholar] [CrossRef]
  248. Ting, E.Y.-C.; Yang, A.C.; Tsai, S.-J. Role of interleukin-6 in depressive disorder. Int. J. Mol. Sci. 2020, 21, 2194. [Google Scholar] [CrossRef]
  249. Hodes, G.E.; Ménard, C.; Russo, S.J. Integrating Interleukin-6 into depression diagnosis and treatment. Neurobiol. Stress 2016, 4, 15–22. [Google Scholar] [CrossRef]
  250. Howren, M.B.; Lamkin, D.M.; Suls, J. Associations of depression with C-reactive protein, IL-1, and IL-6: A meta-analysis. Psychosom. Med. 2009, 71, 171–186. [Google Scholar] [CrossRef]
  251. Dantzer, R.; O’connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef]
  252. Dantzer, R.; O’Connor, J.C.; Lawson, M.A.; Kelley, K.W. Inflammation-associated depression: From serotonin to kynurenine. Psychoneuroendocrinology 2011, 36, 426–436. [Google Scholar] [CrossRef]
  253. Pariante, C.M.; Lightman, S.L. The HPA axis in major depression: Classical theories and new developments. Trends Neurosci. 2008, 31, 464–468. [Google Scholar] [CrossRef]
  254. Kéri, S.; Szabó, C.; Kelemen, O. Expression of Toll-Like Receptors in peripheral blood mononuclear cells and response to cognitive-behavioral therapy in major depressive disorder. Brain Behav. Immun. 2014, 40, 235–243. [Google Scholar] [CrossRef]
  255. Isaković, J.; Gorup, D.; Mitrečić, D. Molecular mechanisms of microglia-and astrocyte-driven neurorestoration triggered by application of electromagnetic fields. Croat. Med. J. 2019, 60, 127–140. [Google Scholar] [CrossRef]
  256. He, G.-L.; Liu, Y.; Li, M.; Chen, C.-H.; Gao, P.; Yu, Z.-P.; Yang, X.-S. The amelioration of phagocytic ability in microglial cells by curcumin through the inhibition of EMF-induced pro-inflammatory responses. J. Neuroinflamm. 2014, 11, 49. [Google Scholar] [CrossRef]
  257. Lopresti, A.L.; Maes, M.; Maker, G.L.; Hood, S.D.; Drummond, P.D. Curcumin for the treatment of major depression: A randomised, double-blind, placebo controlled study. J. Affect. Disord. 2014, 167, 368–375. [Google Scholar] [CrossRef]
  258. Yu, J.-J.; Pei, L.-B.; Zhang, Y.; Wen, Z.-Y.; Yang, J.-L. Chronic supplementation of curcumin enhances the efficacy of antidepressants in major depressive disorder: A randomized, double-blind, placebo-controlled pilot study. J. Clin. Psychopharmacol. 2015, 35, 406–410. [Google Scholar] [CrossRef] [PubMed]
  259. Sanmukhani, J.; Satodiya, V.; Trivedi, J.; Patel, T.; Tiwari, D.; Panchal, B.; Goel, A.; Tripathi, C.B. Efficacy and safety of curcumin in major depressive disorder: A randomized controlled trial. Phytother. Res. 2014, 28, 579–585. [Google Scholar] [CrossRef] [PubMed]
  260. Ramaholimihaso, T.; Bouazzaoui, F.; Kaladjian, A. Curcumin in depression: Potential mechanisms of action and current evidence—A narrative review. Front. Psychiatry 2020, 11, 572533. [Google Scholar] [CrossRef] [PubMed]
  261. Liu, D.; Wang, Z.; Gao, Z.; Xie, K.; Zhang, Q.; Jiang, H.; Pang, Q. Effects of curcumin on learning and memory deficits, BDNF, and ERK protein expression in rats exposed to chronic unpredictable stress. Behav. Brain Res. 2014, 271, 116–121. [Google Scholar] [CrossRef]
  262. Lass, P.; Slawek, J.; Derejko, M.; Rubello, D. Neurological and psychiatric disorders in thyroid dysfunctions. Role Nucl. Med. SPECT PET Imaging. Minerva Endocrinol. 2008, 33, 75–84. [Google Scholar]
  263. Kirkegaard, C.; Faber, J. The role of thyroid hormones in depression. Eur. J. Endocrinol. 1998, 138, 1–9. [Google Scholar] [CrossRef]
