- Review
- Open access
- Published:
A comprehensive review on the impact of polyphenol supplementation and exercise on depression and brain function parameters
Behavioral and Brain Functions volume 21, Article number: 10 (2025)
Abstract
The objective of this review study is to examine the combined antidepressant effects of exercise and polyphenol supplementation, with a focus on specific polyphenolic compounds such as crocin, curcumin, and quercetin, as well as different forms of physical exercise, including aerobic and resistance training. The research examines how these interventions influence depressive-like behaviors, cognitive function, and neurochemical markers in animal models and human participants. The findings demonstrate that both exercise and polyphenols independently contribute to mood enhancement, reduced anxiety, and improved cognitive function through mechanisms such as neurogenesis, neurotransmitter modulation, and anti-inflammatory effects. Notably, the combined interventions showed a synergistic effect, providing more significant benefits in reducing symptoms of depression and anxiety, enhancing cognitive performance, and supporting overall mental well-being. These results suggest that integrating exercise and polyphenol supplementation could be a promising non-pharmacological approach to managing depression and related disorders.
Introduction
Depression is among the most prevalent mental disorders, significantly disrupting emotional control, thought processes, and daily functioning. It is a leading contributor to disability, affecting one in eight individuals globally. Depression manifests through persistent sadness, lack of motivation, and cognitive impairments, requiring effective interventions [1]. In 2020, the COVID-19 pandemic led to a substantial rise in the prevalence of anxiety and depressive disorders. Preliminary data indicate a 26% increase in anxiety disorders and a 28% increase in major depressive disorders within a single year [2]. Although there are effective methods for preventing and treating mental disorders, the majority of affected individuals lack access to appropriate care. Additionally, many people with mental disorders face stigma, discrimination, and human rights violations [3]. Depression is a widespread mental health condition marked by enduring sadness, diminished interest in activities once found enjoyable, and a variety of cognitive and physical symptoms that greatly hinder everyday functioning. As reported by the World Health Organization (WHO), depression impacts more than 280 million individuals globally, establishing it as a major contributor to disability worldwide [4]. The origins of depression are multifaceted, involving a complex interaction of genetic, biological, environmental, and psychological factors. Biological aspects particularly include imbalances in neurotransmitters, inflammation, and oxidative stress, which play significant roles in the development and progression of depression [5]. Brain health includes cognitive abilities, emotional stability, and general mental well-being. It is shaped by a variety of factors, such as lifestyle choices, diet, exercise, and genetic predispositions. Depression significantly influences mood and emotional well-being and negatively impacts cognitive functions, including memory, attention, and executive functioning. Long-term depression is linked to structural and functional alterations in the brain, such as a decrease in hippocampal volume, disrupted neural network connectivity, and reduced neurogenesis [6]. Conventional approaches to treating depression usually involve using antidepressant medications and different types of psychotherapy. Although these therapies can be beneficial, they frequently have drawbacks like side effects, inconsistent effectiveness across individuals, and challenges related to accessibility. As a result, there is an increasing focus on exploring alternative and complementary methods for treating depression, especially those centered around lifestyle adjustments. Notably, dietary changes and consistent physical exercise have become prominent areas of study for their ability to enhance mental health without significant adverse effects [7].
Polyphenols are recognized for their antioxidant properties, which play a crucial role in reducing oxidative stress, a significant factor in the onset of depression and neurodegenerative conditions [8]. These compounds exert neuroprotective effects by influencing signaling pathways related to inflammation, oxidative stress, and the generation of new nerve cells in the brain [9]. Polyphenols have emerged as potential natural interventions for depression, influencing brain function through multiple pathways. Flavonoids, for instance, penetrate the blood-brain barrier and exert neuroprotective effects by enhancing synaptic plasticity, decreasing neuroinflammation, and stimulating neurogenesis [10]. Several studies have underscored the potential of polyphenols to enhance mood and cognitive function. Research conducted by Vauzour, Vafeiadou [11] showed that diets rich in flavonoids can improve memory and learning abilities by influencing the signaling pathways crucial for synaptic plasticity [11]. In a study conducted by Lin et al., it was demonstrated that supplementation with polyphenols notably decreased symptoms associated with anxiety and depression among clinical populations [12]. These results imply that incorporating polyphenols into treatment strategies could offer substantial benefits in managing depression and promoting overall brain health.
Regular physical exercise is widely recognized for its positive impact on mental health, enhancing mood, reducing stress, and improving cognitive function. Exercise triggers the release of neurotrophic factors, endorphins, and various neurotransmitters that play crucial roles in maintaining brain health and regulating emotions [13]. Engaging in regular physical activity stands out as a highly effective lifestyle intervention for enhancing mental health. Exercise not only lowers the likelihood of depression but also alleviates its symptoms while boosting cognitive function [14]. These benefits arise from complex interactions involving physiological, psychological, and neurobiological mechanisms. From a physiological perspective, physical exercise boosts cerebral blood flow, thereby improving the supply of oxygen and essential nutrients crucial for optimal brain performance. Additionally, exercise triggers the release of neurotrophic factors like brain-derived neurotrophic factor (BDNF), which play pivotal roles in fostering neurogenesis and synaptic plasticity [15].
From a psychological perspective, engaging in exercise fosters feelings of accomplishment, diminishes stress levels, and encourages social engagement, all of which collectively enhance mood and overall mental well-being [13]. At the neurobiological level, exercise influences neurotransmitter concentrations, particularly serotonin, dopamine, and norepinephrine, which play pivotal roles in mood regulation. These neurotransmitter changes contribute to secondary benefits, including reduced oxidative stress and neuroinflammation, further promoting brain health and emotional stability [16]. Regarding these advantages, incorporating consistent physical activity into the treatment of depression can markedly improve therapeutic results and promote overall brain well-being. While polyphenols and exercise individually provide substantial benefits for depression and brain health, recent studies indicate that their combined effects could be even more powerful. These interventions intersect in influencing oxidative stress, inflammation, and neurotransmitter function, suggesting synergistic potential in enhancing mental well-being and cognitive function. Given their benefits, integrating polyphenol supplementation with regular exercise presents a promising strategy for optimizing mental health outcomes. These interventions may enhance each other’s effects, improving mood and cognitive function by boosting neuroplasticity, decreasing inflammation, and regulating neurotransmitter systems. This review primarily aims to explore the combined effects of polyphenol supplementation and exercise on depression and brain health parameters. By examining current research, this review aims to clarify how these interventions help reduce depressive symptoms and improve cognitive and emotional well-being.
Depression and brain health
Understanding depression
Depression is a type of mood disorder, influenced by multiple factors, and marked by persistent feelings of sadness and a lack of interest or pleasure in activities [17]. The term “depression” can describe both a symptom and a medical condition. As a symptom, it encompasses a range of feelings from mild sadness to severe, pathological depression. However, as a syndrome, it requires a specific set of diagnostic criteria to be met [18]. Depression is identified by a range of symptoms including ongoing feelings of hopelessness, cognitive decline, physical discomfort, decreased motivation, impaired thinking, disrupted sleep patterns, and reduced verbal and physical activity. Patients with mild depression often exhibit physical dysfunctions, such as weight loss, sleep disturbances, fatigue, and weakness. In contrast, those with severe depression may experience more extreme symptoms, including suicidal behavior [19]. Given the significant burden of these symptoms, depression is recognized as one of the most common and debilitating mental health conditions worldwide. It affects individuals of all ages and backgrounds, significantly impairing daily functioning and quality of life. The WHO reports that depression impacts around 5% of the global adult population, ranking it among the most prevalent mental health disorders worldwide. According to the same report, depression is more common in women, affecting about 6% of women compared to 4% of men at any given time. Moreover, depression affects approximately 5.7% of adults aged 60 and above, underscoring its widespread occurrence across various age demographics [4]. Depressive disorders vary in presentation and severity. The most common form, Major Depressive Disorder (MDD), affects approximately 51.3% of individuals diagnosed with depression. Other subtypes include melancholic depression, psychotic depression (38% prevalence), and bipolar disorder, which alternates between depressive and manic episodes. Additionally, antenatal and postnatal depression, cyclothymic disorder, and seasonal affective disorder are recognized variations with distinct clinical characteristics [20]. During a bout of depression, a person experiences a prolonged state of low mood marked by feelings of sadness, irritability, or emptiness. This period can also include a significant reduction in interest or enjoyment from activities that previously brought happiness or satisfaction [21]. This condition persists throughout most of the day, almost every day, for a minimum of two weeks, clearly setting it apart from normal fluctuations in mood [22]. In addition to feeling depressed, individuals often display a variety of other symptoms. These can encompass challenges with concentration, heightened feelings of guilt or diminished self-esteem, a pervasive feeling of hopelessness regarding the future, recurring thoughts about death or suicide, disruptions in sleep patterns, alterations in appetite resulting in either weight loss or gain, and a persistent sense of fatigue or lack of energy [23, 24]. Depression significantly impacts various aspects of life, including social interactions, productivity at work or school, and participation in community activities [25]. The severity of a depressive episode is classified as mild, moderate, or severe depending on the number and intensity of symptoms and how they disrupt daily activities [26]. Depressive episodes can present in different patterns. For instance, individuals with a single episode depressive disorder experience their initial and singular episode of depression [27]. On the other hand, those with recurrent depressive disorder have a history of encountering at least two separate episodes of depression, typically alternating with periods of normal mood [28]. Bipolar disorder is characterized by alternating episodes of depression and manic symptoms. During manic phases, individuals typically experience elevated mood, heightened activity levels, rapid speech, inflated self-esteem, reduced need for sleep, distractibility, and impulsivity [29].
Diagnosis and treatment
Diagnosing depression involves a thorough evaluation by a healthcare professional, which typically includes a clinical interview, standardized screening tools, and an assessment of the patient’s medical and psychological history. The primary method for diagnosing MDD follows the American Psychiatric Association’s DSM-5 criteria, which require at least five specific symptoms (e.g., depressed mood, loss of interest) to be present over a two-week period. While the diagnosis is primarily clinical, physical exams and laboratory tests may be used to exclude underlying medical conditions that mimic depressive symptoms, such as thyroid dysfunction or vitamin deficiencies [30]. Common tools used to assess depression include the Patient Health Questionnaire-9 (PHQ-9) and the Beck Depression Inventory (BDI). These tools help quantify the severity of depression symptoms [31, 32]. In addition, clinicians may use structured interviews like the SCID-5 (Structured Clinical Interview for DSM-5) for a more detailed evaluation [33]. Getting an accurate diagnosis is crucial to differentiate depression from other mental health conditions and to ensure that the patient receives the most effective treatment. Effective treatment options for depression involve both psychological therapies and medications. Psychological treatments are generally considered the primary approach for managing depression. In cases of moderate to severe depression, these psychological therapies can be supplemented with antidepressant medications [34]. However, for mild depression, medication is usually not necessary. Psychological therapies focus on helping individuals develop new perspectives, coping mechanisms, and interpersonal skills. These approaches typically involve talk therapy sessions with trained professionals or supervised lay therapists, and can take place either face-to-face or through online platforms [35, 36].
Brain health parameters
Cognitive function
Cognitive function encompasses the mental abilities that allow individuals to perform tasks, from the simplest to the most complex. These abilities include memory, attention, language, problem-solving, and decision-making. Maintaining cognitive function is essential for everyday activities and significantly impacts the overall quality of life. As individuals age, a decline in cognitive abilities is often observed, but this process can be influenced by factors such as genetics, lifestyle, and environmental conditions. Studies show that engaging in physical exercise, following a healthy diet, and staying mentally active through activities like reading or solving puzzles can help sustain or even enhance cognitive function [37, 38]. Moreover, progress in neuroimaging technologies such as magnetic resonance imaging (MRI) and positron emission tomography (PET) has offered a more comprehensive understanding of the structural and functional alterations in the brain associated with cognitive decline. These alterations include hippocampal atrophy, reduced gray matter volume in the prefrontal cortex, impaired synaptic density, and disruptions in neural network connectivity. Functionally, cognitive decline is linked to decreased cerebral blood flow, altered neurotransmitter activity, and impaired neurogenesis, which collectively contribute to deficits in memory, attention, and executive functioning [39].
Neuroplasticity
Neuroplasticity refers to the extraordinary capacity of the brain to reshape itself by creating new neural pathways throughout an individual’s lifetime. This flexibility is essential for acquiring new skills, consolidating memories, and recovering from brain injuries [40]. Neuroplasticity plays a crucial role in depression recovery, as it allows the brain to adapt to stress and form new neuronal connections. Deficits in neuroplasticity are linked to cognitive decline and mood disorders, making it a critical target for therapeutic interventions. Several factors can enhance neuroplasticity, including enriched environments, physical exercise, and pharmacological treatments such as selective serotonin reuptake inhibitors (SSRIs), N-methyl-D-aspartate (NMDA) receptor modulators like ketamine, and neurotrophic agents such as BDNF enhancers. These pharmacological interventions promote synaptic remodeling, enhance neuronal survival, and improve cognitive function in individuals with neuropsychiatric disorders. A critical molecule in this process is BDNF, which supports the survival and growth of neurons [41]. Research indicates that aerobic exercise can increase BDNF levels, which in turn enhances cognitive function and emotional health. This demonstrates the brain’s remarkable ability to adapt and recover, even when confronted with damage or disease [42].
Neuroinflammation
Neuroinflammation refers to the activation of the brain’s immune response, which can be initiated by infections, toxic substances, or brain injuries. While acute neuroinflammation acts as a natural and protective mechanism, prolonged neuroinflammation is linked to numerous neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis [43]. Pro-inflammatory cytokines such as interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) play a key role in the inflammatory processes and are associated with the advancement of neurodegenerative diseases. To address neuroinflammation, various strategies are employed, including the use of anti-inflammatory drugs and lifestyle modifications like improved diet and consistent physical activity. These approaches have demonstrated potential in lowering inflammation levels and enhancing overall brain health [44].
Neurotransmitter regulation
Neurotransmitters are essential chemicals that facilitate communication between neurons and play a crucial role in regulating mood and cognitive function. Imbalances in neurotransmitter systems—particularly in serotonin, dopamine, and norepinephrine—are strongly implicated in depression. Low serotonin levels are associated with mood dysregulation, while deficits in dopamine contribute to anhedonia and lack of motivation. Additionally, disruptions in the balance between excitatory neurotransmitters like glutamate and inhibitory neurotransmitters like gamma-aminobutyric acid (GABA) can further exacerbate depressive symptoms. Given these complex interactions, targeting neurotransmitter regulation is a primary focus of depression treatment strategies [45]. Serotonin and dopamine play critical roles in regulating mood, and many psychiatric treatments are designed to correct imbalances in these neurotransmitters [46]. Furthermore, maintaining a proper equilibrium between excitatory neurotransmitters, such as glutamate, and inhibitory neurotransmitters, like GABA, is crucial for ensuring stable neural function. An imbalance in neurotransmitter systems can contribute to various disorders, including anxiety and seizures. Gaining a thorough understanding of these systems is crucial for creating effective treatments and enhancing the overall outcomes for patients [47, 48].
Exercise and mental well-being
Types of exercise
Exercise is fundamental for sustaining and enhancing overall health. Different types of exercise provide unique physiological and psychological benefits, making them important for mental well-being. Aerobic activities, resistance training, and high-intensity interval training (HIIT) each contribute to health improvements in distinct ways, particularly in the context of depression and cognitive function.
Aerobic exercise
Aerobic exercise, often referred to as cardiovascular or endurance training, encompasses activities designed to elevate the heart rate and breathing rate by involving large muscle groups. Examples of aerobic exercise include walking, running, cycling, and swimming. The health advantages of aerobic exercise are widely recognized. Engaging consistently in aerobic activities boosts cardiovascular health, increases lung function, and supports effective weight management [49]. Furthermore, engaging in aerobic exercise has been associated with a lower risk of developing chronic conditions, including type 2 diabetes, high blood pressure, and some types of cancer [50]. The American College of Sports Medicine advises that adults should aim for a minimum of 150 min of moderate-intensity aerobic activity or 75 min of vigorous-intensity exercise each week to gain significant health benefits [51].
Resistance training
Resistance training, also known as strength training, focuses on enhancing muscle strength, endurance, and size through exercises that counteract a force. This type of training can be performed using free weights, resistance bands, various weight machines, or body weight exercises such as calisthenics, pull-ups, push-ups, and squats, which require no external resistance but rely on the individual’s own body weight to build strength [52]. Resistance training is notably effective in combating muscle loss associated with aging and enhancing the ability to perform everyday tasks [53]. The American College of Sports Medicine advises incorporating resistance exercises targeting all major muscle groups at least twice a week into a well-rounded fitness regimen [51].
High-intensity interval training (HIIT)
HIIT involves alternating between brief, vigorous exercise and lower-intensity recovery or rest periods. This training method can encompass various activities like sprinting, cycling, body-weight exercises, and circuit training. HIIT has become increasingly popular because it effectively enhances cardiovascular fitness and reduces body fat more quickly than traditional aerobic exercise [54]. Research has demonstrated that HIIT can boost insulin sensitivity, elevate mitochondrial density, and enhance overall physical performance [55]. Importantly, HIIT also exerts significant mental health benefits, including reductions in depressive symptoms and anxiety levels. Studies suggest that short bursts of intense exercise stimulate endorphin release, enhance serotonin production, and improve cognitive flexibility, making HIIT a valuable intervention for mental well-being [56]. However, it may not be appropriate for everyone, especially individuals with specific medical conditions or those new to exercise. Thus, aerobic exercise, resistance training, and HIIT each provide distinct advantages and can collectively form a comprehensive fitness program. The selection of exercise types should be tailored to personal objectives, current fitness levels, and any existing health issues. Including a mix of these exercises can maximize overall health benefits and improve physical performance.
Effects on depression
Reduction of depressive symptoms
Extensive research has investigated the impact of exercise on depression, consistently demonstrating its effectiveness in alleviating depressive symptoms. Engaging in regular physical activity is known to elevate mood, boost energy levels, and enhance overall well-being, making it a beneficial strategy for individuals dealing with depression [57]. Various forms of exercise, including aerobic training, resistance training, and dance-based therapies, have been studied for their role in mental health improvements. Table 1 provides an overview of these exercise modalities and their specific effects on depressive symptoms and cognitive function, illustrating the most effective approaches for different populations. The studies included span various populations, including adolescents, older adults, and individuals with major depressive disorder or mild cognitive impairment. The data highlights that aerobic exercise, resistance training, and dance-based therapies show significant improvements in mood and cognition. Notably, high-intensity and structured exercise programs yield the most substantial benefits, suggesting that tailored exercise interventions could be an effective non-pharmacological strategy for managing depression and cognitive decline.
