Next Article in Journal
A Comparison of Brain-State Dynamics across Common Anesthetic Agents in Male Sprague-Dawley Rats
Next Article in Special Issue
Rapid Detection of Brettanomyces bruxellensis in Wine by Polychromatic Flow Cytometry
Previous Article in Journal
Methodological Considerations in Development of UV Imaging for Characterization of Intra-Tumoral Injectables Using cAMP as a Model Substance
Previous Article in Special Issue
Lactobacillus paracasei KW3110 Prevents Inflammatory-Stress-Induced Mitochondrial Dysfunction in Mouse Macrophages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Therapeutic Role of Exercise and Probiotics in Stressful Brain Conditions

by
Ismael Martínez-Guardado
1,*,
Silvia Arboleya
2,
Francisco Javier Grijota
1,
Aleksandra Kaliszewska
3,
Miguel Gueimonde
2 and
Natalia Arias
1,3,4,5,*
1
BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
2
Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33003 Oviedo, Spain
3
Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK
4
Health Research Institute of the Principality of Asturias-ISPA, 33011 Oviedo, Spain
5
Institute of Neurosciences of the Principality of Asturias (INEUROPA), 33003 Oviedo, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(7), 3610; https://doi.org/10.3390/ijms23073610
Submission received: 28 February 2022 / Revised: 14 March 2022 / Accepted: 22 March 2022 / Published: 25 March 2022
(This article belongs to the Special Issue Stress Response Effectors and Strategies in Probiotics)

Abstract

:
Oxidative stress has been recognized as a contributing factor in aging and in the progression of multiple neurological disorders such as Parkinson’s disease, Alzheimer’s dementia, ischemic stroke, and head and spinal cord injury. The increased production of reactive oxygen species (ROS) has been associated with mitochondrial dysfunction, altered metal homeostasis, and compromised brain antioxidant defence. All these changes have been reported to directly affect synaptic activity and neurotransmission in neurons, leading to cognitive dysfunction. In this context two non-invasive strategies could be employed in an attempt to improve the aforementioned stressful brain status. In this regard, it has been shown that exercise could increase the resistance against oxidative stress, thus providing enhanced neuroprotection. Indeed, there is evidence suggesting that regular physical exercise diminishes BBB permeability as it reinforces antioxidative capacity, reduces oxidative stress, and has anti-inflammatory effects. However, the differential effects of different types of exercise (aerobic exhausted exercise, anaerobic exercise, or the combination of both types) and the duration of physical activity will be also addressed in this review as likely determinants of therapeutic efficacy. The second proposed strategy is related to the use of probiotics, which can also reduce some biomarkers of oxidative stress and inflammatory cytokines, although their underlying mechanisms of action remain unclear. Moreover, various probiotics produce neuroactive molecules that directly or indirectly impact signalling in the brain. In this review, we will discuss how physical activity can be incorporated as a component of therapeutic strategies in oxidative stress-based neurological disorders along with the augmentation of probiotics intake.

1. Introduction

Physical activity (PA) is a modifiable lifestyle factor associated with improved overall health, reduced premature mortality, and a demonstrable role in the prevention of several chronic conditions [1,2,3]. Exercise promotes brain health by supporting brain’s structural integrity and function, which may improve cognitive performance and counteract at least certain aspects of cognitive ageing.
Several longitudinal studies have suggested that maintaining regular PA is linked to a reduced risk of cognitive impairments in older adults. A meta-analysis of 15 prospective studies has found that both high and moderate-low levels of exercise exert significant protection against cognitive decline in non-demented older individuals [4]. Notably, a recent meta-analysis has demonstrated that habitual PA prevents hippocampal volumetric decreases, which occur over time in humans [5]. The hippocampus is the primary centre of learning and memory, and since age-related hippocampal atrophy is a strong predictor of future cognitive decline [6,7,8], these findings hold promising implications for combating cognitive decline through exercise-induced retention of hippocampal volume.
Moreover, there is evidence to suggest that exercise may have a protective role against the development and progression of neurodegenerative disorders such as Parkinson’s disease (PD) and Alzheimer’s disease (AD). In a recent meta-analysis, Santos-Lozano and colleagues have found that adhering to the international PA guidelines of ≥150 min/week of moderate-intense activity was associated with a 40% reduction of risk for the development of AD [9]. PA was also shown to have cognition-enhancing effects in individuals at high risk of developing AD and in those affected by the disease [10,11,12]. Similarly, a systematic review evaluating the effect of exercise on cognitive function in PD has found evidence to support the role of PA in improving cognition in both animal models and human patients [13].
On the other hand, over the last two decades, the gut microbiota has emerged as an important organ with key functions such as the training of host immunity, digesting food, regulating gut endocrine, and neurological functions [14,15]. Evidence continues to demonstrate that the gut microbiota is particularly implicated in brain physiology and behavior, affecting host mental health [16,17,18]. New advances on the techniques for its analyses have allowed for knowing deeply the gut microbiome on different human conditions (age- or healthy-related) [19,20,21]; however, a “gold standard” reference of a human gut microbiota composition is not yet known. What is known is that the imbalance of the gut microbiota composition or functions can affect physiological homeostasis and alter the signals from the gut to brain, negatively influencing brain health or vice versa [22,23]. Different factors can be affect the gut microbiota, exercise being one of them [24]. Additionally, the modulation of the microbiota for improving cognition has attracted a deal of attention in recent years. The strategy most widely studied to this end is the use of probiotic microorganisms, the so-called “psychobiotics”, selected as a biotherapeutic tool for the maintenance of correct brain function through the microbiota–gut–brain axis [22,25].
Despite the promising potential of PA or probiotics for mitigating cognitive and brain deficits resulting from ageing and neurodegeneration, the mechanisms by which these strategies exert their effects in humans are not fully understood. In this review, we will discuss the effects of PA on improving cerebrovascular function, promoting adult neurogenesis, and reducing neuroinflammation as potential mechanisms linking exercise and brain health. Furthermore, we will describe the current evidence of the beneficial effects of probiotics on brain health, focusing on brain oxidative stress and the human clinical trials carried out evaluating the effects of different bacterial strains on mental health.

2. The Effect of Exercise on Cerebrovascular Function

Cerebrovascular function is an important determinant of brain health. Apart from age and genetic liabilities, the major risk factors for dementia and AD are of vascular nature [26,27]. Whilst the mechanisms linking cardiovascular disorder (CVD) risk factors and neurodegeneration are unclear, there is evidence to suggest that exercise-induced amelioration of CVD factors may be beneficial in the prevention of neurodegenerative disorders and counteracting cognitive aging.

2.1. Cerebral Blood Flow and Angiogenesis

Cerebral blood flow (CBF) supplies the brain with oxygen and nutrients it relies on to function properly. A reduction in CBF, also known as hypoperfusion, is a hallmark of brain ageing—with every decade of life, the CBF has been estimated to diminish by 5% [28]. Hypoperfusion has also been identified as a key pathological feature in neurodegenerative disorders and a major contributing factor in the development of mild cognitive impairment (MCI) and AD [29,30,31]. A lower CBF has been linked to a higher rate of future cognitive deficits in both non-demented elderly individuals and AD patients [30,32,33,34,35]. Notably, in healthy individuals, the association between age and CBF is influenced by cardiorespiratory fitness levels [36], with evidence suggesting that aerobic exercise can mitigate the effect of age on CBF and improve cognition in the healthy ageing population [37,38]. Similarly, engaging in a 12-month aerobic exercise programme was shown to improve CBF in older individuals with MCI, with the magnitude of improvement in the CBF marker directly correlated with improvements in memory function [39]. Whilst meta-analyses are currently lacking, the data from human studies linking aerobic exercise with improved CBF and cognitive performance is very encouraging. However, more long-term data is needed to determine if these beneficial effects persist and identify at which points the intervention is most efficacious.
A potential mechanism mediating the beneficial effects of exercise on cerebrovascular health and cognition is angiogenesis, i.e., the formation of new blood vessels. Exercise promotes the sprouting of new capillaries from pre-existing vessels, increasing capillary density [40]. This may be particularly beneficial in AD and PD, which are associated with a reduced density of microvessels in the brain [41,42]. Angiogenetic processes are regulated by vascular endothelial growth factors (VEGF)—a group of signalling molecules involved in the growth and maintenance of vascular and neuronal cells. Exercise promotes angiogenesis and increases the mRNA and protein levels of VEGF in both young and elderly individuals [43]. VEGF are produced primarily by myocytes and diffuse into the peripheral circuitry but have been shown to cross the blood–brain barrier (BBB) [44,45]. Therefore, it is likely that at least some of the positive effects of exercise are directly related to enhanced cerebral perfusion mediated by angiogenesis.
Indeed, a number of pre-clinical studies have demonstrated exercise-induced angiogenesis and provided evidence to support its cognitive-enhancing effects in-vivo. Prolonged PA—30 days of running on a wheel—was shown to promote angiogenesis in the motor cortex of rats as well as increase blood flow and volume as measured by functional magnetic resonance imaging [46]. Similarly, exercise-induced angiogenesis was reported in a mouse model of PD, with striatal (i.e., the brain region affected in PD) levels of angiogenetic markers VEGF and CD34 significantly increased following four weeks of treadmill training [47]. An increase in VEGF levels in response to exercise was also reported in aged rats [40]. Notably, in a TgCRND8 transgenic mouse model of AD amyloidosis, three months of running normalised hippocampal vascular morphology and preserved spatial memory [48]. Additionally, exercised-induced increase in muscle fibre VEGF levels was positively correlated with hippocampal-based memory and learning and VEGF levels in the hippocampus of rats [49]. Interestingly, VEGF have also been shown to directly enhance neurogenesis and synaptic function [50], which will be discussed in following sections.

2.2. BBB Integrity, Systemic Inflammation, and Clearance of Toxic Protein Aggregates

In the context of the prevention and slowing progression of PD and AD, it is important to note the effects of exercise on diminishing the permeability of BBB. The central nervous system (CNS) is separated from the peripheral tissues by BBB which regulates the entry of nutrients, molecules, and cells from the systemic circulation to the brain and vice-versa. It also plays a critical role in the clearance of cellular metabolites and toxins from the brain [51,52,53]. Disruption of BBB integrity (i.e., reduced barrier tightness and leakiness) occurs in ageing and neurodegeneration, leading to the infiltration of peripheral inflammatory mediators such as cytokines and macrophages and accumulation of toxic protein aggregates [53,54,55]. Loss of BBB integrity is associated with low-grade chronic inflammation, which affects a large proportion of the Western population [56,57] and has been implicated in the pathogenesis of neurodegenerative disorders [58]. In turn, the increase in inflammatory mediators entering the CNS following BBB dysregulation activates glial cells, triggering a second wave of inflammation and resulting in a vicious cycle of inflammation, leading to further damage of the BBB [26]. In this regard, exercise has been shown to exert neuroprotective effects by reducing systemic inflammation and therefore lowering the risk of infiltration into the CNS and improving the constitution of BBB tight junctions.
Regular PA has been reported to reduce the levels of pro-inflammatory cytokines, such as interleukin 6 (IL-6), tumour necrosis factor alpha (TNF-α), and monocyte chemoattractant protein-1 (MCP-1), which are associated with low-grade chronic inflammation [59,60,61,62], as well as increase the levels of anti-inflammatory cytokines [63]. Aranson and colleagues have demonstrated that in a group of healthy middle-aged individuals, the levels of C-reactive protein (CRP), a plasma biomarker of inflammation implicated in the pathogenesis of neurodegenerative disorders [64], decrease continuously with increased levels of physical fitness [65]. Furthermore, Chupel et al. have found combined taurine supplementation and exercise training to reduce levels of pro-inflammatory markers, improve BBB integrity, and increase cognitive performance in a group of elderly women [66]. Similar results have been seen in animal models—five weeks of running training improved the structural components of the BBB in diabetic rats, preserving the levels of claudin-5, a protein related to BBB integrity, and improving non-spatial memory [67]. Interestingly, exercise can also facilitate clearing of toxic metabolites such as amyloid beta (Aβ—the main component of amyloid plaques) by upregulating the expression of low-density lipoprotein receptor-related protein 1 (LRP-1) receptors on BBB, through which Aβ is transported out of the brain [68].
In conclusion, there is a large body of evidence to support the role of PA in maintaining cerebrovascular health. By increasing cerebral perfusion through angiogenesis, diminishing BBB permeability, reducing systemic inflammation, and aiding in the clearance of toxic protein aggregates, PA exerts neuroprotective effects that may counteract cognitive ageing and potentially prevent or delay the development of neurodegenerative disorders.

3. The Effect of Exercise on Neuroinflammation

In addition to the aforementioned effects of exercise on systemic inflammation, PA has also been shown to reduce neuroinflammation, which is a major feature of both AD/PD and ageing [69]. In the Tg2575 mouse model of AD, physically inactive mice were found to have a higher hippocampal expression of pro-inflammatory interleukins IL-1β and TNF-α, with reduced levels of interferon-gamma (IFN-γ) [70]. Three weeks of voluntary wheel running reduced the levels of IL-1β and TNF-α in the hippocampus and restored IFN-γ to wild-type levels, accompanied by a significant reduction in Aβ in the exercised group compared to sedentary animals [70]. PA has also been shown to mediate a phenotypic conversion of microglia, the primary immune cell in the CNS, from M1 (i.e., the classic “inflammatory” activation microglia sub-type) to M2 (i.e., the alternatively activated “neuroprotective” microglial sub-type) in a number of animal models [71,72,73] (see Figure 1). Jiang et al. [72] have shown that PA promoted the polarisation of microglia towards M2 phenotype and improved cognitive function in a rat model of chronic cerebral hypoperfusion. Similarly, in a streptozotocin (STZ)-induced rat model of AD, chronic treadmill exercise significantly inhibited reactive gliosis following STZ injection and shifted activated microglia from M1 phenotype to M2 [74]. This was accompanied by a reduction in pro-inflammatory markers and an upregulation of anti-inflammatory cytokine expression in the hippocampus of those animals as well as a significant preservation of hippocampal-dependent cognitive function [74]. Therefore, PA has the potential to modulate microglial phenotypes and promote anti-inflammatory effects in the brain, particularly the hippocampus, which at least in animal models appears to improve cognitive function.

4. The Effect of Exercise on Neurogenesis

Neurogenesis—the process of generating new functional neurons from neural stem and progenitor cells, is crucial for learning and memory function [75]. In the adult brain neurogenesis is largely restricted to two proliferative niches—the subventricular zone and the subgranular zone of the hippocampal dentate gyrus [76]. In animal models, ablation of hippocampal neurogenesis is associated with deficits in a range of hippocampus-dependent cognitive processes such as spatial memory, contextualised fear conditioning, and object recognition [77,78,79,80,81,82]. The rate of neurogenesis slows with age in both humans and laboratory animals [83,84,85,86,87], with impairments in the production of new neurons also reported in AD and PD as well as pre-clinical models of the diseases [88,89,90], thus suggesting that pro-neurogenic interventions may potentially mitigate cognitive deficits associated with ageing and neurodegeneration, which are likely to be translatable between pre-clinical models, and human conditions.
Exercise has been shown to promote neurogenesis in rodents with evidence suggesting the same may be true for humans [91,92,93]. Part of PA’s pro-neurogenic effects are thought to be due to its ability to reduce neuroinflammation, which is known to inhibit adult hippocampal neurogenesis (AHN) [94,95]. On the other hand, exercise-induced metabolic factors (i.e., lactate) and muscle-derived myokines (cathepsin-B and irisin) have been shown to stimulate the production of brain-derived neurotrophic factor (BDNF) [96,97,98,99,100,101,102], a growth factor involved in neuronal plasticity, synaptogenesis, and neurogenesis [103] (see Figure 1). In humans, the blood and brain levels of BDNF are reduced in AD and PD patients [104,105,106], with reports of a positive correlation between BDNF levels and cognitive performance [107,108]. Exercise is a potent stimulator of BDNF production—even a single session of intense PA has been shown to enhance the production of BDNF in both non-demented, healthy adults [109] and AD patients [109,110]. Similarly, a recent meta-analysis has demonstrated that PA interventions significantly elevate BDNF levels in PD patients, regardless of exercise type [111].
In rodents, wheel running is associated with an increase in mRNA and protein levels of BDNF within the hippocampus [92,112,113,114,115]. Furthermore, an elevation in hippocampal BDNF has been mechanistically linked to exercise-induced improvements in learning and memory mediated by the upregulation of signalling molecules cAMP response-element-binding protein (CREB) and synapsin I [116,117]. Indeed, inhibiting BDNF action in vivo blocks the beneficial effects of exercise on cognition—learning and memory function in exercised animals receiving BDNF blocker were reduced to sedentary control levels, highlighting the central role of BDNF in mediating exercise-induced cognitive benefits [118]. Notably, in a recent pre-clinical study, Choi and colleagues have found that inducing hippocampal neurogenesis via pharmacological or genetical means alone failed to improve cognitive function in a mouse model of AD [119]. In contrast, behavioural symptoms were ameliorated when hippocampal neurogenesis was induced by exercise and accompanied by an elevation in BDNF, interleukin 6 (IL-6), and synaptic markers. Interestingly, stimulating AHN in conjunction with the overexpression of BDNF mimicked exercised-induced improvements in cognition [119]. This suggests that promoting AHN can confer beneficial effects on cognition in AD, but only in the presence of a healthier brain environment optimal for the production of neurotrophins such as BDNF, which can be created through exercise, thus, further supporting the validity of PA as a non-invasive intervention with potent cognitive-enhancing effects. However, it is important to note that due to the inaccessibility of the human brain, most of what we know about the pro-neurogenic effects of exercise is drawn from animal models and indirect measures of human neurogenesis based on levels of BDNF and cognitive performance.