  264. Ittermann, T.; Völzke, H.; Baumeister, S.E.; Appel, K.; Grabe, H.J. Diagnosed thyroid disorders are associated with depression and anxiety. Soc. Psychiatry Psychiatr. Epidemiol. 2015, 50, 1417–1425. [Google Scholar] [CrossRef]
  265. Bauer, M.; Goetz, T.; Glenn, T.; Whybrow, P. The thyroid-brain interaction in thyroid disorders and mood disorders. J. Neuroendocrinol. 2008, 20, 1101–1114. [Google Scholar] [CrossRef]
  266. Nuguru, S.P.; Rachakonda, S.; Sripathi, S.; Khan, M.I.; Patel, N.; Meda, R.T. Hypothyroidism and depression: A narrative review. Cureus 2022, 14, e28201. [Google Scholar] [CrossRef]
  267. Loh, H.H.; Lim, L.L.; Yee, A.; Loh, H.S. Association between subclinical hypothyroidism and depression: An updated systematic review and meta-analysis. BMC Psychiatry 2019, 19, 12. [Google Scholar] [CrossRef]
  268. Cleare, A.; McGregor, A.; O’keane, V. Neuroendocrine evidence for an association between hypothyroidism, reduced central 5-HT activity and depression. Clin. Endocrinol. 1995, 43, 713–719. [Google Scholar] [CrossRef] [PubMed]
  269. Duval, F.; Mokrani, M.-C.; Erb, A.; Danila, V.; Lopera, F.G.; Foucher, J.R.; Jeanjean, L.C. Thyroid axis activity and dopamine function in depression. Psychoneuroendocrinology 2021, 128, 105219. [Google Scholar] [CrossRef]
  270. Swann, A. Thyroid hormone and norepinephrine: Effects on alpha-2, beta, and reuptake sites in cerebral cortex and heart. J. Neural Transm. 1988, 71, 195–205. [Google Scholar] [CrossRef]
  271. Haggerty Jr, J.J.; Prange Jr, A.J. Borderline hypothyroidism and depression. Annu. Rev. Med. 1995, 46, 37–46. [Google Scholar] [CrossRef]
  272. Sullivan, P.; Wilson, D.; Mulder, R.; Joyce, P. The hypothalamic-pituitary-thyroid axis in major depression. Acta Psychiatr. Scand. 1997, 95, 370–378. [Google Scholar] [CrossRef]
  273. Hage, M.P.; Azar, S.T. The link between thyroid function and depression. J. Thyroid. Res. 2011, 2012, 590648. [Google Scholar] [CrossRef]
  274. Hein, M.D.; Jackson, I.M. Thyroid function in psychiatric illness. Gen. Hosp. Psychiatry 1990, 12, 232–244. [Google Scholar] [CrossRef] [PubMed]
  275. Kotkowska, Z.; Strzelecki, D. Depression and Autoimmune Hypothyroidism—Their Relationship and the Effects of Treating Psychiatric and Thyroid Disorders on Changes in Clinical and Biochemical Parameters Including BDNF and Other Cytokines—A Systematic Review. Pharmaceuticals 2022, 15, 391. [Google Scholar] [CrossRef]
  276. Betsy, A.; Binitha, M.; Sarita, S. Zinc deficiency associated with hypothyroidism: An overlooked cause of severe alopecia. Int. J. Trichology 2013, 5, 40–42. [Google Scholar]
  277. Rayman, M.P. Multiple nutritional factors and thyroid disease, with particular reference to autoimmune thyroid disease. Proc. Nutr. Soc. 2019, 78, 34–44. [Google Scholar] [CrossRef]
  278. Ventura, M.; Melo, M.; Carrilho, F. Selenium and thyroid disease: From pathophysiology to treatment. Int. J. Endocrinol. 2017, 2017, 1297658. [Google Scholar] [CrossRef]
  279. Trifu, S.; Popescu, A.; Dragoi, A.; Trifu, A. Thyroid hormones as a third line of augmentation medication in treatment-resistant depression. Acta Endocrinológica 2020, 16, 256. [Google Scholar] [CrossRef]
  280. Bauer, M.; Heinz, A.; Whybrow, P. Thyroid hormones, serotonin and mood: Of synergy and significance in the adult brain. Mol. Psychiatry 2002, 7, 140–156. [Google Scholar] [CrossRef]
  281. Sarris, J.; O’Neil, A.; Coulson, C.E.; Schweitzer, I.; Berk, M. Lifestyle medicine for depression. BMC Psychiatry 2014, 14, 107. [Google Scholar] [CrossRef]