Exercise alleviates depressive symptoms through several neurochemical pathways. One key mechanism is the release of endorphins, which are natural pain relievers that induce feelings of euphoria, commonly known as the ‘runner’s high’ [58, 59]. Another crucial factor is the enhancement of neurotransmitter activity. Regular physical activity increases serotonin levels, which helps regulate mood, emotions, and sleep, thereby reducing depressive symptoms [57]. Additionally, exercise boosts dopamine production, improving reward processing and motivation—factors often impaired in individuals with depression [60]. These neurochemical changes collectively contribute to the antidepressant effects of physical activity. A meta-analysis by Morres et al. revealed that individuals who consistently participated in regular exercise had considerably lower levels of depressive symptoms compared to their sedentary counterparts. This meta-analysis reviewed research on aerobic exercise programs. The results indicate that while the specific type of exercise may not be the most important factor, maintaining regularity and intensity in physical activity is crucial for reducing depressive symptoms [61]. Exercise not only induces biochemical alterations but also fosters psychological benefits. Participating in physical activity often leads to a sense of achievement and boosts self-esteem, aspects that are frequently diminished in individuals suffering from depression [62]. Structured exercise routines provide a sense of regularity and purpose, which can be particularly advantageous for individuals dealing with depressive symptoms [63]. Another way exercise alleviates depression is by encouraging behavioral activation, a key psychological intervention for mood improvement. Individuals with depression often withdraw from social interactions, worsening feelings of isolation and sadness [64]. Engaging in structured physical activities, particularly in group settings such as fitness classes or sports teams, provides opportunities for social connection. These interactions not only enhance mood but also reinforce a sense of community and belonging, which are crucial for mental well-being [65]. Participating in group fitness classes, joining sports teams, or being part of walking clubs creates opportunities for social interaction. These activities can significantly enhance mood and contribute positively to overall mental well-being [66]. Moreover, engaging in physical activity can act as a helpful diversion from negative thinking and repetitive worries that often accompany depression. The concentration needed for exercise helps shift focus away from troubling thoughts, offering temporary relief from the mental strains of depression. With regular practice, this shift in focus can lead to a noticeable decrease in the severity of depressive symptoms [67]. Crucially, the positive effects of physical activity on depression extend beyond those already diagnosed with the condition. Studies indicate that regular exercise can also help prevent the development of depression in individuals who are at risk [13]. Research conducted by Harvey et al. revealed that increased physical activity is linked to a decreased likelihood of developing depression over time. This finding underscores the value of exercise as a practical and affordable approach for promoting mental health and preventing depressive disorders [68]. Hence, exercise offers a broad range of benefits for managing and preventing depression. Its positive impact on mood regulation, biochemical processes, psychological well-being, and social interaction highlights its effectiveness as a comprehensive approach to mental health. Consistent engagement in physical activity, no matter the form, can substantially alleviate symptoms of depression and enhance the overall quality of life for those experiencing this condition [69, 70].
Impact on anxiety and stress
Exercise not only helps with depressive symptoms but also significantly influences anxiety and stress. Engaging in physical activity can serve as a natural remedy for anxiety, alleviating both its physical and psychological symptoms. Exercise’s impact on anxiety can be attributed, in part, to its influence on the hypothalamic-pituitary-adrenal (HPA) axis, a key regulator of the body’s stress response [71]. This system controls the release of cortisol, a hormone crucial for managing stress. When stress becomes chronic, it can lead to persistent activation of the HPA axis, causing consistently high levels of cortisol. This prolonged exposure to elevated cortisol can contribute to increased anxiety and various other health problems [72]. Engaging in consistent physical activity has been demonstrated to lower cortisol levels, which helps stabilize the body’s response to stress. Research conducted by Hill et al. revealed that individuals who regularly participated in aerobic exercises exhibited notably reduced cortisol levels when faced with stressors, compared to those who led a sedentary lifestyle. This decrease in cortisol helps counteract the detrimental effects of prolonged stress and contributes to better mental well-being [73]. Additionally, physical activity promotes the release of neurotrophic factors like BDNF, which plays a crucial role in supporting neuronal growth and function. Elevated levels of BDNF have been linked to lower levels of anxiety and enhanced cognitive performance, underscoring the positive impact of exercise on brain health and function [74]. Another significant effect of exercise on anxiety and stress is its positive influence on sleep quality. Anxiety and stress often lead to poor sleep, and physical activity has been demonstrated to improve both the duration and quality of sleep [75]. Brand et al. conducted a study revealing that adolescents engaging in regular physical activity experienced better sleep quality and lower anxiety levels than their less active counterparts. Enhanced sleep quality contributes to improved emotional regulation and reduced anxiety, establishing a beneficial cycle that promotes overall mental well-being [76].
Behavioral mechanisms significantly contribute to the anxiety-reducing effects of physical activity. Engaging in exercise offers a healthy way to release accumulated energy and tension, often experienced by those with anxiety. The rhythmic movements involved in activities like running, cycling, or swimming can induce a meditative state, aiding in calming the mind and alleviating anxiety symptoms [63, 77]. Social engagement is another crucial element. Joining group exercises or team sports can cultivate a sense of community and provide social support, which is vital for handling anxiety and stress [78]. Being socially connected offers emotional backing, diminishes feelings of loneliness, and creates opportunities for positive social interaction. This structured environment can be especially advantageous for individuals dealing with social anxiety, as it facilitates safe and supportive social experiences [79, 80].
Mechanisms of action
Neurotransmitter modulation
Depression and anxiety are closely linked to dysfunctions in neurotransmitter systems, particularly the monoaminergic, neurotrophic, and neuropeptide pathways. Among these, serotonin, dopamine, and noradrenaline are central to mood regulation and are frequently dysregulated in depressive disorders [81]. Additionally, BDNF and neuropeptide Y (NPY) contribute to neurogenesis and neuronal health, playing crucial roles in emotional resilience [82]. Excitatory and inhibitory neurotransmitter systems, such as glutamate and GABA, also influence stress response and cognitive function. Moreover, neuropeptides like orexins regulate sleep-wake cycles and interact with monoaminergic pathways, further implicating their role in depression pathophysiology [83, 84]. Balancing the catecholaminergic, monoaminergic, glutamatergic, and orexinergic systems is crucial for maintaining mental health. Unfortunately, many current antidepressants take a long time to start working and are not always effective, highlighting the need for quicker, more efficient treatments with fewer side effects.
Research indicates that lifestyle choices, such as regular exercise, can positively influence these neurotransmitter systems, providing significant benefits for mental well-being [85]. Various forms of physical activity, including aerobic exercise, resistance training, and HIIT, influence neurotransmitter regulation and neurobiological pathways in distinct ways, leading to different mental health outcomes. These variations are linked to changes in key neurochemical systems, including serotonin, dopamine, endorphins, GABA, and BDNF, which contribute to mood enhancement, cognitive function, and emotional resilience (Table 2).
Serotonin plays a critical role in mood regulation, sleep patterns, and appetite control, and its dysregulation is strongly linked to depression and anxiety [86]. In individuals with depression, serotonin levels are frequently lower than normal, leading to mood disturbances and cognitive impairments. One well-established way to enhance serotonin function is through physical activity, which has been shown to boost serotonin production and release in the brain. This process leads to increased sensitivity of serotonin receptors and stimulation of hippocampal neurogenesis, contributing to long-term improvements in mood and cognitive abilities [87, 88].
The impact of exercise on serotonin regulation in the brain varies depending on exercise type, intensity, and duration. Aerobic training, such as running at 19 m/min for 8 weeks, has been shown to promote hippocampal neurogenesis and counteract serotonin receptor deficits associated with chronic stress-induced depression [89, 90]. Swimming-based exercise models further confirm this effect, with studies showing increased 5-HT2 receptor responsiveness and postsynaptic 5-HT1A receptor function in the brain after four weeks of training [91]. The duration and intensity of exercise influence serotonin metabolism in distinct brain regions: acute swimming (1 h) increases 5-HT and 5-HIAA levels specifically in the brainstem, while prolonged swimming (4 weeks) enhances serotonin turnover in the cerebral cortex, with effects persisting for up to a week post-exercise. However, chronic swimming results in a transient reduction of hippocampal and hypothalamic 5-HT levels, followed by a rebound effect after a week of rest [92].
Beyond direct serotonin modulation, exercise influences stress-related biochemical pathways. In models of chronic stress-induced depression, swimming has been shown to reduce serum corticosterone (CORT) levels and proinflammatory cytokines (IFN-γ, TNF-α), while simultaneously increasing serotonin levels in the prefrontal cortex, reinforcing its antidepressant effects [93]. Additionally, exercise contributes to better sleep quality by modulating striatal serotonin and norepinephrine levels in sleep-deprived mice, potentially alleviating symptoms such as fatigue, mood instability, and impaired cognitive function [94]. These findings highlight the multifaceted role of exercise in serotonin regulation, suggesting that its benefits extend beyond serotonin alone and involve broader neurobiological mechanisms.
Endorphins, a different set of neurotransmitters, are released during vigorous exercise, excitement, and in response to pain, functioning as natural analgesics and mood boosters [95]. These neuropeptides, which include β-endorphin, enkephalins, and dynorphins, play a crucial role in enhancing mood, lowering anxiety levels, and promoting a general sense of well-being [6]. Notably, β-endorphin has been found to exert substantial antidepressant effects by influencing dopamine neurons involved in motivation and pleasure [96]. The influence of exercise on depressive symptoms varies based on exercise modality and intensity. In elderly individuals, a six-week structured aerobic regimen was associated with significant reductions in Beck Depression Inventory (BDI) scores, particularly in somatic symptoms, suggesting that physical activity contributes to emotional well-being and mood stabilization [67]. Similarly, research suggests that resistance training may induce a more pronounced increase in β-endorphins compared to aerobic exercise, pointing to distinct neurochemical responses across exercise types [97]. Furthermore, HIIT has been demonstrated to trigger the acute release of plasma β-endorphins, supporting both stress reduction and mood enhancement. While peripheral β-endorphins are often linked to analgesia and mood regulation, their direct influence on central nervous system activity remains debated. This effect is particularly notable in HIIT due to the rapid energy demands and metabolic stress, leading to both immediate and long-term psychological benefits. Importantly, the interaction between endorphins and other neurotransmitter systems, such as dopamine and serotonin, suggests a broader, integrated mechanism by which exercise supports mental well-being. This highlights the synergistic effects of exercise-induced neurotransmitter modulation, reinforcing the role of physical activity as an effective non-pharmacological intervention for depression and anxiety [98].
Intense exercise has been shown to increase plasma β-endorphin and leu-enkephalin levels, which may contribute to mood stabilization and relaxation. While plasma β-endorphin levels correlate with stress resilience, central β-endorphin release in the brain remains a subject of ongoing investigation [95]. Unlike β-endorphins and enkephalins, dynorphins act on kappa opioid receptors (KORs) and are often released in response to stress. While dynorphins have been associated with anxiety and depressive symptoms in certain contexts, some studies indicate that they can enhance serotonin levels, providing potential antidepressant effects [99]. Exercise-induced dynorphin release is linked to increased BDNF expression, which supports neuroplasticity and may contribute to antidepressant effects [100]. In animal models, voluntary exercise has been shown to upregulate dynorphin mRNA, further suggesting a potential therapeutic role for physical activity in depression management [101].
Dopamine is a vital neurotransmitter that plays a key role in reward, mood regulation, and attention. Physical activity stimulates the mesolimbic dopamine pathway, increasing dopamine levels and receptor sensitivity, which are essential for both mood stabilization and cognitive function [102]. Importantly, dopamine does not act in isolation but interacts with serotonin, BDNF, and other neurochemical systems to regulate emotional resilience. Exercise enhances dopaminergic transmission by activating D1 and D2 receptors, leading to improved synaptic plasticity and neurogenesis [103]. These mechanisms collectively contribute to the antidepressant effects of exercise. Dopamine receptor activation facilitates cyclic AMP (cAMP) signaling and the activation of protein kinase A (PKA), which in turn enhances the expression of brain-derived neurotrophic factor (BDNF)—a key regulator of neurogenesis and cognitive enhancement. Additionally, dopamine modulates hippocampal neurogenesis by promoting the proliferation of neural progenitor cells in the dentate gyrus, a process essential for learning and memory [104]. The activation of dopamine pathways by exercise has also been shown to improve working memory and executive function by strengthening prefrontal cortex activity, which plays a crucial role in cognitive flexibility and emotional regulation [105].
Treadmill running (10 m/min, 5 days/week for 6 weeks) increases dopamine D2 receptor-like binding in the dorsomedial caudate putamen, ventrolateral caudate putamen, ventromedial caudate putamen, and olfactory tubercle, supporting its potential antidepressant effects [106]. Voluntary exercise enhances dopamine release, but blocking D2 receptors eliminates these benefits, indicating its key role in mood regulation [107]. Exercise also restores dopamine receptor function in addiction models, highlighting its therapeutic potential [108]. A combination of aerobic and resistance training increases dopamine levels more effectively than aerobic exercise alone. Studies show that mixed training improves mental health and dopamine production, reducing depression symptoms [109]. Exercise stimulates dopamine release via BDNF, enhancing neuroplasticity and cognitive function. Voluntary running boosts BDNF and dopamine levels in the striatum, with sustained effects even after a week of rest [110].
GABA is an inhibitory neurotransmitter essential for reducing neuronal excitability and promoting relaxation [111]. Physical activity enhances GABAergic transmission by increasing receptor expression and synthesis, with different exercise modalities influencing this system in distinct ways [112, 113]. For instance, aerobic exercise significantly increases GABA receptor subunit expression (α2, α5, γ2) in the prefrontal cortex and hippocampus, contributing to improved mood regulation [114]. Long-term voluntary running also elevated GAD67 enzyme expression in the forebrain, facilitating greater GABA production and modulating the GABAergic system to counteract depression [115]. Meanwhile, anaerobic and high-intensity circuit training induce acute GABAergic responses through lactate metabolism, which serves as a precursor for GABA synthesis in the sensorimotor cortex [116]. These differences underscore the importance of tailoring exercise interventions for optimal mental health benefits.
Yoga has been found to significantly boost plasma GABA levels, with some studies suggesting parallel increases in brain GABA concentrations by 27%. Moreover, a positive correlation exists between reduced anxiety levels and increased GABAergic activity in the thalamus, suggesting that yoga could serve as an effective intervention for anxiety and depression [117, 118]. These findings highlight the different types of exercise that influence GABA regulation in distinct ways and the importance of tailoring exercise interventions for optimizing mental health benefits and suggest that a combination of aerobic, anaerobic, and mind-body exercises may provide the most effective outcomes.
Glutamate, the primary excitatory neurotransmitter, plays a crucial role in synaptic plasticity and neurogenesis [119]. Engaging in physical activity transforms the brain’s pro-inflammatory state into an anti-inflammatory one, enhancing glutamate function and alleviating depressive symptoms. The exercise-induced boost in neurotransmitters and blood flow in the hippocampus and prefrontal cortex amplifies these positive effects [120, 121]. Moreover, physical activity influences glutamatergic neurotransmission, which is vital in mitigating the neurotoxic effects linked to elevated levels of quinolinic acid. By regulating the balance between excitatory and inhibitory neurotransmitters, exercise helps prevent neuronal damage and supports overall brain health [122]. Moreover, physical activity influences glutamatergic neurotransmission, which is vital in mitigating the neurotoxic effects linked to elevated levels of quinolinic acid. Quinolinic acid, a downstream metabolite of the kynurenine pathway, acts as an N-methyl-D-aspartate (NMDA) receptor agonist, leading to excessive excitotoxicity, oxidative stress, and neuronal damage, which have been implicated in neurodegenerative diseases and psychiatric disorders, including depression and anxiety [123]. Exercise regulates glutamatergic signaling, promoting synaptic balance and protecting against excitotoxic damage. Notably, the kynurenine pathway is a key mediator of exercise-induced glutamate modulation. Regular physical activity enhances the conversion of kynurenine into kynurenic acid, a neuroprotective metabolite that mitigates the toxic effects of quinolinic acid, reducing neuroinflammation and excitotoxicity [124].
Aerobic training has been shown to reduce excessive glutamate levels in neurodegenerative conditions such as Alzheimer’s disease by increasing receptor sensitivity rather than merely lowering glutamate concentration, contributing to improved mood and cognitive function [125]. High-intensity training (above 80% of maximum heart rate) elicits an immediate increase in brain glutamate and its precursor glutamine in the visual cortex and anterior cingulate cortex, regions involved in cognitive processing and mood regulation [126, 127]. This transient glutamate may contribute to the antidepressant effects of exercise, enhancing neuroplasticity and counteracting neuroinflammatory pathways that contribute to depression and schizophrenia [121]. While aerobic and high-intensity training primarily influence acute glutamate dynamics, resistance exercise may contribute to long-term glutamate balance by promoting BDNF-mediated neuroprotection. Strength training has been associated with improved glutamatergic signaling, reducing neuroinflammation and oxidative stress, key contributors to depression and cognitive decline [128]. Different exercise modalities influence glutamate regulation through distinct mechanisms. Understanding these differential effects could help optimize exercise-based interventions for neuropsychiatric disorders, including depression and neurodegeneration.
Irisin, a hormone produced by muscles during physical activity, creates a direct link between the muscles and the brain. It significantly boosts levels of BDNF and IGF-1, which encourages neurogenesis and provides antidepressant benefits [129]. Irisin also plays a role in reducing oxidative stress and inflammation, thereby improving overall brain health. By regulating energy metabolism and offering neuroprotection, this hormone becomes essential in the fight against depression and anxiety [130]. Swimming for four weeks has been found to alleviate hypothyroidism-related depression by stimulating irisin-mediated pathways that enhance serotonin activity and reduce oxidative stress. The long-term benefits of aerobic training include improved metabolic health, increased resistance to neuroinflammation, and enhanced mitochondrial function, all contributing to reduced depressive symptoms [131, 132].
Orexins, produced in the hypothalamus, play a key role in regulating wakefulness, motivation, and mood. Physical activity boosts orexin levels, which in turn enhance neurogenesis and hippocampal function. This improvement leads to better cognition and mood, helping to alleviate depressive symptoms [133, 134]. Additionally, orexin’s influence on increasing physical activity and energy expenditure further reinforces its antidepressant effects [135]. Aerobic exercise significantly elevates plasma orexin-A levels, enhancing hippocampal neurogenesis and cognitive function. Human studies show that moderate-intensity aerobic exercise (e.g., 30 min of cycling or walking at 4.5 km/h, three times per week) leads to a notable increase in orexin secretion, contributing to improved mood and reduced depressive symptoms. Additionally, engaging in aerobic activity with a heart rate of 120–140 bpm for 45 min stimulates orexin release, reinforcing its role in energy regulation and emotional resilience. While orexin generally supports mood regulation and cognitive enhancement, its effects vary across brain regions. In the amygdala, an upregulation of orexin expression has been associated with depression, while exercise-induced suppression of orexin in this region exerts antidepressant effects [136, 137].
Neuropeptide Y (NPY) plays a crucial role in regulating mood, managing stress responses, and maintaining energy balance. Physical activity elevates NPY levels, which possess antidepressant qualities and promotes the proliferation of hippocampal cells. NPY influences stress and depressive symptoms by affecting neuroplasticity and maintaining energy balance. Its ability to regulate mood and lessen negative emotional experiences underscores its significance in mental health [138, 139].