5. The Inhibition of Oxidative Stress through Exercise: The Influence of Different Exercise Type Programmes

Regular exercise is known to play an important preventive and therapeutic role in many diseases, as well as being beneficial to brain function [120,121,122]. Indeed, the increment of pumping and oxygenation of blood mediated by exercise creates an important environment to improve neuroprotective impact on brain function [123]. Moreover, previous studies have demonstrated that physical exercise (resistance and aerobic training) influence mitochondrial function and immune–inflammatory responses [124] through an important cellular process called mitochondria quality control [125]. This process includes the modification of mitochondrial proteins, mitochondrial dynamics, and autophagy [126], which leads to the control of the production of free radicals by creating antioxidant networks [127] (see Figure 2). As previously discussed, BDNF signalling through exercise plays a major role in neurogenesis, and the maintenance of cerebral BDNF levels is important for effective neural function [128]. However, it has been demonstrated that other neurotrophic factors such as vascular endothelial growth factor (VEGF) are involved in neurogenesis [129] (see Figure 2). In this line, Rich et al. [130] reported that exercise is an effective strategy to improve neurogenesis and angiogenesis in the hippocampus via VEGF production. However, it seems that the increased VEGF expression is transient after an acute bout of exercise (7–10 days), returning to baseline levels in a month [131]. Regardless, PA could be postulated as an important physiological stimulus capable of initiating protective brain mechanisms.
It is important to note that mitochondrial-induced ROS production during PA could have negative health consequences that are associated with oxidative stress in exercise practitioners [132]. In this line, the production of ROS is the result of the percentage of oxygen that the mitochondria has not been able to reduce during energy production [133]. However, it has been demonstrated that PA causes certain positive adaptations in the organism, such as the moderate elevations of ROS, which are involved in protective redox signalling and regulation [134]. Moreover, the elevation of these molecules by PA can regulate signalling or act as a signalling to muscular adaption [135] and produce adaptive responses to withstand further stress [136]. In addition, it has been shown that interindividual variability and training status can influence the levels of oxidative stress produced during exercise [137]. In this line, Margaritelis et al. [138] reported that different oxidative stress responses were observed in ninety-eight young men after five sets of eight eccentric maximal voluntary contractions, with an angular velocity of 60°/s with 2-minute rest intervals between sets. Thus, as a non-pharmacological strategy, PA has great potential to trigger regulatory responses that delay increased oxidative stress and decreased mitochondrial enzymatic activities [139].
This muscular adaptation and its reduction of oxidative stress shed light into the muscle–brain axis from a humeral point of view. In this line, a recent review [140] mentioned that a potential mechanism of PA benefits for the brain is the mitochondrial reprogramming, which is related to the increase of some systemic parameters (i.e., temperature, blood pressure, or oxygen supply, among others) in different tissues such as the muscle. Those parameters lead to the enhancement of clearance mechanisms of harmful substances, which reduce mitochondrial damage along the muscle–brain axis. In this line, Pereira et al. [141] have shown that increasing the blood flow in the dentate gyrus by cardiovascular training during 12 weeks improves the learning rate of an hippocampal-dependent task. Thus, an inadequate supply of oxygen would compromise mitochondrial function, negatively affecting brain function due to the increased oxidative stress [142].
Moreover, Erickson et al. [143] reported that PA training improves spatial memory function and reduces age-related hippocampal tissue loss in older adults. Furthermore, it is well established that PA is a potent stimulus to induce different signalling pathways that produce phenotypic changes in the mitochondria, leading to greater muscle health [126]. However, different methodological aspects of PA such as intensity and duration can exert different mitochondrial changes [140]. Thus, it has been shown that short-duration interval exercises with maximal and supramaximal intensities (high intensity interval training; HIIT or sprint interval training; or SIT) have a greater capacity to induce mitochondrial biogenesis than exercises performed at moderate intensity. Accordingly, MacInnis and Gibala [144] reported that 6–7 sessions of both HIIT and SIT produced 25–35% increasement of mitochondrial content. In addition, several studies have reported that high-intensity programmes elicited an increase in skeletal muscle mitochondrial respiration [145] and content [146] in comparison with moderate-intensity programmes. In conclusion, although endurance training was considered the primary means of achieving mitochondrial adaptation, performing alternative exercise training modalities could be an interesting strategy to induce similar improvements in mitochondria with a similar workload into a shorter period [133].
As previously described, oxidative stress can have different pathophysiological impacts [147]. Indeed, several studies have linked ROS’ reactivity to cellular damages such as lipoperoxidation, oxidation of protein sulfhydryl groups, and oxidation of purine and pyrimidine bases, which lead to the development of pathologies [148]. Indeed, the excessive production of ROS has been correlated to AD, vascular pathologies, and PD, among others [149].
Another complication of the increased ROS is the damage caused to mitochondria. ROS increasement has been proved to severely damage the mitochondria´s membranes, which leads to the reduction in the biogenesis of the mitochondria, which translates into a reduction in the production of ATP [150].
Exercise may increase the level, activation, and mRNA expression of endogenous antioxidant systems in the brain, and it has been shown to down-regulate the levels of the oxidative damage [151,152,153,154,155]. So, exercise has been implicated in reducing the risk of brain oxidative damage, but this response depends on the type of exercise used [155]. In this line, it has been shown that a single session of exhaustive exercise causes oxidative damage in untrained people. Indeed, the ROS released after a training session can cause damage at the lipid, protein, or DNA levels [156], whereas, in trained subjects these effects are not observed due to a greater resistance of these subjects to oxidative stress [157]. Additionally, harmful effects such as reduced force generation and increased muscle atrophy occur after non-regular strenuous exercise, whereas regular training has positive effects by influencing cellular processes leading to increased antioxidant expression [158]. These results proved evidence that different exercise interventions lead to different ROS responses.

5.1. The Relationship between Exercise and Increments of Oxidative Stress

Strong increases in ROS concentrations after strenuous exercise can cause contractile dysfunction and muscle atrophy, which promote muscle weakness and fatigue [159]. Other studies have shown that regardless of the type of acute exercise performed, aerobic [160] or anaerobic [161], there is an increase in ROS concentrations. ROS is naturally generated by the skeletal muscles when contracting; however, intense or prolonged exercise can result in increased oxidative damage production such as superoxide, hydrogen peroxide, and hydroxyl radicals.
Superoxide is formed primarily as an intermediate in many biochemical reactions. This anion is negatively charged and relatively impermeable to the membranes; however, compared to other free radicals, superoxide has a relatively long half-life, allowing its diffusion within the cell and, thus, increasing the number of potential targets to damage [162]. Regarding this, hydrogen peroxide (H2O2) is a reactive compound that leads to the generation of free radicals such as hydroxyl radicals. Hydrogen peroxide is stable, permeable to membranes with a relatively long half-life within the cell to which it is cytotoxic but with a relatively weak oxidizing agent [162]. Finally, hydroxyl radicals (OH-) are highly reactive with a strong oxidizing potential. Those features imply that when generated, they damage molecules near to their site of generation, although they are not permeable to membranes. The hydroxyl radicals are considered the most harmful ROS [162].
Several types of exercises have been linked to the production of the abovementioned ROS; we will shortly approach the most mentioned, such as resistance exercise and aerobic, anaerobic, and mixed exercises.
Resistance exercise has been linked to a rapid oxidative response after performing exercise [163,164], while others show hardly any effects [165]. Hudson et al. [166] have studied the relationship between acute moderate and high-intensity back squat exercise and the oxidative damage response as assessed by the biomarker protein carbonyls. Acute exercise has long been associated with a transient oxidative stress response [167]. The magnitude of oxidative stress following aerobic-type exercise is generally proportional to exercise intensity [160,168]. Additionally, evidence indicates that high-intensity resistance exercise, involving a large muscle mass such as squat exercise, consistently elicit a measurable blood oxidative stress [148,149,161,169]. Nonetheless, Hoffman et al. [137] demonstrated that plasma malondialdehyde values were correlated with an indirect marker of tissue oxygenation during the hypertrophy protocol as compared with the strength protocol. Those results are opposite to those of Hudson et al. [166] where the magnitude of carbonyl elevation was higher in the strength protocol than the hypertrophy protocol. Although both studies are supporting the positive relationship between intensity and oxidative stress, differences could be explained by the time course to complete and recover from the two protocols.
Regarding anaerobic exercise, an increasement in ROS production, lactic acid, acidosis, catecholamines, and post-exercise inflammation have been reported [170]. Precisely, this type of exercise significantly enhances purine catabolism and causes rapid deoxygenation named ischemia reperfusion phenomenon. These two phenomena are known to increase the activity of xanthine oxidase, which accelerates the production of ROS, more specifically O2 and H2O2 [171].
Similar results linking intense exercise to the increase in ROS have been found in mice. Aguiar et al. [154] found that intense exercise promoted brain mitochondrial dysfunction as well as an increase in the frontal cortex thiobarbituric acid-reactive substance levels in exercised mice. In agreement to the above-mentioned study, Somani et al. [172] observed that different brain areas contained different activities of antioxidant enzymes, as well as glutathione peroxidise and oxidized glutathione (GPx and GSSG) levels, which were preferentially altered as a result of exercise training to cope with oxidative stress.
It is also important to note that exercise-induced oxidative stress has wide interindividual variability [163]. Kawamura and Muraoka [173] studied 98 subjects performing eccentric quadriceps exercise and red blood cell, plasma, and urine samples were collected immediately after exercise and two days post-exercise. In the study, three biomarkers related to oxidative stress were analysed, such as F2-isoprostanes, protein carbonyls and glutathione. A considerable number of the participants exhibited changes in biomarker levels in the opposite direction to the group average. Indeed, 13% of the participants exhibited a decrease in F2-isoprostanes and protein carbonyls, whereas 10% of the participants showed an increase in glutathione levels. Furthermore, one out of three individuals showed unexpected or negligible responses (0% to ± 5%) to exercise in at least one redox biomarker [164]. These data highlighted the importance of inter-individual variability. This variability could be influenced by the training levels of the subjects, their level of rest, and the intake of antioxidants or even psychological variables, all of which should be taken into account when performing these studies.

5.2. Exercise as a Strategy to Reduce Oxidative Stress

Moderate aerobic training or simply voluntary exercise, such as running on a wheel, ameliorates antioxidant capacity [174,175,176,177,178,179,180], and regular moderate exercise improves brain function [177], memory [176], proteasome activation, and up-regulation of the antioxidant system [181]. Furthermore, daily moderate exercise has been shown to reduce damage of hippocampal slices from Wistar rats exposed to in vitro ischemia [182,183]. As previously described, anaerobic exercise in a progressive exercise programme can also improve different activities of antioxidant enzymes in the brain [172]. Similarly, anaerobic exercise with 10 s (short) or 40 s (long) rest intervals increased the antioxidant capacity from different tissues [184] at the same time that running on a treadmill until exhaustion did not induce lipid peroxidation by oxidative stress in the hippocampus [185]. Surprisingly, some other studies in which rats were overtrained in long terms of strenuous exercise or when the duration increased abruptly did not induce brain oxidative stress [186,187,188], and neither did similarly acute and chronic exercise promote oxidant stress in the prefrontal cortex, striatum, and hippocampus [189]. Those results are opposite to the abovementioned studies, where aerobic extenuation or anaerobic programs have been found to increase oxidative stress, pointing out the need to further investigate the key factors attenuating those oxidative effects.
The literature supporting a positive relationship between aerobic exercise and the reduction of oxidative stress is much clearer [167,190]. It has been shown that aerobic exercise leads to an increase in maximal oxygen consumption (VO2max) and an increase in ROS production [168]. However, if aerobic exercise intensities do not exceed 50% of VO2max, ROS production is reduced to minimal values, as demonstrated in studies carried out by Ashton et al. [191] or by Chevion et al. [192]. Additionally, aerobic exercise has been shown to promote a positive effect on SOD levels in 100% of the cases and improved lipid peroxidation in 90% of the studies [176,178,179,180,183,188,189,193].
In conclusion, aerobic exhausted exercise, anaerobic exercise, or the combination of both types of training still report confusing findings in relation to the production of oxidative stress. In contrast, regular moderate aerobic exercise appears to be highly contrasted to protect against brain oxidative stress. More research into factors causing inter-individual variability when performing each type of training would shed light into the relationship between exercise and oxidative stress.