  282. Wong, V.W.-H.; Ho, F.Y.-Y.; Shi, N.-K.; Sarris, J.; Chung, K.-F.; Yeung, W.-F. Lifestyle medicine for depression: A meta-analysis of randomized controlled trials. J. Affect. Disord. 2021, 284, 203–216. [Google Scholar] [CrossRef]
  283. Gómez-Gómez, I.; Bellón, J.; Resurrección, D.M.; Cuijpers, P.; Moreno-Peral, P.; Rigabert, A.; Maderuelo-Fernández, J.; Motrico, E. Effectiveness of universal multiple-risk lifestyle interventions in reducing depressive symptoms: Systematic review and meta-analysis. Prev. Med. 2020, 134, 106067. [Google Scholar] [CrossRef]
  284. Lopresti, A.L.; Hood, S.D.; Drummond, P.D. A review of lifestyle factors that contribute to important pathways associated with major depression: Diet, sleep and exercise. J. Affect. Disord. 2013, 148, 12–27. [Google Scholar] [CrossRef]
  285. Berk, M.; Sarris, J.; Coulson, C.E.; Jacka, F.N. Lifestyle management of unipolar depression. Acta Psychiatr. Scand. 2013, 127, 38–54. [Google Scholar] [CrossRef]
  286. Binnewies, J.; Nawijn, L.; van Tol, M.-J.; van der Wee, N.J.; Veltman, D.J.; Penninx, B.W. Associations between depression, lifestyle and brain structure: A longitudinal MRI study. NeuroImage 2021, 231, 117834. [Google Scholar] [CrossRef]
  287. Wang, X.; Arafa, A.; Liu, K.; Eshak, E.S.; Hu, Y.; Dong, J.-Y. Combined healthy lifestyle and depressive symptoms: A meta-analysis of observational studies. J. Affect. Disord. 2021, 289, 144–150. [Google Scholar] [CrossRef]
  288. Van Dammen, L.; Wekker, V.; De Rooij, S.; Groen, H.; Hoek, A.; Roseboom, T. A systematic review and meta-analysis of lifestyle interventions in women of reproductive age with overweight or obesity: The effects on symptoms of depression and anxiety. Obes. Rev. 2018, 19, 1679–1687. [Google Scholar] [CrossRef] [PubMed]
  289. Bruins, J.; Jörg, F.; Bruggeman, R.; Slooff, C.; Corpeleijn, E.; Pijnenborg, M. The Effects of Lifestyle Interventions on (Long-Term) Weight Management, Cardiometabolic Risk and Depressive Symptoms in People with Psychotic Disorders: A Meta-Analysis. PLoS ONE 2014, 9, e112276. [Google Scholar] [CrossRef] [PubMed]
  290. Pinniger, R.; Brown, R.F.; Thorsteinsson, E.B.; McKinley, P. Argentine tango dance compared to mindfulness meditation and a waiting-list control: A randomised trial for treating depression. Complement. Ther. Med. 2012, 20, 377–384. [Google Scholar] [CrossRef] [PubMed]
  291. Pots, W.T.; Meulenbeek, P.A.; Veehof, M.M.; Klungers, J.; Bohlmeijer, E.T. The efficacy of mindfulness-based cognitive therapy as a public mental health intervention for adults with mild to moderate depressive symptomatology: A randomized controlled trial. PLoS ONE 2014, 9, e109789. [Google Scholar] [CrossRef] [PubMed]
  292. McCarney, R.W.; Schulz, J.; Grey, A.R. Effectiveness of mindfulness-based therapies in reducing symptoms of depression: A meta-analysis. Eur. J. Psychother. Couns. 2012, 14, 279–299. [Google Scholar] [CrossRef]
  293. Gee, B.; Orchard, F.; Clarke, E.; Joy, A.; Clarke, T.; Reynolds, S. The effect of non-pharmacological sleep interventions on depression symptoms: A meta-analysis of randomised controlled trials. Sleep Med. Rev. 2019, 43, 118–128. [Google Scholar] [CrossRef] [PubMed]
  294. Siah, C.J.R.; Goh, Y.S.; Lee, J.; Poon, S.N.; Ow Yong, J.Q.Y.; Tam, W.-S.W. The effects of forest bathing on psychological well-being: A systematic review and meta-analysis. Int. J. Ment. Health Nurs. 2023, 32, 1038–1054. [Google Scholar] [CrossRef]
