Next Article in Journal
Locomotor Coordination, Visual Perception and Head Stability during Running
Next Article in Special Issue
Increased Time Difference between Imagined and Physical Walking in Older Adults at a High Risk of Falling
Previous Article in Journal
Neurobiological Trajectories Involving Social Isolation in PTSD: A Systematic Review
Previous Article in Special Issue
Mental Imagery and Acute Exercise on Episodic Memory Function
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Contribution of Functional Magnetic Resonance Imaging to the Understanding of the Effects of Acute Physical Exercise on Cognition

1
Research Group Neuroprotection, German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany
2
Department of Neurology, Medical Faculty, Otto von Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
3
Institute III, Department of Sport Science, Otto von Guericke University Magdeburg, Zschokkestr. 32, 39104 Magdeburg, Germany
4
Center for Behavioral Brain Sciences (CBBS), Brenneckestraße 6, 39118 Magdeburg, Germany
*
Author to whom correspondence should be addressed.
Submission received: 3 December 2019 / Revised: 4 March 2020 / Accepted: 13 March 2020 / Published: 18 March 2020
(This article belongs to the Special Issue Exercising against Age-Effects on the Brain)

Abstract

:
The fact that a single bout of acute physical exercise has a positive impact on cognition is well-established in the literature, but the neural correlates that underlie these cognitive improvements are not well understood. Here, the use of neuroimaging techniques, such as functional magnetic resonance imaging (fMRI), offers great potential, which is just starting to be recognized. This review aims at providing an overview of those studies that used fMRI to investigate the effects of acute physical exercises on cerebral hemodynamics and cognition. To this end, a systematic literature survey was conducted by two independent reviewers across five electronic databases. The search returned 668 studies, of which 14 studies met the inclusion criteria and were analyzed in this systematic review. Although the findings of the reviewed studies suggest that acute physical exercise (e.g., cycling) leads to profound changes in functional brain activation, the small number of available studies and the great variability in the study protocols limits the conclusions that can be drawn with certainty. In order to overcome these limitations, new, more well-designed trials are needed that (i) use a more rigorous study design, (ii) apply more sophisticated filter methods in fMRI data analysis, (iii) describe the applied processing steps of fMRI data analysis in more detail, and (iv) provide a more precise exercise prescription.