Aerobic endurance exercise has been shown to significantly increase NPY expression in both animal and human models. In a rat model, a 6-week swimming exercise protocol (60 min/day) increased hippocampal NPY levels and mitigated depressive behaviors. However, these effects were attenuated when exercise was combined with nandrolone decanoate administration, suggesting an interaction between anabolic steroid exposure and exercise-induced neuropeptide regulation [139]. Similarly, in humans, endurance activities like a 25 km swimming competition significantly increased plasma NPY levels, which correlated with metabolic adjustments related to energy homeostasis and leptin regulation [140]. A 45-minute aerobic exercise session (cycling at 2 W/kg lean body mass) significantly elevated plasma NPY levels in individuals with bulimia nervosa, particularly when combined with the anti-lipolytic drug acipimox. This response, which was more pronounced in BN patients, highlights the role of NPY in appetite regulation and metabolic adaptation to energy expenditure. However, the modulation of plasma NPY by different exercise modalities, including resistance training and prolonged high-intensity exercise, requires further investigation [141].
NPY’s antidepressant potential has been demonstrated in forced swim test (FST) studies. In rats, intracerebroventricular (i.c.v.) injections of NPY over 24 h increased swimming and reduced immobility in a dose-dependent manner, mimicking serotonergic antidepressants. This suggests that NPY modulates stress-related behaviors and may contribute to exercise-induced antidepressant effects by enhancing neuroplasticity and stress resilience. These findings highlight the need to explore how different exercise types, durations, and intensities regulate NPY signaling to optimize mental health benefits [142]. Central administration of NPY in the brain via intracerebroventricular (i.c.v.) injections has been shown to exert antidepressant-like effects in forced swim test (FST) studies. In rats, i.c.v. injections of NPY into the lateral ventricle at 0, 8, and 23 h post-preswim increased swimming and reduced immobility in a dose-dependent manner, mimicking the effects of serotonergic antidepressants [143].
Tryptophan, a precursor to serotonin, is essential for regulating mood and emotional stability. Physical activity increases tryptophan availability, enhancing serotonin synthesis and neurotransmission [144, 145]. Exercise helps regulate tryptophan metabolism, thereby alleviating depressive symptoms and supporting mental health. These benefits are observed in both central and peripheral tryptophan levels, underscoring the antidepressant effects of regular physical activity [87]. Aerobic exercise at moderate intensity (cycling at 60 rpm) decreases plasma tryptophan by 12%, increases kynurenine by 6%, and correlates with neopterin, an immune activation marker. This suggests exercise-mediated depression relief through immune pathways [146]. In elderly individuals, 60 min of treadmill running increased the plasma tryptophan-to-BCAA ratio by 78%, which is associated with enhanced brain tryptophan availability and potentially improved mood [147]. In rats, chronic treadmill running (30 min/day, 28 days) increased serotonin receptor (5-HT1A) and tryptophan hydroxylase (TPH) levels, reducing forced swim test immobility, confirming its antidepressant effects [148, 149].
Postpartum depression, linked to serotonergic dysfunction, improves with treadmill running (30 min/day, 2 weeks), leading to increased serotonin and TPH expression and enhanced exploratory behavior in postpartum rats [150]. Additionally, exercise reduces maternal separation-induced depression by promoting hippocampal neurogenesis [151]. While aerobic exercise primarily enhances serotonin metabolism, anaerobic and HIIT exercise may modulate mood through noradrenergic and neurogenesis-related pathways. In rats, moderate to vigorous-intensity exercise (3 days/week) increased hippocampal neurogenesis and noradrenaline while preventing serotonin and tryptophan depletion-related memory impairments. While serotonin plays a critical role in mood regulation, emerging evidence suggests that exercise exerts antidepressant effects through multiple interconnected mechanisms, including neurotransmitter modulation, neurogenesis, and inflammatory response reduction [152].
Cholinergic receptors, which include nicotinic and muscarinic receptors, play key roles in neurotransmission and mood regulation [153]. Physical activity enhances acetylcholine levels and receptor activity, leading to improved cognitive function and better mood. Nicotinic receptors, specifically, are considered potential therapeutic targets for treating depression. The modulation of cholinergic transmission through exercise mimics the antidepressant effects of nicotine, thereby alleviating depressive symptoms [154, 155].
Thus, exercise provides a multi-faceted antidepressant effect by modulating serotonin, dopamine, endorphins, GABA, glutamate, and neuropeptides such as irisin, orexins, and NPY. These neurochemical adaptations enhance neurogenesis, synaptic plasticity, and emotional resilience, reinforcing the role of exercise as a non-pharmacological intervention for depression and anxiety. Future research should further explore the differential effects of exercise modalities on neurotransmitter regulation to optimize clinical applications in mental health treatment.
Neurogenesis
Regular physical activity has been demonstrated to boost adult neurogenesis, especially in the hippocampus, a region vital for learning and memory [156]. Studies in both humans and animals indicate that consistent exercise is associated with enhanced cognitive abilities and a decrease in depressive symptoms [128, 157,158,159]. Research shows that individuals who participate in moderate exercise demonstrate superior memory retention and learning abilities compared to those who lead a sedentary lifestyle [160, 161]. This positive impact is also observed in animals, where physical activity promotes the growth and longevity of new neurons in the hippocampus, a key region involved in memory and mood regulation [162, 163]. The antidepressant effects of exercise are similar to those obtained from pharmacological treatments, with the benefits continuing for months even after stopping the exercise regimen. This indicates that exercise has a long-lasting positive impact on brain health [164, 165]. These benefits are primarily driven by an increase in neurotrophic factors like BDNF, IGF-1, and VEGF, which are crucial for neurogenesis and brain plasticity [166]. Exercise stimulates the proliferation of neural progenitor cells, especially type 2 progenitor cells, which eventually mature and become integrated into the hippocampal network. This process is supported by both the proliferation and survival phases of neurogenesis, with exercise having a stronger effect on the survival of new neurons [167]. The benefits of exercise-induced neurogenesis are not confined to young animals; older animals also experience increased neuron production, suggesting that physical activity can help counteract age-related declines in neurogenesis [168]. Beyond encouraging cell proliferation, exercise also triggers structural changes in the hippocampus. These changes include modifications in dendritic structure and an increase in synaptic density, especially in the dentate gyrus and CA1 regions. These alterations are essential for synaptic plasticity, a key factor in learning and memory [169, 170]. IGF-1 plays a significant role in this process, not only by promoting cell proliferation but also by increasing dendritic spine density and enhancing overall neuronal connectivity [171]. Inhibiting IGF-1 signaling can counteract these benefits, emphasizing its critical importance in the neurogenic and neuroplastic effects of exercise [172]. The connection between stress and hippocampal plasticity highlights the advantages of exercise. Chronic stress is known to decrease neurogenesis and cause dendritic atrophy, especially in the CA3 region of the hippocampus. This structural degradation is linked to impaired cognitive function and a heightened risk of depression [173]. Conversely, exercise helps counteract these stress-related alterations by fostering resilience and improving hippocampal function [174]. This indicates that physical activity not only boosts neurogenesis but also shields the brain from structural and functional harm caused by chronic stress [174].
Overall, the enhancement of neurogenesis and structural plasticity in the hippocampus due to exercise is driven by a complex interaction of growth factors and neurotrophic signals. The increase in BDNF and IGF-1 levels in response to physical activity is especially important, as these factors help new neurons survive and integrate into existing networks, boost synaptic plasticity, and lead to better cognitive function and mood. These findings highlight the promise of physical activity as a non-drug approach to enhancing mental health, especially in addressing depression and cognitive decline associated with aging.
Anti-inflammatory effects
Engaging in physical exercise has gained recognition as an effective approach to managing depression and anxiety, largely due to its impact on neuro-inflammation. The beneficial mechanisms are diverse and intricate, primarily centered around the reduction of pro-inflammatory activities within the body [175]. Research has shown that engaging in physical activity can reduce the levels of circulating (plasma) pro-inflammatory cytokines, including IL-1β, TNF-α, IL-6, IL-8, and IL-10. These cytokines are commonly elevated in individuals with chronic illnesses and depression [176, 177]. Engaging in moderate-intensity physical activity has been found to lower blood concentrations of pro-inflammatory cytokines, which is associated with reductions in stress and depressive symptoms. This decrease in inflammatory markers, particularly IL-1β, has been associated with reductions in stress and depressive symptoms. Notably, IL-1β is also implicated in sleep regulation, suggesting that exercise-induced reductions in this cytokine may contribute to improved sleep quality, which is often disrupted in depression [178].
Exercise influences the regulation of the HPA axis and glucocorticoid levels, which are frequently imbalanced in depression. Since prolonged glucocorticoid activation contributes to oxidative stress and mitochondrial dysfunction, exercise plays a crucial role in restoring homeostasis. By enhancing mitochondrial function in the hippocampus, exercise counteracts glucocorticoid-induced neurotoxicity and reduces systemic inflammation, thereby improving mental health outcomes [179,180,181,182].
Exercise has shown promising therapeutic benefits, especially for individuals with treatment-resistant depression (TRD), a condition often characterized by elevated pro-inflammatory cytokines. Accordingly, studies have found that increased circulating levels of TNF-α correlate positively with the antidepressant effects observed after a 12-week aerobic exercise regimen. Moreover, the reduction in IL-1β levels correlates with a decrease in depressive symptoms, indicating that exercise may help alleviate the severe chronic inflammation frequently associated with TRD. This reduction in inflammation is crucial, as it is often a contributing factor to the poor overall health observed in individuals with TRD [183].
The anti-inflammatory effects of exercise are partly attributed to a decrease in visceral fat, a known source of pro-inflammatory cytokines such as TNF-α and IL-6. A key factor in this inflammatory modulation is IL-6, which exhibits dual roles depending on its source. While IL-6 secreted from adipose tissue is linked to chronic low-grade inflammation, exercise-induced IL-6 functions as a myokine with predominantly anti-inflammatory effects [184]. This exercise-induced IL-6 promotes the secretion of IL-10, an anti-inflammatory cytokine, and suppresses TNF-α production, thereby contributing to the resolution of inflammation [185,186,187,188]. This dual role of IL-6 highlights the nuanced relationship between physical exercise and immune modulation, reinforcing the importance of regular physical activity in managing inflammation-related disorders such as depression.
The anti-inflammatory effects of exercise extend beyond cytokine modulation, involving pathways that regulate mitochondrial function and neuroinflammation. A central regulator of this process is peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), which promotes mitochondrial biogenesis and reduces inflammatory signaling. Furthermore, PGC-1α plays a key role in the kynurenine metabolic pathway, facilitating the conversion of neurotoxic kynurenine into kynurenic acid, a metabolite that protects against neuroinflammation. This conversion is crucial in reducing the harmful effects of quinolinic acid, a metabolite that worsens neuroinflammation and can lead to neuronal damage [189,190,191]. In depressive disorders, the regulation of these neurotransmitters is often disrupted, and physical activity helps to rebalance their levels, thereby reducing depressive symptoms. Collectively, these anti-inflammatory mechanisms—including cytokine regulation, mitochondrial support, and kynurenine metabolism—highlight the integral role of exercise in modulating neuroimmune interactions. Through these pathways, exercise exerts antidepressant and anxiolytic effects, reinforcing its importance as a non-pharmacological strategy for improving mental well-being (Fig. 1).
Polyphenols and mental well-being
Introduction to polyphenols
In recent years, consumer interest in polyphenol-rich foods has increased significantly due to their diverse health benefits. Polyphenols are a diverse class of secondary metabolites characterized by multiple hydroxyl groups attached to aromatic rings, contributing to their strong antioxidant, anti-inflammatory, and neuroprotective properties [186, 192,193,194]. Historically referred to as “vegetable tannins” due to their role in leather tanning, polyphenols are now classified based on their chemical structure and biological function [195]. The primary categories include flavonoids, phenolic acids, lignans, tannins, and stilbenes, each exhibiting distinct physiological effects [196].
Flavonoids, the most abundant subgroup, include flavonols (e.g., quercetin), flavones (e.g., apigenin), and anthocyanins (e.g., cyanidin), known for their antioxidant and anti-inflammatory properties. Phenolic acids, derived from benzoic and cinnamic acids, are prevalent in plant-based foods and contribute to oxidative stress modulation. Other key subgroups, such as lignans (found in flaxseeds) and stilbenes (e.g., resveratrol in red wine), have been linked to neuroprotection and cardiovascular benefits [197,198,199].
Tannins, a distinct polyphenol subclass, are divided into hydrolyzable and condensed tannins. Hydrolyzable tannins, found in foods like pomegranates and tea, degrade into bioactive phenolic acids (gallic and ellagic acid), which exhibit antimicrobial, antioxidant, and cardioprotective effects [200]. Condensed tannins, or proanthocyanidins, are polymers of flavan-3-ols (e.g., catechin, epicatechin) that resist enzymatic hydrolysis, instead undergoing microbial metabolism to produce bioactive compounds that support metabolic and cardiovascular health [201].
The functional differences between these tannin types influence their physiological effects—hydrolyzable tannins provide rapid benefits through hydrolysis-derived phenolic acids, while condensed tannins exert sustained effects via gut microbiota interactions. Both classes contribute to food preservation, nutraceuticals, and therapeutic applications, underscoring their relevance in modern health sciences [195, 202].
Hence, polyphenols constitute a highly complex and diverse category of compounds known for their substantial health advantages, largely attributed to their antioxidant capabilities. Beyond their general health benefits, emerging research highlights their neuroprotective and antidepressant properties, which stem from their ability to modulate oxidative stress, neuroinflammation, and key neurotransmitter pathways. These mechanisms position polyphenols as promising candidates for supporting mental health and mitigating depressive symptoms.
Antidepressant properties
Polyphenols, a diverse set of naturally occurring compounds, have attracted considerable attention for their potential antidepressant effects. This interest largely arises from their capability to influence various biochemical pathways that are crucial for regulating mood (Fig. 2) [203]. Table 3 provides an overview of the effects of different polyphenols, including crocin, curcumin, and quercetin, on depressive symptoms and cognitive functions. These polyphenols exert neuroprotective effects by modulating neuroinflammation, neurotransmitter function, and oxidative stress pathways.
One of the most extensively researched mechanisms is the inhibition of serotonin reuptake. Serotonin, a critical neurotransmitter that plays a major role in regulating mood, is frequently observed at lower levels in people suffering from depression. By preventing the reuptake of serotonin, polyphenols boost its presence in the synaptic cleft, thus enhancing serotonergic communication and potentially lifting mood [204, 205]. Polyphenols inhibit serotonin reuptake through multiple mechanisms, primarily by interacting with the serotonin transporter (SERT), reducing its efficiency, and increasing extracellular serotonin levels in the brain [206, 207]. Additionally, they suppress monoamine oxidase-A (MAO-A), the enzyme responsible for serotonin degradation, thereby prolonging serotonin’s availability [208]. Some polyphenols, like curcumin and crocin, also enhance serotonin biosynthesis by upregulating TPH, the key enzyme in serotonin production [209, 210]. Beyond direct effects on serotonin metabolism, polyphenols regulate SERT expression through their anti-inflammatory and antioxidant properties further supporting serotonin homeostasis by mitigating stress-induced increases in SERT activity [211, 212]. Beyond their direct influence on serotonin metabolism, polyphenols exert additional antidepressant effects by modulating neurotrophic factors. Notably, their ability to enhance BDNF supports serotonergic signaling, neuronal plasticity, and long-term synaptic adaptations that contribute to mood stabilization [213].
Through these combined actions—direct inhibition of SERT, suppression of MAO-A, increased serotonin synthesis, and modulation of inflammation and epigenetics—polyphenols effectively enhance serotonergic neurotransmission. This mechanism resembles the action of selective SSRIs, a widely used class of antidepressants [210, 214]. Additionally, polyphenols interact with other neurotransmitter systems, including dopamine and noradrenaline, which play crucial roles in motivation, pleasure, and cognitive function [215, 216]. Animal studies have demonstrated that compounds such as curcumin (from turmeric), resveratrol (from grapes), and baicalin (from Scutellaria baicalensis) significantly reduce depressive-like behaviors in models using the Forced Swim Test (FST) and Tail Suspension Test (TST) [217,218,219,220,221,222]. Polyphenols typically decrease immobility and enhance sucrose preference, indicating their potential to counteract both despair and anhedonia [210, 223].
Another essential pathway affected by polyphenols is the hypothalamic-pituitary-adrenal (HPA) axis, which regulates the body’s response to stress. Chronic stress can lead to excessive cortisol production, negatively impacting brain regions such as the hippocampus, which is critical for mood regulation. Polyphenols have been suggested to modulate the HPA axis by reducing cortisol levels, thereby alleviating stress-induced depressive symptoms [224,225,226].
Additionally, polyphenols are known for their potent antioxidant capabilities, which play a crucial role in reducing oxidative stress-a state where there is an overabundance of free radicals in the body. This condition is associated with the development of depression, as it can lead to brain cell damage and promote neuroinflammation. By neutralizing free radicals and decreasing oxidative stress, polyphenols help safeguard neural tissues from harm. This protective effect can help maintain cognitive function and potentially ease symptoms of depression [227,228,229,230]. In addition to reducing oxidative stress, polyphenols possess anti-inflammatory properties. Chronic inflammation has been increasingly linked to depression, as pro-inflammatory cytokines such as IL-6 and TNF-α can interfere with neurotransmitter synthesis and disrupt neural pathways. Polyphenols mitigate these effects by lowering the levels of pro-inflammatory cytokines and promoting anti-inflammatory signaling, thereby restoring normal brain function [8, 231]. Polyphenols also influence the gut-brain axis by modulating the composition of gut microbiota. They promote the growth of beneficial bacteria while suppressing harmful ones, leading to the production of neuroactive compounds that positively affect mood and cognition. Some polyphenols also enhance the synthesis of short-chain fatty acids, which possess anti-inflammatory and neuroprotective properties [232, 233].
Together, these mechanisms highlight the promising role of polyphenols in mental health. By influencing neurotransmitter systems, reducing oxidative stress and inflammation, regulating the HPA axis, and promoting neurogenesis, polyphenols offer a multifaceted approach to depression management. While much of the existing evidence is derived from preclinical studies, these findings underscore the need for further human clinical trials to validate their efficacy and long-term safety.
Combined effects of exercise and polyphenols on depression and mental health
The integration of regular physical exercise and polyphenol supplementation presents a promising strategy for enhancing mental well-being. Both interventions independently exert antidepressant effects through distinct but complementary mechanisms, including neurotransmitter modulation, neurogenesis stimulation, oxidative stress reduction, and inflammation control [234, 235]. Emerging research suggests that their combination may have synergistic benefits in mitigating depression, anxiety, and cognitive decline [236, 237].