  295. Souter, M.A.; Miller, M.D. Do Animal-Assisted Activities Effectively Treat Depression? A Meta-Analysis. Anthrozoös 2007, 20, 167–180. [Google Scholar] [CrossRef]
  296. Siette, J.; Cassidy, M.; Priebe, S. Effectiveness of befriending interventions: A systematic review and meta-analysis. BMJ Open 2017, 7, e014304. [Google Scholar] [CrossRef]
  297. Stice, E.; Burton, E.; Kate Bearman, S.; Rohde, P. Randomized trial of a brief depression prevention program: An elusive search for a psychosocial placebo control condition. Behav. Res. Ther. 2007, 45, 863–876. [Google Scholar] [CrossRef]
  298. Cregg, D.R.; Cheavens, J.S. Gratitude Interventions: Effective Self-help? A Meta-analysis of the Impact on Symptoms of Depression and Anxiety. J. Happiness Stud. 2021, 22, 413–445. [Google Scholar] [CrossRef]
  299. Kisely, S.; Li, A.; Warren, N.; Siskind, D. A systematic review and meta-analysis of deep brain stimulation for depression. Depress. Anxiety 2018, 35, 468–480. [Google Scholar] [CrossRef] [PubMed]
  300. Tai, Y.; Obayashi, K.; Yamagami, Y.; Kurumatani, N.; Saeki, K. Association Between Passive Body Heating by Hot Water Bathing Before Bedtime and Depressive Symptoms Among Community-Dwelling Older Adults. Am. J. Geriatr. Psychiatry 2022, 30, 161–170. [Google Scholar] [CrossRef] [PubMed]
  301. de Oliveira, G.D.; Oancea, S.C.; Nucci, L.B.; Vogeltanz-Holm, N. The association between physical activity and depression among individuals residing in Brazil. Soc. Psychiatry Psychiatr. Epidemiol. 2018, 53, 373–383. [Google Scholar] [CrossRef] [PubMed]
  302. Stubbs, B.; Koyanagi, A.; Schuch, F.B.; Firth, J.; Rosenbaum, S.; Veronese, N.; Solmi, M.; Mugisha, J.; Vancampfort, D. Physical activity and depression: A large cross-sectional, population-based study across 36 low- and middle-income countries. Acta Psychiatr. Scand. 2016, 134, 546–556. [Google Scholar] [CrossRef]
  303. Pearce, M.; Garcia, L.; Abbas, A.; Strain, T.; Schuch, F.B.; Golubic, R.; Kelly, P.; Khan, S.; Utukuri, M.; Laird, Y.; et al. Association between Physical Activity and Risk of Depression: A Systematic Review and Meta-analysis. JAMA Psychiatry 2022, 79, 550–559. [Google Scholar] [CrossRef] [PubMed]
  304. Laird, E.; Rasmussen, C.L.; Kenny, R.A.; Herring, M.P. Physical Activity Dose and Depression in a Cohort of Older Adults in The Irish Longitudinal Study on Ageing. JAMA Netw. Open 2023, 6, e2322489. [Google Scholar] [CrossRef] [PubMed]
  305. Schuch, F.B.; Vancampfort, D.; Richards, J.; Rosenbaum, S.; Ward, P.B.; Stubbs, B. Exercise as a treatment for depression: A meta-analysis adjusting for publication bias. J. Psychiatr. Res. 2016, 77, 42–51. [Google Scholar] [CrossRef] [PubMed]
  306. Krogh, J.; Hjorthøj, C.; Speyer, H.; Gluud, C.; Nordentoft, M. Exercise for patients with major depression: A systematic review with meta-analysis and trial sequential analysis. BMJ Open 2017, 7, e014820. [Google Scholar] [CrossRef]
  307. Dunn, A.L.; Trivedi, M.H.; Kampert, J.B.; Clark, C.G.; Chambliss, H.O. Exercise treatment for depression: Efficacy and dose response. Am. J. Prev. Med. 2005, 28, 1–8. [Google Scholar] [CrossRef]
  308. Meyer, J.D.; Koltyn, K.F.; Stegner, A.J.; Kim, J.S.; Cook, D.B. Influence of Exercise Intensity for Improving Depressed Mood in Depression: A Dose-Response Study. Behav. Ther. 2016, 47, 527–537. [Google Scholar] [CrossRef] [PubMed]