6. The Beneficial Effects of Probiotics on Brain Health

The central nervous system is functionally communicated with the gastrointestinal tract. While brain signals different from motor, sensory, and secretory functions have an impact on the gastrointestinal system, signals from the gut also influence brain function [194,195]. This bidirectional communication, known as the “gut-brain axis”, includes the conjunction of the central nervous system, the enteric nervous system, the parasympathetic and sympathetic nervous systems, the endocrine-immune system, and the hypothalamus-pituitary-adrenal axis, in addition to the circulatory system [22]. Over the past few years, the gut microbiota has emerged as another key player in the interaction between gut and brain [196,197], leading to the coining of the phrase: “the microbiota-gut-brain axis”. The gut microbiota is a complex and dynamic community of microorganisms including bacteria, viruses, protozoa, fungi, and archaea that has co-evolved with the carrier host [198]. A total of 1014 microorganisms was reported in the gut, comprising 150 times more genes than the total number of human genes [107], which is called the “microbiome”. This “forgotten organ” [199] exerts essential functions for the host with effects beyond the gastrointestinal environment and is a milestone for physiological homeostasis maintenance [14]. Among those functions, intestinal microbiota trains the host immune system, participates actively in the gut barrier integrity and metabolism, and produces and controls the production of vitamins, hormones, and neurotransmitters [200,201]. Recently, it has been demonstrated that gut microbiota impact hypothalamus and amygdala functions [202,203] that are specially implicated in stress [204]. Evidence continues to demonstrate that the gut microbiota is particularly implicated in brain physiology and behaviour, affecting host mental health [17,18,22]. The dynamic nature of the gut microbiota makes it highly responsive to external factors. It is powerfully demonstrated that the age of the subject, the dietary pattern, exercise habits, medications, or stress affect the microbiota composition [205,206]. The imbalance of the gut microbiota, a status known as “dysbiosis” (despite to be not known yet the exactly definition of this microbiota alteration), can involve an altered signalling from the gut to brain, negatively influencing brain health or vice versa [22] (see Figure 3).
Accordingly, the gut microbiota is being increasingly used as a key target for both dietary and therapeutic strategies interventions to finally modulate the gut-brain axis. One promising approach entails the use of probiotics, which are defined as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [207]. Last two decades’ research have evidenced the impact of probiotics on gut microbiota physiology, establishing a clear strain-specificity and population- or diseases-specificity and knocking over the traditional concept of the “golden-strain” [208]. In recent years, different and specific probiotics strains have emerged by their beneficial effects in particular stages of life or diseases [16,208,209]. With respect to mechanisms of action, they have been described in different ways by which probiotics work, including the enhancement of the epithelial barrier, increased adhesion to intestinal mucosa and concomitant inhibition of pathogen adhesion, bacteriocins and acids production, the inhibition of bacterial translocation, anti-inflammatory substances production and immune system modulation, vitamins and neurotransmitters production, effect on calcium-dependent potassium channels in intestinal sensory neurons, and the induction of opioid and cannabinoid receptors in intestinal epithelial cells, among others [209,210] (see Figure 3).
Taken this together, it is clear that certain probiotics strains can modulate various features of the microbiota–gut–brain axis affecting positively on brain health. Most of the research was conducted in pre-clinical models (see Table 1), but there is also a significant body of evidence resulting from human studies. The reported beneficial findings of psychobiotics range from effects on physiological stress to anxiety, depression, mood, pain, or cognition [22]. In the Table 2, the majority of the clinical studies with probiotic strains showing effects on brain health have been compiled. Most of these studies did not set out to test the specific biological mechanisms that could be underlying the positive behavioural or mental health. However, as it was stated before, there is extended literature showing different hypotheses. Some biomarkers of inflammation and oxidative stress including interleukin 6, tumour necrosis factor alpha, catalase, or superoxide dismutase (SOD) could be of interest and very informative about this yet unknown process. Indeed, in two clinical trials with AD´s patients examining different probiotics combinations [211,212], an improvement in the Mini-mental state examination score were observed together with a significant increase in total antioxidant capacity and total glutathione (GSH) and a significant reduction in high sensitivity C-reactive protein. It is also worthy to mention that not all the probiotics tested in clinical trials entailed positive effects, even using the same strain or combination of strains in different populations. For example, the combination of the strains Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 showed positive effects on anxiety and depression parameters in three different studies [213,214,215]; however, Romjin et al. [216] did not showed any evidence that the same probiotic formulation was effective in treating low mood. Limosilactobacillus reuteri DSM 17938 (former Lactobacillus reuteri DSM 17938) showed effectiveness in the alleviation of pain in a cohort of children aged at 4–18 years with functional abdominal pain and inflammatory bowel disease [217]; however, no effects on neurocognitive and sensory outcomes were observed in a cohort of very low birth-weight preterm infants [218].
Table 1. Effects of exercise on brain health in pre-clinical studies.
Table 1. Effects of exercise on brain health in pre-clinical studies.
StudyType of ExerciseType of NeurodegenerativeEffects on Brain Health
Pre-Clinical
Ding et al. [129]Aerobic exerciseHealthyAngiogenesis
Swain et al. [46]Aerobic exerciseHealthyAngiogenesis in the motor cortex
Increase blood flow and volume
Maliszewska-Cyna et al. [48]Aerobic exerciseADNormalise hippocampal vascular morphology and preserved spatial memory
Karakilic et al. [49]Aerobic exerciseHealthyIncrease in muscle fibre VEGF levels was positively correlated with hippocampal-based memory and learning, and VEGF levels in the hippocampus
De Senna et al. [67]Aerobic exerciseDiabetic RatsImproves Non-Spatial Memory, Locomotor Skills, and the BBB
Herring et al. [68]Aerobic exerciseADFacilitate clearing of toxic metabolites such as amyloid beta (Aβ) by upregulating the expression of low-density lipoprotein receptor-related protein 1 (LRP-1) receptors on BBB through which Aβ is transported out of the brain
Nichol et al. [70]Aerobic exerciseADReduced the levels of IL-1β and TNF-α in the hippocampus and restored IFN-γ to wild-type levels, accompanied by a significant reduction in Aβ
He et al. [71]Aerobic exerciseHealthyPromotes Glymphatic Clearance of Aβ and Reduces the Activation of Astrocytes and Microglia
Jiang et al. [72]Aerobic exerciseHealthyImproves cognitive function together with microglia phenotype modulation and remyelination in chronic cerebral hypoperfusion
Kohman et al. [73]Aerobic exerciseHealthyReduces activation of microglia isolated from hippocampus and brain
Lu et al. [74]Aerobic exerciseADInhibite reactive gliosis following STZ injection and shifted activated microglia from M1 phenotype to M2.
Preservation of hippocampal-dependent cognitive function
Van Praag et al. [91]Aerobic exerciseHealthyIncreases cell proliferation and neurogenesis
Liu and Nusslock [92]Aerobic exerciseHealthyMediated neurogenesis in the hippocampus via BDNF
Moon et al. [96]Aerobic exerciseHealthyInduce systemic Cathepsin B secretion is associated with memory function
Wrann et al. [97]Aerobic exerciseHealthyExercise induces hippocampal BDNF through a PGC-1α/FNDC5 pathway
Lourenco et al. [98]Aerobic exerciseADIncrease hippocampal FNDC5/irisin in patients at risk of developing AD or in patients
already exhibiting cognitive impairment
Hayek et al. [99]Aerobic exerciseHealthyExercise induces the Mus musculus Bdnf gene and promotes learning and memory formation
Oliff et al. [112]Aerobic exerciseHealthyIncrease in mRNA and protein levels of BDNF within the hippocampus
Van Hoomissen et al. [113]
Adlard et al. [114]
Lee and Soya [115]
Vaynman et al. [116]Aerobic exerciseHealthyExercise induces improvements in learning and memory mediated by upregulation of signalling molecules cAMP response-element-binding protein (CREB) and synapsin I
Choi et al. [119]Aerobic exerciseADBehavioural symptoms were ameliorated when hippocampal neurogenesis was induced by exercise and accompanied by elevation in BDNF, interleukin 6 (IL-6) and synaptic markers
Cotman and Engesser [128]Aerobic exerciseHealthyExercise increases in brain-derived neurotrophic factor, a molecule that increases neuronal survival, enhances learning and protects against cognitive decline
Rich et al. [130]Aerobic exerciseHealthyExercise is an effective strategy to improve neurogenesis and angiogenesis in the hippocampus via VEGF production
Pereira et al. [141]Aerobic exerciseHealthyImproves the learning rate of an hippocampal-dependent task
Reduce mitochondrial damage along the muscle–brain axis
Um et al. [151]Aerobic exerciseADExercise may increase the level, activation, and mRNA expression of endogenous antioxidant systems in the brain, and it has been shown to down-regulate the levels of the oxidative damage
Aguiar et al. [152]Aerobic exercise (downhill training)HealthyDownhill running is as effective as level running in increasing hippocampal BDNF protein levels; BDNF protein is elevated in the striatum after downhill physical training.
Aguiar et al. [153]High-Intensity Physical ExerciseHealthyCellular signalling disturbances were associated with poor antioxidant response in the basal ganglia and with implicit memory impairment.
Aguiar et al. [154]Short bouts of mild-intensity physical exerciseHealthyImprovement of age-
related spatial memory déficitsIncreased hippocampal plasticity via AKT, CREB, and BDNF signalling
Tuon et al. [155]Aerobic exercisePDProtective effect on PD-induced 6-OHDA, possibly due the ability of exercise to modulate the brain redox state and preserve the content of the proteins that are important for normal brain function
Somani et al. [172]Aerobic exerciseHealthyExercise training causes more oxidative stress in the brainstem (BS) and corpus striatum (CS) regions, or has a better ability to induce antioxidant enzymes to cope with the superoxides formed. BS and CS may be more sensitive to oxidative stress
Somani et al. [172]Anaerobic exerciseHealthyImprove different activities of antioxidant enzymes in brain
Radák et al. [157]Aerobic exerciseHealthyImprove memory function
Radák et al. [177]Aerobic exerciseHealthyImprove brain function
Radák et al. [181]Aerobic exerciseHealthyImprove proteasome activation, and up-regulation of the antioxidant system
Scopel et al. [182]Aerobic exerciseHealthyReduce damage of hippocampal rats exposed to in vitro ischemia
Cechetti et al. [183]
Acikgoz et al. [185]Anaerobic exerciseHealthyRunning on a treadmill until exhaustion did not induce lipid peroxidation by oxidative stress in the hippocampus
Fry et al. [186]Moderate to strenous exerciseHealthyNot induce brain oxidative stress
Petibois et al. [187]Moderate to strenous exerciseHealthyNot induce brain oxidative stress
Ogonovszky et al. [188]Moderate to strenous exerciseHealthyNot induce brain oxidative stress
Aksu et al. [189]Acute and chronic exerciseHealthyAcute and chronic exercise neither promoted oxidant stress in prefrontal cortex, striatum, and hippocampus
AD, Alzheimer’s disease; PD, Parkinson.
Table 2. Effects of exercise on brain health in clinical studies.
Table 2. Effects of exercise on brain health in clinical studies.
StudyType of ExerciseType of NeurodegenerativeEffects on Brain Health
Clinical
Santos-Lozano et al. [9]≥150 min/week of moderate-intense activityAD40% reduction of risk for development of AD
Zimmerman et al. [36]Aerobic exerciseHealthyMitigate the effect of age on CBF
Ainslie et al. [37]
Lucas et al. [38]
Thomas et al. [39]Aerobic exerciseMCIMitigate the effect of age on CBF
Improvements in memory function
Gavin et al. [43]Aerobic exerciseHealthyIncreases the mRNA and protein levels of VEGF
Aronson et al. [65]Different levels of PAHealthyLevels of CRP decrease continuously with increased levels of physical fitness
Chupel et al. [66]Combined exercise training programme (resistance training and aerobic exercise)HealthyReduce levels of pro-inflammatory markers, improve BBB integrity, and increase cognitive performance
Coelho et al. [109]Aerobic exerciseHealthyEnhance the production of BDNF
Kwak [110]Aerobic exerciseADImprovements on BDNF Peripheral Levels and Cognition
Ruiz-González et al. [111]Aerobic exercisePDElevate BDNF levels
Rashid et al. [123]Aerobic exerciseADNeuroprotective impact on brain function
Increase in angiogenesis, neurogenesis, and synaptogenesis mainly due to an increase in blood flow, brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), hormones, and second messengers.
Murphy et al. [142]Different types of exerciseHealthyExercise improves spatial memory function and reduced age-related hippocampal tissue loss in older adults
AD, Alzheimer’s disease; PD, Parkinson’s disease; MCI, Mild cognitive impairment.
In the gut-brain axis context, a new definition for probiotics was raised. “Live organism that, when ingested in adequate amounts, produces a health benefit in patients suffering from psychiatric illness” was coined as “phychobiotics” by Dinan et al. [25]. This definition was later expanded to include prebiotics and also human populations at risk [219]. Nowadays, defining the action mode of psychobiotics at a cellular level, and how it mediates beneficial effects at a mechanistic level, is a challenge and hot topic field for neuroscientists and microbiologists, and it is a multidisciplinary field with high potential health benefits. To date, different ways or mechanisms through which probiotics can affect the gut-brain axis have been proposed (Figure 3). It is known that certain probiotics strains can produce neurochemicals, such as gamma-aminobutyric (GABA) [220] or acetylcholine [221], important neurotransmitters in the human brain. Intestinal bacterial was also observed to be involved in the production or metabolism of serotonin, norepinephrine, or dopamine [25]. Thus, a huge range of neurotransmitters can be produced by gut bacteria, being some of them key players in gastrointestinal and brain health. Moreover, inflammatory mediators such as cytokines or hormones such as cortisol, which can be modulated by intestinal bacteria or probiotics, are involved in this bidirectional communication [222]. In vivo studies have shown how the ingestion of certain probiotic strains decreased the levels of corticosterone and pro-inflammatory cytokines, with brain health benefits for the host [22]. On the other hand, some probiotics or psychobiotic strains produce short chain fatty acids (SCFA), which can cross the blood–brain barrier and exert neuroprotective and antidepressant properties [223], but it was also observed how SCFA reverted morphology deficits in microglia cells and enhanced stress levels in mice [22]. In a key study, Bravo et al. [210] demonstrated another communication path between the gut and brain, the vagus nerve, after vagotomy experiments with a probiotic strain (see Table 3).
It is also worthy to mention that only a limited range of probiotics have been tested as psychobiotics, with strains belonged to the genus Bifidobacterium and Lactobacillus being the most used [219], and most of them were administered in different mixes or combinations. Moreover, most of those strains were not selected based on their specific properties for gut-brain axis modulation but by their commercial availability or by other physiological beneficial effects. Accordingly, future studies are needed to select specific and targeted psychobiotic strains intended to specific disorders, and the use of one strain should not be generalized to another destination without its validation by independent studies. Finally, studies focusing on the mechanisms underlying the beneficial mental health action of the probiotics are required.
Table 3. Studies evaluating the effects of probiotics on brain health.
Table 3. Studies evaluating the effects of probiotics on brain health.
Probiotic UsedHuman CohortBeneficial Effects Reference
Anxiety and depression
Lactobacillus casei strain ShirotaChronic fatigue syndrome patientsDecreased anxiety symptomsRao et al. [224]
Lactobacillus helveticus R0052, Bifidobacterium longum R0175Healthy adult volunteersAlleviated psychological distress. Better anxiety, depression, anger-hostility, and problem-solving parameters Messaoudi et al. [213]
L. helveticus R0052, B. longum R0175Adults with UFC <50 ng/ml at baselineAlleviated psychological distress. Decreased anxiety, depression parametersMessaoudi et al. [214]
Lactobacillus acidophilus CUL60 (NCIMB 30157), L. acidophilus CUL21 (NCIMB 30156), Bifidobacterium animalis ssp. lactis (B. lactis) CUL34 (NCIMB 30172), Bifidobacterium bifidum CUL20 (NCIMB 30153)Healthy adult volunteersDecreased anxiety scores Owen et al. [225]
L. acidophilus, L. casei, B. bifidumAdult patients with major depressive disorderDecreased Beck´s Depression Inventory total scoresAkkasheh et al. [226]
Probiotic A: L. acidophilus LA5, B. lactis BB12. Probiotic B: L. casei, L. acidophilus, Lactobacillus rhamnosus, Lactobacillus delbrueckii ssp. bulgaricus (L. bulgaricus), Bifidobacterium breve, B. longum, Streptococcus thermophilus plus fructo-oligosaccharidesHealthy adult volunteersImprovement in depression and anxiety scoresMohammadi et al. [227]
L. casei strain ShirotaHealthy adult volunteersPrevention of cortisol hyper-secretion and physical symptoms under stressful conditionsTakada et al. [228]
L. acidophilus, B. bifidum, S. thermophilusResistant depression adult patients being currently depressedDecreased depression scoresBambling et al. [229]
S. thermophilus (CNCM I-1630), L. bulgaricus (CNCM I-1632, I-1519), Lactococcus lactis ssp. lactis (CNCM I-1631), L. acidophilus, S. thermophilus, Lactobacillus plantarum, B. lactis (CNCM I-2494), Lactobacillus reuteri (DSM 17938) (in combination with maltodextrin, silica, casein, lactose and gluten) Healthy adult volunteersDecreased Hamilton´s anxiety scoresColica et al. [230]
B. longum NCC3001Patients with IBS and diarrhoeaDecreased depression scoresPinto-Sanchez et al. [231]
L. rhamnosus HN001Pregnant womenLower depression and anxiety scores in
postpartum period
Slykerman et al. [232]
L. casei W56, L. acidophilus W22, Lactobacillus paracasei W20, B. lactis W51, Lactobacillus salivarius W24, Lactococcus lactis W19, B. lactis W52, L. plantarum W62 and B. bifidum W23Healthy adult volunteersImprovement in depression and anxiety scores Bagga et al. [233]
L. casei, L acidophilus, L bulgarigus, L rhamnosus, B. breve, B. longum, S. thermophilus (in combination with antidepressants and fructooligosaccharides)Moderate depression adult patientsDecreased Hamilton rating scale for
depression
Ghorbani et al. [234]
L. helveticus Rosell-52 (R005), B. longum Rosell-175 (R0175)Adult patients with major depressive disorderImprovement in depression scoresKazemi et al. [215]
Bacillus coagulans MTCC 5856 Adult IBS patients with major depressive disorderDecreased depression scoresMajeed et al. [235]
Clostridium butyricum MIYAIRI 588 (CBM588) (in combination with antidepressants)Treatment-resistant major depressive disorder patientsImprovement in depression scoresMiyaoka et al. [236]
B. breve strain A-1Schizophrenia patients Improvement in depression and anxiety scoresOkubo et al. [237]
L. plantarum P8Stressed adults Reduced anxiety scores Lew et al. [238]
B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W37, Lactobacillus brevis W63, L. casei W56, L. salivarius W24, Lactococcus lactis W19, Lactococcus lactis W58Female health care workers employed on a rotating-shift scheduleImprovement in anxiety and fatigueSmith-Ryan et al. [239]
Mood
L. casei strain Shirota in milk drinkHealthy adult volunteersMood improvementBenton et al. [240]
B. animalis ssp lactis (CNCM I-2494), S. thermophilus (CNCM I-1630), L. bulgaricus CNCM I-1632, L. bulgaricus I-1519, Lactococcus lactis ssp lactis (CNCM I-1631) in fermented milk Healthy women volunteersChanges in activity of brain regions controlling emotion and sensationTillisch et al. [241]
B. bifidum W23, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, Lactococcus lactis W19, Lactococcus lactis W58Healthy adult volunteersReduced cognitive reactivity to sad mood (rumination and aggressive thoughts)Steenbergen et al. [242]
Lactobacillus fermentum LF16, L. rhamnosus LR06, L. plantarum LP01, B. longum BL04Healthy adult volunteersImprovement depressive mood state, anger, fatigue, and sleep qualityMarotta et al. [243]
Stress
L. acidophilus Rosell-52, B. longum Rosell-175Stressed adults volunteersReduced two stress-induced gastrointestinal symptoms (abdominal pain and nausea/vomiting)Diop et al. [244]
B.longum 1714Healthy adult volunteersReduced daily reported stress Allen et al. [245]
L. casei strain ShirotaHealthy adult students volunteersDecreased stress-associated responses of abdominal dysfunction measured by feelings of stress and salivary cortisol levels Kato-Kataoka et al. [246]
Probiotic A: L. acidophilus LA5, B. lactis BB12. Probiotic B: L. casei, L. acidophilus, L. rhamnosus, L. bulgaricus, B. breve, B. longum, S. thermophilus (in combination with fructo-oligosaccharides)Healthy adult volunteersImprovement in stress scoresMohammadi et al. [227]
L. plantarum DR7Stressed adults volunteersReduced symptoms of stress and anxietyChong et al. [247]
L. plantarum P8Stressed adults volunteersReduced stress scores Lew et al. [238]
Cognition
L. acidophilus CUL60 (NCIMB 30157), L. acidophilus CUL21 (NCIMB 30156), B. lactis CUL34 (NCIMB 30172), B. bifidum CUL20 (NCIMB 30153)Healthy adult volunteersImprovement in attention tasksOwen et al. [225]
L. acidophilus, L. casei, B. bifidum, L. fermentum in milk drinkAlzheimer’s disease patientsImproed performance in the Mini-mental state examination scoreAkbari et al. [211]
B. longum 1714Healthy adult volunteersImproved hippocampal-dependent visuospatial memory performance and enhanced frontal midline electroencephalographic mobilityAllen et al. [245]
L. plantarum DSM 24730, S. thermophilus DSM 24731, B. breve DSM 24732, L. paracasei DSM 24733, L. delbrueckii ssp. bulgaricus DSM 24734, L. acidophilus DSM 24735, B. longum DSM 24736, Bifidobacterium longum ssp. infantis (B. infantis) DSM 24737HIV-1 infected patientsImprovement in several neurocognitive testsCeccarelli et al. [248]
L. casei W56, L. acidophilus W22, Lactobacillus paracasei W20, B. lactis W51, Lactobacillus salivarius W24, Lactococcus lactis W19, B. lactis W52, L. plantarum W62, B. bifidum W23Healthy adult volunteersImprovement in emotional memory and emotional decision-making tasksBagga et al. [233]
L. casei W56, L. acidophilus W22, L. paracasei W20, B. lactis W51, L. salivarius W24, Lactococcus lactis W19, B. lactis W52, L. plantarum W62, B. bifidum W23Healthy adult volunteersBehaviour modulation and a shift towards efficient
attentional control
Bagga et al. [249]
B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, L. lactis W19, Lactococcus lactis W58Adult patients with mild to severe depressionReduced cognitive reactivity towards sad moodChahwan et al. [250]
L. plantarum DR7Stressed adults volunteersImproved cognitive and memory functions in adults >30 years oldChong et al. [247]
L. helveticus IDCC380Healthy older volunteersImprovement in the performance of cognitive testsChung et al. [251]
B. breve A1Older adults with mild cognitive impairment or memory complaintsImprovement in several neurocognitive testsKobayashi et al. [252,253]
L. plantarum P8Stressed adults Improvement in memory and cognitive traits (social emotional cognition, verbal learning, and memory upon application)Lew et al. [238]
Lactobacillus gasseri CP2305Healthy young adults exposed to chronic stressReduced anxiety and sleep disturbanceNishida et al. [254]
B. bifidum W23, B. lactis W51, B. lactis W52, L. acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, Lactococcus lactis W19, Lactococcus lactis W58Healthy women volunteersCognitive improvement under induced acute stressPapalini et al. [255]
L. plantarum 299v (in combination with antidepressants)Adult patients with major depressive disorderImprovement in cognitive performance (attention, perceptivity, and verbal learning) Rudzki et al. [256]
L. acidophilus, B. bifidum, B. longum (in combination with selenium)Alzheimer’s disease patientsImprovement Mini-mental state examination scoreTamtaji et al. [212]
B. longum 1714 Healthy adult volunteersReduced mental fatigue and modulated neural responses during social stressWang et al. [257]
Pain
B. infantis M-63, B. breve M-16V, B. longum BB536Children with IBS (age 8–17.9 years) and children with FD (age 8–16.6 years)Improvement in abdominal pain and frequency in IBS children Giannetti et al. [258]
L. reuteri DSM 17938Children (age 4-18 years) with treatment of functional abdominal pain and IBS in children.Reduced intensity and duration of pain Jadrešin et al. [217]
ASD
L. acidophilus (strain Rosell-11)Children with autism (age 4–10 years)Improvement in the ability of concentration and carrying out ordersKałużna-Czaplińska et al. [259]
L. acidophilus, L. case, Lactobacillus delbruecki, B. longum, B. bifidum formulated with the immunomodulator Del-Immune V® (L. rhamnosus V lysate)Children with autism Improvement all autism treatment evaluation checklist domains (speech, language, communication, sociability, sensory, cognitive awareness, and health, physical, and behaviour)West et al. [260]
L. acidophilus, L. rhamnosus, B. longumChildren with autism (age 5–9 years)Improvements in the severity of autismShaaban et al. [261]
L. plantarum PS128Children with autism (age 7–15 years)Ameliorated opposition/defiance behaviours
total score. Improvement in attention
deficit hyperactivity disorder and oppositional defiant disorder in younger children (aged 7–12 years)
Liu et al. [262]
UFC: urinary free cortisol; IBS: irritable bowel syndrome; HIV-1: immunodeficiency virus-1; FD: functional dyspepsia; ASD: autism syndrome disorder. Taxonomy from Lactobacillus genus was recently revised (Zheng et al. 2020) and the former species: Lactobacillus brevis, Lactobacillus casei, Lactobacillus fermentum, Lactobacillus paracasei, Lactobacillus plantarum, Lactobacillus reuteri, Lactobacillus rhamnosus and Lactobacillus salivarius are now referred to as Secundilactobacillus collinoides, Lacticaseibacillus casei, Limosilactobacillus fermentum, Lacticaseibacillus paracasei, Lactiplantibacillus plantarum, Limosilactobacillus reuteri, Lacticaseibacillus rhamnosus and Ligilactobacillus salivarius, respectively.