Synergistic effects on neurotransmitter regulation
Several studies indicate that combining polyphenols with exercise influences key neurotransmitter systems, including serotonin, dopamine, and noradrenaline. Ghalandari-Shamami, Nourizade [234] investigated the combined antidepressant effects of crocin (a polyphenol in saffron) and voluntary exercise in adolescent rats subjected to chronic stress. Their findings revealed that the combination of crocin and exercise significantly improved behavioral outcomes and structural changes in the prefrontal cortex. These improvements were linked to increased serotonin and dopamine levels and reduced corticosterone concentrations, which are commonly elevated in stress-related disorders [234].
The ability of polyphenols and exercise to influence neurotransmitter balance extends beyond depression models. For instance, Azarian, Farsi [236] investigated endurance training and crocin supplementation in a rodent model of Alzheimer’s disease, revealing improvements in anxiety-like behaviors and aerobic capacity. These findings suggest that the neuromodulatory effects of polyphenols and exercise may also be relevant for neurodegenerative conditions, broadening their therapeutic potential [236].
Effects on neuroinflammation and oxidative stress
Chronic inflammation and oxidative stress contribute significantly to the pathophysiology of depression. Research indicates that polyphenols and exercise independently reduce circulating (plasma) pro-inflammatory cytokines such as IL-1β and TNF-α while also modulating brain-derived neuroinflammatory markers (e.g., microglial activation and astrocytic response in hippocampal tissue), contributing to improved cognitive and mood-related outcomes [238, 239]. In an experimental model of methamphetamine-induced neurotoxicity, Nourolapour, Abbassi Daloii [239] demonstrated that aerobic exercise and crocin supplementation significantly upregulated hippocampal neuroprotective gene expression, including BDNF and TrkB, which are critical for neuronal survival and plasticity. Notably, the greatest enhancement was observed in the group that received both interventions, supporting the hypothesis that polyphenols amplify the neuroprotective effects of exercise [239].
Amoei, Meshkati [238] examined the combined impact of aerobic exercise and curcumin supplementation on anxiety behaviors and locomotor activity in mice exposed to lead nitrate, a neurotoxic agent. The intervention modulated hippocampal oxidative stress markers (e.g., SOD and MDA levels) and reduced plasma corticosterone, a key indicator of HPA axis activity [238].
Noruzi, Meshkati [237] explored the combined impact of HIIT and curcumin supplementation on cognitive function and oxidative stress markers in male BALB/c mice exposed to lead. The results showed significant cognitive improvements and reductions in oxidative damage in groups that received either HIIT or curcumin alone, but the greatest neuroprotection was observed in the combined intervention group. These findings suggest that curcumin enhances the antioxidant and anti-inflammatory properties of HIIT, which may be particularly beneficial in stress-induced cognitive impairment [237].
Cognitive and behavioral benefits
The cognitive-enhancing effects of polyphenols and exercise have been well-documented, particularly in stress-induced and neurodegenerative conditions [235, 240]. In a study on rats with lipopolysaccharide (LPS)-induced depression, Abdallah, El-Gohary [235] found that quercetin supplementation combined with treadmill exercise significantly reduced oxidative stress, inflammation, and apoptosis, while increasing serotonin and BDNF levels. This suggests that exercise and polyphenols interact through mitochondrial regulation to support cognitive function and mental health.
Osali and Rostami [240] conducted a clinical trial investigating the effects of aerobic exercise and nanocurcumin supplementation in women aged 60–65 with metabolic syndrome. The results indicated that the combination of exercise and nanocurcumin significantly reduced plasma IL-1β and nitric oxide (NO) levels, correlating with improvements in depression scores [240]. Both polyphenols and exercise have demonstrated cognitive-enhancing effects, particularly in stress-induced and neurodegenerative conditions. Moghadam, Bagheri [241] investigated the impact of resistance training in combination with saffron supplementation on mood and peripheral neurochemical markers in young men. Their findings indicated that this dual intervention led to increased plasma serotonin, dopamine, and β-endorphins, which are associated with improved mood and stress resilience [241].
In a randomized controlled trial, Johnston [242] assessed the impact of HIIT combined with curcumin supplementation on mental health and exercise performance. While no significant differences in anxiety and depression scores were observed between groups, the study emphasized the need for further research with larger sample sizes and optimized supplementation protocols [242].
Mechanistic insights into the synergistic effects of exercise and polyphenols on brain function
The combination of exercise and polyphenol supplementation presents a promising strategy for enhancing brain function and mitigating depression through overlapping and complementary mechanisms. Both interventions influence neurotrophic signaling, neurotransmitter regulation, neuroinflammation, oxidative stress, mitochondrial function, and synaptic plasticity. These pathways converge to promote neurogenesis, synaptic integrity, and cognitive resilience, while counteracting neurodegeneration and mood disorders. The synergistic interaction between exercise and polyphenols amplifies these benefits beyond what either intervention achieves alone, making this combined approach a compelling avenue for mental health improvement.
A central mechanism through which exercise and polyphenols exert their beneficial effects is the upregulation of BDNF, a key regulator of synaptic plasticity, neuronal survival, and mood stabilization (Tables 2 and 3). Exercise stimulates BDNF expression through activation of PGC-1α and the cyclic AMP response element-binding protein (CREB) pathway, which enhances synaptic remodeling and neurogenesis, particularly in the hippocampus and prefrontal cortex [129]. Polyphenols such as curcumin, quercetin, and resveratrol complement this effect by increasing histone acetylation and DNA methylation, epigenetic modifications that promote BDNF transcription. Through these pathways, the combined action of exercise and polyphenols enhances cognitive function and confers resilience against stress-induced neuronal damage [243, 244] (Fig. 3).
The impact of exercise and polyphenols on neurotransmitter regulation further highlights their synergistic potential in combating depression. Exercise increases plasma tryptophan availability, facilitating its transport into the brain, where it is converted into serotonin and increasing the activity of TPH, the rate-limiting enzyme in serotonin synthesis [245]. Simultaneously, polyphenols inhibit monoamine oxidase (MAO), an enzyme responsible for serotonin degradation, thereby prolonging serotonin’s availability in the synaptic cleft [210]. This dual modulation of serotonin metabolism results in sustained improvements in mood and emotional resilience. Similarly, dopamine and norepinephrine levels are elevated through exercise-induced activation of the mesolimbic reward pathway, which enhances motivation, focus, and stress resilience [246]. Polyphenols contribute to the stabilization of dopaminergic neurons by reducing oxidative stress and inflammatory damage, thereby preventing dopaminergic deficits that are commonly associated with mood disorders [247, 248]. Additionally, both interventions modulate GABA and glutamate levels, restoring the balance between inhibitory and excitatory neurotransmission, which is often disrupted in depression and cognitive dysfunction.
A key contributor to mood disorders and neurodegeneration is chronic neuroinflammation, which is mitigated by both exercise and polyphenols through distinct but complementary pathways. Exercise exerts anti-inflammatory effects by suppressing the activation of NF-κB and reducing levels of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β [249]. Polyphenols enhance these effects by inhibiting microglial activation and downregulating inflammatory cascades driven by oxidative stress [249]. The ability of polyphenols to cross the blood-brain barrier enables them to directly modulate neuroinflammation, reinforcing the neuroprotective effects of exercise [250]. This combined regulation of neuroimmune signaling fosters a protective brain environment, reducing the risk of neuropsychiatric disorders.
Another major factor in cognitive decline and mood disorders is oxidative stress, which is markedly attenuated through the synergistic action of exercise and polyphenols. Exercise enhances the endogenous antioxidant defense system by increasing the expression of SOD, CAT, and glutathione peroxidase (GPx), enzymes that neutralize reactive oxygen species (ROS) and prevent oxidative damage [251]. Polyphenols further reinforce these antioxidant defenses by scavenging free radicals and activating nuclear factor erythroid 2-related factor 2 (Nrf2), a master regulator of oxidative stress response genes [252]. The combined effect of exercise-induced and polyphenol-mediated antioxidant activity reduces neuronal apoptosis, maintains mitochondrial integrity, and preserves overall brain function.
The regulation of mitochondrial function is another crucial pathway through which exercise and polyphenols synergistically enhance brain health. Mitochondrial dysfunction is a hallmark of neurodegenerative and mood disorders, often resulting in impaired ATP production and increased oxidative stress. Exercise stimulates mitochondrial biogenesis through PGC-1α, nuclear respiratory factors (Nrf1 and Nrf2), and mitochondrial transcription factor A (TFAM), leading to enhanced energy metabolism and improved neuronal survival [253, 254]. Polyphenols contribute to mitochondrial protection by preventing mitochondrial fragmentation, supporting efficient electron transport chain function, and reducing the accumulation of damaged mitochondria [255]. This synergy between exercise and polyphenols ensures optimal energy production, thereby sustaining neuronal activity and cognitive performance.
The kynurenine pathway, a metabolic route that diverts tryptophan from serotonin synthesis to neurotoxic metabolites, is also modulated by exercise and polyphenols. Under conditions of chronic stress and inflammation, tryptophan is preferentially converted into quinolinic acid, a neurotoxin that overactivates N-methyl-D-aspartate (NMDA) receptors, leading to excitotoxicity and neuronal death [256]. Exercise counteracts this process by promoting the conversion of kynurenine into kynurenic acid, a neuroprotective metabolite, thereby shifting the pathway away from neurotoxicity [257, 258]. Polyphenols further reinforce this protective mechanism by inhibiting indoleamine 2,3-dioxygenase (IDO), the enzyme responsible for initiating tryptophan degradation, thus preserving serotonin availability and reducing the harmful effects of excessive glutamatergic activity [259,260,261].
Beyond molecular mechanisms, behavioral outcomes associated with exercise and polyphenols substantiate their combined benefits. Both interventions have been shown to reduce anxiety-like and depression-like behaviors in preclinical and clinical studies, with measurable improvements in emotional regulation and cognitive flexibility. The engagement of the prefrontal cortex and hippocampus, key regions for executive function and memory, is enhanced through the structural and functional modifications induced by both exercise and polyphenol consumption. By maintaining neural circuit integrity, reinforcing neuroplasticity, and mitigating stress-induced impairments, the combination of exercise and polyphenols provides a robust defense against cognitive decline and mood disorders.
Thus, the synergistic effects of exercise and polyphenol supplementation on brain function are mediated through the modulation of neurotrophic signaling, neurotransmitter balance, inflammatory responses, oxidative stress, mitochondrial function, and kynurenine metabolism. These mechanisms collectively enhance neurogenesis, synaptic plasticity, and cognitive performance while offering protection against mood disorders and neurodegeneration. By integrating regular physical activity with polyphenol-rich diets, individuals can target multiple mechanistic pathways, including neurotransmitter modulation, neurotrophic support, inflammation control, and mitochondrial protection. This combined approach offers a promising strategy for enhancing cognitive resilience, reducing depressive symptoms, and mitigating neurodegenerative risks. Further research should aim to optimize the type, duration, and intensity of exercise, as well as the specific polyphenol compounds and dosages, to maximize these synergistic effects in clinical settings.
Practical guidelines for exercise and polyphenol supplementation in depression management and brain health
To enhance the practical relevance and applicability of our findings, we propose specific guidelines for the application of exercise and polyphenol supplementation in the management of depression and the enhancement of brain function.
To optimize mental health benefits, a dual-intervention approach combining structured exercise with targeted polyphenol supplementation is recommended. Each component contributes uniquely to neuroprotection, mood stabilization, and cognitive resilience, making their combined application particularly effective. A well-structured exercise regimen should include a combination of aerobic and resistance training to maximize the neuroprotective and antidepressant effects. Aerobic exercise, such as brisk walking, jogging, cycling, or swimming, should be performed at moderate to vigorous intensity, maintaining 60–80% of maximum heart rate. A minimum of 150 min per week of moderate-intensity exercise or 75 min per week of vigorous-intensity exercise is recommended, aligning with guidelines from the American College of Sports Medicine. Sessions should ideally be spread across at least three to five days per week to ensure consistent neurobiological benefits. Resistance training, involving weightlifting, bodyweight exercises, or resistance bands, should be incorporated at least twice a week, targeting major muscle groups to enhance neurotrophic factor expression and neurotransmitter balance. HIIT can also be beneficial, particularly in individuals with lower adherence to longer-duration aerobic programs, as it provides substantial cognitive and emotional benefits in shorter timeframes.
Polyphenol supplementation should be tailored to optimize its synergistic effects with exercise. The selection of polyphenols should focus on those with well-documented neuroprotective and antidepressant properties, including crocin, curcumin, quercetin, resveratrol, and flavonoids from green tea and citrus fruits. Dosage recommendations vary depending on the polyphenol; for instance, crocin supplementation at 30 mg/day has been shown to alleviate depressive symptoms, while curcumin at 100–200 mg/day enhances neurotransmitter balance and reduces inflammation. Quercetin supplementation at 500–1000 mg/day has demonstrated efficacy in modulating oxidative stress and neuroinflammation, while resveratrol at 100–500 mg/day supports mitochondrial function and neuroplasticity. These polyphenols can be consumed in either dietary form, through foods such as saffron, turmeric, onions, berries, and red wine, or as standardized supplements to ensure bioavailability. To maximize absorption and efficacy, polyphenols should be consumed with healthy fats, such as those from olive oil or avocados, or in formulations that enhance bioavailability, such as curcumin with piperine.
To achieve optimal results, exercise and polyphenol supplementation should be consistently integrated into daily routines. Morning exercise sessions may be preferable for enhancing dopamine and serotonin levels throughout the day, while polyphenol intake should be distributed across meals to maintain steady plasma concentrations. The long-term adherence to this combined intervention is crucial for sustained improvements in mood, cognitive function, and overall brain health. The personalized adjustment of exercise intensity and polyphenol dosage, based on individual metabolic responses and mental health status, can further enhance outcomes. While these practical recommendations provide a framework for integrating exercise and polyphenol supplementation into mental health interventions, several research gaps remain. To ensure widespread applicability, future studies must refine dosing strategies, optimize intervention protocols, and establish long-term efficacy.
Critical analysis, research gaps, and future directions
The existing body of research highlights the potential of both exercise and polyphenol supplementation in alleviating depressive symptoms and enhancing cognitive function. However, despite promising results, there remain substantial gaps in understanding the precise mechanisms, dose-response relationships, and long-term effectiveness of these interventions in human populations. While numerous studies have demonstrated the antidepressant and neuroprotective effects of exercise and polyphenols individually, their combined effects remain underexplored, particularly in clinical populations. The existing evidence is primarily derived from preclinical models or short-term human trials, which limits the generalizability of findings to diverse populations with varying degrees of depression severity. Furthermore, while several studies indicate a synergistic effect between exercise and polyphenols, the biological pathways underlying this interaction require further elucidation.
A significant limitation in current research is the lack of standardization in the methodologies used to assess depressive symptoms, cognitive improvements, and biological markers. Variability in exercise protocols (e.g., type, intensity, and duration) and polyphenol dosages, along with inconsistencies in study populations (e.g., age, sex, health status), complicates direct comparisons across studies. A major limitation in translating polyphenol research into clinical practice is their poor bioavailability. Many polyphenols, including curcumin and quercetin, undergo rapid metabolism, reducing their therapeutic efficacy. Future research should focus on optimizing formulations (e.g., nano-encapsulation, co-administration with bioavailability enhancers like piperine) to enhance absorption and prolong systemic circulation.
Another critical aspect that warrants further investigation is the role of individual differences in response to exercise and polyphenol supplementation. Genetic and metabolic variability significantly influence individual responses to exercise and polyphenol supplementation. Variations in neurotransmitter-related genes (e.g., COMT, BDNF Val66Met, SERT polymorphisms) may alter the efficacy of these interventions. Additionally, baseline inflammatory profiles and metabolic differences impact polyphenol metabolism, necessitating a precision medicine approach in future research to tailor interventions based on individual biological profiles. Additionally, while much of the existing literature has focused on aerobic exercise, less attention has been given to the differential effects of resistance training, HIIT, and mind-body exercises like yoga or tai chi on depression and cognitive function. More comparative studies are needed to determine the most effective exercise modalities in combination with polyphenol supplementation.
Beyond mechanistic and methodological gaps, there is also a need for long-term clinical trials to evaluate the sustainability of these interventions. Despite strong evidence supporting the short-term benefits of exercise and polyphenol supplementation, long-term studies remain scarce. Future trials should prioritize longitudinal designs with extended follow-ups to assess whether these interventions provide sustained neuroprotection and reduce depression recurrence. Additionally, the potential interactions between polyphenols and commonly prescribed antidepressants have not been sufficiently explored. Understanding whether polyphenols can serve as adjuncts to conventional pharmacotherapy without adverse interactions is crucial for integrating these natural compounds into mainstream clinical practice.
Furthermore, the role of lifestyle factors, such as diet quality, sleep patterns, and stress levels, in mediating the effects of exercise and polyphenol supplementation should be examined in more detail. Given that mental health is influenced by multiple interconnected biological and behavioral factors, future research should adopt a multi-dimensional approach that integrates diet, exercise, and psychological interventions to maximize therapeutic outcomes.
Thus, while the current literature provides compelling evidence supporting the use of exercise and polyphenols for mental health, more rigorous, well-controlled human studies are needed to refine dosing strategies, optimize intervention protocols, and establish long-term efficacy. Future research should prioritize standardized methodologies, personalized treatment approaches, and real-world applicability to bridge the gap between experimental findings and clinical implementation. By addressing these research gaps, future studies can refine clinical guidelines, develop precision-targeted interventions, and establish standardized methodologies that maximize the therapeutic benefits of exercise and polyphenol supplementation for depression and cognitive resilience.
Conclusions
The combined effects of exercise and polyphenol supplementation offer a promising, non-pharmacological approach for managing depression and enhancing brain function. Both interventions modulate key biological pathways, including neurotransmitter regulation, neurogenesis, oxidative stress reduction, and inflammation control, contributing to improved mood, cognitive function, and overall mental well-being. Evidence suggests that polyphenols such as crocin, curcumin, and quercetin, when paired with structured physical activity, may yield synergistic benefits beyond their individual effects. However, further clinical studies are needed to optimize dosage, duration, and exercise modalities, ensuring the most effective implementation in depression management. Future research should also explore long-term effects and potential interactions with conventional therapies to establish their broader clinical relevance.
Data availability
No datasets were generated or analysed during the current study.
References
WHO WHO. Mental disorders. 2022 [updated 2022; cited]; Available from: https://www.who.int/news-room/fact-sheets/detail/mental-disorders
WHO WHO. Mental Health and COVID-19: Early evidence of the pandemic’s impact: Scientific brief, 2 March 2022. Geneva: World Health Organization. 2022 [updated 2022; cited]; Available from: https://www.who.int/publications/i/item/WHO-2019-nCoV-Sci_Brief-Mental_health-2022.1
Kazdin AE, Rabbitt SM. Novel models for delivering mental health services and reducing the burdens of mental illness. Clin Psychol Sci. 2013;1:170–91.