  309. Lavebratt, C.; Herring, M.P.; Liu, J.J.; Wei, Y.B.; Bossoli, D.; Hallgren, M.; Forsell, Y. Interleukin-6 and depressive symptom severity in response to physical exercise. Psychiatry Res. 2017, 252, 270–276. [Google Scholar] [CrossRef] [PubMed]
  310. Furuyashiki, A.; Tabuchi, K.; Norikoshi, K.; Kobayashi, T.; Oriyama, S. A comparative study of the physiological and psychological effects of forest bathing (Shinrin-yoku) on working age people with and without depressive tendencies. Environ. Health Prev. Med. 2019, 24, 46. [Google Scholar] [CrossRef] [PubMed]
  311. Li, Q.; Ochiai, H.; Ochiai, T.; Takayama, N.; Kumeda, S.; Miura, T.; Aoyagi, Y.; Imai, M. Effects of forest bathing (shinrin-yoku) on serotonin in serum, depressive symptoms and subjective sleep quality in middle-aged males. Environ. Health Prev. Med. 2022, 27, 44. [Google Scholar] [CrossRef] [PubMed]
  312. Phillips, W.M. Purpose in life, depression, and locus of control. J. Clin. Psychol. 1980, 36, 661–667. [Google Scholar] [CrossRef] [PubMed]
  313. Robak, R.W.; Griffin, P.W. Purpose in life: What is its relationship to happiness, depression, and grieving? N. Am. J. Psychol. 2000, 2, 113–119. [Google Scholar]
  314. Jones, E. COVID-19 and the Blitz compared: Mental health outcomes in the UK. Lancet Psychiatry 2021, 8, 708–716. [Google Scholar] [CrossRef] [PubMed]
  315. Coverdale, B.J. Evaluating the Effectiveness of Upward Bound Programs. Master’s Thesis, The Ohio State University, Columbus, OH, USA, 2009. [Google Scholar]
  316. Rodiek, S. Influence of an outdoor garden on mood and stress in older persons. J. Ther. Hortic. 2002, 13, 13–21. [Google Scholar]
  317. Dinas, P.; Koutedakis, Y.; Flouris, A. Effects of exercise and physical activity on depression. Ir. J. Med. Sci. 2011, 180, 319–325. [Google Scholar] [CrossRef]
  318. Ewert, A. The Effects of Outdoor Adventure Activities Upon Self-Concept. Master’s Thesis, Eastern Washington University, Cheney, WA, USA, 1977. [Google Scholar]
  319. Emmons, R.A.; Crumpler, C.A. Gratitude as a Human Strength: Appraising the Evidence. J. Soc. Clin. Psychol. 2000, 19, 56–69. [Google Scholar] [CrossRef]
  320. Chen, Y.; Ishak, Z. Gratitude Diary: The Impact on Depression Symptoms. Psychology 2022, 13, 443–453. [Google Scholar] [CrossRef]
  321. Feng, L.; Yin, R. Social Support and Hope Mediate the Relationship Between Gratitude and Depression Among Front-Line Medical Staff During the Pandemic of COVID-19. Front. Psychol. 2021, 12, 623873. [Google Scholar] [CrossRef] [PubMed]
  322. Bryan, J.L.; Young, C.M.; Lucas, S.; Quist, M.C. Should I say thank you? Gratitude encourages cognitive reappraisal and buffers the negative impact of ambivalence over emotional expression on depression. Personal. Individ. Differ. 2018, 120, 253–258. [Google Scholar] [CrossRef]
  323. Kaniuka, A.R.; Kelliher Rabon, J.; Brooks, B.D.; Sirois, F.; Kleiman, E.; Hirsch, J.K. Gratitude and suicide risk among college students: Substantiating the protective benefits of being thankful. J. Am. Coll. Health 2021, 69, 660–667. [Google Scholar] [CrossRef]
  324. Gogo, A.; Osta, A.; McClafferty, H.; Rana, D.T. Cultivating a way of being and doing: Individual strategies for physician well-being and resilience. Curr. Probl. Pediatr. Adolesc. Health Care 2019, 49, 100663. [Google Scholar] [CrossRef]
  325. Davis, D.E.; Choe, E.; Meyers, J.; Wade, N.; Varjas, K.; Gifford, A.; Quinn, A.; Hook, J.N.; Van Tongeren, D.R.; Griffin, B.J.; et al. Thankful for the little things: A meta-analysis of gratitude interventions. J. Couns. Psychol. 2016, 63, 20–31. [Google Scholar] [CrossRef] [PubMed]