7. Probiotics as a Therapeutic Tool against Brain Oxidative Stress

As previously stated, oxidative stress and the associated damage constitute a common trait in different disorders, and it is associated with ageing. Therefore, maintaining the redox balance results are critical for host homeostasis in general and for brain function in particular. This has attracted the attention of the research community towards the development of intervention strategies aimed at reducing this oxidative stress as a key step for health maintenance. The use of dietary antioxidant compounds has been the most studied approach, but others have also been evaluated. To this regard, although not one of the most commonly explored mechanisms, the use of probiotics for their antioxidative properties has been assessed.
The seminal studies in this area were performed with microorganisms of the genus Lactobacillus more than two decades ago by selecting strains well suited to deal with reactive oxygen species [263]. A strain selected in these early studies, Limosilactobacillus fermentum (formerly Lactobacillus fermentum) strain ME3 (DSM14241), was reported to improve markers of oxidative stress, such as total antioxidative activity (TAC) or glutathione red/ox ratio in healthy volunteers [264]. These results prompted later studies with this and other strains of lactobacilli [265], demonstrating the ability of some of these microorganisms to uptake and reduce the oxidised glutathione from the environment, contributing to the GSH/GSSG balance [263,266]. Moreover, these observations promoted the study of the application of antioxidant lactobacilli in different conditions, with positive results in the reduction of inflammation and myeloperoxidase activity in an animal model of induced colitis [267] or, more recently, the improvement of vascular endothelial damage in a model of chronic nitric oxide (NO) synthase inhibition [268]. Different mechanisms have been proposed for these effects of lactobacilli administration, including the ability to reduce glutathione or to produce enzymes such as feruloyl esterases able to release antioxidant hydroxycinnamic acids from undigested dietary compounds [263,266,269]. In addition to the abovementioned mechanisms, the bacterial metabolites, mainly SCFA, may also play a role. Acetate and butyrate, two of the main SCFA produced in the colon, seem to be able to downregulate the expression of G protein-coupled receptors (GPRs), increasing NO and reducing the production of ROS, thus mediating an antioxidative effect [270]. To this regard, in a PD model, the administration of a probiotic mix increased the levels of butyrate and neurotrohic factors in the brain, attenuating induced neuronal loss in the nigrostriatal pathway [271].
The increasing interest in the development of intervention tools for the prevention of age-related diseases and neurodegeneration has led to an increasing number of studies aiming at reducing brain oxidative stress by using lactobacilli or other probiotics such as bifidobacteria. The number of human intervention studies in this area is still limited with some data suggesting a potentially beneficial role for probiotics in AD with reductions on the levels of a lipid peroxidation product such as malondialdehyde (MAD), a common marker of oxidative stress, without detecting differences in TAC or NO [211]. Similarly, human studies have also been conducted on Parkinson’s patients or persons with mild cognitive impairment. A recent systematic review on the topic [272] concludes that although probiotics must be promising more RCT trials are needed.
Therefore, to date, most evidence in this area comes from animal studies. Different animal models have been used in the different studies. For example, by using a D-Galactose-induced aging mice model, Li et al. [273] demonstrated that the administration of a L. fermentum strain was able to induce the expression of antioxidant enzymes in the brain through the activation of the Nrf2 signalling pathway. Similarly, Lactiplantiibacillus plantarum strain Dp189 was recently reported to prevent cognitive decline and the hyperfosforilation of tau protein in a similar model [274]. The same strain has also been shown to downregulate oxidative damage in a PD model [275]. Moreover, the administration of L. plantarum GKM3 to senescence-accelerated mice (SAMP8) was found to reduce the bran levels of two oxidative markers: the 8-hydroxy-2′-deoxyguanosine and the lipid peroxidation by-products TBARS [273]. It is important to point out that, although the most studied, lactobacilli are not the only probiotics that have been tested. Bacterial mixes, also containing bifidobacterial strains, have also been reported to pose beneficial effects by reducing MDA and SOD levels in a β-amyloid injection model [276] or by limiting neuronal damage and lipid peroxidation in a 6-hydroxydopamine-induced PD model [277]. Not just bacteria but also the yeast Saccharomyces boulardii, a commonly used probiotic microorganism, has demonstrated a positive effect on brain oxidative stress in a mice model of antibiotic-induced dysbiosis [278]. The administration of this yeast was found to reduce MDA levels and acetylcholine esterase and myeloperoxidase activities in the brain whist it increased GSH and SOD activity.

8. Conclusions

Multiple lines of evidence provide strong support for the involvement of oxidative stress in relation to PA. Moreover, the dual role of oxidative stress in essential neuroprotective cellular mechanisms versus detrimental effects of increased uncontrolled ROS production should be carefully considered while developing strategies to mitigate oxidative stress under PA conditions. Indeed, we have discussed how the preventative and therapeutic effects of exercise are associated with the modality as well as the duration and intensity. Besides, moderate to vigorous intensity, along with the long duration and high frequency of exercise, has better benefits. The positive impacts of exercise on brain health might be associated with an improved mitochondrial function, enhanced production of BDNF, increased cerebral perfusion, and neurogenesis.
Moreover, we want to point out the relevance of the microbiota–gut–brain axis, which links the health of the gut microbiota and the central nervous system. Any imbalance in the commensal gut microbiota leads to aberrant endocrine, immunological, and neuronal signals that ultimately harm neuronal development and aggravate the age-related and neurodegenerative disease’s symptoms. Biotherapy using “psycobiotics” shows immense potential as therapeutic or prophylactic agents against oxidative stress and, also, due to their role to reinstate balance to the microbiota and the corresponding pathways that link microbial metabolism and brain functions.

Author Contributions

Conceptualization, N.A., I.M.-G., and A.K.; investigation, N.A., I.M.-G., S.A., F.J.G., A.K., and M.G; writing—original draft preparation N.A., I.M.-G., S.A., F.J.G., A.K., and M.G.; writing—review and editing, N.A., I.M.-G., and A.K.; supervision, N.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Science, Innovation and Universities (PSI2017-83893-R), the Ministry of Economy and Business (PSI2017-90806-REDT) and the Ministry of Economy and Competitivity (PID2020-117259RB-I00). S.A. was the recipient of a postdoctoral Juan de la Cierva contract (Ministry of Science and Innovation, Ref. IJCI-2017-32156).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analysed in this study. Data sharing is not applicable to this article.