WHO WHO. Depressive disorder (depression). 2023 [updated 2023; cited]; Available from: https://www.who.int/news-room/fact-sheets/detail/depression
Mandal PK, Gaur S, Roy RG, Samkaria A, Ingole R, Goel A, Schizophrenia. Bipolar and major depressive disorders: overview of clinical features, neurotransmitter alterations, Pharmacological interventions, and impact of oxidative stress in the disease process. ACS Chem Neurosci. 2022;13:2784–802.
Malhi GS, Mann JJ, Depression. Lancet. 2018;392:2299–312.
Jacka FN, O’Neil A, Opie R, Itsiopoulos C, Cotton S, Mohebbi M, et al. A randomised controlled trial of dietary improvement for adults with major depression (the ‘SMILES’ trial). BMC Med. 2017;15:23.
Winiarska-Mieczan A, Kwiecień M, Jachimowicz-Rogowska K, Donaldson J, Tomaszewska E, Baranowska-Wójcik E. Anti-Inflammatory, antioxidant, and neuroprotective effects of Polyphenols—Polyphenols as an element of diet therapy in depressive disorders. Int J Mol Sci. 2023;24:2258.
Scalbert A, Johnson IT, Saltmarsh M. Polyphenols: antioxidants and beyond. Am J Clin Nutr. 2005;81:S215–7.
Williams RJ, Spencer JP. Flavonoids, cognition, and dementia: actions, mechanisms, and potential therapeutic utility for alzheimer disease. Free Radic Biol Med. 2012;52:35–45.
Vauzour D, Vafeiadou K, Rodriguez-Mateos A, Rendeiro C, Spencer JP. The neuroprotective potential of flavonoids: a multiplicity of effects. Genes Nutr. 2008;3:115–26.
Lin K, Li Y, Toit ED, Wendt L, Sun J. Effects of polyphenol supplementations on improving depression, anxiety, and quality of life in patients with depression. Front Psychiatry. 2021;12:765485.
Mammen G, Faulkner G. Physical activity and the prevention of depression: A systematic review of prospective studies. Am J Prev Med. 2013;45:649–57.
Knapen J, Vancampfort D, Moriën Y, Marchal Y. Exercise therapy improves both mental and physical health in patients with major depression. Disabil Rehabil. 2015;37:1490–5.
Erickson KI, Voss MW, Prakash RS, Basak C, Szabo A, Chaddock L, et al. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci. 2011;108:3017–22.
Dishman RK, Berthoud HR, Booth FW, Cotman CW, Edgerton VR, Fleshner MR, et al. Neurobiology of exercise. Obesity. 2006;14:345–56.
Watson R, Harvey K, McCabe C, Reynolds S. Understanding anhedonia: a qualitative study exploring loss of interest and pleasure in adolescent depression. Eur Child Adolesc Psychiatry. 2020;29:489–99.
Paykel ES. Basic concepts of depression. Dialog Clin Neurosci. 2008;10:279–89.
Willner P, Scheel-Krüger J, Belzung C. The neurobiology of depression and antidepressant action. Neurosci Biobehavioral Reviews. 2013;37:2331–71.
Mekonnen D, Kebede Y. The prevalence of suicidal ideation and attempts among individuals attending an adult psychiatry out-patient clinic in Gondar. Ethiopia Afr Health Sci. 2011;11.
Charlton BG. A model for self-treatment of four sub-types of symptomatic ‘depression’ using non-prescription agents: neuroticism (anxiety and emotional instability); malaise (fatigue and painful symptoms); demotivation (anhedonia) and seasonal affective disorder ‘SAD’. Med Hypotheses. 2009;72:1–7.
Wirz-Justice A. Diurnal variation of depressive symptoms. Dialog Clin Neurosci. 2008;10:337–43.
Kanter JW, Busch AM, Weeks CE, Landes SJ. The nature of clinical depression: symptoms, syndromes, and behavior analysis. Behav Analyst. 2008;31:1–21.
Keenan K, Feng X, Hipwell A, Klostermann S. Depression begets depression: comparing the predictive utility of depression and anxiety symptoms to later depression. J Child Psychol Psychiatry. 2009;50:1167–75.
Lee H, Ang S. Productive activities and risk of cognitive impairment and depression: does the association vary by gender?? Sociol Perspect. 2020;63:608–29.
Zimmerman M, Morgan TA, Stanton K. The severity of psychiatric disorders. World Psychiatry. 2018;17:258–75.
Baddeley JL, Pennebaker JW, Beevers CG. Everyday social behavior during a major depressive episode. Social Psychol Personality Sci. 2012;4:445–52.
Solomon DA, Keller MB, Leon AC, Mueller TI, Lavori PW, Shea MT, et al. Multiple recurrences of major depressive disorder. Am J Psychiatry. 2000;157:229–33.
Tondo L, Vazquez H, Baldessarini GJ. Depression and mania in bipolar disorder. Curr Neuropharmacol. 2017;15:353–8.
American Psychiatric Association. Diagnostic and statistical manual of mental disorders: DSM-5™. 5th ed. Arlington, VA, US: American Psychiatric Publishing, Inc.; 2013.
Kroenke K, Spitzer RL, Williams JBW. The PHQ-9. J Gen Intern Med. 2001;16:606–13.
Beck AT, Steer RA, Brown G. Beck depression inventory–II. Psychological assessment; 1996.
First MB, Williams JB, Karg RS, Spitzer RL. SCID-5-RV. 2015.
Craighead WE, Dunlop BW. Combination psychotherapy and antidepressant medication treatment for depression: for whom, when, and how. Ann Rev Psychol. 2014;65:267–300.
Hollon SD, Thase ME, Markowitz JC. Treatment and prevention of depression. Psychol Sci Public Interest. 2002;3:39–77.
Khan A, Shrivastava R, Tugnawat D, Singh A, Dimidjian S, Patel V, et al. Design and development of a digital program for training non-specialist health workers to deliver an evidence-based psychological treatment for depression in primary care in India. J Technol Behav Sci. 2020;5:402–15.
Ngandu T, Lehtisalo J, Solomon A, Levälahti E, Ahtiluoto S, Antikainen R, et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet. 2015;385:2255–63.
Livingston G, Sommerlad A, Orgeta V, Costafreda SG, Huntley J, Ames D, et al. Dementia prevention, intervention, and care. Lancet. 2017;390:2673–734.
Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018;14:535–62.
Mishra A, Patni P, Hegde S, Aleya L, Tewari D. Neuroplasticity and environment: A pharmacotherapeutic approach toward preclinical and clinical Understanding. Curr Opin Environ Sci Health. 2021;19:100210.
Gómez-Pinilla F. Brain foods: the effects of nutrients on brain function. Nat Rev Neurosci. 2008;9:568–78.
Dinoff A, Herrmann N, Swardfager W, Gallagher D, Lanctôt KL. The effect of exercise on resting concentrations of peripheral brain-derived neurotrophic factor (BDNF) in major depressive disorder: A meta-analysis. J Psychiatr Res. 2018;105:123–31.
Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell. 2010;140:918–34.
Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14:463–77.
Teleanu RI, Niculescu A-G, Roza E, Vladâcenco O, Grumezescu AM, Teleanu DM. Neurotransmitters—Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. Journal [serial on the Internet]. 2022 Date; 23(11).
Millan MJ. Multi-target strategies for the improved treatment of depressive States: conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther. 2006;110:135–370.
Brambilla P, Perez J, Barale F, Schettini G, Soares JC. GABAergic dysfunction in mood disorders. Mol Psychiatry. 2003;8:721–37.
Sanacora G, Treccani G, Popoli M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology. 2012;62:63–77.
Kramer AF, Erickson KI. Effects of physical activity on cognition, well-being, and brain: human interventions. Alzheimer’s Dement. 2007;3:S45–51.
Bullard T, Ji M, An R, Trinh L, Mackenzie M, Mullen SP. A systematic review and meta-analysis of adherence to physical activity interventions among three chronic conditions: cancer, cardiovascular disease, and diabetes. BMC Public Health. 2019;19:636.
American College of Sports Medicine. ACSM’s guidelines for exercise testing and prescription. Lippincott williams & wilkins; 2013.
Westcott WL. Resistance training is medicine: effects of strength training on health. Curr Sports Med Rep. 2012;11:209–16.
Borde R, Hortobágyi T, Granacher U. Dose–Response relationships of resistance training in healthy old adults: A systematic review and Meta-Analysis. Sports Med. 2015;45:1693–720.
Gillen JB, Gibala MJ. Is high-intensity interval training a time-efficient exercise strategy to improve health and fitness? Appl Physiol Nutr Metab. 2014;39:409–12.
Burgomaster KA, Howarth KR, Phillips SM, Rakobowchuk M, MacDonald MJ, McGee SL, et al. Similar metabolic adaptations during exercise after low volume sprint interval and traditional endurance training in humans. J Physiol. 2008;586:151–60.
Riebe D, Ehrman JK, Liguori G, Magal M. ACSM’s Guidelines for Exercise Testing and Prescription. Wolters Kluwer; 2018.
Schuch FB, Vancampfort D, Firth J, Rosenbaum S, Ward PB, Silva ES, et al. Physical activity and incident depression: A Meta-Analysis of prospective cohort studies. Am J Psychiatry. 2018;175:631–48.
Harber VJ, Sutton JR. Endorphins and exercise. Sports Med. 1984;1:154–71.
Siebers M, Biedermann SV, Fuss J. Do endocannabinoids cause the runner’s high?? Evidence and open questions. Neuroscientist. 2022;29:352–69.
Dishman RK, McDowell CP, Herring MP. Customary physical activity and odds of depression: a systematic review and meta-analysis of 111 prospective cohort studies. Br J Sports Med. 2021;55:926.
Morres ID, Hatzigeorgiadis A, Stathi A, Comoutos N, Arpin-Cribbie C, Krommidas C, et al. Aerobic exercise for adult patients with major depressive disorder in mental health services: A systematic review and meta-analysis. Depress Anxiety. 2019;36:39–53.
Ryan MP. The antidepressant effects of physical activity: mediating self-esteem and self-efficacy mechanisms. Psychol Health. 2008;23:279–307.
Salmon P. Effects of physical exercise on anxiety, depression, and sensitivity to stress: A unifying theory. Clin Psychol Rev. 2001;21:33–61.
Pepin R, Stevens CJ, Choi NG, Feeney SM, Bruce ML. Modifying behavioral activation to reduce social isolation and loneliness among older adults. Am J Geriatric Psychiatry. 2021;29:761–70.
Franke T, Sims-Gould J, Nettlefold L, Ottoni C, McKay HA. It makes me feel not so alone: features of the choose to move physical activity intervention that reduce loneliness in older adults. BMC Public Health. 2021;21:312.
Barton J, Griffin M, Pretty J. Exercise-, nature- and socially interactive-based initiatives improve mood and self-esteem in the clinical population. Perspect Public Health. 2011;132:89–96.
Craft LL, Perna FM. The benefits of exercise for the clinically depressed. Prim Care Companion J Clin Psychiatry. 2004;6:104–11.
Harvey SB, Øverland S, Hatch SL, Wessely S, Mykletun A, Hotopf M. Exercise and the prevention of depression: results of the HUNT cohort study. Am J Psychiatry. 2017;175:28–36.
Kandola A, Ashdown-Franks G, Hendrikse J, Sabiston CM, Stubbs B. Physical activity and depression: towards Understanding the antidepressant mechanisms of physical activity. Neurosci Biobehavioral Reviews. 2019;107:525–39.
Mikkelsen K, Stojanovska L, Polenakovic M, Bosevski M, Apostolopoulos V. Exercise and mental health. Maturitas. 2017;106:48–56.
Duclos M, Tabarin A. Exercise and the Hypothalamo-Pituitary-Adrenal axis. Sports Endocrinology: S.Karger AG; 2016. p. 0.
Hinds JA, Sanchez ER. The Role of the Hypothalamus–Pituitary–Adrenal (HPA) Axis in Test-Induced Anxiety: Assessments, Physiological Responses, and Molecular Details. Journal [serial on the Internet]. 2022 Date; 2(1).
Hill EE, Zack E, Battaglini C, Viru M, Viru A, Hackney AC. Exercise and Circulating cortisol levels: the intensity threshold effect. J Endocrinol Investig. 2008;31:587–91.
Jasińska-Mikołajczyk A, Drews K, Domaszewska K, Kolasa G, Konofalska M, Jowik K et al. The effect of physical activity on neurotrophin concentrations and cognitive control in patients with a depressive episode. Front Psychiatry. 2022;13.
Ji C, Yang J, Lin L, Chen S. Physical exercise ameliorates anxiety, depression and sleep quality in college students: experimental evidence from exercise intensity and frequency. Behav Sci. 2022;12:61.
Brand S, Gerber M, Beck J, Hatzinger M, Pühse U, Holsboer-Trachsler E. High exercise levels are related to favorable sleep patterns and psychological functioning in adolescents: A comparison of athletes and controls. J Adolesc Health. 2010;46:133–41.
Herring MP, O’Connor PJ, Dishman RK. The effect of exercise training on anxiety symptoms among patients: a systematic review. Arch Intern Med. 2010;170:321–31.
Rees T, Hardy L. An investigation of the social support experiences of High-Level sports performers. Sport Psychol. 2000;14:327–47.
Rebar AL, Stanton R, Geard D, Short C, Duncan MJ, Vandelanotte C. A meta-meta-analysis of the effect of physical activity on depression and anxiety in non-clinical adult populations. Health Psychol Rev. 2015;9:366–78.
Levinson CA, Rodebaugh TL, Menatti AR, Weeks JW. Development and validation of the social exercise and anxiety measure (SEAM): assessing fears, avoidance, and importance of social exercise. J Psychopathol Behav Assess. 2013;35:244–53.
Haenisch B, Bönisch H. Depression and antidepressants: insights from knockout of dopamine, serotonin or noradrenaline re-uptake transporters. Pharmacol Ther. 2011;129:352–68.
Numakawa T, Odaka H, Adachi N. Actions of brain-Derived neurotrophin factor in the neurogenesis and neuronal function, and its involvement in the pathophysiology of brain diseases. Int J Mol Sci. 2018;19:3650.
Lee MT, Peng W-H, Kan H-W, Wu C-C, Wang D-W, Ho Y-C. Neurobiology of depression: chronic stress alters the glutamatergic system in the Brain—Focusing on AMPA receptor. Biomedicines. 2022;10:1005.
Nollet M, Leman S. Role of orexin in the pathophysiology of depression: potential for Pharmacological intervention. CNS Drugs. 2013;27:411–22.
Ren J, Xiao H. Exercise for mental Well-Being: exploring Neurobiological advances and intervention effects in depression. Life. 2023;13:1505.
Holmes A, Murphy DL, Crawley JN. Abnormal behavioral phenotypes of serotonin transporter knockout mice: parallels with human anxiety and depression. Biol Psychiatry. 2003;54:953–9.
Phillips C, Fahimi A. Immune and neuroprotective effects of physical activity on the brain in depression. Front NeuroSci. 2018;12.
Pietrelli A, Matković L, Vacotto M, Lopez-Costa JJ, Basso N, Brusco A. Aerobic exercise upregulates the BDNF-Serotonin systems and improves the cognitive function in rats. Neurobiol Learn Mem. 2018;155:528–42.
Kondo M, Nakamura Y, Ishida Y, Shimada S. The 5-HT3 receptor is essential for exercise-induced hippocampal neurogenesis and antidepressant effects. Mol Psychiatry. 2015;20:1428–37.
Kim MH, Leem YH. Chronic exercise improves repeated restraint stress-induced anxiety and depression through 5HT1A receptor and cAMP signaling in hippocampus. J Exerc Nutr Biochem. 2014;18:97–104.
Dey S. Physical exercise as a novel antidepressant agent: possible role of serotonin receptor subtypes. Physiol Behav. 1994;55:323–9.
Dey S, Singh RH, Dey PK. Exercise training: significance of regional alterations in serotonin metabolism of rat brain in relation to antidepressant effect of exercise. Physiol Behav. 1992;52:1095–9.
Liu W, Sheng H, Xu Y, Liu Y, Lu J, Ni X. Swimming exercise ameliorates depression-like behavior in chronically stressed rats: relevant to Proinflammatory cytokines and IDO activation. Behav Brain Res. 2013;242:110–6.
Daniele TMC, de Bruin PFC, Rios ERV, de Bruin VMS. Effects of exercise on depressive behavior and striatal levels of norepinephrine, serotonin and their metabolites in sleep-deprived mice. Behav Brain Res. 2017;332:16–22.
Dishman RK, O’Connor PJ. Lessons in exercise neurobiology: the case of endorphins. Ment Health Phys Act. 2009;2:4–9.
Merenlender-Wagner A, Dikshtein Y, Yadid G. The β-Endorphin role in Stress-Related psychiatric disorders. Curr Drug Targets. 2009;10:1096–108.
Balchin R, Linde J, Blackhurst D, Rauch HGL, Schönbächler G. Sweating away depression? The impact of intensive exercise on depression. J Affect Disord. 2016;200:218–21.
Dinas PC, Koutedakis Y, Flouris AD. Effects of exercise and physical activity on depression. Ir J Med Sci. 2011;180:319–25.
Knoll AT, Carlezon WA. Dynorphin, stress, and depression. Brain Res. 2010;1314:56–73.
Bjørnebekk A, Mathé AA, Brené S. The antidepressant effect of running is associated with increased hippocampal cell proliferation. Int J Neuropsychopharmacol. 2005;8:357–68.
Werme M, Thorén P, Olson L, Brené S. Running and cocaine both upregulate dynorphin mRNA in medial caudate putamen. Eur J Neurosci. 2000;12:2967–74.
Ruiz-Tejada A, Neisewander J, Katsanos CS. Regulation of voluntary physical activity behavior: A review of evidence involving dopaminergic pathways in the brain. Brain Sci. 2022;12:333.
Foley TE, Fleshner M. Neuroplasticity of dopamine circuits after exercise: implications for central fatigue. Neuromol Med. 2008;10:67–80.
Mishra A, Singh S, Tiwari V, Parul, Shukla S. Dopamine D1 receptor activation improves adult hippocampal neurogenesis and exerts anxiolytic and antidepressant-like effect via activation of Wnt/β-catenin pathways in rat model of Parkinson’s disease. Neurochem Int. 2019;122:170–86.
Matta Mello Portugal E, Cevada T, Sobral Monteiro-Junior R, Teixeira Guimarães T, da Cruz Rubini E, Lattari E, et al. Neuroscience of exercise: from neurobiology mechanisms to mental health. Neuropsychobiology. 2013;68:1–14.
Robison LS, Swenson S, Hamilton J, Thanos PK. Exercise reduces dopamine D1R and increases D2R in rats: implications for addiction. Med Sci Sports Exerc. 2018;50:1596–602.
Chen C, Nakagawa S, Kitaichi Y, An Y, Omiya Y, Song N, et al. The role of medial prefrontal corticosterone and dopamine in the antidepressant-like effect of exercise. Psychoneuroendocrinology. 2016;69:1–9.