  326. Liu, S.; Sheng, J.; Li, B.; Zhang, X. Recent advances in non-invasive brain stimulation for major depressive disorder. Front. Hum. Neurosci. 2017, 11, 526. [Google Scholar] [CrossRef]
  327. Brononi, A.R.; Sampaio-Junior, B.; Moffa, A.H.; Aparicio, L.; Gordon, P.; Klein, I.; Rios, R.M. Noninvasive brain stimulation in psychiatric disorders: A primer. Braz. J. Psychiatry 2019, 4, 70–81. [Google Scholar] [CrossRef] [PubMed]
  328. Dunlop, K.; Hanlon, C.A.; Downar, J. Noninvasive brain stimulation treatments for addiction and major depression. Ann. N. Y. Acad. Sci. 2017, 1394, 31–54. [Google Scholar] [CrossRef]
  329. Mutz, J.; Edgcumbe, D.R.; Brunoni, A.R.; Fu, C.H. Efficacy and acceptability of non-invasive brain stimulation for the treatment of adult unipolar and bipolar depression: A systematic review and meta-analysis of randomised sham-controlled trials. Neurosci. Biohevioral Rev. 2018, 92, 291–303. [Google Scholar] [CrossRef]
  330. McClure, D.; Greenman, S.C.; Koppulu, S.S.; Varvara, M.; Yaseen, Z.S.; Galynker, I.I. A pilot study of safety and efficacy of cranial electrotherapy stimulation in treatment of bipolar II depression. J. Nerv. Ment. Dis. 2015, 203, 827–835. [Google Scholar] [CrossRef] [PubMed]
  331. Hanusch, K.U.; Janssen, C.W. The impact of whole-body hyperthermia interventions on mood and depression—Are we ready for recommendations for clinical application? Int. J. Hyperth. 2019, 36, 573–581. [Google Scholar] [CrossRef] [PubMed]
  332. Hussain, J.; Cohen, M. Clinical effects of regular dry sauna bathing: A systematic review. Evid.-Based Complement. Altern. Med. 2018, 2018, 1857413. [Google Scholar] [CrossRef] [PubMed]
  333. Laukkanen, J.A.; Laukkanen, T.; Kunustor, S.K. Cardiovascular and other health benefits of sauna bathing: A review of the evidence. Mayo Clin. Proc. 2018, 93, 1111–1121. [Google Scholar] [CrossRef] [PubMed]
  334. Laukkanen, T.; Khan, H.; Zaccardi, F.; Laukkanen, J.A. Association between sauna bathing and fatal cardiovascular and all-cuase mortality. JAMA Intern. Med. 2015, 175, 542–548. [Google Scholar] [CrossRef] [PubMed]
  335. Laukkanen, T.; Kunutsor, S.; Kauhanen, J.; Laukkanen, J.A. Sauna bathing is inversely associated with dementia and Alzheimer’s disease in middle-aged Finnish men. Age Ageing 2017, 46, 245–249. [Google Scholar] [CrossRef] [PubMed]
  336. Kunutsor, S.K.; Khan, H.; Laukkanen, T.; Laukkanen, J.A. Joint associations of sauna bathing and cardiorespiratory fitness on cardiovascular and all-cause mortality risk: A long-term prospective cohort study. Ann. Med. 2018, 50, 139–146. [Google Scholar] [CrossRef] [PubMed]
  337. Scoon, G.S.; Hopkins, W.G.; Mayhew, S.; Cotter, J.D. Effect of post-exercise sauna bathing on the endurance performance of competitive male runners. J. Sci. Med. Sport. 2007, 10, 259–262. [Google Scholar] [CrossRef] [PubMed]
  338. Flux, M.C.; Smith, D.G.; Allen, J.J.B.; Mehl, M.R.; Medrano, A.; Begay, T.K.; Middlemist, B.H.; Marquart, B.M.; Cole, S.P.; Sauder, C.J.; et al. Association of plasma cytokines and antidepressant response following mild-intensity whole-body hyperthermia in major depressive disorder. Transl. Psychiatry 2023, 13, 132. [Google Scholar] [CrossRef]
  339. Amano, K.; Yanagihori, R.; Tei, C. Waon therapyis effective as the treatment of myalgic encephalomyelitis/Chronic fatigue syndrome. J. Jpn. Soc. Balneol. Clim. Phys. Med. 2015, 78, 285–302. [Google Scholar]