Acknowledgments

We would like to thank M Jesus Pioz and Elena Mocholi at OTRI and Alvaro Bustinduy at the University of Nebrija who have supported us during the preparation of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Reiner, M.; Niermann, C.; Jekauc, D.; Woll, A. Long-term health benefits of physical activity—A systematic review of longitudinal studies. BMC Public Health 2013, 13, 813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tikkanen-Dolenc, H.; Wadén, J.; Forsblom, C.; Harjutsalo, V.; Thorn, L.M.; Saraheimo, M.; Elonen, N.; Tikkanen, H.O.; Groop, P.-H. FinnDiane Study Group. Physical activity reduces risk of premature mortality in patients with type 1 diabetes with and without kidney disease. Diabetes Care 2017, 40, 1727–1732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Marques, A.; Santos, T.; Martins, J.; Matos, M.G.D.; Valeiro, M.G. The association between physical activity and chronic diseases in european adults. Eur. J. Sport Sci. 2018, 18, 140–149. [Google Scholar] [CrossRef] [PubMed]
  4. Sofi, F.; Valecchi, D.; Bacci, D.; Abbate, R.; Gensini, G.F.; Casini, A.; Macchi, C. Physical activity and risk of cognitive decline: A meta-analysis of prospective studies. J. Intern. Med. 2011, 269, 107–117. [Google Scholar] [CrossRef]
  5. Firth, J.; Stubbs, B.; Vancampfort, D.; Schuch, F.; Lagopoulos, J.; Rosenbaum, S.; Ward, P.B. Effect of aerobic exercise on hippocampal volume in humans: A systematic review and meta-analysis. NeuroImage 2018, 166, 230–238. [Google Scholar] [CrossRef]
  6. Goukasian, N.; Porat, S.; Blanken, A.; Avila, D.; Zlatev, D.; Hurtz, S.; Hwang, K.S.; Pierce, J.; Joshi, S.H.; Woo, E.; et al. Cognitive correlates of hippocampal atrophy and ventricular enlargement in adults with or without mild cognitive impairment. Dement. Geriatr. Cogn. Disord. Extra 2019, 9, 281–293. [Google Scholar] [CrossRef]
  7. Apostolova, L.G.; Mosconi, L.; Thompson, P.M.; Green, A.E.; Hwang, K.S.; Ramirez, A.; Mistur, R.; Tsui, W.H.; de Leon, M.J. Subregional hippocampal atrophy predicts alzheimer’s dementia in the cognitively normal. Neurobiol. Aging 2010, 31, 1077–1088. [Google Scholar] [CrossRef] [Green Version]
  8. Tan, Z.S.; Spartano, N.L.; Beiser, A.S.; DeCarli, C.; Auerbach, S.H.; Vasan, R.S.; Seshadri, S. Physical activity, brain volume, and dementia risk: The framingham study. J. Gerontol. Ser. A 2017, 72, 789–795. [Google Scholar] [CrossRef] [Green Version]
  9. Santos-Lozano, A.; Pareja-Galeano, H.; Sanchis-Gomar, F.; Quindós-Rubial, M.; Fiuza-Luces, C.; Cristi-Montero, C.; Emanuele, E.; Garatachea, N.; Lucia, A. Physical activity and alzheimer disease: A protective association. Mayo Clin. Proc. 2016, 91, 999–1020. [Google Scholar] [CrossRef] [Green Version]
  10. Du, Z.; Li, Y.; Li, J.; Zhou, C.; Li, F.; Yang, X. Physical activity can improve cognition in patients with alzheimer’s disease: A systematic review and meta-analysis of randomized controlled trials. Clin. Interv. Aging 2018, 13, 1593–1603. [Google Scholar] [CrossRef] [Green Version]
  11. Jia, R.; Liang, J.; Xu, Y.; Wang, Y. Effects of physical activity and exercise on the cognitive function of patients with alzheimer disease: A meta-analysis. BMC Geriatr. 2019, 19, 181. [Google Scholar] [CrossRef] [PubMed]
  12. Ahlskog, J.E.; Geda, Y.E.; Graff-Radford, N.R.; Petersen, R.C. Physical exercise as a preventive or disease-modifying treatment of dementia and brain aging. Mayo Clin. Proc. 2011, 86, 876–884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Murray, D.K.; Sacheli, M.A.; Eng, J.J.; Stoessl, A.J. The effects of exercise on cognition in parkinson’s disease: A systematic review. Transl. Neurodegener. 2014, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  14. De Vos, W.M.; Tilg, H.; Van Hul, M.; Cani, P.D. Gut microbiome and health: Mechanistic insights. Gut 2022, 0, 1–13. [Google Scholar] [CrossRef] [PubMed]
  15. Arias, N.; Arboleya, S.; Allison, J.; Kaliszewska, A.; Higarza, S.G.; Gueimonde, M.; Arias, J.L. The relationship between choline bioavailability from diet, intestinal microbiota composition, and its modulation of human diseases. Nutrients 2020, 12, 2340. [Google Scholar] [CrossRef] [PubMed]
  16. Higarza, S.G.; Arboleya, S.; Arias, J.L.; Gueimonde, M.; Arias, N. Akkermansia muciniphila and environmental enrichment reverse cognitive impairment associated with high-fat high-cholesterol consumption in rats. Gut Microbes 2021, 13, 1–20. [Google Scholar] [CrossRef] [PubMed]
  17. De Palma, G.; Collins, S.M.; Bercik, P.; Verdu, E.F. The microbiota-gut-brain axis in gastrointestinal disorders: Stressed bugs, stressed brain or both? J. Physiol. 2014, 592, 2989–2997. [Google Scholar] [CrossRef]
  18. Cryan, J.F.; Dinan, T.G. Gut microbiota: Microbiota and neuroimmune signalling-metchnikoff to microglia. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 494–496. [Google Scholar] [CrossRef]
  19. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  20. Fan, P.; Nelson, C.D.; Driver, J.D.; Elzo, M.A.; Peñagaricano, F.; Jeong, K.C. Host genetics exerts lifelong effects upon hindgut microbiota and its association with bovine growth and immunity. ISME J. 2021, 15, 2306–2321. [Google Scholar] [CrossRef]
  21. Allison, J.; Kaliszewska, A.; Uceda, S.; Reiriz, M.; Arias, N. Targeting DNA methylation in the adult brain through diet. Nutrients 2021, 13, 3979. [Google Scholar] [CrossRef] [PubMed]
  22. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  23. Kaliszewska, A.; Allison, J.; Martini, M.; Arias, N. Improving age-related cognitive decline through dietary interventions targeting mitochondrial dysfunction. Int. J. Mol. Sci. 2021, 22, 3574. [Google Scholar] [CrossRef]
  24. Clarke, N.D.; Kornilios, E.; Richardson, D.L. Carbohydrate and caffeine mouth rinses do not affect maximum strength and muscular endurance performance. J. Strength Cond. Res. 2015, 10, 2926–2931. [Google Scholar] [CrossRef] [Green Version]
  25. Dinan, T.G.; Stanton, C.; Cryan, J.F. Psychobiotics: A novel class of psychotropic. Biol. Psychiatry 2013, 74, 720–726. [Google Scholar] [CrossRef]
  26. Kelleher, R.J.; Soiza, R.L. Evidence of endothelial dysfunction in the development of alzheimer’s disease: Is alzheimer’s a vascular disorder? Am. J. Cardiovasc. Dis. 2013, 3, 197–226. [Google Scholar]
  27. Potashkin, J.; Huang, X.; Becker, C.; Chen, H.; Foltynie, T.; Marras, C. Understanding the links between cardiovascular disease and parkinson’s disease. Mov. Disord. 2020, 35, 55–74. [Google Scholar] [CrossRef]
  28. Tarumi, T.; Zhang, R. Cerebral blood flow in normal aging adults: Cardiovascular determinants, clinical implications, and aerobic fitness. J. Neurochem. 2018, 144, 595–608. [Google Scholar] [CrossRef] [Green Version]
  29. Leeuwis, A.E.; Benedictus, M.R.; Kuijer, J.P.A.; Binnewijzend, M.A.A.; Hooghiemstra, A.M.; Verfaillie, S.C.J.; Koene, T.; Scheltens, P.; Barkhof, F.; Prins, N.D.; et al. Lower cerebral blood flow is associated with impairment in multiple cognitive domains in alzheimer’s disease. Alzheimers Dement. 2017, 13, 531–540. [Google Scholar] [CrossRef]
  30. Wolters, F.J.; Zonneveld, H.I.; Hofman, A.; van der Lugt, A.; Koudstaal, P.J.; Vernooij, M.W.; Ikram, M.A. Cerebral perfusion and the risk of dementia. Circulation 2017, 136, 719–728. [Google Scholar] [CrossRef] [Green Version]
  31. Austin, B.P.; Nair, V.A.; Meier, T.B.; Xu, G.; Rowley, H.A.; Carlsson, C.M.; Johnson, S.C.; Prabhakaran, V. Effects of hypoperfusion in alzheimer’s disease. J. Alzheimers Dis. JAD 2011, 26 (Suppl. 3), 123–133. [Google Scholar] [CrossRef] [PubMed]
  32. Benedictus, M.R.; Leeuwis, A.E.; Binnewijzend, M.A.A.; Kuijer, J.P.A.; Scheltens, P.; Barkhof, F.; van der Flier, W.M.; Prins, N.D. Lower cerebral blood flow is associated with faster cognitive decline in alzheimer’s disease. Eur. Radiol. 2017, 27, 1169–1175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Chao, L.L.; Buckley, S.T.; Kornak, J.; Schuff, N.; Madison, C.; Yaffe, K.; Miller, B.L.; Kramer, J.H.; Weiner, M.W. ASL perfusion MRI predicts cognitive decline and conversion from MCI to dementia. Alzheimer Dis. Assoc. Disord. 2010, 24, 19–27. [Google Scholar] [CrossRef]
  34. De Vis, J.B.; Peng, S.-L.; Chen, X.; Li, Y.; Liu, P.; Sur, S.; Rodrigue, K.M.; Park, D.C.; Lu, H. Arterial-Spin-Labeling (ASL) perfusion MRI Predicts cognitive function in elderly individuals: A 4-year longitudinal study. J. Magn. Reson. Imaging 2018, 48, 449–458. [Google Scholar] [CrossRef] [PubMed]
  35. Xekardaki, A.; Rodriguez, C.; Montandon, M.-L.; Toma, S.; Tombeur, E.; Herrmann, F.R.; Zekry, D.; Lovblad, K.-O.; Barkhof, F.; Giannakopoulos, P.; et al. Arterial spin labeling may contribute to the prediction of cognitive deterioration in healthy elderly individuals. Radiology 2015, 274, 490–499. [Google Scholar] [CrossRef]
  36. Zimmerman, B.; Sutton, B.; Low, K.; Fletcher, M.; Tan, C.H.; Schneider-Garces, N.; Li, Y.; Ouyang, C.; Maclin, E.; Gratton, G.; et al. Cardiorespiratory fitness mediates the effects of aging on cerebral blood flow. Front. Aging Neurosci. 2014, 6, 59. [Google Scholar] [CrossRef] [Green Version]
  37. Ainslie, P.N.; Cotter, J.D.; George, K.P.; Lucas, S.; Murrell, C.; Shave, R.; Thomas, K.N.; Williams, M.J.A.; Atkinson, G. Elevation in cerebral blood flow velocity with aerobic fitness throughout healthy human ageing. J. Physiol. 2008, 586, 4005–4010. [Google Scholar] [CrossRef]
  38. Lucas, S.J.E.; Ainslie, P.N.; Murrell, C.J.; Thomas, K.N.; Franz, E.A.; Cotter, J.D. Effect of age on exercise-induced alterations in cognitive executive function: Relationship to cerebral perfusion. Exp. Gerontol. 2012, 47, 541–551. [Google Scholar] [CrossRef]
  39. Thomas, B.P.; Tarumi, T.; Sheng, M.; Tseng, B.; Womack, K.B.; Cullum, C.M.; Rypma, B.; Zhang, R.; Lu, H. Brain perfusion change in patients with mild cognitive impairment after 12 months of aerobic exercise training. J. Alzheimers Dis. 2020, 75, 617–631. [Google Scholar] [CrossRef]
  40. Ding, Y.-H.; Li, J.; Zhou, Y.; Rafols, J.A.; Clark, J.C.; Ding, Y. Cerebral angiogenesis and expression of angiogenic factors in aging rats after exercise. Curr. Neurovasc. Res. 2006, 3, 15–23. [Google Scholar] [CrossRef]
  41. De Silva, T.M.; Faraci, F.M. Microvascular dysfunction and cognitive impairment. Cell. Mol. Neurobiol. 2016, 36, 241–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Iturria-Medina, Y.; Sotero, R.C.; Toussaint, P.J.; Mateos-Pérez, J.M.; Evans, A.C. Early role of vascular dysregulation on late-onset alzheimer’s disease based on multifactorial data-driven analysis. Nat. Commun. 2016, 7, 11934. [Google Scholar] [CrossRef] [PubMed]
  43. Gavin, T.P.; Ruster, R.S.; Carrithers, J.A.; Zwetsloot, K.A.; Kraus, R.M.; Evans, C.A.; Knapp, D.J.; Drew, J.L.; McCartney, J.S.; Garry, J.P.; et al. No Difference in the skeletal muscle angiogenic response to aerobic exercise training between young and aged men. J. Physiol. 2007, 585, 231–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Fournier, N.M.; Duman, R.S. Role of Vascular endothelial growth factor in adult hippocampal neurogenesis: Implications for the pathophysiology and treatment of depression. Behav. Brain Res. 2012, 227, 440–449. [Google Scholar] [CrossRef] [Green Version]
  45. Ballard, H.J. Exercise makes your brain bigger: Skeletal muscle VEGF and hippocampal neurogenesis. J. Physiol. 2017, 595, 5721–5722. [Google Scholar] [CrossRef]
  46. Swain, R.A.; Harris, A.B.; Wiener, E.C.; Dutka, M.V.; Morris, H.D.; Theien, B.E.; Konda, S.; Engberg, K.; Lauterbur, P.C.; Greenough, W.T. Prolonged exercise induces angiogenesis and increases cerebral blood volume in primary motor cortex of the rat. Neuroscience 2003, 117, 1037–1046. [Google Scholar] [CrossRef]
  47. Al-Jarrah, M.; Jamous, M.; Al Zailaey, K.; Bweir, S.O. Endurance exercise training promotes angiogenesis in the brain of chronic/progressive mouse model of parkinson’s disease. NeuroRehabilitation 2010, 26, 369–373. [Google Scholar] [CrossRef]
  48. Maliszewska-Cyna, E.; Oore, J.; Xhima, K.; Thomason, L.A.M.; Steinman, J.; McLaurin, J.; Sled, J.G.; Stefanovic, B.; Aubert, I. P1-015: Evaluation of effects of physical exercise on vascular and cerebral pathology, plasticity and function in a mouse model of Alzheimer’s disease. Alzheimers Dement. 2016, 12, P404–P405. [Google Scholar] [CrossRef]
  49. Karakilic, A.; Yuksel, O.; Kizildag, S.; Hosgorler, F.; Topcugil, B.; Ilgin, R.; Gumus, H.; Guvendi, G.; Koc, B.; Kandis, S.; et al. Regular aerobic exercise increased VEGF levels in both soleus and gastrocnemius muscles correlated with hippocampal learning and VEGF levels. Acta Neurobiol. Exp. 2021, 81, 1–9. [Google Scholar] [CrossRef]
  50. De Rossi, P.; Harde, E.; Dupuis, J.P.; Martin, L.; Chounlamountri, N.; Bardin, M.; Watrin, C.; Benetollo, C.; Pernet-Gallay, K.; Luhmann, H.J.; et al. A critical role for VEGF and VEGFR2 in NMDA receptor synaptic function and fear-related behavior. Mol. Psychiatry 2016, 21, 1768–1780. [Google Scholar] [CrossRef]
  51. Cockerill, I.; Oliver, J.-A.; Xu, H.; Fu, B.M.; Zhu, D. Blood-brain barrier integrity and clearance of amyloid-β from the BBB. Adv. Exp. Med. Biol. 2018, 1097, 261–278. [Google Scholar] [CrossRef] [PubMed]
  52. Rhea, E.M.; Banks, W.A. Role of the blood-brain barrier in central nervous system insulin resistance. Front. Neurosci. 2019, 13, 521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kadry, H.; Noorani, B.; Cucullo, L. A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef] [PubMed]
  54. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood–brain barrier breakdown in alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef]
  55. Noe, C.R.; Noe-Letschnig, M.; Handschuh, P.; Noe, C.A.; Lanzenberger, R. Dysfunction of the blood-brain barrier—A key step in neurodegeneration and dementia. Front. Aging Neurosci. 2020, 12, 185. [Google Scholar] [CrossRef]
  56. Flegal, K.M.; Kruszon-Moran, D.; Carroll, M.D.; Fryar, C.D.; Ogden, C.L. Trends in obesity among adults in the United States, 2005 to 2014. JAMA 2016, 315, 2284–2291. [Google Scholar] [CrossRef] [Green Version]
  57. Chiappetta, S.; Sharma, A.M.; Bottino, V.; Stier, C. COVID-19 and the role of chronic inflammation in patients with obesity. Int. J. Obes. 2020, 44, 1790–1792. [Google Scholar] [CrossRef]
  58. Walker, K.A. Inflammation and neurodegeneration: Chronicity matters. Aging 2021, 11, 3–4. [Google Scholar] [CrossRef]
  59. Esposito, K.; Pontillo, A.; Di Palo, C.; Giugliano, G.; Masella, M.; Marfella, R.; Giugliano, D. Effect of weight loss and lifestyle changes on vascular inflammatory markers in obese women: A randomized trial. JAMA 2003, 289, 1799–1804. [Google Scholar] [CrossRef] [Green Version]
  60. Goldhammer, E.; Tanchilevitch, A.; Maor, I.; Beniamini, Y.; Rosenschein, U.; Sagiv, M. Exercise training modulates cytokines activity in coronary heart disease patients. Int. J. Cardiol. 2005, 100, 93–99. [Google Scholar] [CrossRef]
  61. Taaffe, D.R.; Harris, T.B.; Ferrucci, L.; Rowe, J.; Seeman, T.E. Cross-sectional and prospective relationships of interleukin-6 and c-reactive protein with physical performance in elderly persons: MacArthur studies of successful aging. J. Gerontol. Ser. A 2000, 55, M709–M715. [Google Scholar] [CrossRef] [PubMed]
  62. Trøseid, M.; Lappegård, K.T.; Claudi, T.; Damås, J.K.; Mørkrid, L.; Brendberg, R.; Mollnes, T.E. Exercise reduces plasma levels of the chemokines MCP-1 and IL-8 in subjects with the metabolic syndrome. Eur. Heart J. 2004, 25, 349–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Petersen, A.M.W.; Pedersen, B.K. The anti-inflammatory effect of exercise. J. Appl. Physiol. 2005, 98, 1154–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cardoso, I.L.; Leal, F. C-reactive protein and neurodegenerative diseases. In Clinical Significance of C-Reactive Protein; Ansar, W., Ghosh, S., Eds.; Springer: Singapore, 2020; pp. 35–57. ISBN 9789811567872. [Google Scholar]
  65. Aronson, D.; Sheikh-Ahmad, M.; Avizohar, O.; Kerner, A.; Sella, R.; Bartha, P.; Markiewicz, W.; Levy, Y.; Brook, G.J. C-Reactive protein is inversely related to physical fitness in middle-aged subjects. Atherosclerosis 2004, 176, 173–179. [Google Scholar] [CrossRef] [PubMed]
  66. Chupel, M.U.; Minuzzi, L.G.; Furtado, G.; Santos, M.L.; Hogervorst, E.; Filaire, E.; Teixeira, A.M. Exercise and taurine in inflammation, cognition, and peripheral markers of blood-brain barrier integrity in older women. Appl. Physiol. Nutr. Metab. 2018, 43, 733–741. [Google Scholar] [CrossRef] [Green Version]
  67. De Senna, P.N.; Xavier, L.L.; Bagatini, P.B.; Saur, L.; Galland, F.; Zanotto, C.; Bernardi, C.; Nardin, P.; Gonçalves, C.A.; Achaval, M. Physical training improves non-spatial memory, locomotor skills and the blood brain barrier in diabetic rats. Brain Res. 2015, 1618, 75–82. [Google Scholar] [CrossRef]
  68. Herring, A.; Yasin, H.; Ambrée, O.; Sachser, N.; Paulus, W.; Keyvani, K. Environmental enrichment counteracts alzheimer’s neurovascular dysfunction in TgCRND8 mice. Brain Pathol. 2008, 18, 32–39. [Google Scholar] [CrossRef]
  69. Cribbs, D.H.; Berchtold, N.C.; Perreau, V.; Coleman, P.D.; Rogers, J.; Tenner, A.J.; Cotman, C.W. Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: A microarray study. J. Neuroinflammation 2012, 9, 179. [Google Scholar] [CrossRef] [Green Version]
  70. Nichol, K.E.; Poon, W.W.; Parachikova, A.I.; Cribbs, D.H.; Glabe, C.G.; Cotman, C.W. Exercise alters the immune profile in Tg2576 alzheimer mice toward a response coincident with improved cognitive performance and decreased amyloid. J. Neuroinflammation 2008, 5, 13. [Google Scholar] [CrossRef] [Green Version]
  71. He, X.; Liu, D.; Zhang, Q.; Liang, F.; Dai, G.; Zeng, J.; Pei, Z.; Xu, G.; Lan, Y. Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front. Mol. Neurosci. 2017, 10, 144. [Google Scholar] [CrossRef] [Green Version]
  72. Jiang, T.; Zhang, L.; Pan, X.; Zheng, H.; Chen, X.; Li, L.; Luo, J.; Hu, X. Physical exercise improves cognitive function together with microglia phenotype modulation and remyelination in chronic cerebral hypoperfusion. Front. Cell. Neurosci. 2017, 11, 404. [Google Scholar] [CrossRef] [PubMed]