Robertson CL, Ishibashi K, Chudzynski J, Mooney LJ, Rawson RA, Dolezal BA, et al. Effect of exercise training on striatal dopamine D2/D3 receptors in methamphetamine users during behavioral treatment. Neuropsychopharmacology. 2016;41:1629–36.
He Q, Wu J, Wang X, Luo F, Yan K, Yu W, et al. Exercise intervention can reduce the degree of drug dependence of patients with amphetamines/addiction by improving dopamine level and immunity and reducing negative emotions. Am J Transl Res. 2021;13:1779–88.
Bastioli G, Arnold JC, Mancini M, Mar AC, Gamallo-Lana B, Saadipour K, et al. Voluntary exercise boosts striatal dopamine release: evidence for the necessary and sufficient role of BDNF. J Neurosci. 2022;42:4725.
Foster AC, Kemp JA. Glutamate- and GABA-based CNS therapeutics. Curr Opin Pharmacol. 2006;6:7–17.
Maejima H, Ninuma S, Okuda A, Inoue T, Hayashi M. Exercise and low-level GABAA receptor Inhibition modulate locomotor activity and the expression of BDNF accompanied by changes in epigenetic regulation in the hippocampus. Neurosci Lett. 2018;685:18–23.
Ignácio ZM, da Silva RS, Plissari ME, Quevedo J, Réus GZ. Physical exercise and neuroinflammation in major depressive disorder. Mol Neurobiol. 2019;56:8323–35.
Lu CY, Liu DX, Jiang H, Pan F, Ho CSH, Ho RCM. Effects of traumatic stress induced in the juvenile period on the expression of Gamma-Aminobutyric acid receptor type A subunits in adult rat brain. Neural Plast. 2017;2017:5715816.
Hill LE, Droste SK, Nutt DJ, Linthorst ACE, Reul J. Voluntary exercise alters GABAA receptor subunit and glutamic acid decarboxylase-67 gene expression in the rat forebrain. J Psychopharmacol. 2008;24:745–56.
Coxon JP, Cash RFH, Hendrikse JJ, Rogasch NC, Stavrinos E, Suo C, et al. GABA concentration in sensorimotor cortex following high-intensity exercise and relationship to lactate levels. J Physiol. 2018;596:691–702.
Streeter CC, Jensen JE, Perlmutter RM, Cabral HJ, Tian H, Terhune DB, et al. Yoga Asana sessions increase brain GABA levels: A pilot study. J Altern Complement Med. 2007;13:419–26.
Streeter CC, Whitfield TH, Owen L, Rein T, Karri SK, Yakhkind A, et al. Effects of yoga versus walking on mood, anxiety, and brain GABA levels: A randomized controlled MRS study. J Altern Complement Med. 2010;16:1145–52.
Jansson LC, Åkerman KE. The role of glutamate and its receptors in the proliferation, migration, differentiation and survival of neural progenitor cells. J Neural Transm. 2014;121:819–36.
Mee-inta O, Zhao Z-W, Kuo Y-M. Physical exercise inhibits inflammation and microglial activation. Cells. 2019;8:691.
Baskerville R, McGrath T, Castell L. The effects of physical activity on glutamate neurotransmission in neuropsychiatric disorders. Front Sports Act Living. 2023;5.
Valente-Silva P, Ruas JL. Tryptophan-kynurenine metabolites in exercise and mental health. Hormones, metabolism and the benefits of exercise. 2017:83–91.
Hestad K, Alexander J, Rootwelt H, Aaseth JO. The Role of Tryptophan Dysmetabolism and Quinolinic Acid in Depressive and Neurodegenerative Diseases. Journal [serial on the Internet]. 2022 Date; 12(7).
Zhao L, Du M, Zhang R. The improvement effect of exercise on rats with chronic stress-induced depression-like behavior: a discussion based on the kynurenine pathway. J Int Med Res. 2024;52:03000605241261986.
Bashiri H, Enayati M, Bashiri A, Salari A-A. Swimming exercise improves cognitive and behavioral disorders in male NMRI mice with sporadic Alzheimer-like disease. Physiol Behav. 2020;223:113003.
Maddock RJ, Casazza GA, Buonocore MH, Tanase C. Vigorous exercise increases brain lactate and Glx (glutamate + glutamine): A dynamic 1H-MRS study. NeuroImage. 2011;57:1324–30.
Maddock RJ, Casazza GA, Fernandez DH, Maddock MI. Acute modulation of cortical glutamate and GABA content by physical activity. J Neurosci. 2016;36:2449.
Lima CB, Soares GSF, Vitor SM, Andrade-da-Costa BLS, Castellano B, Guedes RCA. Spreading depression features and Iba1 immunoreactivity in the cerebral cortex of developing rats submitted to treadmill exercise after treatment with monosodium glutamate. Int J Dev Neurosci. 2014;33:98–105.
Reddy I, Yadav Y, Dey CS. Cellular and molecular regulation of Exercise—A neuronal perspective. Cell Mol Neurobiol. 2023;43:1551–71.
Mahalakshmi B, Maurya N, Lee S-D, Bharath Kumar V. Possible neuroprotective mechanisms of physical exercise in neurodegeneration. Int J Mol Sci. 2020;21:5895.
Ezzat W, Abd-El Hamid MS. Aerobic exercise mediated increase in BDNF expression ameliorates depression in Propylthiouracil-Induced hypothyroidism in adult rats. J Affect Disorders Rep. 2021;6:100268.
Cokar D, Polat MG, Timurtas E, Sertbas Y, Sogut İ. Neuroprotective and metabotropic effect of aerobic exercise training in female patients with type 2 diabetes mellitus. Turk J Biochem. 2022;47:741–8.
Shariq AS, Rosenblat JD, Alageel A, Mansur RB, Rong C, Ho RC, et al. Evaluating the role of orexins in the pathophysiology and treatment of depression: A comprehensive review. Prog Neuropsychopharmacol Biol Psychiatry. 2019;92:1–7.
Kunugi H. Depression and lifestyle: focusing on nutrition, exercise, and their possible relevance to molecular mechanisms. J Neuropsychiatry Clin Neurosci. 2023;77:420–33.
Sargin D. The role of the orexin system in stress response. Neuropharmacology. 2019;154:68–78.
Kakizaki M, Tsuneoka Y, Takase K, Kim SJ, Choi J, Ikkyu A, et al. Differential roles of each orexin receptor signaling in obesity. iScience. 2019;20:1–13.
Monda V, Sessa F, Ruberto M, Carotenuto M, Marsala G, Monda M, et al. Aerobic exercise and metabolic syndrome: the role of sympathetic activity and the redox system. Diabetes Metabolic Syndrome Obes. 2020;13:2433–42.
Morales-Medina JC, Dumont Y, Quirion R. A possible role of neuropeptide Y in depression and stress. Brain Res. 2010;1314:194–205.
Joksimovic J, Selakovic D, Matovic M, Zaletel I, Puskas N, Rosic G. The role of neuropeptide-Y in nandrolone decanoate-induced Attenuation of antidepressant effect of exercise. PLoS ONE. 2017;12:e0178922.
Karamouzis I, Karamouzis M, Vrabas IS, Christoulas K, Kyriazis N, Giannoulis E, et al. The effects of marathon swimming on serum leptin and plasma neuropeptide. Y Levels. 2002;40:132–6.
Smitka K, Papezova H, Vondra K, Hill M, Hainer V, Nedvidkova J. A higher response of plasma neuropeptide Y, growth hormone, leptin levels and extracellular glycerol levels in subcutaneous abdominal adipose tissue to acipimox during exercise in patients with bulimia nervosa: single-blind, randomized, Microdialysis study. Nutr Metabolism. 2011;8:81.
Stogner KA, Holmes PV. Neuropeptide-Y exerts antidepressant-like effects in the forced swim test in rats. Eur J Pharmacol. 2000;387:R9–10.
Alizadeh Pahlavani H. Possible role of exercise therapy on depression: effector neurotransmitters as key players. Behav Brain Res. 2024;459:114791.
Strasser B, Fuchs D. Role of physical activity and diet on mood, behavior, and cognition. Neurol Psychiatry Brain Res. 2015;21:118–26.
Jenkins TA, Nguyen JC, Polglaze KE, Bertrand PP. Influence of Tryptophan and serotonin on mood and cognition with a possible role of the gut-brain axis. Nutrients. 2016;8:56.
Strasser B, Geiger D, Schauer M, Gatterer H, Burtscher M, Fuchs D. Effects of exhaustive aerobic exercise on Tryptophan-Kynurenine metabolism in trained athletes. PLoS ONE. 2016;11:e0153617.
Melancon MO, Lorrain D, Dionne IJ. Exercise increases Tryptophan availability to the brain in older men age 57–70 years. Med Sci Sports Exerc. 2012;44:881–7.
Kim TW, Lim BV, Baek D, Ryu DS, Seo JH. Stress-Induced depression is alleviated by aerobic exercise through Up-Regulation of 5-Hydroxytryptamine 1A receptors in rats. Int Neurourol J. 2015;19:27–33.
Shin MS, Park SS, Lee JM, Kim TW, Kim YP. Treadmill exercise improves depression-like symptoms by enhancing serotonergic function through upregulation of 5-HT(1A) expression in the olfactory bulbectomized rats. J Exerc Rehabil. 2017;13:36–42.
Ji E-S, Lee J-M, Kim T-W, Kim Y-M, Kim Y-S, Kim K. Treadmill exercise ameliorates depressive symptoms through increasing serotonin expression in postpartum depression rats. J Exerc Rehabil. 2017;13:130–5.
Baek S-S, Jun T-W, Kim K-J, Shin M-S, Kang S-Y, Kim C-J. Effects of postnatal treadmill exercise on apoptotic neuronal cell death and cell proliferation of maternal-separated rat pups. Brain Develop. 2012;34:45–56.
Lee H, Ohno M, Ohta S, Mikami T. Regular moderate or intense exercise prevents Depression-Like behavior without change of hippocampal Tryptophan content in chronically Tryptophan-Deficient and stressed mice. PLoS ONE. 2013;8:e66996.
Dulawa SC, Janowsky DS. Cholinergic regulation of mood: from basic and clinical studies to emerging therapeutics. Mol Psychiatry. 2019;24:694–709.
Zong B, Yu F, Zhang X, Zhao W, Sun P, Li S et al. Understanding how physical exercise improves Alzheimer’s disease: cholinergic and monoaminergic systems. Front Aging Neurosci. 2022;14.
Basso JC, Suzuki WA. The effects of acute exercise on mood, cognition, neurophysiology, and neurochemical pathways: a review. Brain Plast. 2017;2:127–52.
Lafenetre P, Leske O, Wahle P, Heumann R. The beneficial effects of physical activity on impaired adult neurogenesis and cognitive performance. Front NeuroSci. 2011;5:51.
Morgan JA, Singhal G, Corrigan F, Jaehne EJ, Jawahar MC, Baune BT. The effects of aerobic exercise on depression-like, anxiety-like, and cognition-like behaviours over the healthy adult lifespan of C57BL/6 mice. Behav Brain Res. 2018;337:193–203.
Liang X, Tang J, Chao F-l, Zhang Y, Chen L-m, Wang F-f, et al. Exercise improves depressive symptoms by increasing the number of excitatory synapses in the hippocampus of CUS-Induced depression model rats. Behav Brain Res. 2019;374:112115.
Déry N, Pilgrim M, Gibala M, Gillen J, Wojtowicz JM, MacQueen G, et al. Adult hippocampal neurogenesis reduces memory interference in humans: opposing effects of aerobic exercise and depression. Front NeuroSci. 2013;7:66.
Stroth S, Hille K, Spitzer M, Reinhardt R. Aerobic endurance exercise benefits memory and affect in young adults. Neuropsychological Rehabilitation. 2009;19:223–43.
Netz Y. Is there a preferred mode of exercise for cognition enhancement in older age?—a narrative review. Front Med. 2019;6:57.
Neeper SA, Gómez-Pinilla F, Choi J, Cotman CW. Physical activity increases mRNA for brain-derived neurotrophic factor and nerve growth factor in rat brain. Brain Res. 1996;726:49–56.
van Praag H, Shubert T, Zhao C, Gage FH. Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci. 2005;25:8680.
Babyak M, Blumenthal JA, Herman S, Khatri P, Doraiswamy M, Moore K, et al. Exercise treatment for major depression: maintenance of therapeutic benefit at 10 months. Psychosom Med. 2000;62:633–8.
Helgadóttir B, Forsell Y, Hallgren M, Möller J, Ekblom Ö. Long-term effects of exercise at different intensity levels on depression: A randomized controlled trial. Prev Med. 2017;105:37–46.
Sun L, Sun Q, Qi J. Adult hippocampal neurogenesis: an important target associated with antidepressant effects of exercise. Rev Neurosci. 2017;28:693–703.
Wu C-W, Chang Y-T, Yu L, Chen H-i, Jen CJ, Wu S-Y, et al. Exercise enhances the proliferation of neural stem cells and neurite growth and survival of neuronal progenitor cells in dentate gyrus of middle-aged mice. J Appl Physiol. 2008;105:1585–94.
Saraulli D, Costanzi M, Mastrorilli V, Farioli-Vecchioli S. The long run: neuroprotective effects of physical exercise on adult neurogenesis from youth to old age. Curr Neuropharmacol. 2017;15:519–33.
Cassilhas RC, Tufik S, de Mello MT. Physical exercise, neuroplasticity, Spatial learning and memory. Cell Mol Life Sci. 2016;73:975–83.
Shih P-C, Yang Y-R, Wang R-Y. Effects of exercise intensity on Spatial memory performance and hippocampal synaptic plasticity in transient brain ischemic rats. PLoS ONE. 2013;8:e78163.
Martín-Rodríguez JF, Ramos-Herrero VD, Parras GG, Flores-Martínez Á, Madrazo-Atutxa A, Cano DA, et al. Chronic adult-onset of growth hormone/IGF-I hypersecretion improves cognitive functions and LTP and promotes neuronal differentiation in adult rats. Acta Physiol. 2020;229:e13293.
Glasper ER, Llorens-Martin MV, Leuner B, Gould E, Trejo JL. Blockade of insulin-like growth factor-I has complex effects on structural plasticity in the hippocampus. Hippocampus. 2010;20:706–12.
Gulyaeva NV. Functional neurochemistry of the ventral and dorsal hippocampus: stress, depression, dementia and remote hippocampal damage. Neurochem Res. 2019;44:1306–22.
Arida RM, Teixeira-Machado L. The contribution of physical exercise to brain resilience. Front Behav Neurosci. 2021;14:626769.
Carek PJ, Laibstain SE, Carek SM. Exercise for the treatment of depression and anxiety. Int J Psychiatry Med. 2011;41:15–28.
da Cunha LL, Feter N, Alt R, Rombaldi AJ. Effects of exercise training on inflammatory, neurotrophic and immunological markers and neurotransmitters in people with depression: A systematic review and meta-analysis. J Affect Disord. 2023;326:73–82.
Nimmo MA, Leggate M, Viana JL, King JA. The effect of physical activity on mediators of inflammation. Diabetes Obes Metabolism. 2013;15:51–60.
Rethorst CD, Greer TL, Toups MSP, Bernstein I, Carmody TJ, Trivedi MH. IL-1β and BDNF are associated with improvement in hypersomnia but not insomnia following exercise in major depressive disorder. Translational Psychiatry. 2015;5:e611–e.
Daniela M, Catalina L, Ilie O, Paula M, Daniel-Andrei I, Ioana B. Effects of exercise training on the autonomic nervous system with a focus on Anti-Inflammatory and antioxidants effects. Antioxidants. 2022;11:350.
Hassamal S. Chronic stress, neuroinflammation, and depression: an overview of pathophysiological mechanisms and emerging anti-inflammatories. Front Psychiatry. 2023;14:1130989.
Kang J, Wang D, Duan Y, Zhai L, Shi L, Guo F. Aerobic exercise prevents depression via alleviating hippocampus injury in chronic stressed depression rats. Brain Sci. 2021;11:9.
Sallam N, Laher I. Exercise modulates oxidative stress and inflammation in aging and cardiovascular diseases. Oxidative Med Cell Longev. 2016;2016:7239639.
Rethorst CD, Toups MS, Greer TL, Nakonezny PA, Carmody TJ, Grannemann BD, et al. Pro-inflammatory cytokines as predictors of antidepressant effects of exercise in major depressive disorder. Mol Psychiatry. 2013;18:1119–24.
García-Juárez M, Camacho-Morales A. Defining the role of Anti- and Pro-inflammatory outcomes of Interleukin-6 in mental health. Neuroscience. 2022;492:32–46.
Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo MA. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat Rev Immunol. 2011;11:607–15.
Petersen AMW, Pedersen BK. The anti-inflammatory effect of exercise. J Appl Physiol. 2005;98:1154–62.
Leal LG, Lopes MA, Batista ML Jr. Physical exercise-induced myokines and muscle-adipose tissue crosstalk: a review of current knowledge and the implications for health and metabolic diseases. Front Physiol. 2018;9:1307.
Islam H, Neudorf H, Mui AL, Little JP. Interpreting ‘anti-inflammatory’ cytokine responses to exercise: focus on interleukin-10. J Physiol. 2021;599:5163–77.
Handschin C, Choi CS, Chin S, Kim S, Kawamori D, Kurpad AJ, et al. Abnormal glucose homeostasis in skeletal muscle–specific PGC-1α knockout mice reveals skeletal muscle–pancreatic Β cell crosstalk. J Clin Investig. 2007;117:3463–74.
Neto IVS, Pinto AP, Muñoz VR, de Cássia Marqueti R, Pauli JR, Ropelle ER, et al. Pleiotropic and multi-systemic actions of physical exercise on PGC-1α signaling during the aging process. Ageing Res Rev. 2023;87:101935.
Agudelo LZ, Femenía T, Orhan F, Porsmyr-Palmertz M, Goiny M, Martinez-Redondo V, et al. Skeletal muscle PGC-1α1 modulates kynurenine metabolism and mediates resilience to stress-induced depression. Cell. 2014;159:33–45.
Gupta E, Mishra P. Functional food with some health benefits, so called superfood: A review. Curr Nutr Food Sci. 2021;17:144–66.
de Araújo FF, de Paulo Farias D, Neri-Numa IA, Pastore GM. Polyphenols and their applications: an approach in food chemistry and innovation potential. Food Chem. 2021;338:127535.
Rasouli H, Farzaei MH, Khodarahmi R. Polyphenols and their benefits: A review. Int J Food Prop. 2017;20:1700–41.
Auad P, Spier F, Gutterres M. Vegetable tannin composition and its association with the leather tanning effect. Chem Eng Commun. 2020;207:722–32.
Metsämuuronen S, Sirén H. Bioactive phenolic compounds, metabolism and properties: a review on valuable chemical compounds in Scots pine and Norway Spruce. Phytochem Rev. 2019;18:623–64.