  340. Soejima, Y.; Munemoto, T.; Masuda, A.; Uwatoko, Y.; Miyata, M.; Tei, C. Effects of Waon therapy on chronic fatigue syndrome: A pilot study. Intern. Med. 2015, 54, 333–338. [Google Scholar] [CrossRef] [PubMed]
  341. Lowry, C.A.; Hale, M.W.; Evans, A.K.; Heerkens, J.; Staub, D.R.; Gasser, P.J.; Shekhar, A. Serotonergic systems, anxiety, and affective disorder: Focus on the dorsomedial part of the dorsal raphe nucleus. Ann. N. Y Acad. Sci. 2008, 1148, 86–94. [Google Scholar] [CrossRef]
  342. Hanusch, K.U.; Janssen, C.H.; Billheimer, D.; Jenkins, I.; Spurgeon, E.; Lowry, C.A.; Raison, C.L. Whole-body hyperthermia for the treatment of major depression: Associations with thermoregulatory cooling. Am. J. Psychiatry 2013, 170, 802–804. [Google Scholar] [CrossRef] [PubMed]
  343. Janssen, C.W.; Lowry, C.A.; Mehl, M.R.; Allen, J.J.; Kelly, K.L. Whole-body hyperthermia for the treatment of major depressive disorder.A randomized Clinical Trial. JAMA Psychiatry 2016, 73, 789–795. [Google Scholar] [CrossRef] [PubMed]
  344. Drevets, W.C.; Bogers, W.; Raichle, M.E. Functional anatomical correlates of antidepressant drug treatment assessed using PET measures of regional glucose metabolism. Eur. Neuropsychopharmacol. 2002, 12, 527–544. [Google Scholar] [CrossRef] [PubMed]
  345. Bansal, Y.; Kuhad, A. Mitochondrial Dysfunction in Depression. Curr. Neuropharmacol. 2016, 14, 610–618. [Google Scholar] [CrossRef] [PubMed]
  346. Gardner, A.; Johansson, A.; Wibom, R.; Nennesmo, I.; von Döbeln, U.; Hagenfeldt, L.; Hällström, T. Alterations of mitochondrial function and correlations with personality traits in selected major depressive disorder patients. J. Affect. Disord. 2003, 76, 55–68. [Google Scholar] [CrossRef] [PubMed]
  347. Rezin, G.T.; Cardoso, M.R.; Gonçalves, C.L.; Scaini, G.; Fraga, D.B.; Riegel, R.E.; Comim, C.M.; Quevedo, J.; Streck, E.L. Inhibition of mitochondrial respiratory chain in brain of rats subjected to an experimental model of depression. Neurochem. Int. 2008, 53, 395–400. [Google Scholar] [CrossRef] [PubMed]
  348. Karabatsiakis, A.; Böck, C.; Salinas-Manrique, J.; Kolassa, S.; Calzia, E.; Dietrich, D.E.; Kolassa, I.T. Mitochondrial respiration in peripheral blood mononuclear cells correlates with depressive subsymptoms and severity of major depression. Transl. Psychiatry 2014, 4, e397. [Google Scholar] [CrossRef]
  349. Hroudová, J.; Fišar, Z.; Kitzlerová, E.; Zvěřová, M.; Raboch, J. Mitochondrial respiration in blood platelets of depressive patients. Mitochondrion 2013, 13, 795–800. [Google Scholar] [CrossRef]
  350. Askalsky, P.; Losifescu, D.V. Transcranial photobiomodulation for the management of depression: Current perspectives. Neuropsychiatr. Dis. Treat. 2019, 15, 3255–3272. [Google Scholar] [CrossRef] [PubMed]
  351. Hamblin, M.R. Shining light on the head: Photobiomodulation for brain disorders. BBA Clin. 2016, 6, 113–124. [Google Scholar] [CrossRef] [PubMed]
  352. Salehpour, F.; Ahmadian, N.; Rasta, S.H.; Farhoudi, M.; Karimi, P.; Sadigh-Eteghad, S. Transcranial low-level laser therapy improves brain mitochondrial function and cognitive impairment in D-galactose-induced aging mice. Neurobiol. Aging 2017, 58, 140–150. [Google Scholar] [CrossRef] [PubMed]
  353. Wang, X.; Tian, F.; Reddy, D.D.; Nalawade, S.S.; Barrett, D.W.; Gonzalez-Lima, F.; Liu, H. Up-regulation of cerebral cytochrome-c-oxidase and hemodynamics by transcranial infrared laser stimulation: A broadband near-infrared spectroscopy study. J. Cereb. Blood Flow. Metab. 2017, 37, 3789–3802. [Google Scholar] [CrossRef] [PubMed]