  73. Kohman, R.A.; Bhattacharya, T.K.; Wojcik, E.; Rhodes, J.S. Exercise reduces activation of microglia isolated from hippocampus and brain of aged mice. J. Neuroinflammation 2013, 10, 885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Lu, Y.; Dong, Y.; Tucker, D.; Wang, R.; Ahmed, M.E.; Brann, D.; Zhang, Q. Treadmill exercise exerts neuroprotection and regulates microglial polarization and oxidative stress in a streptozotocin-induced rat model of sporadic alzheimer’s disease. J. Alzheimers Dis. JAD 2017, 56, 1469–1484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Deng, W.; Aimone, J.B.; Gage, F.H. New neurons and new memories: How does adult hippocampal neurogenesis affect learning and memory? Nat. Rev. Neurosci. 2010, 11, 339–350. [Google Scholar] [CrossRef]
  76. Ming, G.; Song, H. Adult neurogenesis in the mammalian brain: Significant answers and significant questions. Neuron 2011, 70, 687–702. [Google Scholar] [CrossRef] [Green Version]
  77. Farioli-Vecchioli, S.; Saraulli, D.; Costanzi, M.; Pacioni, S.; Cinà, I.; Aceti, M.; Micheli, L.; Bacci, A.; Cestari, V.; Tirone, F. The timing of differentiation of adult hippocampal neurons is crucial for spatial memory. PLOS Biol. 2008, 6, e246. [Google Scholar] [CrossRef]
  78. Saxe, M.D.; Battaglia, F.; Wang, J.-W.; Malleret, G.; David, D.J.; Monckton, J.E.; Garcia, A.D.R.; Sofroniew, M.V.; Kandel, E.R.; Santarelli, L.; et al. Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc. Natl. Acad. Sci.USA 2006, 103, 17501–17506. [Google Scholar] [CrossRef] [Green Version]
  79. Winocur, G.; Wojtowicz, J.M.; Sekeres, M.; Snyder, J.S.; Wang, S. Inhibition of neurogenesis interferes with hippocampus-dependent memory function. Hippocampus 2006, 16, 296–304. [Google Scholar] [CrossRef]
  80. Dupret, D.; Revest, J.-M.; Koehl, M.; Ichas, F.; Giorgi, F.D.; Costet, P.; Abrous, D.N.; Piazza, P.V. Spatial relational memory requires hippocampal adult neurogenesis. PLoS ONE 2008, 3, e1959. [Google Scholar] [CrossRef] [Green Version]
  81. Snyder, J.S.; Hong, N.S.; McDonald, R.J.; Wojtowicz, J.M. A role for adult neurogenesis in spatial long-term memory. Neuroscience 2005, 130, 843–852. [Google Scholar] [CrossRef]
  82. Goodman, T.; Trouche, S.; Massou, I.; Verret, L.; Zerwas, M.; Roullet, P.; Rampon, C. Young hippocampal neurons are critical for recent and remote spatial memory in adult mice. Neuroscience 2010, 171, 769–778. [Google Scholar] [CrossRef] [PubMed]
  83. Heine, V.M.; Maslam, S.; Joëls, M.; Lucassen, P.J. Prominent decline of newborn cell proliferation, differentiation, and apoptosis in the aging dentate gyrus, in absence of an age-related hypothalamus–pituitary–adrenal axis activation. Neurobiol. Aging 2004, 25, 361–375. [Google Scholar] [CrossRef]
  84. Kang, W.; Hébert, J.M. FGF signaling is necessary for neurogenesis in young mice and sufficient to reverse its decline in old mice. J. Neurosci. 2015, 35, 10217–10223. [Google Scholar] [CrossRef] [PubMed]
  85. Rao, M.S.; Hattiangady, B.; Shetty, A.K. The window and mechanisms of major age-related decline in the production of new neurons within the dentate gyrus of the hippocampus. Aging Cell 2006, 5, 545–558. [Google Scholar] [CrossRef] [PubMed]
  86. Kuhn, H.G.; Dickinson-Anson, H.; Gage, F.H. Neurogenesis in the dentate gyrus of the adult rat: Age-related decrease of neuronal progenitor proliferation. J. Neurosci. 1996, 16, 2027–2033. [Google Scholar] [CrossRef] [Green Version]
  87. Lichtenwalner, R.J.; Forbes, M.E.; Bennett, S.A.; Lynch, C.D.; Sonntag, W.E.; Riddle, D.R. Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis. Neuroscience 2001, 107, 603–613. [Google Scholar] [CrossRef]
  88. Moon, M.; Cha, M.-Y.; Mook-Jung, I. Impaired hippocampal neurogenesis and its enhancement with ghrelin in 5XFAD mice. J. Alzheimers Dis. JAD 2014, 41, 233–241. [Google Scholar] [CrossRef]
  89. Perry, E.K.; Johnson, M.; Ekonomou, A.; Perry, R.H.; Ballard, C.; Attems, J. Neurogenic abnormalities in alzheimer’s disease differ between stages of neurogenesis and are partly related to cholinergic pathology. Neurobiol. Dis. 2012, 47, 155–162. [Google Scholar] [CrossRef] [Green Version]
  90. Marxreiter, F.; Regensburger, M.; Winkler, J. Adult neurogenesis in parkinson’s disease. Cell. Mol. Life Sci. 2013, 70, 459–473. [Google Scholar] [CrossRef]
  91. Van Praag, H.; Kempermann, G.; Gage, F.H. Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat. Neurosci. 1999, 2, 266–270. [Google Scholar] [CrossRef]
  92. Liu, P.Z.; Nusslock, R. Exercise-mediated neurogenesis in the hippocampus via BDNF. Front. Neurosci. 2018, 12, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bergmann, O.; Spalding, K.L.; Frisén, J. Adult neurogenesis in humans. Cold Spring Harb. Perspect. Biol. 2015, 7, a018994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Monje, M.L.; Toda, H.; Palmer, T.D. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003, 302, 1760–1765. [Google Scholar] [CrossRef] [PubMed]
  95. Ekdahl, C.T.; Claasen, J.-H.; Bonde, S.; Kokaia, Z.; Lindvall, O. Inflammation is detrimental for neurogenesis in adult brain. Proc. Natl. Acad. Sci. USA 2003, 100, 13632–13637. [Google Scholar] [CrossRef] [Green Version]
  96. Moon, H.Y.; Becke, A.; Berron, D.; Becker, B.; Sah, N.; Benoni, G.; Janke, E.; Lubejko, S.T.; Greig, N.H.; Mattison, J.A.; et al. Running-induced systemic cathepsin b secretion is associated with memory function. Cell Metab. 2016, 24, 332–340. [Google Scholar] [CrossRef] [Green Version]
  97. Wrann, C.D.; White, J.P.; Salogiannnis, J.; Laznik-Bogoslavski, D.; Wu, J.; Ma, D.; Lin, J.D.; Greenberg, M.E.; Spiegelman, B.M. Exercise induces hippocampal bdnf through a PGC-1α/FNDC5 pathway. Cell Metab. 2013, 18, 649–659. [Google Scholar] [CrossRef] [Green Version]
  98. Lourenco, M.V.; Frozza, R.L.; de Freitas, G.B.; Zhang, H.; Kincheski, G.C.; Ribeiro, F.C.; Gonçalves, R.A.; Clarke, J.R.; Beckman, D.; Staniszewski, A.; et al. Exercise-linked FNDC5/Irisin rescues synaptic plasticity and memory defects in alzheimer’s models. Nat. Med. 2019, 25, 165–175. [Google Scholar] [CrossRef]
  99. Hayek, L.E.; Khalifeh, M.; Zibara, V.; Assaad, R.A.; Emmanuel, N.; Karnib, N.; El-Ghandour, R.; Nasrallah, P.; Bilen, M.; Ibrahim, P.; et al. Lactate mediates the effects of exercise on learning and memory through SIRT1-dependent activation of hippocampal brain-derived neurotrophic factor (BDNF). J. Neurosci. 2019, 39, 2369–2382. [Google Scholar] [CrossRef] [Green Version]
  100. Margineanu, M.B.; Mahmood, H.; Fiumelli, H.; Magistretti, P.J. L-lactate regulates the expression of synaptic plasticity and neuroprotection genes in cortical neurons: A transcriptome analysis. Front. Mol. Neurosci. 2018, 11, 375. [Google Scholar] [CrossRef]
  101. Schiffer, T.; Schulte, S.; Sperlich, B.; Achtzehn, S.; Fricke, H.; Strüder, H.K. Lactate infusion at rest increases BDNF blood concentration in humans. Neurosci. Lett. 2011, 488, 234–237. [Google Scholar] [CrossRef]
  102. Severinsen, M.C.K.; Pedersen, B.K. Muscle–organ crosstalk: The emerging roles of myokines. Endocr. Rev. 2020, 41, 594–609. [Google Scholar] [CrossRef] [PubMed]
  103. Miranda, M.; Morici, J.F.; Zanoni, M.B.; Bekinschtein, P. Brain-derived neurotrophic factor: A key molecule for memory in the healthy and the pathological brain. Front. Cell. Neurosci. 2019, 13, 363. [Google Scholar] [CrossRef] [PubMed]
  104. Howells, D.W.; Porritt, M.J.; Wong, J.Y.; Batchelor, P.E.; Kalnins, R.; Hughes, A.J.; Donnan, G.A. Reduced BDNF MRNA expression in the parkinson’s disease substantia nigra. Exp. Neurol. 2000, 166, 127–135. [Google Scholar] [CrossRef] [PubMed]
  105. Passaro, A.; Dalla Nora, E.; Morieri, M.L.; Soavi, C.; Sanz, J.M.; Zurlo, A.; Fellin, R.; Zuliani, G. Brain-derived neurotrophic factor plasma levels: Relationship with dementia and diabetes in the elderly population. J. Gerontol. Ser. A 2015, 70, 294–302. [Google Scholar] [CrossRef] [Green Version]
  106. Peng, S.; Wuu, J.; Mufson, E.J.; Fahnestock, M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of alzheimer’s disease. J. Neurochem. 2005, 93, 1412–1421. [Google Scholar] [CrossRef]
  107. Qin, X.-Y.; Cao, C.; Cawley, N.X.; Liu, T.-T.; Yuan, J.; Loh, Y.P.; Cheng, Y. Decreased peripheral brain-derived neurotrophic factor levels in alzheimer’s disease: A meta-analysis study (N = 7277). Mol. Psychiatry 2017, 22, 312–320. [Google Scholar] [CrossRef]
  108. Palareti, G.; Legnani, C.; Cosmi, B.; Antonucci, E.; Erba, N.; Poli, D.; Testa, S.; Tosetto, A.; DULCIS (D-dimer-ULtrasonography in Combination Italian Study) Investigators (See Appendix); DULCIS (D-dimer-ULtrasonography in Combination Italian Study) Investigators. Comparison between different D-Dimer cutoff values to assess the individual risk of recurrent venous thromboembolism: Analysis of results obtained in the DULCIS Study. Int. J. Lab. Hematol. 2016, 38, 42–49. [Google Scholar] [CrossRef]
  109. Coelho, F.G.M.; Vital, T.M.; Stein, A.M.; Arantes, F.J.; Rueda, A.V.; Camarini, R.; Teodorov, E.; Santos-Galduróz, R.F. Acute aerobic exercise increases brain-derived neurotrophic factor levels in elderly with alzheimer’s disease. J. Alzheimers Dis. 2014, 39, 401–408. [Google Scholar] [CrossRef]
  110. Kwak, K.-P. P3-235: Exercise training in very old adults with mild cognitive impairment: Improvements on BDNF peripheral levels and cognition. Alzheimers Dement. 2015, 11, P722. [Google Scholar] [CrossRef]
  111. Ruiz-González, D.; Hernández-Martínez, A.; Valenzuela, P.L.; Morales, J.S.; Soriano-Maldonado, A. Effects of physical exercise on plasma brain-derived neurotrophic factor in neurodegenerative disorders: A systematic review and meta-analysis of randomized controlled trials. Neurosci. Biobehav. Rev. 2021, 128, 394–405. [Google Scholar] [CrossRef]
  112. Oliff, H.S.; Berchtold, N.C.; Isackson, P.; Cotman, C.W. Exercise-induced regulation of brain-derived neurotrophic factor (bdnf) transcripts in the rat hippocampus. Mol. Brain Res. 1998, 61, 147–153. [Google Scholar] [CrossRef]
  113. Van Hoomissen, J.D.; Chambliss, H.O.; Holmes, P.V.; Dishman, R.K. Effects of chronic exercise and imipramine on MRNA for BDNF after olfactory bulbectomy in rat. Brain Res. 2003, 974, 228–235. [Google Scholar] [CrossRef]
  114. Adlard, P.A.; Perreau, V.M.; Cotman, C.W. The exercise-induced expression of BDNF within the Hippocampus Varies Across Life-Span. Neurobiol. Aging 2005, 26, 511–520. [Google Scholar] [CrossRef]
  115. Lee, M.; Soya, H. Effects of acute voluntary loaded wheel running on BDNF expression in the rat hippocampus. J. Exerc. Nutr. Biochem. 2017, 21, 52–57. [Google Scholar] [CrossRef] [PubMed]
  116. Vaynman, S.; Ying, Z.; Gomez-Pinilla, F. Hippocampal BDNF mediates the efficacy of exercise on synaptic plasticity and cognition. Eur. J. Neurosci. 2004, 20, 2580–2590. [Google Scholar] [CrossRef]
  117. Vaynman, S.; Ying, Z.; Gómez-Pinilla, F. Exercise induces BDNF and synapsin i to specific hippocampal subfields. J. Neurosci. Res. 2004, 76, 356–362. [Google Scholar] [CrossRef]
  118. Jiang, L.; Zhang, H.; Wang, C.; Ming, F.; Shi, X.; Yang, M. Serum level of brain-derived neurotrophic factor in parkinson’s disease: A meta-analysis. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 88, 168–174. [Google Scholar] [CrossRef]
  119. Choi, S.H.; Bylykbashi, E.; Chatila, Z.K.; Lee, S.W.; Pulli, B.; Clemenson, G.D.; Kim, E.; Rompala, A.; Oram, M.K.; Asselin, C.; et al. Combined adult neurogenesis and bdnf mimic exercise effects on cognition in an alzheimer’s mouse model. Science 2018, 361, eaan8821. [Google Scholar] [CrossRef] [Green Version]
  120. De Felice, F.G.; Ferreira, S.T. Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to alzheimer disease. Diabetes 2014, 63, 2262–2272. [Google Scholar] [CrossRef] [Green Version]
  121. Arias, N.; Fidalgo, C.; Vallejo, G.; Arias, J.L. Brain network function during shifts in learning strategies in portal hypertension animals. Brain Res. Bull. 2014, 104, 52–59. [Google Scholar] [CrossRef]
  122. Arias, N.; Méndez, M.; Arias, J.L. Differential contribution of the hippocampus in two different demanding tasks at early stages of hepatic encephalopathy. Neuroscience 2015, 284, 1–10. [Google Scholar] [CrossRef] [PubMed]
  123. Rashid, M.H.; Zahid, M.F.; Zain, S.; Kabir, A.; Hassan, S.U. The neuroprotective effects of exercise on cognitive decline: A preventive approach to alzheimer disease. Cureus 2020, 12, e6958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Peeri, M.; Amiri, S. Protective effects of exercise in metabolic disorders are mediated by inhibition of mitochondrial-derived sterile inflammation. Med. Hypotheses 2015, 85, 707–709. [Google Scholar] [CrossRef] [PubMed]
  125. Philp, A.M.; Saner, N.J.; Lazarou, M.; Ganley, I.G.; Philp, A. The influence of aerobic exercise on mitochondrial quality control in skeletal muscle. J. Physiol. 2021, 599, 3463–3476. [Google Scholar] [CrossRef] [PubMed]
  126. Sorriento, D.; Di Vaia, E.; Iaccarino, G. Physical exercise: A novel tool to protect mitochondrial health. Front. Physiol. 2021, 12, 526. [Google Scholar] [CrossRef] [PubMed]
  127. Marques-Aleixo, I.; Oliveira, P.J.; Moreira, P.I.; Magalhães, J.; Ascensão, A. Physical exercise as a possible strategy for brain protection: Evidence from mitochondrial-mediated mechanisms. Prog. Neurobiol. 2012, 99, 149–162. [Google Scholar] [CrossRef]
  128. Cotman, C.W.; Engesser-Cesar, C. Exercise enhances and protects brain function. Exerc. Sport Sci. Rev. 2002, 30, 75–79. [Google Scholar] [CrossRef]
  129. Ding, Y.H.; Li, J.; Yao, W.X.; Rafols, J.A.; Clark, J.C.; Ding, Y. Exercise preconditioning upregulates cerebral integrins and enhances cerebrovascular integrity in ischemic rats. Acta Neuropathol. 2006, 112, 74–84. [Google Scholar] [CrossRef]
  130. Rich, B.; Scadeng, M.; Yamaguchi, M.; Wagner, P.D.; Breen, E.C. Skeletal myofiber vascular endothelial growth factor is required for the exercise training-induced increase in dentate gyrus neuronal precursor cells. J. Physiol. 2017, 595, 5931–5943. [Google Scholar] [CrossRef] [Green Version]
  131. Gustafsson, T.; Knutsson, A.; Puntschart, A.; Kaijser, L.; Nordqvist, A.-C.S.; Sundberg, C.J.; Jansson, E. Increased expression of vascular endothelial growth factor in human skeletal muscle in response to short-term one-legged exercise training. Pflugers Arch. 2002, 444, 752–759. [Google Scholar] [CrossRef]
  132. Powers, S.K.; Deminice, R.; Ozdemir, M.; Yoshihara, T.; Bomkamp, M.P.; Hyatt, H. Exercise-induced oxidative stress: Friend or foe? J. Sport Health Sci. 2020, 9, 415–425. [Google Scholar] [CrossRef] [PubMed]
  133. Peternelj, T.-T.; Coombes, J.S. Antioxidant supplementation during exercise training: Beneficial or detrimental? Sports Med. 2011, 41, 1043–1069. [Google Scholar] [CrossRef] [PubMed]
  134. Sies, H.; Jones, D.P. Reactive Oxygen Species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
  135. Thirupathi, A.; Wang, M.; Lin, J.K.; Fekete, G.; István, B.; Baker, J.S.; Gu, Y. Effect of different exercise modalities on oxidative stress: A systematic review. BioMed Res. Int. 2021, 2021, e1947928. [Google Scholar] [CrossRef] [PubMed]
  136. Bayod, S.; Guzmán-Brambila, C.; Sanchez-Roige, S.; Lalanza, J.; Kaliman, P.; Ortuño-Sahagun, D.; Escorihuela, R.; Pallàs, M. Voluntary exercise promotes beneficial anti-aging mechanisms in SAMP8 female brain. J. Mol. Neurosci. 2015, 55, 525–532. [Google Scholar] [CrossRef] [Green Version]
  137. Hoffman, J.R.; Im, J.; Kang, J.; Maresh, C.M.; Kraemer, W.J.; French, D.; Nioka, S.; Kime, R.; Rundell, K.W.; Ratamess, N.A.; et al. Comparison of low- and high-intensity resistance exercise on lipid peroxidation: Role of muscle oxygenation. J. Strength Cond. Res. 2007, 21, 118–122. [Google Scholar] [CrossRef]
  138. Margaritelis, N.V.; Kyparos, A.; Paschalis, V.; Theodorou, A.A.; Panayiotou, G.; Zafeiridis, A.; Dipla, K.; Nikolaidis, M.G.; Vrabas, I.S. Reductive stress after exercise: The issue of redox individuality. Redox Biol. 2014, 2, 520–528. [Google Scholar] [CrossRef] [Green Version]
  139. Radak, Z.; Hart, N.; Sarga, L.; Koltai, E.; Atalay, M.; Ohno, H.; Boldogh, I. Exercise plays a preventive role against alzheimer’s disease. J. Alzheimers Dis. JAD 2010, 20, 777–783. [Google Scholar] [CrossRef] [Green Version]
  140. Burtscher, J.; Millet, G.P.; Place, N.; Kayser, B.; Zanou, N. The muscle-brain axis and neurodegenerative diseases: The key role of mitochondria in exercise-induced neuroprotection. Int. J. Mol. Sci. 2021, 22, 6479. [Google Scholar] [CrossRef]
  141. Pereira, A.C.; Huddleston, D.E.; Brickman, A.M.; Sosunov, A.A.; Hen, R.; McKhann, G.M.; Sloan, R.; Gage, F.H.; Brown, T.R.; Small, S.A. An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc. Natl. Acad. Sci. USA 2007, 104, 5638–5643. [Google Scholar] [CrossRef] [Green Version]
  142. Murphy, R.M.; Watt, M.J.; Febbraio, M.A. Metabolic communication during exercise. Nat. Metab. 2020, 2, 805–816. [Google Scholar] [CrossRef] [PubMed]
  143. Erickson, K.I.; Voss, M.W.; Prakash, R.S.; Basak, C.; Szabo, A.; Chaddock, L.; Kim, J.S.; Heo, S.; Alves, H.; White, S.M.; et al. Exercise training increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci. USA 2011, 108, 3017–3022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. MacInnis, M.J.; Gibala, M.J. Physiological adaptations to interval training and the role of exercise intensity. J. Physiol. 2017, 595, 2915–2930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Daussin, F.N.; Zoll, J.; Dufour, S.P.; Ponsot, E.; Lonsdorfer-Wolf, E.; Doutreleau, S.; Mettauer, B.; Piquard, F.; Geny, B.; Richard, R. Effect of interval versus continuous training on cardiorespiratory and mitochondrial functions: Relationship to aerobic performance improvements in sedentary subjects. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 295, R264–R272. [Google Scholar] [CrossRef] [Green Version]
  146. MacInnis, M.J.; Zacharewicz, E.; Martin, B.J.; Haikalis, M.E.; Skelly, L.E.; Tarnopolsky, M.A.; Murphy, R.M.; Gibala, M.J. Superior mitochondrial adaptations in human skeletal muscle after interval compared to continuous single-leg cycling matched for total work. J. Physiol. 2017, 595, 2955–2968. [Google Scholar] [CrossRef]
  147. Cunningham, G.M.; Roman, M.G.; Flores, L.C.; Hubbard, G.B.; Salmon, A.B.; Zhang, Y.; Gelfond, J.; Ikeno, Y. The paradoxical role of thioredoxin on oxidative stress and aging. Arch. Biochem. Biophys. 2015, 576, 32–38. [Google Scholar] [CrossRef]
  148. Punchard, N.; Kelly, F. Free Radicals: A Practical Approach; IRL Press: Oxford, UK, 1996. [Google Scholar]
  149. Pincemail, J. Free radicals and antioxidants in human diseases. In Analysis of Free Radicals in Biological Systems; Springer: Berlin/Heidelberg, Germany, 1995; pp. 83–98. [Google Scholar]