Shen N, Wang T, Gan Q, Liu S, Wang L, Jin B. Plant flavonoids: classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022;383:132531.
Kumar N, Goel N. Phenolic acids: natural versatile molecules with promising therapeutic applications. Biotechnol Rep. 2019;24:e00370.
Rodríguez-García C, Sánchez-Quesada C, Toledo E, Delgado-Rodríguez M, Gaforio JJ. Naturally Lignan-Rich Foods: A Dietary Tool for Health Promotion? Journal [serial on the Internet]. 2019; 24(5).
Oluwole O, Fernando WB, Lumanlan J, Ademuyiwa O, Jayasena V. Role of phenolic acid, tannins, Stilbenes, lignans and flavonoids in human health– a review. Int J Food Sci Technol. 2022;57:6326–35.
Smeriglio A, Barreca D, Bellocco E, Trombetta D. Proanthocyanidins and hydrolysable tannins: occurrence, dietary intake and Pharmacological effects. Br J Pharmacol. 2017;174:1244–62.
Haslam E, Cai Y. Plant polyphenols (vegetable tannins): Gallic acid metabolism. Nat Prod Rep. 1994;11:41–66.
Trebatická J, Ďuračková Z. Psychiatric disorders and polyphenols: can they be helpful in therapy? Oxid Med Cell Longev. 2015;2015:248529.
Asadi S, Gholami MS, Siassi F, Qorbani M, Sotoudeh G. Beneficial effects of nano-curcumin supplement on depression and anxiety in diabetic patients with peripheral neuropathy: A randomized, double-blind, placebo-controlled clinical trial. Phytother Res. 2020;34:896–903.
Dimpfel W. Rat electropharmacograms of the flavonoids Rutin and Quercetin in comparison to those of moclobemide and clinically used reference drugs suggest antidepressive and/or neuroprotective action. Phytomedicine. 2009;16:287–94.
Wang R, Xu Y, Wu H-L, Li Y-B, Li Y-H, Guo J-B, et al. The antidepressant effects of Curcumin in the forced swimming test involve 5-HT1 and 5-HT2 receptors. Eur J Pharmacol. 2008;578:43–50.
Dreiseitel A, Korte G, Schreier P, Oehme A, Locher S, Domani M, et al. Berry anthocyanins and their Aglycons inhibit monoamine oxidases A and B. Pharmacol Res. 2009;59:306–11.
Topaz GR, March A, Smith VE, Stieglitz KA. Inhibition of monoamine oxidase (MAO) via Green tea extracts: structural insights of Catechins as potential inhibitors of MAO. Frontiers in Natural Product Chemistry: Volume 5: Bentham Science Publishers; 2019. pp. 1–38.
Ahmad S, Azhar A, Tikmani P, Rafique H, Khan A, Mesiya H et al. A randomized clinical trial to test efficacy of chamomile and saffron for neuroprotective and anti-inflammatory responses in depressive patients. Heliyon. 2022;8.
Abd-Rabo MM, Georgy GS, Saied NM, Hassan WA. Involvement of the serotonergic system and neuroplasticity in the antidepressant effect of Curcumin in ovariectomized rats: comparison with oestradiol and Fluoxetine. Phytother Res. 2019;33:387–96.
Oh D-R, Choi C, Kim MJ, Mun BY, Ko H, Oh K-N, et al. Antidepressant effects of p-coumaric acid isolated from vaccinium bracteatum leaves extract on chronic restraint stress mouse model and antagonism of serotonin 6 receptor in vitro. Phytomedicine. 2023;116:154871.
Oh D-R, Yoo J-S, Kim Y, Kang H, Lee H, Lm SJ et al. Vaccinium bracteatum leaf extract reverses chronic restraint Stress-Induced Depression-Like behavior in mice: regulation of Hypothalamic-Pituitary-Adrenal axis, serotonin turnover systems, and ERK/Akt phosphorylation. Front Pharmacol. 2018;9.
Xu Y, Ku B, Cui L, Li X, Barish PA, Foster TC, et al. Curcumin reverses impaired hippocampal neurogenesis and increases serotonin receptor 1A mRNA and brain-derived neurotrophic factor expression in chronically stressed rats. Brain Res. 2007;1162:9–18.
Bortolotto VC, Pinheiro FC, Araujo SM, Poetini MR, Bertolazi BS, de Paula MT, et al. Chrysin reverses the depressive-like behavior induced by hypothyroidism in female mice by regulating hippocampal serotonin and dopamine. Eur J Pharmacol. 2018;822:78–84.
Antunes MS, Cattelan Souza L, Ladd FVL, Ladd AABL, Moreira AL, Bortolotto VC, et al. Hesperidin ameliorates Anxiety-Depressive-Like behavior in 6-OHDA model of Parkinson’s disease by regulating striatal cytokine and neurotrophic factors levels and dopaminergic innervation loss in the striatum of mice. Mol Neurobiol. 2020;57:3027–41.
Xu Y, Wang Z, You W, Zhang X, Li S, Barish PA, et al. Antidepressant-like effect of trans-resveratrol: involvement of serotonin and noradrenaline system. Eur Neuropsychopharmacol. 2010;20:405–13.
da Silva Marques JG, Antunes FTT, da Silva Brum LF, Pedron C, de Oliveira IB, de Barros Falcão Ferraz A, et al. Adaptogenic effects of Curcumin on depression induced by moderate and unpredictable chronic stress in mice. Behav Brain Res. 2021;399:113002.
Fu H, Liu L, Tong Y, Li Y, Zhang X, Gao X, et al. The antidepressant effects of hesperidin on chronic unpredictable mild stress-induced mice. Eur J Pharmacol. 2019;853:236–46.
Madiha S, Haider S. Curcumin restores rotenone induced depressive-like symptoms in animal model of neurotoxicity: assessment by social interaction test and sucrose preference test. Metab Brain Dis. 2019;34:297–308.
Shen J, Qu C, Xu L, Sun H, Zhang J. Resveratrol exerts a protective effect in chronic unpredictable mild stress–induced depressive-like behavior: involvement of the AKT/GSK3β signaling pathway in hippocampus. Psychopharmacology. 2019;236:591–602.
Chen J-J, Shen J-X, Yu Z-H, Pan C, Han F, Zhu X-L, et al. The antidepressant effects of Resveratrol are accompanied by the Attenuation of dendrite/dendritic spine loss and the upregulation of BDNF/p-cofilin1 levels in chronic restraint mice. Neurochem Res. 2021;46:660–74.
Jia Z, Yang J, Cao Z, Zhao J, Zhang J, Lu Y, et al. Baicalin ameliorates chronic unpredictable mild stress-induced depression through the BDNF/ERK/CREB signaling pathway. Behav Brain Res. 2021;414:113463.
Shukla P, Akotkar L, Aswar U. Resveratrol attenuates early life stress induced depression in rats: behavioural and neurochemical evidence. Neurosci Lett. 2024;820:137606.
Zheng X, Cheng Y, Chen Y, Yue Y, Li Y, Xia S, et al. Ferulic acid improves Depressive-Like behavior in Prenatally-Stressed offspring rats via Anti-Inflammatory activity and HPA axis. Int J Mol Sci. 2019;20:493.
Platero JL, Cuerda-Ballester M, Sancho-Cantus D, Benlloch M, Ceron JJ, Peres Rubio C, et al. The impact of Epigallocatechin gallate and coconut oil treatment on cortisol activity and depression in multiple sclerosis patients. Life. 2021;11:353.
Tsang C, Hodgson L, Bussu A, Farhat G, Al-Dujaili E. Effect of Polyphenol-Rich dark chocolate on salivary cortisol and mood in adults. Antioxidants. 2019;8:149.
Yang W, Cui K, Li X, Zhao J, Zeng Z, Song R, et al. Effect of polyphenols on cognitive function: evidence from Population-based studies and clinical trials. J Nutr Health Aging. 2021;25:1190–204.
Farag S, Tsang C, Al-Dujaili EAS, Murphy PN. Effect of polyphenol supplementation on memory functioning in overweight and obese adults: A systematic review and Meta-Analysis. Nutrients. 2024;16:474.
Rodrigo-Gonzalo MJ, González-Manzano S, Mendez-Sánchez R, Santos-Buelga C, Recio-Rodríguez JI. Effect of Polyphenolic Complements on Cognitive Function in the Elderly: A Systematic Review. Journal [serial on the Internet]. 2022; 11(8).
Wang J, Wang S, Guo H, Li Y, Jiang Z, Gu T, et al. Rosmarinic acid protects rats against post-stroke depression after transient focal cerebral ischemic injury through enhancing antioxidant response. Brain Res. 2021;1757:147336.
Tayab MA, Islam MN, Chowdhury KAA, Tasnim FM. Targeting neuroinflammation by polyphenols: A promising therapeutic approach against inflammation-associated depression. Biomed Pharmacother. 2022;147:112668.
Zhou N, Gu X, Zhuang T, Xu Y, Yang L, Zhou M. Gut microbiota: A pivotal hub for polyphenols as antidepressants. J Agric Food Chem. 2020;68:6007–20.
Hao W, Gan H, Wang L, Huang J, Chen J. Polyphenols in edible herbal medicine: targeting gut-brain interactions in depression-associated neuroinflammation. Crit Rev Food Sci Nutr. 2023;63:12207–23.
Ghalandari-Shamami M, Nourizade S, Yousefi B, Vafaei AA, Pakdel R, Rashidy-Pour A. Beneficial effects of physical activity and Crocin against adolescent stress induced anxiety or Depressive-Like symptoms and dendritic morphology remodeling in prefrontal cortex in adult male rats. Neurochem Res. 2019;44:917–29.
Abdallah HM, El-Gohary RM, Khattab H, Farghal EE, Ghabrial MM, Hashad AF. Potential synergestic effect of Quercetin and exercise on depressive like behavior in male albino rats: targeting oxidative stress, inflammation, neural apoptosis and mitophagy. Bull Egypt Soc Physiological Sci. 2024;44:54–68.
Azarian F, Farsi S, Hosseini SA, Azarbayjani MA. The effect of endurance training and Crocin consumption on Anxiety-like behaviors and aerobic power in rats with Alzheimer’s. Iran J Psychiatry Behav Sci. 2019;13:e89011.
Noruzi S, Meshkati Z, Badami R, Nasiri R. The effect of high intensity interval training (HIIT) and dietary supplement Curcumin on cognitive function and the level of stress markers in the brain of male Balb/C mice exposed to lead nitrate. Motor Behav. 2023;15:69–100.
Amoei M, Meshkati Z, Nasiri R, Dakhili A. The effect of aerobic exercise and Curcumin diet supplement on anxiety behavior and locomotor activity of female Balb/C mice exposed to lead nitrate using the Hole-Board test. Armaghane Danesh. 2021;26:596–610.
Nourolapour S, Abbassi Daloii A, Ziaolhagh SJ, Barari A. The effect of aerobic exercise and Crocin consumption on the gene expression of BDNF, TrKB, dopamine, and serotonin in the cerebral cortex of rats induced with methamphetamine. J Sport Exerc Physiol. 2024;17.
Osali A, Rostami A. Effect of 6 weeks of aerobic training with Nanocurcumin consumption on IL1β, nitric oxide, and depression in women with metabolic syndrome. Int J Diabetes Developing Ctries. 2023;43:1007–14.
Moghadam BH, Bagheri R, Roozbeh B, Ashtary-Larky D, Gaeini AA, Dutheil F, et al. Impact of saffron (Crocus sativus Linn) supplementation and resistance training on markers implicated in depression and happiness levels in untrained young males. Physiol Behav. 2021;233:113352.
Johnston NJ. The Effects of Combined Curcumin Supplementation and High Intensity Interval Training on Time Trial Performance in Moderately Active Individuals. 2019.
Gangisetty O, Murugan S. Epigenetic modifications in neurological diseases: natural products as epigenetic modulators a treatment strategy. In: Essa MM, Akbar M, Guillemin G, editors. The benefits of natural products for neurodegenerative diseases. Cham: Springer International Publishing; 2016. pp. 1–25.
Prasanth MI, Sivamaruthi BS, Cheong CS, Verma K, Tencomnao T, Brimson JM et al. Role of Epigenetic Modulation in Neurodegenerative Diseases: Implications of Phytochemical Interventions. Journal [serial on the Internet]. 2024 Date; 13(5).
Langfort J, Barańczuk E, Pawlak D, Chalimoniuk M, Lukačova N, Maršala J, et al. The effect of endurance training on regional serotonin metabolism in the brain during early stage of detraining period in the female rat. Cell Mol Neurobiol. 2006;26:1325–40.
Hiraga T, Hata T, Soya S, Shimoda R, Takahashi K, Soya M, et al. Light-exercise-induced dopaminergic and noradrenergic stimulation in the dorsal hippocampus: using a rat physiological exercise model. FASEB J. 2024;38:e70215.
Mohammed HS, Khadrawy YA, El-Sherbini TM, Amer HM. Electrocortical and biochemical evaluation of antidepressant efficacy of formulated Nanocurcumin. Appl Biochem Biotechnol. 2019;187:1096–112.
Gu Z, Chu L, Han Y. Therapeutic effect of Resveratrol on mice with depression. Exp Ther Med. 2019;17:3061–4.
Scheffer DL, Latini A. Exercise-induced immune system response: Anti-inflammatory status on peripheral and central organs. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165823.
Velásquez-Jiménez D, Corella-Salazar DA, Zuñiga-Martínez BS, Domínguez-Avila JA, Montiel-Herrera M, Salazar-López NJ, et al. Phenolic compounds that cross the blood–brain barrier exert positive health effects as central nervous system antioxidants. Food Funct. 2021;12:10356–69.
Powers SK, Goldstein E, Schrager M, Ji LL. Exercise Training and Skeletal Muscle Antioxidant Enzymes: An Update. Journal [serial on the Internet]. 2023 Date; 12(1).
Bajpai VK, Alam MB, Ju M-K, Kwon K-R, Huh YS, Han Y-K, et al. Antioxidant mechanism of polyphenol-rich nymphaea Nouchali leaf extract protecting DNA damage and attenuating oxidative stress-induced cell death via Nrf2-mediated heme-oxygenase-1 induction coupled with ERK/p38 signaling pathway. Biomed Pharmacother. 2018;103:1397–407.
Jodeiri Farshbaf M, Ghaedi K, Megraw TL, Curtiss J, Shirani Faradonbeh M, Vaziri P, et al. Does PGC1α/FNDC5/BDNF elicit the beneficial effects of exercise on neurodegenerative disorders?? Neuromol Med. 2016;18:1–15.
Taylor DF, Bishop DJ. Transcription Factor Movement and Exercise-Induced Mitochondrial Biogenesis in Human Skeletal Muscle: Current Knowledge and Future Perspectives. Journal [serial on the Internet]. 2022 Date; 23(3).
Sandoval-Acuña C, Ferreira J, Speisky H. Polyphenols and mitochondria: an update on their increasingly emerging ROS-scavenging independent actions. Arch Biochem Biophys. 2014;559:75–90.
Zádori D, Klivényi P, Szalárdy L, Fülöp F, Toldi J, Vécsei L. Mitochondrial disturbances, excitotoxicity, neuroinflammation and kynurenines: novel therapeutic strategies for neurodegenerative disorders. J Neurol Sci. 2012;322:187–91.
Martin KS, Azzolini M, Lira Ruas J. The kynurenine connection: how exercise shifts muscle Tryptophan metabolism and affects energy homeostasis, the immune system, and the brain. Am J Physiology-Cell Physiol. 2020;318:C818–30.
Joisten N, Walzik D, Metcalfe AJ, Bloch W, Zimmer P. Physical exercise as kynurenine pathway modulator in chronic diseases: implications for immune and energy homeostasis. Int J Tryptophan Res. 2020;13:1178646920938688.
Mallik SB, Mudgal J, Kinra M, Hall S, Grant GD, Anoopkumar-Dukie S, et al. Involvement of indoleamine 2, 3-dioxygenase (IDO) and brain-derived neurotrophic factor (BDNF) in the neuroprotective mechanisms of ferulic acid against depressive-like behaviour. Metab Brain Dis. 2023;38:2243–54.
Gualdoni GA, Fuchs D, Zlabinger GJ, Gostner JM. Resveratrol intake enhances indoleamine-2,3-dioxygenase activity in humans. Pharmacol Rep. 2016;68:1065–8.
Hajiluian G, Karegar SJ, Shidfar F, Aryaeian N, Salehi M, Lotfi T, et al. The effects of ellagic acid supplementation on neurotrophic, inflammation, and oxidative stress factors, and indoleamine 2, 3-dioxygenase gene expression in multiple sclerosis patients with mild to moderate depressive symptoms: A randomized, triple-blind, placebo-controlled trial. Phytomedicine. 2023;121:155094.
Carter T, Guo B, Turner D, Morres I, Khalil E, Brighton E, et al. Preferred intensity exercise for adolescents receiving treatment for depression: a pragmatic randomised controlled trial. BMC Psychiatry. 2015;15:247.
Crews DJ, Lochbaum MR, Landers DM. Aerobic physical activity effects on psychological well-being in low-income Hispanic children. Percept Mot Skills. 2004;98:319–24.
Daley AJ, Copeland RJ, Wright NP, Roalfe A, Wales JKH. Exercise therapy as a treatment for psychopathologic conditions in obese and morbidly obese adolescents: A randomized, controlled trial. Pediatrics. 2006;118:2126–34.
Goldfield GS, Kenny GP, Alberga AS, Prud’homme D, Hadjiyannakis S, Gougeon R, et al. Effects of aerobic training, resistance training, or both on psychological health in adolescents with obesity: the HEARTY randomized controlled trial. J Consult Clin Psychol. 2015;83:1123.
Hughes CW, Barnes S, Barnes C, DeFina LF, Nakonezny P, Emslie GJ. Depressed adolescents treated with exercise (DATE): A pilot randomized controlled trial to test feasibility and Establish preliminary effect sizes. Ment Health Phys Act. 2013;6:119–31.
Jeong Y-J, Hong S-C, Lee MS, Park M-C, Kim Y-K, Suh C-M, DANCE MOVEMENT THERAPY IMPROVES EMOTIONAL RESPONSES AND MODULATES NEUROHORMONES IN ADOLESCENTS WITH MILD DEPRESSION. Int J Neurosci. 2005;115:1711–20.
Petty KH, Davis CL, Tkacz J, Young-Hyman D, Waller JL. Exercise effects on depressive symptoms and Self-Worth in overweight children: A randomized controlled trial**. J Pediatr Psychol. 2009;34:929–39.
Zhao Y, Wang W, Wang M, Gao F, Hu C, Cui B et al. Personalized individual-based exercise prescriptions are effective in treating depressive symptoms of college students during the COVID-19: A randomized controlled trial in China. Front Psychiatry. 2023;13.
Chang J, Zhu W, Zhang J, Yong L, Yang M, Wang J et al. The effect of Chinese square dance exercise on cognitive function in older women with mild cognitive impairment: the mediating effect of mood status and quality of life. Front Psychiatry. 2021;12.