  354. Sanderson, T.H.; Wider, J.M.; Lee, I.; Reynolds, C.A.; Liu, J.; Lepore, B.; Tousignant, R.; Bukowski, M.J.; Johnston, H.; Fite, A.; et al. Inhibitory modulation of cytochrome c oxidase activity with specific near-infrared light wavelengths attenuates brain ischemia/reperfusion injury. Sci. Rep. 2018, 8, 3481. [Google Scholar] [CrossRef]
  355. Oron, U.; Ilic, S.; De Taboada, L.; Streeter, J. Ga-As (808 nm) laser irradiation enhances ATP production in human neuronal cells in culture. Photomed. Laser Surg. 2007, 25, 180–182. [Google Scholar] [CrossRef] [PubMed]
  356. Wu, Q.; Xuan, W.; Ando, T.; Xu, T.; Huang, L.; Huang, Y.; Dai, T.; Dhital, S.; Sharma, S.K.; Whalen, M.J.; et al. Low-level laser therapy for closed-head traumatic brain injury in mice: Effect of different wavelengths. Lasers Surg. Med. 2012, 44, 218–226. [Google Scholar] [CrossRef] [PubMed]
  357. Gabel, C.P.; Petrie, S.R.; Mischoulon, D.; Hamblin, M.R.; Yeung, A.; Sangermano, L.; Cassano, P. A case control series for the effect of photobiomodulation in patients with low back pain and concurrent depression. Laser Ther. 2018, 27, 167–173. [Google Scholar] [CrossRef]
  358. Oron, A.; Oron, U. Low-Level Laser Therapy to the Bone Marrow Ameliorates Neurodegenerative Disease Progression in a Mouse Model of Alzheimer’s Disease: A Minireview. Photomed. Laser Surg. 2016, 34, 627–630. [Google Scholar] [CrossRef]
  359. Real, T. I Don’t Want to Talk about It: Overcoming the Secret Legacy of Male Depression; Simon and Schuster: New York, NY, USA, 1998. [Google Scholar]
Table 2. A summary of lifestyle interventions and their impacts on depression.
Table 2. A summary of lifestyle interventions and their impacts on depression.
InterventionEffect
DanceAntidepressant impact (SMD = 0.50, p = 0.01) [290]
MindfulnessDecreases in depressive symptoms (SMD = 0.31–0.56) [291,292]
SleepImproved sleep quality decreases depressive symptoms (SMD = −0.45 [−0.55, −0.36])
[293]
Natural environmentsIncreases positive mood and lowers feelings of depression SMD = −0.67 [−0.99, −0.35] [294]
Time with animalsReduction in depressive symptoms (SMD = 0.61 [0.03, 1.19]) [295].
SocializationSignificant improvement in depressive scores SMD = 0.18 [−0.00 to 0.36] [296]
JournalingPositive impact (SMD = 0.61 [0.19, 1.02]) [297]
GratitudeAssociated with positive mental health, including alleviating depression (SMD = Reduction in depressive symptoms 0.29 [−0.37, −0.23]) [298]
Deep brain stimulationReduction in mean depression score SMD = –0.42 [–0.72, −0.12] [299]
Sauna/whole body hyperthermiaReduced odds of depressive symptoms for people using sauna
OR = 0.60 [0.39, 0.90] [300]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Halma, M.; Plothe, C.; Marik, P.E. Integrative Interventions for Improving Outcomes in Depression: A Narrative Review. Psychol. Int. 2024, 6, 550-577. https://doi.org/10.3390/psycholint6020033

AMA Style

Halma M, Plothe C, Marik PE. Integrative Interventions for Improving Outcomes in Depression: A Narrative Review. Psychology International. 2024; 6(2):550-577. https://doi.org/10.3390/psycholint6020033

Chicago/Turabian Style

Halma, Matthew, Christof Plothe, and Paul E. Marik. 2024. "Integrative Interventions for Improving Outcomes in Depression: A Narrative Review" Psychology International 6, no. 2: 550-577. https://doi.org/10.3390/psycholint6020033

Article Metrics

Back to TopTop