  150. García-Mesa, Y.; Colie, S.; Corpas, R.; Cristòfol, R.; Comellas, F.; Nebreda, A.R.; Giménez-Llort, L.; Sanfeliu, C. Oxidative stress is a central target for physical exercise neuroprotection against pathological brain aging. J. Gerontol. Ser. Biomed. Sci. Med. Sci. 2016, 71, 40–49. [Google Scholar] [CrossRef]
  151. Um, H.S.; Kang, E.B.; Leem, Y.H.; Cho, I.H.; Yang, C.H.; Chae, K.R.; Hwang, D.Y.; Cho, J.Y. Exercise training acts as a therapeutic strategy for reduction of the pathogenic phenotypes for alzheimer’s disease in an NSE/APPsw-transgenic model. Int. J. Mol. Med. 2008, 22, 529–539. [Google Scholar]
  152. Aguiar, A.S.; Speck, A.E.; Prediger, R.D.S.; Kapczinski, F.; Pinho, R.A. Downhill training upregulates mice hippocampal and striatal brain-derived neurotrophic factor levels. J. Neural Transm. Vienna Austria 1996 2008, 115, 1251–1255. [Google Scholar] [CrossRef]
  153. Aguiar, A.S.; Boemer, G.; Rial, D.; Cordova, F.M.; Mancini, G.; Walz, R.; de Bem, A.F.; Latini, A.; Leal, R.B.; Pinho, R.A.; et al. High-intensity physical exercise disrupts implicit memory in mice: Involvement of the striatal glutathione antioxidant system and intracellular signaling. Neuroscience 2010, 171, 1216–1227. [Google Scholar] [CrossRef]
  154. Aguiar, A.S.; Castro, A.A.; Moreira, E.L.; Glaser, V.; Santos, A.R.S.; Tasca, C.I.; Latini, A.; Prediger, R.D.S. Short bouts of mild-intensity physical exercise improve spatial learning and memory in aging rats: Involvement of hippocampal plasticity via AKT, CREB and BDNF signaling. Mech. Ageing Dev. 2011, 132, 560–567. [Google Scholar] [CrossRef] [PubMed]
  155. Tuon, T.; Valvassori, S.S.; Lopes-Borges, J.; Luciano, T.; Trom, C.B.; Silva, L.A.; Quevedo, J.; Souza, C.T.; Lira, F.S.; Pinho, R.A. Physical training exerts neuroprotective effects in the regulation of neurochemical factors in an animal model of parkinson’s disease. Neuroscience 2012, 227, 305–312. [Google Scholar] [CrossRef] [PubMed]
  156. Dizdaroglu, M.; Jaruga, P. Mechanisms of free radical-induced damage to DNA. Free Radic. Res. 2012, 46, 382–419. [Google Scholar] [CrossRef] [PubMed]
  157. Radak, Z.; Taylor, A.W.; Ohno, H.; Goto, S. Adaptation to exercise-induced oxidative stress: From muscle to brain. Exerc. Immunol. Rev. 2001, 7, 90–107. [Google Scholar]
  158. Steinbacher, P.; Eckl, P. Impact of oxidative stress on exercising skeletal muscle. Biomolecules 2015, 5, 356–377. [Google Scholar] [CrossRef]
  159. Reid, M.B. Free Radicals and Muscle Fatigue: Of ROS, Canaries, and the IOC. Free Radic. Biol. Med. 2008, 44, 169–179. [Google Scholar] [CrossRef]
  160. Marzatico, F.; Pansarasa, O.; Bertorelli, L.; Somenzini, L.; Della Valle, G. Blood free radical antioxidant enzymes and lipid peroxides following long-distance and lactacidemic performances in highly trained aerobic and sprint athletes. J. Sports Med. Phys. Fit. 1997, 37, 235–239. [Google Scholar]
  161. Groussard, C.; Machefer, G.; Rannou, F.; Faure, H.; Zouhal, H.; Sergent, O.; Chevanne, M.; Cillard, J.; Gratas-Delamarche, A. Physical fitness and plasma non-enzymatic antioxidant status at rest and after a wingate test. Can. J. Appl. Physiol. Rev. Can. Physiol. Appl. 2003, 28, 79–92. [Google Scholar] [CrossRef]
  162. Powers, S.K.; Jackson, M.J. Exercise-induced oxidative stress: Cellular mechanisms and impact on muscle force production. Physiol. Rev. 2008, 88, 1243–1276. [Google Scholar] [CrossRef] [Green Version]
  163. Bloomer, R.J.; Falvo, M.J.; Fry, A.C.; Schilling, B.K.; Smith, W.A.; Moore, C.A. Oxidative stress response in trained men following repeated squats or sprints. Med. Sci. Sports Exerc. 2006, 38, 1436–1442. [Google Scholar] [CrossRef] [Green Version]
  164. Bloomer, R.J.; Fry, A.C.; Falvo, M.J.; Moore, C.A. Protein carbonyls are acutely elevated following single set anaerobic exercise in resistance trained men. J. Sci. Med. Sport 2007, 10, 411–417. [Google Scholar] [CrossRef] [PubMed]
  165. McAnulty, S.R.; McAnulty, L.S.; Nieman, D.C.; Morrow, J.D.; Utter, A.C.; Dumke, C.L. Effect of resistance exercise and carbohydrate ingestion on oxidative stress. Free Radic. Res. 2005, 39, 1219–1224. [Google Scholar] [CrossRef] [PubMed]
  166. Hudson, M.B.; Hosick, P.A.; McCaulley, G.O.; Schrieber, L.; Wrieden, J.; McAnulty, S.R.; Triplett, N.T.; McBride, J.M.; Quindry, J.C. The effect of resistance exercise on humoral markers of oxidative stress. Med. Sci. Sports Exerc. 2008, 40, 542–548. [Google Scholar] [CrossRef] [PubMed]
  167. Clarkson, P.M. Antioxidants and physical performance. Crit. Rev. Food Sci. Nutr. 1995, 35, 131–141. [Google Scholar] [CrossRef] [PubMed]
  168. Alessio, H.M. Exercise-induced oxidative stress. Med. Sci. Sports Exerc. 1993, 25, 218–224. [Google Scholar] [CrossRef]
  169. Chen, Z.; Zhong, C. Oxidative stress in alzheimer’s disease. Neurosci. Bull. 2014, 30, 271–281. [Google Scholar] [CrossRef]
  170. Sahlin, K.; Cizinsky, S.; Warholm, M.; Höberg, J. Repetitive static muscle contractions in humans—A trigger of metabolic and oxidative stress? Eur. J. Appl. Physiol. 1992, 64, 228–236. [Google Scholar] [CrossRef]
  171. Heunks, L.M.; Viña, J.; van Herwaarden, C.L.; Folgering, H.T.; Gimeno, A.; Dekhuijzen, P.N. Xanthine oxidase is involved in exercise-induced oxidative stress in chronic obstructive pulmonary disease. Am. J. Physiol. 1999, 277, R1697–R1704. [Google Scholar] [CrossRef]
  172. Somani, S.M.; Ravi, R.; Rybak, L.P. Effect of exercise training on antioxidant system in brain regions of rat. Pharmacol. Biochem. Behav. 1995, 50, 635–639. [Google Scholar] [CrossRef]
  173. Kawamura, T.; Muraoka, I. Exercise-induced oxidative stress and the effects of antioxidant intake from a physiological viewpoint. Antioxidants 2018, 7, 119. [Google Scholar] [CrossRef] [Green Version]
  174. Ohkuwa, T.; Sato, Y.; Naoi, M. Glutathione status and reactive oxygen generation in tissues of young and old exercised rats. Acta Physiol. Scand. 1997, 159, 237–244. [Google Scholar] [CrossRef] [PubMed]
  175. Itoh, H.; Ohkuwa, T.; Yamamoto, T.; Sato, Y.; Miyamura, M.; Naoi, M. Effects of endurance physical training on hydroxyl radical generation in rat tissues. Life Sci. 1998, 63, 1921–1929. [Google Scholar] [CrossRef]
  176. Radák, Z.; Kaneko, T.; Tahara, S.; Nakamoto, H.; Pucsok, J.; Sasvári, M.; Nyakas, C.; Goto, S. Regular exercise improves cognitive function and decreases oxidative damage in rat brain. Neurochem. Int. 2001, 38, 17–23. [Google Scholar] [CrossRef]
  177. Radak, Z.; Toldy, A.; Szabo, Z.; Siamilis, S.; Nyakas, C.; Silye, G.; Jakus, J.; Goto, S. The effects of training and detraining on memory, neurotrophins and oxidative stress markers in rat brain. Neurochem. Int. 2006, 49, 387–392. [Google Scholar] [CrossRef]
  178. Navarro, A.; Gomez, C.; López-Cepero, J.M.; Boveris, A. Beneficial effects of moderate exercise on mice aging: Survival, behavior, oxidative stress, and mitochondrial electron transfer. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 286, R505–R511. [Google Scholar] [CrossRef] [Green Version]
  179. Vollert, C.; Zagaar, M.; Hovatta, I.; Taneja, M.; Vu, A.; Dao, A.; Levine, A.; Alkadhi, K.; Salim, S. Exercise prevents sleep deprivation-associated anxiety-like behavior in rats: Potential role of oxidative stress mechanisms. Behav. Brain Res. 2011, 224, 233–240. [Google Scholar] [CrossRef]
  180. Falone, S.; D’Alessandro, A.; Mirabilio, A.; Petruccelli, G.; Cacchio, M.; Di Ilio, C.; Di Loreto, S.; Amicarelli, F. Long term running biphasically improves methylglyoxal-related metabolism, redox homeostasis and neurotrophic support within adult mouse brain cortex. PLoS ONE 2012, 7, e31401. [Google Scholar] [CrossRef] [Green Version]
  181. Radák, Z.; Sasvári, M.; Nyakas, C.; Taylor, A.W.; Ohno, H.; Nakamoto, H.; Goto, S. Regular training modulates the accumulation of reactive carbonyl derivatives in mitochondrial and cytosolic fractions of rat skeletal muscle. Arch. Biochem. Biophys. 2000, 383, 114–118. [Google Scholar] [CrossRef] [Green Version]
  182. Scopel, D.; Fochesatto, C.; Cimarosti, H.; Rabbo, M.; Belló-Klein, A.; Salbego, C.; Netto, C.A.; Siqueira, I.R. Exercise intensity influences cell injury in rat hippocampal slices exposed to oxygen and glucose deprivation. Brain Res. Bull. 2006, 71, 155–159. [Google Scholar] [CrossRef]
  183. Cechetti, F.; Rhod, A.; Simão, F.; Santin, K.; Salbego, C.; Netto, C.A.; Siqueira, I.R. Effect of treadmill exercise on cell damage in rat hippocampal slices submitted to oxygen and glucose deprivation. Brain Res. 2007, 1157, 121–125. [Google Scholar] [CrossRef]
  184. Qiao, D.; Hou, L.; Liu, X. Influence of intermittent anaerobic exercise on mouse physical endurance and antioxidant components. Br. J. Sports Med. 2006, 40, 214–218. [Google Scholar] [CrossRef] [PubMed]
  185. Acikgoz, O.; Aksu, I.; Topcu, A.; Kayatekin, B.M. Acute exhaustive exercise does not alter lipid peroxidation levels and antioxidant enzyme activities in rat hippocampus, prefrontal cortex and striatum. Neurosci. Lett. 2006, 406, 148–151. [Google Scholar] [CrossRef] [PubMed]
  186. Fry, R.W.; Morton, A.R.; Keast, D. Overtraining in athletes. An update. Sports Med. 1991, 12, 32–65. [Google Scholar] [CrossRef] [PubMed]
  187. Petibois, C.; Cazorla, G.; Poortmans, J.-R.; Déléris, G. Biochemical aspects of overtraining in endurance sports: The metabolism alteration process syndrome. Sports Med. 2003, 33, 83–94. [Google Scholar] [CrossRef] [PubMed]
  188. Ogonovszky, H.; Berkes, I.; Kumagai, S.; Kaneko, T.; Tahara, S.; Goto, S.; Radák, Z. The effects of moderate-, strenuous- and over-training on oxidative stress markers, DNA repair, and memory, in rat brain. Neurochem. Int. 2005, 46, 635–640. [Google Scholar] [CrossRef]
  189. Aksu, I.; Topcu, A.; Camsari, U.M.; Acikgoz, O. Effect of acute and chronic exercise on oxidant-antioxidant equilibrium in rat hippocampus, prefrontal cortex and striatum. Neurosci. Lett. 2009, 452, 281–285. [Google Scholar] [CrossRef]
  190. Leeuwenburgh, C.; Hansen, P.A.; Holloszy, J.O.; Heinecke, J.W. Hydroxyl radical generation during exercise increases mitochondrial protein oxidation and levels of urinary dityrosine. Free Radic. Biol. Med. 1999, 27, 186–192. [Google Scholar] [CrossRef]
  191. Ashton, T.; Rowlands, C.C.; Jones, E.; Young, I.S.; Jackson, S.K.; Davies, B.; Peters, J.R. Electron spin resonance spectroscopic detection of oxygen-centred radicals in human serum following exhaustive exercise. Eur. J. Appl. Physiol. 1998, 77, 498–502. [Google Scholar] [CrossRef]
  192. Chevion, S.; Moran, D.S.; Heled, Y.; Shani, Y.; Regev, G.; Abbou, B.; Berenshtein, E.; Stadtman, E.R.; Epstein, Y. Plasma antioxidant status and cell injury after severe physical exercise. Proc. Natl. Acad. Sci. USA 2003, 100, 5119–5123. [Google Scholar] [CrossRef] [Green Version]
  193. Liu, J.; Yeo, H.C.; Overvik-Douki, E.; Hagen, T.; Doniger, S.J.; Chyu, D.W.; Brooks, G.A.; Ames, B.N.; Chu, D.W. Chronically and acutely exercised rats: Biomarkers of oxidative stress and endogenous antioxidants. J. Appl. Physiol. 2000, 89, 21–28. [Google Scholar] [CrossRef]
  194. Mayer, E.A. Gut feelings: The emerging biology of gut-brain communication. Nat. Rev. Neurosci. 2011, 12, 453–466. [Google Scholar] [CrossRef] [PubMed]
  195. Collins, S.M.; Bercik, P. Gut microbiota: Intestinal bacteria influence brain activity in healthy humans. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 326–327. [Google Scholar] [CrossRef] [PubMed]
  196. Collins, S.M.; Denou, E.; Verdu, E.F.; Bercik, P. The putative role of the intestinal microbiota in the irritable bowel syndrome. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2009, 41, 850–853. [Google Scholar] [CrossRef]
  197. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70 (Suppl. 1), S38–S44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef] [Green Version]
  200. Sommer, F.; Bäckhed, F. The Gut Microbiota—Masters of Host development and physiology. Nat. Rev. Microbiol. 2013, 11, 227–238. [Google Scholar] [CrossRef]
  201. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [Green Version]
  202. Stilling, R.M.; Ryan, F.J.; Hoban, A.E.; Shanahan, F.; Clarke, G.; Claesson, M.J.; Dinan, T.G.; Cryan, J.F. Microbes & neurodevelopment—Absence of microbiota during early life increases activity-related transcriptional pathways in the amygdala. Brain. Behav. Immun. 2015, 50, 209–220. [Google Scholar] [CrossRef]
  203. Van de Wouw, M.; Stilling, R.M.; Peterson, V.L.; Ryan, F.J.; Hoban, A.E.; Shanahan, F.; Clarke, G.; Claesson, M.J.; Dinan, T.G.; Cryan, J.F.; et al. Host microbiota regulates central nervous system serotonin receptor 2C editing in rodents. ACS Chem. Neurosci. 2019, 10, 3953–3960. [Google Scholar] [CrossRef]
  204. Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef] [PubMed]
  205. Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L. The first microbial colonizers of the human gut: Composition, activities, and health implications of the infant gut microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the healthy gut microbiota composition? a changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. the international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  208. Arboleya, S.; González, S.; Salazar, N.; Ruas-Madiedo, P.; de los Reyes-Gavilán, C.G.; Gueimonde, M. Development of probiotic products for nutritional requirements of specific human populations. Eng. Life Sci. 2012, 12, 368–376. [Google Scholar] [CrossRef]
  209. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2017, 61, 1600240. [Google Scholar] [CrossRef] [Green Version]
  210. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of lactobacillus strain regulates emotional behavior and central gaba receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [Green Version]
  211. Akbari, E.; Asemi, Z.; Daneshvar Kakhaki, R.; Bahmani, F.; Kouchaki, E.; Tamtaji, O.R.; Hamidi, G.A.; Salami, M. Effect of probiotic supplementation on cognitive function and metabolic status in alzheimer’s disease: A randomized, double-blind and controlled trial. Front. Aging Neurosci. 2016, 8, 256. [Google Scholar] [CrossRef] [Green Version]
  212. Tamtaji, O.R.; Heidari-Soureshjani, R.; Mirhosseini, N.; Kouchaki, E.; Bahmani, F.; Aghadavod, E.; Tajabadi-Ebrahimi, M.; Asemi, Z. Probiotic and selenium co-supplementation, and the effects on clinical, metabolic and genetic status in alzheimer’s disease: A randomized, double-blind, controlled trial. Clin. Nutr. 2019, 38, 2569–2575. [Google Scholar] [CrossRef]
  213. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
  214. Messaoudi, M.; Violle, N.; Bisson, J.-F.; Desor, D.; Javelot, H.; Rougeot, C. Beneficial psychological effects of a probiotic formulation (lactobacillus helveticus R0052 and Bifidobacterium Longum R0175) in healthy human volunteers. Gut Microbes 2011, 2, 256–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Kazemi, A.; Noorbala, A.A.; Azam, K.; Eskandari, M.H.; Djafarian, K. Effect of Probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clin. Nutr. Edinb. Scotl. 2019, 38, 522–528. [Google Scholar] [CrossRef] [PubMed]
  216. Romijn, A.R.; Rucklidge, J.J.; Kuijer, R.G.; Frampton, C. A double-blind, randomized, placebo-controlled trial of lactobacillus helveticus and bifidobacterium longum for the symptoms of depression. Aust. N. Z. J. Psychiatry 2017, 51, 810–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Jadrešin, O.; Hojsak, I.; Mišak, Z.; Kekez, A.J.; Trbojevic, T.; Ivkovic, L.; Kolacek, S. Lactobacillus reuteri DSM 17938 in the treatment of functional abdominal pain in children: RCT study. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 925–929. [Google Scholar] [CrossRef] [PubMed]
  218. Akar, M.; Eras, Z.; Oncel, M.Y.; Arayici, S.; Guzoglu, N.; Canpolat, F.E.; Uras, N.; Oguz, S.S. Impact of oral probiotics on neurodevelopmental outcomes in preterm infants. J. Matern. Fetal Neonatal Med. 2017, 30, 411–415. [Google Scholar] [CrossRef] [PubMed]
  219. Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W.J. Psychobiotics and the manipulation of bacteria-gut-brain signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
  220. Barrett, E.; Ross, R.P.; O’Toole, P.W.; Fitzgerald, G.F.; Stanton, C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef]
  221. Roshchina, V.V. Evolutionary considerations of neurotransmitters in microbial, plant, and animal cells. Microb. Endocrinol. 2010, 874, 17–52. [Google Scholar]
  222. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  223. Han, A.; Sung, Y.-B.; Chung, S.-Y.; Kwon, M.-S. Possible additional antidepressant-like mechanism of sodium butyrate: Targeting the hippocampus. Neuropharmacology 2014, 81, 292–302. [Google Scholar] [CrossRef]
  224. Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Owen, L.; Reinders, M.; Narramore, R.; Marsh, A.R.; Lui, F.G.; Baron, R.; Plummer, S.; Corfe, B.M. A double blind, placebo controlled, randomised pilot trial examining the effects of probiotic administration on mood and cognitive function. Proc. Nutr. Soc. 2014, 73, E29. [Google Scholar] [CrossRef] [Green Version]
  226. Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315–320. [Google Scholar] [CrossRef] [PubMed]
  227. Mohammadi, A.A.; Jazayeri, S.; Khosravi-Darani, K.; Solati, Z.; Mohammadpour, N.; Asemi, Z.; Adab, Z.; Djalali, M.; Tehrani-Doost, M.; Hosseini, M.; et al. The effects of probiotics on mental health and hypothalamic-pituitary-adrenal axis: A randomized, double-blind, placebo-controlled trial in petrochemical workers. Nutr. Neurosci. 2016, 19, 387–395. [Google Scholar] [CrossRef]
  228. Takada, M.; Nishida, K.; Kataoka-Kato, A.; Gondo, Y.; Ishikawa, H.; Suda, K.; Kawai, M.; Hoshi, R.; Watanabe, O.; Igarashi, T.; et al. Probiotic lactobacillus casei strain shirota relieves stress-associated symptoms by modulating the gut-brain interaction in human and animal models. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2016, 28, 1027–1036. [Google Scholar] [CrossRef] [Green Version]
  229. Bambling, M.; Edwards, S.C.; Hall, S.; Vitetta, L. A combination of probiotics and magnesium orotate attenuate depression in a small ssri resistant cohort: An intestinal anti-inflammatory response is suggested. Inflammopharmacology 2017, 25, 271–274. [Google Scholar] [CrossRef]
  230. Colica, C.; Avolio, E.; Bollero, P.; Costa de Miranda, R.; Ferraro, S.; Sinibaldi Salimei, P.; De Lorenzo, A.; Di Renzo, L. Evidences of a new psychobiotic formulation on body composition and anxiety. Mediators Inflamm. 2017, 2017, 5650627. [Google Scholar] [CrossRef]