Wang L, Wu B, Tao H, Chai N, Zhao X, Zhen X, et al. Effects and mediating mechanisms of a structured limbs-exercise program on general cognitive function in older adults with mild cognitive impairment: A randomized controlled trial. Int J Nurs Stud. 2020;110:103706.
Lazarou I, Parastatidis T, Tsolaki A, Gkioka M, Karakostas A, Douka S, et al. International ballroom dancing against neurodegeneration: A randomized controlled trial in Greek Community-Dwelling elders with mild cognitive impairment. Am J Alzheimer’s Disease Other Dementias®. 2017;32:489–99.
Zhu Y, Wu H, Qi M, Wang S, Zhang Q, Zhou L, et al. Effects of a specially designed aerobic dance routine on mild cognitive impairment. Clin Interv Aging. 2018;13:1691–700.
Tremont G, Davis J, Ott BR, Uebelacker L, Kenney L, Gillette T, et al. Feasibility of a yoga intervention for individuals with mild cognitive impairment: A randomized controlled trial. J Integr Complement Med. 2022;28:250–60.
Eyre HA, Siddarth P, Acevedo B, Van Dyk K, Paholpak P, Ercoli L, et al. A randomized controlled trial of Kundalini yoga in mild cognitive impairment. Int Psychogeriatr. 2017;29:557–67.
Yang H, Leaver AM, Siddarth P, Paholpak P, Ercoli L, St. Cyr NM et al. Neurochemical and neuroanatomical plasticity following memory training and yoga interventions in older adults with mild cognitive impairment. Front Aging Neurosci. 2016;8.
Yu DJ, Yu AP, Bernal JDK, Fong DY, Chan DKC, Cheng CP et al. Effects of exercise intensity and frequency on improving cognitive performance in middle-aged and older adults with mild cognitive impairment: A pilot randomized controlled trial on the minimum physical activity recommendation from WHO. Front Physiol. 2022;13.
Barzroodi pour M, Bayat M, Golab F, Eftekharzadeh M, Katebi M, Soleimani M, et al. The effect of exercise on GABA signaling pathway in the model of chemically induced seizures. Life Sci. 2019;232:116667.
Clark PJ, Amat J, McConnell SO, Ghasem PR, Greenwood BN, Maier SF, et al. Running reduces uncontrollable Stress-Evoked serotonin and potentiates Stress-Evoked dopamine concentrations in the rat dorsal striatum. PLoS ONE. 2015;10:e0141898.
Kim T-K, Kim J-E, Park J-Y, Lee J-E, Choi J, Kim H, et al. Antidepressant effects of exercise are produced via suppression of hypocretin/orexin and melanin-concentrating hormone in the basolateral amygdala. Neurobiol Dis. 2015;79:59–69.
Messina G, Di Bernardo G, Viggiano A, De Luca V, Monda V, Messina A et al. Exercise increases the level of plasma orexin A in humans. 2016;27:611-6.
Martínez-Damas MG, Genis-Mendoza AD, Cruz VP-dl, Canela-Tellez GD, Jiménez-Estrada I, Sanchez JHN, et al. Epicatechin treatment generates resilience to chronic mild stress-induced depression in a murine model through a modulatory effect on KAT. Physiol Behav. 2021;238:113466.
Sharma P, Sharma S, Singh D. Apigenin reverses behavioural impairments and cognitive decline in kindled mice via CREB-BDNF upregulation in the hippocampus. Nutr Neurosci. 2020;23:118–27.
Bijani S, Dizaji R, Sharafi A, Hosseini M-J. Neuroprotective effect of apigenin on Depressive-Like behavior: mechanistic approach. Neurochem Res. 2022;47:644–55.
Du Y, Li W, Li Y, Yang J, Wang X, Yin S, et al. Repeated Arctigenin treatment produces antidepressant- and anxiolytic-like effects in mice. Brain Res Bull. 2019;146:79–86.
Yao Y, Man L, Du J, Wu D, Yang L, Peng F, et al. Astilbin ameliorates depressive-like behavior caused by postnatal immune activation through Menin-regulated astrocyte inflammation. J Affect Disord. 2022;301:87–98.
Singh N, Bansal Y, Bhandari R, Marwaha L, Singh R, Chopra K, et al. Naringin reverses neurobehavioral and biochemical alterations in intracerebroventricular Collagenase-Induced intracerebral hemorrhage in rats. Pharmacology. 2017;100:172–87.
Achour M, Ferdousi F, Sasaki K, Isoda H. Luteolin modulates neural stem cells fate determination: in vitro study on human neural stem cells, and in vivo study on LPS-Induced depression mice model. Front Cell Dev Biology. 2021;9.
Tana NT. Luteolin ameliorates depression-like behaviors by suppressing ER stress in a mouse model of Alzheimer’s disease. Biochem Biophys Res Commun. 2022;588:168–74.
Parashar A, Mehta V, Udayabanu M. Rutin alleviates chronic unpredictable stress-induced behavioral alterations and hippocampal damage in mice. Neurosci Lett. 2017;656:65–71.
Olugbemide AS, Ben-Azu B, Bakre AG, Ajayi AM, Femi-Akinlosotu O, Umukoro S. Naringenin improves depressive- and anxiety-like behaviors in mice exposed to repeated hypoxic stress through modulation of oxido-inflammatory mediators and NF-kB/BDNF expressions. Brain Res Bull. 2021;169:214–27.
Bansal Y, Singh R, Saroj P, Sodhi RK, Kuhad A. Naringenin protects against oxido-inflammatory aberrations and altered Tryptophan metabolism in olfactory bulbectomized-mice model of depression. Toxicol Appl Pharmcol. 2018;355:257–68.
Huang X, Li W, You B, Tang W, Gan T, Feng C, et al. Serum metabonomic study on the Antidepressant-like effects of ellagic acid in a chronic unpredictable mild Stress-Induced mouse model. J Agric Food Chem. 2020;68:9546–56.
Sasaki K, Iwata N, Ferdousi F, Isoda H. Antidepressant-Like effect of ferulic acid via promotion of energy metabolism activity. Mol Nutr Food Res. 2019;63:1900327.
Wang Y, Wang B, Lu J, Shi H, Gong S, Wang Y, et al. Fisetin provides antidepressant effects by activating the Tropomyosin receptor kinase B signal pathway in mice. J Neurochem. 2017;143:561–8.
Can ÖD, Turan N, Demir Özkay Ü, Öztürk Y. Antidepressant-like effect of Gallic acid in mice: dual involvement of serotonergic and catecholaminergic systems. Life Sci. 2017;190:110–7.
Abbaszade-Cheragheali A, Beheshti F, Kakhki S, Khatibi SR, Dehnokhalaji F, Akbari E, et al. Crocin, the main active saffron (Crocus sativus L.) constituent, as a potential candidate to prevent anxiety and depressive-like behaviors induced by unpredictable chronic mild stress. Neurosci Lett. 2022;791:136912.
Xiao Q, Shu R, Wu C, Tong Y, Xiong Z, Zhou J, et al. Crocin-I alleviates the depression-like behaviors probably via modulating microbiota-gut-brain axis in mice exposed to chronic restraint stress. J Affect Disord. 2020;276:476–86.
Zhang L, Previn R, Lu L, Liao R-F, Jin Y, Wang R-K. Crocin, a natural product attenuates lipopolysaccharide-induced anxiety and depressive-like behaviors through suppressing NF-kB and NLRP3 signaling pathway. Brain Res Bull. 2018;142:352–9.
Xie Y, He Q, Chen H, Lin Z, Xu Y, Yang C. Crocin ameliorates chronic obstructive pulmonary disease-induced depression via PI3K/Akt mediated suppression of inflammation. Eur J Pharmacol. 2019;862:172640.
Wang Y, Zhou S, Song X, Ding S, Wang B, Wen J, et al. Study on antidepressant effect and mechanism of Crocin mediated by the mTOR signaling pathway. Neurochem Res. 2022;47:3126–36.
Fang K, Li H-R, Chen X-X, Gao X-R, Huang L-L, Du A-Q et al. Quercetin alleviates LPS-Induced Depression-Like behavior in rats via regulating BDNF-Related imbalance of Copine 6 and TREM1/2 in the hippocampus and PFC. Front Pharmacol. 2020;10.
Bicca DF, Spiazzi CC, Ramalho JB, Soares MB, Cibin FWS. A subchronic low-dose exposure of a glyphosate-based herbicide induces depressive and anxious-like behavior in mice: Quercetin therapeutic approach. Environ Sci Pollut Res. 2021;28:67394–403.
Guan Y, Wang J, Wu X, Song L, Wang Y, Gong M, et al. Quercetin reverses chronic unpredictable mild stress-induced depression-like behavior in vivo by involving nuclear factor-E2-related factor 2. Brain Res. 2021;1772:147661.
Sun Y, Zhang H, Wu Z, Yu X, Yin Y, Qian S, et al. Quercitrin rapidly alleviated Depression-like behaviors in Lipopolysaccharide-Treated mice: the involvement of PI3K/AKT/NF-κB signaling suppression and CREB/BDNF signaling restoration in the hippocampus. ACS Chem Neurosci. 2021;12:3387–96.
Ugwu PI, Ben-Azu B, Ugwu SU, Uruaka CI, Nworgu CC, Okorie PO, et al. Preventive putative mechanisms involved in the psychopathologies of mice passively coping with psychosocial defeat stress by Quercetin. Brain Res Bull. 2022;183:127–41.
Donoso F, Egerton S, Bastiaanssen TFS, Fitzgerald P, Gite S, Fouhy F, et al. Polyphenols selectively reverse early-life stress-induced behavioural, neurochemical and microbiota changes in the rat. Psychoneuroendocrinology. 2020;116:104673.
Wang G, Li Y, Lei C, Lei X, Zhu X, Yang L, et al. Quercetin exerts antidepressant and cardioprotective effects in Estrogen receptor α-deficient female mice via BDNF-AKT/ERK1/2 signaling. J Steroid Biochem Mol Biol. 2021;206:105795.
Anggreini P, Ardianto C, Rahmadi M, Khotib J. Quercetin attenuates acute predator stress exposure-evoked innate fear and behavioral perturbation. 2019;30.
Ma Z-X, Zhang R-Y, Rui W-J, Wang Z-Q, Feng X. Quercetin alleviates chronic unpredictable mild stress-induced depressive-like behaviors by promoting adult hippocampal neurogenesis via FoxG1/CREB/ BDNF signaling pathway. Behav Brain Res. 2021;406:113245.
Ardianto C, Budiatin AS, Sumartha INB, Nurrahmi N, Rahmadi M, Khotib J. Resveratrol ameliorates physical and psychological stress-induced depressive-like behavior. J Basic Clin Physiol Pharmacol. 2021;32:335–40.
Fahim AT, Abd El-Fattah AA, Sadik NAH, Ali BM. Resveratrol and dimethyl fumarate ameliorate testicular dysfunction caused by chronic unpredictable mild stress-induced depression in rats. Arch Biochem Biophys. 2019;665:152–65.
Abd El-Fattah AA, Fahim AT, Sadik NAH, Ali BM. Resveratrol and dimethyl fumarate ameliorate depression-like behaviour in a rat model of chronic unpredictable mild stress. Brain Res. 2018;1701:227–36.
Finnell JE, Lombard CM, Melson MN, Singh NP, Nagarkatti M, Nagarkatti P, et al. The protective effects of Resveratrol on social stress-induced cytokine release and depressive-like behavior. Brain Behav Immun. 2017;59:147–57.
Yu Y-C, Li J, Zhang M, Pan J-C, Yu Y, Zhang J-B et al. Resveratrol improves Brain-Gut axis by regulation of 5-HT-Dependent signaling in the rat model of irritable bowel syndrome. Front Cell Neurosci. 2019;13.
Yazir Y, Demirtaş Şahin T, Furat Rençber S, Gacar G, Halbutoğulları ZS, Utkan T, et al. Restorative effect of Resveratrol on expression of endothelial and neuronal nitric oxide synthase in cavernous tissues of chronic unpredictable mild stress-exposed rats: an impact of inflammation. Int J Impot Res. 2018;30:318–26.
Chen W-J, Du J-K, Hu X, Yu Q, Li D-X, Wang C-N, et al. Protective effects of Resveratrol on mitochondrial function in the hippocampus improves inflammation-induced depressive-like behavior. Physiol Behav. 2017;182:54–61.
Kortam MA, Ali BM, Fathy N. The deleterious effect of stress-induced depression on rat liver: protective role of Resveratrol and dimethyl fumarate via inhibiting the MAPK/ERK/JNK pathway. J Biochem Mol Toxicol. 2021;35:e22627.
Hao W, Zhu X, Liu Z, Song Y, Wu S, Lu X, et al. Resveratrol alleviates aluminum-induced intestinal barrier dysfunction in mice. Environ Toxicol. 2022;37:1373–81.
Zhu X, Li W, Li Y, Xu W, Yuan Y, Zheng V, et al. The antidepressant- and anxiolytic-like effects of Resveratrol: involvement of phosphodiesterase-4D Inhibition. Neuropharmacology. 2019;153:20–31.
Fan C, Li Y, Lan T, Wang W, Mao X, Yu SY. Prophylactic treatment of Curcumin in a rat model of depression by attenuating hippocampal synaptic loss. Food Funct. 2021;12:11202–13.
Khadrawy YA, Hosny EN, Magdy M, Mohammed HS. Antidepressant effects of curcumin-coated iron oxide nanoparticles in a rat model of depression. Eur J Pharmacol. 2021;908:174384.
Fidelis EM, Savall ASP, da Luz Abreu E, Carvalho F, Teixeira FEG, Haas SE, et al. Curcumin-Loaded nanocapsules reverses the Depressant-Like behavior and oxidative stress induced by β-Amyloid in mice. Neuroscience. 2019;423:122–30.
Abd-Elhakim YM, Abdel-Motal SM, Malhat SM, Mostafa HI, Moselhy AAA, Beheiry RR, et al. Curcumin mitigates neurotoxic and neurobehavioral changes of gentamicin and sodium salicylate in rats by adjusting oxidative stress and apoptosis. Life Sci. 2021;265:118824.
Rubab S, Naeem K, Rana I, Khan N, Afridi M, Ullah I, et al. Enhanced neuroprotective and antidepressant activity of curcumin-loaded nanostructured lipid carriers in lipopolysaccharide-induced depression and anxiety rat model. Int J Pharm. 2021;603:120670.
Afzal A, Batool Z, Sadir S, Liaquat L, Shahzad S, Tabassum S, et al. Therapeutic potential of Curcumin in reversing the depression and associated pseudodementia via modulating stress hormone, hippocampal neurotransmitters, and BDNF levels in rats. Neurochem Res. 2021;46:3273–85.
Fan C, Song Q, Wang P, Li Y, Yang M, Yu SY. Neuroprotective effects of Curcumin on IL-1β-Induced neuronal apoptosis and Depression-Like behaviors caused by chronic stress in rats. Front Cell Neurosci. 2019;12.
Abu-Taweel GM, Rudayni HA. Curcumin ameliorated the mercuric chloride induced depression and anxiety in female mice offspring. Environ Res. 2022;204:112031.
Liao D, Lv C, Cao L, Yao D, Wu Y, Long M, et al. Curcumin attenuates chronic unpredictable mild stress-Induced Depressive-Like behaviors via restoring changes in oxidative stress and the activation of Nrf2 signaling pathway in rats. Oxidative Med Cell Longev. 2020;2020:9268083.
Qi X-J, Liu X-Y, Tang L-M-Y, Li P-F, Qiu F, Yang A-H. Anti-depressant effect of curcumin-loaded guanidine-chitosan thermo-sensitive hydrogel by nasal delivery. Pharm Dev Technol. 2020;25:316–25.
Guo L-T, Wang S-Q, Su J, Xu L-X, Ji Z-Y, Zhang R-Y, et al. Baicalin ameliorates neuroinflammation-induced depressive-like behavior through Inhibition of toll-like receptor 4 expression via the PI3K/AKT/FoxO1 pathway. J Neuroinflamm. 2019;16:95.
Liu L, Dong Y, Shan X, Li L, Xia B, Wang H. Anti-Depressive Effectiveness of Baicalin In Vitro and In Vivo. Journal [serial on the Internet]. 2019 Date; 24(2).
Xiao Z, Cao Z, Yang J, Jia Z, Du Y, Sun G, et al. Baicalin promotes hippocampal neurogenesis via the Wnt/β-catenin pathway in a chronic unpredictable mild stress-induced mouse model of depression. Biochem Pharmacol. 2021;190:114594.
Yu H, Zhang F, Guan X. Baicalin reverse depressive-like behaviors through regulation SIRT1-NF-kB signaling pathway in olfactory bulbectomized rats. Phytother Res. 2019;33:1480–9.
Fang A, Li Y, Wu X, Wu B, Zhang Y. Baicalin attenuates inflammatory pain associated depressive symptoms via Akt-mediated adult hippocampal neurogenesis. Metab Brain Dis. 2020;35:1085–93.
Zhang K, He M, Wang F, Zhang H, Li Y, Yang J et al. Revealing antidepressant mechanisms of Baicalin in hypothalamus through systems approaches in Corticosterone- induced depressed mice. Front NeuroSci. 2019;13.
Liu X, Liu C. Baicalin ameliorates chronic unpredictable mild stress-induced depressive behavior: involving the Inhibition of NLRP3 inflammasome activation in rat prefrontal cortex. Int Immunopharmacol. 2017;48:30–4.
Zhang C-Y-Y, Zeng M-J, Zhou L-P, Li Y-Q, Zhao F, Shang Z-Y, et al. Baicalin exerts neuroprotective effects via inhibiting activation of GSK3β/NF-κB/NLRP3 signal pathway in a rat model of depression. Int Immunopharmacol. 2018;64:175–82.
Lu Y, Sun G, Yang F, Guan Z, Zhang Z, Zhao J, et al. Baicalin regulates depression behavior in mice exposed to chronic mild stress via the Rac/LIMK/cofilin pathway. Biomed Pharmacother. 2019;116:109054.
Gao C, Du Q, Li W, Deng R, Wang Q, Xu A, et al. Baicalin modulates APPL2/Glucocorticoid receptor signaling cascade, promotes neurogenesis, and attenuates emotional and olfactory dysfunctions in chronic Corticosterone-Induced depression. Mol Neurobiol. 2018;55:9334–48.
Acknowledgements
None.
Funding
No funding.
Author information
Authors and Affiliations
Contributions
Shihong Jie, Aili Fu, Chuan Wang, and Sogand Rajabi helped in the investigation, design, writing, conception, and drafting of the manuscript. All authors approved the final version for paper. All authors reviewed the manuscript.”
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
N/A.
Consent for publication
N/A.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Jie, S., Fu, A., Wang, C. et al. A comprehensive review on the impact of polyphenol supplementation and exercise on depression and brain function parameters. Behav Brain Funct 21, 10 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12993-025-00273-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12993-025-00273-2