  231. Pinto-Sanchez, M.I.; Hall, G.B.; Ghajar, K.; Nardelli, A.; Bolino, C.; Lau, J.T.; Martin, F.-P.; Cominetti, O.; Welsh, C.; Rieder, A.; et al. Probiotic bifidobacterium longum ncc3001 reduces depression scores and alters brain activity: A pilot study in patients with irritable bowel syndrome. Gastroenterology 2017, 153, 448–459.e8. [Google Scholar] [CrossRef]
  232. Slykerman, R.F.; Hood, F.; Wickens, K.; Thompson, J.M.D.; Barthow, C.; Murphy, R.; Kang, J.; Rowden, J.; Stone, P.; Crane, J.; et al. Effect of lactobacillus rhamnosus hn001 in pregnancy on postpartum symptoms of depression and anxiety: A randomised double-blind placebo-controlled trial. EBioMedicine 2017, 24, 159–165. [Google Scholar] [CrossRef] [Green Version]
  233. Bagga, D.; Reichert, J.L.; Koschutnig, K.; Aigner, C.S.; Holzer, P.; Koskinen, K.; Moissl-Eichinger, C.; Schöpf, V. Probiotics drive gut microbiome triggering emotional brain signatures. Gut Microbes 2018, 9, 486–496. [Google Scholar] [CrossRef]
  234. Ghorbani, Z.; Nazari, S.; Etesam, F.; Nourimajd, S.; Ahmadpanah, M.; Jahromi, S.R. The effect of synbiotic as an adjuvant therapy to fluoxetine in moderate depression: A randomized multicenter trial. Arch. Neurosci 2018, 5, e60507. [Google Scholar] [CrossRef] [Green Version]
  235. Majeed, M.; Nagabhushanam, K.; Arumugam, S.; Majeed, S.; Ali, F. Bacillus coagulans MTCC 5856 for the management of major depression with irritable bowel syndrome: A randomised, double-blind, placebo controlled, multi-centre, pilot clinical study. Food Nutr. Res. 2018, 62, 1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Miyaoka, T.; Kanayama, M.; Wake, R.; Hashioka, S.; Hayashida, M.; Nagahama, M.; Okazaki, S.; Yamashita, S.; Miura, S.; Miki, H.; et al. Clostridium butyricum MIYAIRI 588 as Adjunctive therapy for treatment-resistant major depressive disorder: A prospective open-label trial. Clin. Neuropharmacol. 2018, 41, 151–155. [Google Scholar] [CrossRef] [PubMed]
  237. Okubo, R.; Koga, M.; Katsumata, N.; Odamaki, T.; Matsuyama, S.; Oka, M.; Narita, H.; Hashimoto, N.; Kusumi, I.; Xiao, J.; et al. Effect of bifidobacterium breve a-1 on anxiety and depressive symptoms in schizophrenia: A proof-of-concept study. J. Affect. Disord. 2019, 245, 377–385. [Google Scholar] [CrossRef] [PubMed]
  238. Lew, L.-C.; Hor, Y.-Y.; Yusoff, N.A.; Choi, S.-B.; Yusoff, M.S.; Roslan, N.S.; Ahmad, A.; Mohammad, J.A.; Abdullah, M.F.I.; Zakaria, N. Probiotic lactobacillus plantarum p8 alleviated stress and anxiety while enhancing memory and cognition in stressed adults: A randomised, double-blind, placebo-controlled study. Clin. Nutr. 2019, 38, 2053–2064. [Google Scholar] [CrossRef] [PubMed]
  239. Smith-Ryan, A.E.; Mock, M.G.; Trexler, E.T.; Hirsch, K.R.; Blue, M.N.M. Influence of a multistrain probiotic on body composition and mood in female occupational shift workers. Appl. Physiol. Nutr. Metab. 2019, 44, 765–773. [Google Scholar] [CrossRef]
  240. Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007, 61, 355–361. [Google Scholar] [CrossRef] [Green Version]
  241. Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain-Raspaud, S.; Trotin, B.; Naliboff, B.; et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 1394–1401.e4. [Google Scholar] [CrossRef] [Green Version]
  242. Steenbergen, L.; Sellaro, R.; van Hemert, S.; Bosch, J.A.; Colzato, L.S. A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain. Behav. Immun. 2015, 48, 258–264. [Google Scholar] [CrossRef] [Green Version]
  243. Marotta, A.; Sarno, E.; Del Casale, A.; Pane, M.; Mogna, L.; Amoruso, A.; Felis, G.E.; Fiorio, M. Effects of probiotics on cognitive reactivity, mood, and sleep quality. Front. Psychiatry 2019, 10, 164. [Google Scholar] [CrossRef]
  244. Diop, L.; Guillou, S.; Durand, H. Probiotic food supplement reduces stress-induced gastrointestinal symptoms in volunteers: A double-blind, placebo-controlled, randomized trial. Nutr. Res. 2008, 28, 1–5. [Google Scholar] [CrossRef] [PubMed]
  245. Allen, A.P.; Hutch, W.; Borre, Y.E.; Kennedy, P.J.; Temko, A.; Boylan, G.; Murphy, E.; Cryan, J.F.; Dinan, T.G.; Clarke, G. Bifidobacterium longum 1714 as a translational psychobiotic: Modulation of stress, electrophysiology and neurocognition in healthy volunteers. Transl. Psychiatry 2016, 6, e939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Kato-Kataoka, A.; Nishida, K.; Takada, M.; Kawai, M.; Kikuchi-Hayakawa, H.; Suda, K.; Ishikawa, H.; Gondo, Y.; Shimizu, K.; Matsuki, T.; et al. Fermented milk containing lactobacillus casei strain shirota preserves the diversity of the gut microbiota and relieves abdominal dysfunction in healthy medical students exposed to academic stress. Appl. Environ. Microbiol. 2016, 82, 3649–3658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Chong, H.X.; Yusoff, N.A.A.; Hor, Y.-Y.; Lew, L.-C.; Jaafar, M.H.; Choi, S.-B.; Yusoff, M.S.B.; Wahid, N.; Abdullah, M.F.I.L.; Zakaria, N.; et al. Lactobacillus plantarum DR7 alleviates stress and anxiety in adults: A randomised, double-blind, placebo-controlled study. Benef. Microbes 2019, 10, 355–373. [Google Scholar] [CrossRef]
  248. Ceccarelli, G.; Brenchley, J.M.; Cavallari, E.N.; Scheri, G.C.; Fratino, M.; Pinacchio, C.; Schietroma, I.; Fard, S.N.; Scagnolari, C.; Mezzaroma, I.; et al. Impact of high-dose multi-strain probiotic supplementation on neurocognitive performance and central nervous system immune activation of HIV-1 infected individuals. Nutrients 2017, 9, 1269. [Google Scholar] [CrossRef] [Green Version]
  249. Bagga, D.; Aigner, C.S.; Reichert, J.L.; Cecchetto, C.; Fischmeister, F.P.S.; Holzer, P.; Moissl-Eichinger, C.; Schöpf, V. Influence of 4-week multi-strain probiotic administration on resting-state functional connectivity in healthy volunteers. Eur. J. Nutr. 2019, 58, 1821–1827. [Google Scholar] [CrossRef] [Green Version]
  250. Chahwan, B.; Kwan, S.; Isik, A.; van Hemert, S.; Burke, C.; Roberts, L. Gut feelings: A randomised, triple-blind, placebo-controlled trial of probiotics for depressive symptoms. J. Affect. Disord. 2019, 253, 317–326. [Google Scholar] [CrossRef]
  251. Chung, Y.-C.; Jin, H.-M.; Cui, Y.; Jung, J.M.; Park, J.-I.; Jung, E.-S.; Choi, E.-K.; Chae, S.-W. Fermented milk of lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J. Funct. Foods 2014, 10, 465–474. [Google Scholar] [CrossRef]
  252. Kobayashi, Y.; Kinoshita, T.; Matsumoto, A.; Yoshino, K.; Saito, I.; Xiao, J.-Z. Bifidobacterium breve A1 supplementation improved cognitive decline in older adults with mild cognitive impairment: An open-label, single-arm study. J. Prev. Alzheimers Dis. 2019, 6, 70–75. [Google Scholar] [CrossRef]
  253. Kobayashi, Y.; Kuhara, T.; Oki, M.; Xiao, J.-Z. Effects of bifidobacterium breve A1 on the cognitive function of older adults with memory complaints: A randomised, double-blind, placebo-controlled trial. Benef. Microbes 2019, 10, 511–520. [Google Scholar] [CrossRef]
  254. Nishida, K.; Sawada, D.; Kuwano, Y.; Tanaka, H.; Rokutan, K. Health benefits of lactobacillus gasseri CP2305 tablets in young adults exposed to chronic stress: A randomized, double-blind, placebo-controlled study. Nutrients 2019, 11, 1859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Papalini, S.; Michels, F.; Kohn, N.; Wegman, J.; van Hemert, S.; Roelofs, K.; Arias-Vasquez, A.; Aarts, E. Stress matters: Randomized controlled trial on the effect of probiotics on neurocognition. Neurobiol. Stress 2019, 10, 100141. [Google Scholar] [CrossRef] [PubMed]
  256. Rudzki, L.; Ostrowska, L.; Pawlak, D.; Małus, A.; Pawlak, K.; Waszkiewicz, N.; Szulc, A. Probiotic lactobacillus plantarum 299v decreases kynurenine concentration and improves cognitive functions in patients with major depression: A double-blind, randomized, placebo controlled study. Psychoneuroendocrinology 2019, 100, 213–222. [Google Scholar] [CrossRef]
  257. Wang, H.; Braun, C.; Murphy, E.F.; Enck, P. Bifidobacterium longum 1714TM strain modulates brain activity of healthy volunteers during social stress. Am. J. Gastroenterol. 2019, 114, 1152–1162. [Google Scholar] [CrossRef] [PubMed]
  258. Giannetti, E.; Maglione, M.; Alessandrella, A.; Strisciuglio, C.; De Giovanni, D.; Campanozzi, A.; Miele, E.; Staiano, A. A Mixture of 3 bifidobacteria decreases abdominal pain and improves the quality of life in children with irritable bowel syndrome: A multicenter, randomized, double-blind, placebo-controlled, crossover trial. J. Clin. Gastroenterol. 2017, 51, e5–e10. [Google Scholar] [CrossRef] [PubMed]
  259. Kałużna-Czaplińska, J.; Błaszczyk, S. The level of arabinitol in autistic children after probiotic therapy. Nutrition 2012, 28, 124–126. [Google Scholar] [CrossRef]
  260. West, R.; Roberts, E.; Sichel, L.S.; Sichel, J. Improvements in gastrointestinal symptoms among children with autism spectrum disorder receiving the Delpro® probiotic and immunomodulator formulation. J. Prob. Health 2013, 1, 1–6. [Google Scholar]
  261. Shaaban, S.Y.; El Gendy, Y.G.; Mehanna, N.S.; El-Senousy, W.M.; El-Feki, H.S.A.; Saad, K.; El-Asheer, O.M. The role of probiotics in children with autism spectrum disorder: A prospective, open-label study. Nutr. Neurosci. 2018, 21, 676–681. [Google Scholar] [CrossRef]
  262. Liu, Y.-W.; Liong, M.T.; Chung, Y.-C.E.; Huang, H.-Y.; Peng, W.-S.; Cheng, Y.-F.; Lin, Y.-S.; Wu, Y.-Y.; Tsai, Y.-C. Effects of lactobacillus plantarum PS128 on children with autism spectrum disorder in taiwan: A randomized, double-blind, placebo-controlled trial. Nutrients 2019, 11, 820. [Google Scholar] [CrossRef] [Green Version]
  263. Kullisaar, T.; Songisepp, E.; Aunapuu, M.; Kilk, K.; Arend, A.; Mikelsaar, M.; Rehema, A.; Zilmer, M. Complete glutathione system in probiotic lactobacillus fermentum ME-3. Prikl. Biokhim. Mikrobiol. 2010, 46, 527–531. [Google Scholar] [CrossRef]
  264. Songisepp, E.; Kals, J.; Kullisaar, T.; Mändar, R.; Hütt, P.; Zilmer, M.; Mikelsaar, M. Evaluation of the functional efficacy of an antioxidative probiotic in healthy volunteers. Nutr. J. 2005, 4, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Chooruk, A.; Piwat, S.; Teanpaisan, R. Antioxidant activity of various oral lactobacillus strains. J. Appl. Microbiol. 2017, 123, 271–279. [Google Scholar] [CrossRef] [PubMed]
  266. Surya, A.; Liu, X.; Miller, M.J. Glutathione utilization in lactobacillus fermentum CECT 5716. J. Agric. Food Chem. 2018, 66, 12651–12656. [Google Scholar] [CrossRef] [PubMed]
  267. Peran, L.; Camuesco, D.; Comalada, M.; Nieto, A.; Concha, A.; Adrio, J.L.; Olivares, M.; Xaus, J.; Zarzuelo, A.; Galvez, J. Lactobacillus fermentum, a probiotic capable to release glutathione, prevents colonic inflammation in the TNBS model of rat colitis. Int. J. Colorectal Dis. 2006, 21, 737–746. [Google Scholar] [CrossRef]
  268. Robles-Vera, I.; Toral, M.; de la Visitación, N.; Sánchez, M.; Romero, M.; Olivares, M.; Jiménez, R.; Duarte, J. The probiotic lactobacillus fermentum prevents dysbiosis and vascular oxidative stress in rats with hypertension induced by chronic nitric oxide blockade. Mol. Nutr. Food Res. 2018, 62, e1800298. [Google Scholar] [CrossRef]
  269. Russo, M.; Fabersani, E.; Abeijón-Mukdsi, M.C.; Ross, R.; Fontana, C.; Benítez-Páez, A.; Gauffin-Cano, P.; Medina, R.B. Lactobacillus fermentum CRL1446 ameliorates oxidative and metabolic parameters by increasing intestinal feruloyl esterase activity and modulating microbiota in caloric-restricted mice. Nutrients 2016, 8, 415. [Google Scholar] [CrossRef] [Green Version]
  270. Robles-Vera, I.; Toral, M.; de la Visitación, N.; Aguilera-Sánchez, N.; Redondo, J.M.; Duarte, J. Protective effects of short-chain fatty acids on endothelial dysfunction induced by angiotensin II. Front. Physiol. 2020, 11, 277. [Google Scholar] [CrossRef] [Green Version]
  271. Srivastav, S.; Neupane, S.; Bhurtel, S.; Katila, N.; Maharjan, S.; Choi, H.; Hong, J.T.; Choi, D.-Y. Probiotics mixture increases butyrate, and subsequently rescues the nigral dopaminergic neurons from mptp and rotenone-induced neurotoxicity. J. Nutr. Biochem. 2019, 69, 73–86. [Google Scholar] [CrossRef] [Green Version]
  272. Xiang, S.; Ji, J.-L.; Li, S.; Cao, X.-P.; Xu, W.; Tan, L.; Tan, C.-C. Efficacy and safety of probiotics for the treatment of alzheimer’s disease, mild cognitive impairment, and parkinson’s disease: A systematic review and meta-analysis. Front. Aging Neurosci. 2022, 14, 730036. [Google Scholar] [CrossRef]
  273. Li, F.; Huang, H.; Wu, Y.; Lu, Z.; Zhou, X.; Tan, F.; Zhao, X. Lactobacillus fermentum HFY06 attenuates D-galactose-induced oxidative stress and inflammation in male kunming mice. Food Funct. 2021, 12, 12479–12489. [Google Scholar] [CrossRef]
  274. Song, X.; Zhao, Z.; Zhao, Y.; Wang, Z.; Wang, C.; Yang, G.; Li, S. Lactobacillus plantarum DP189 prevents cognitive dysfunction in D-Galactose/AlCl3 induced mouse model of alzheimer’s disease via modulating gut microbiota and PI3K/Akt/GSK-3β signaling pathway. Nutr. Neurosci. 2021, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
  275. Wang, L.; Zhao, Z.; Zhao, L.; Zhao, Y.; Yang, G.; Wang, C.; Gao, L.; Niu, C.; Li, S. Lactobacillus plantarum DP189 reduces α-SYN aggravation in MPTP-induced parkinson’s disease mice via regulating oxidative damage, inflammation, and gut microbiota disorder. J. Agric. Food Chem. 2022, 70, 1163–1173. [Google Scholar] [CrossRef] [PubMed]
  276. Athari Nik Azm, S.; Djazayeri, A.; Safa, M.; Azami, K.; Ahmadvand, B.; Sabbaghziarani, F.; Sharifzadeh, M.; Vafa, M. Lactobacilli and bifidobacteria ameliorate memory and learning deficits and oxidative stress in β-Amyloid (1-42) Injected rats. Appl. Physiol. Nutr. Metab. 2018, 43, 718–726. [Google Scholar] [CrossRef] [PubMed]
  277. Alipour Nosrani, E.; Tamtaji, O.R.; Alibolandi, Z.; Sarkar, P.; Ghazanfari, M.; Azami Tameh, A.; Taghizadeh, M.; Banikazemi, Z.; Hadavi, R.; Naderi Taheri, M. Neuroprotective effects of probiotics bacteria on animal model of parkinson’s disease induced by 6-hydroxydopamine: A behavioral, biochemical, and histological study. J. Immunoass. Immunochem. 2021, 42, 106–120. [Google Scholar] [CrossRef]
  278. Roy Sarkar, S.; Mitra Mazumder, P.; Chatterjee, K.; Sarkar, A.; Adhikary, M.; Mukhopadhyay, K.; Banerjee, S. Saccharomyces Boulardii ameliorates gut dysbiosis associated cognitive decline. Physiol. Behav. 2021, 236, 113411. [Google Scholar] [CrossRef]
Figure 1. Mechanisms mediating the beneficial effects of exercise on brain health. Physical activity improves cognitive function in ageing and neurodegenerative diseases by improving cerebrovascular function, reducing neuroinflammation, and promoting adult hippocampal neurogenesis. Figure created using BioRender software.
Figure 1. Mechanisms mediating the beneficial effects of exercise on brain health. Physical activity improves cognitive function in ageing and neurodegenerative diseases by improving cerebrovascular function, reducing neuroinflammation, and promoting adult hippocampal neurogenesis. Figure created using BioRender software.
Ijms 23 03610 g001
Figure 2. Mechanisms mediating the beneficial effects of specific training programmes on the muscle–brain axis. Resistance and aerobic training improves mitochondria function, immune system, and inflammatory response by improving mitochondria quality control, which leads to muscular adaptation, brain neurogenesis, and angiogenesis. Figure created using BioRender software.
Figure 2. Mechanisms mediating the beneficial effects of specific training programmes on the muscle–brain axis. Resistance and aerobic training improves mitochondria function, immune system, and inflammatory response by improving mitochondria quality control, which leads to muscular adaptation, brain neurogenesis, and angiogenesis. Figure created using BioRender software.
Ijms 23 03610 g002
Figure 3. Routes of communications between gut microbiota and brain and mechanisms through probiotics keep homeostasis between both systems. CNS: central nervous system; SNS: sympathetic nervous system; PSNS: parasympathetic nervous system; ENS: enteric nervous system; HPA: hypothalamic-pituitary-adrenal; CS: circulatory system; GABA: α-aminobutyric acid; SCFA: Short chain fatty acids; 5HT: serotonin; NE: norepinephrine; NH4+: ammonium; GLP-1: glucagon-like peptide -1; PYY: peptide YY; CCK: cholecystokinin; IL: interleukin; TNF-α: tumor necrosis factor α; TLR: toll-like receptors; FXR: bile acid farnesoid X receptor; TGR5: bile acid Takeda G-protein-coupled receptor 5; GPR41/43: G protein-coupled receptors 41/43; 5HTa R: serotonin receptor. Figure created using BioRender software.
Figure 3. Routes of communications between gut microbiota and brain and mechanisms through probiotics keep homeostasis between both systems. CNS: central nervous system; SNS: sympathetic nervous system; PSNS: parasympathetic nervous system; ENS: enteric nervous system; HPA: hypothalamic-pituitary-adrenal; CS: circulatory system; GABA: α-aminobutyric acid; SCFA: Short chain fatty acids; 5HT: serotonin; NE: norepinephrine; NH4+: ammonium; GLP-1: glucagon-like peptide -1; PYY: peptide YY; CCK: cholecystokinin; IL: interleukin; TNF-α: tumor necrosis factor α; TLR: toll-like receptors; FXR: bile acid farnesoid X receptor; TGR5: bile acid Takeda G-protein-coupled receptor 5; GPR41/43: G protein-coupled receptors 41/43; 5HTa R: serotonin receptor. Figure created using BioRender software.
Ijms 23 03610 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martínez-Guardado, I.; Arboleya, S.; Grijota, F.J.; Kaliszewska, A.; Gueimonde, M.; Arias, N. The Therapeutic Role of Exercise and Probiotics in Stressful Brain Conditions. Int. J. Mol. Sci. 2022, 23, 3610. https://doi.org/10.3390/ijms23073610

AMA Style

Martínez-Guardado I, Arboleya S, Grijota FJ, Kaliszewska A, Gueimonde M, Arias N. The Therapeutic Role of Exercise and Probiotics in Stressful Brain Conditions. International Journal of Molecular Sciences. 2022; 23(7):3610. https://doi.org/10.3390/ijms23073610

Chicago/Turabian Style

Martínez-Guardado, Ismael, Silvia Arboleya, Francisco Javier Grijota, Aleksandra Kaliszewska, Miguel Gueimonde, and Natalia Arias. 2022. "The Therapeutic Role of Exercise and Probiotics in Stressful Brain Conditions" International Journal of Molecular Sciences 23, no. 7: 3610. https://doi.org/10.3390/ijms23073610

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

Article Metrics

Back to TopTop