1. Introduction

In Western societies, technological advances constantly lower the need to be physically active in every-day life situations (e.g., transportation to the workplace or physical demands during work) [1,2,3,4]. Hence, it is not surprising that the time spent physically inactive and sedentary has increased dramatically [5,6,7]. In the long term, rising levels of physical inactivity are expected to cause serious health problems [8]. For instance, sedentary behavior [9,10] and physical inactivity [11,12] are associated with worsened cognitive functions and a higher risk of neurodegenerative diseases (e.g., dementia) in the aging population [13,14,15,16,17,18,19,20,21,22]. In order to avoid periods of prolonged physical inactivity and sedentary behavior (e.g., sedentism during office working day), it is advised to take physical activity breaks ([23,24,25]. There is substantial evidence in the literature showing that acute (single) bouts of physical exercise (defined as planned and structured form of distinct physical activities [26,27,28,29]) improve cognitive domains, such as attention and/or cognitive control substantially, albeit only transiently [30,31,32,33]. However, the underlying neurobiological mechanisms of these effects are not yet fully understood. In this regard, the use of neuroimaging methods offers great potential for acquiring a deeper understanding of physical exercise-induced changes in the neural correlates of cognition, such as changes in functional brain activation [33,34,35]. The most common methods used to investigate effects on functional brain activation are functional near-infrared spectroscopy (fNIRS) [34] and electroencephalography (EEG) [36,37]; however, also functional magnetic resonance imaging (fMRI) has recently been applied in the context of acute physical exercise and cognition [38,39]. The strengths of fNIRS and EEG compared to fMRI are a higher temporal resolution, greater portability, and applicability in almost all cohorts (e.g., for individuals with metallic implants or claustrophobia) [34,40,41]. However, fNIRS and EEG have a limited spatial resolution and only allow for the evaluation of brain activation patterns in cortical areas [41,42,43]. In comparison to fNIRS and EEG, fMRI enables the assessment of brain activation changes in cortical and subcortical areas and offers a higher spatial resolution, which results in superior source localization [42,43,44]. Hence, fMRI is well suited to study the influence of acute physical exercise on subcortical structures, such as the hippocampus, which have a crucial role in cognitive processes (e.g., memory) [45,46,47,48,49]. Since hippocampal activation during cognitive tasks can distinguish between individuals with cognitive impairment and healthy controls [50,51,52], the investigation of the influence of acute physical exercise on these subcortical structures may help to develop more efficient physical intervention strategies with regard to neuroprotection. Furthermore, given the ability of fMRI to measure both subcortical and cortical activation, the application of fMRI prior to and after an acute bout of physical exercise is also useful to complement the results obtained by EEG and/or fNIRS.
fMRI brain (neural) activity is indirectly assessed by using the magnitude of venous blood oxygenation level-dependent contrast (BOLD) [53,54,55,56,57,58,59]. While BOLD contrast has been shown to reflect neural activity [60,61], this activity is also influenced by blood volume, oxygenation, and changes in regional blood flow [57,59,62]. An increase in neuronal activity (e.g., as a consequence of solving a cognitive task) induces higher neuronal energy metabolism, in which oxygen is consumed to produce energy in order to satisfy the energetic demands of neural tissue (neurometabolic coupling). This leads to a local decrease in the concentration of oxygenated hemoglobin and a local increase in the concentration of deoxygenated hemoglobin [58,63,64,65,66,67]. Furthermore, an increase in neural activity also stimulates local changes in cerebral hemodynamics, which trigger intensified blood flow to the activated brain regions (neurovascular coupling). An increase in the local cerebral blood flow leads to an increased washout of deoxygenated hemoglobin and ensures that an appropriate level of oxygen is supplied to the activated neuronal tissue [65,66,67,68,69]. A schematic illustration of these neurobiological mechanisms is provided in Figure 1.
Based on the paramagnetic properties of deoxyhemoglobin, neural activity-dependent changes in the concentration of deoxygenated hemoglobin influence the local reduction in main field homogeneity and, thus, the magnitude of the BOLD contrast [57,58,70]. Changes in the BOLD signal can be analyzed with different methods, such as task-based fMRI analyses (e.g., BOLD changes obtained during a cognitive task) or resting-state fMRI analyses (e.g., BOLD changes obtained while performing no explicit task). Recently, several studies using task-based fMRI analyses [38,71,72] and resting-state fMRI analyses [73] were performed to investigate the changes in neural correlates driving cognitive improvements after acute physical exercise. While there are systematic reviews available that provide recommendations with regard to acute exercise [33] or fMRI [62,74,75], no systematic review has pooled the methodology and findings of fMRI studies dealing with acute physical exercise and cognition. As the application of fMRI in the field of acute physical exercise and cognition poses specific challenges to the study design and analysis of fMRI signals (e.g., the influence of systemic artifacts, the influence of temporal window between exercise cessation, and an fMRI scan), previously published recommendations might not be fully transferable. Hence, this literature survey aims to systematically summarize the methodological details and findings of all studies using fMRI to investigate the influence of acute physical exercise on cognition. Based on the results of this systematic review, recommendations for future studies are derived.

2. Methods

In this systematic review, we followed the recommendations provided in the PRISMA statement (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) [76]. Two independent researchers performed a systematic literature search in the following five electronic databases (applied specifications): Pubmed (all fields), Scopus (title, abstract, keywords), Web of Science (all fields), PsycInfo (all fields), and SportDiscus (all fields) on 3rd March 2020. In this process, the following terms were used as search strings:
Search strategy
  • “acute exercis*” OR “acute aerobic” OR “acute strength” OR “acute resistance” OR “acute effects of exercis*” OR “single bout of exercis*” OR “acute bout of physical activity” OR “single bout of physical activity” OR “acute physical activity” OR “physical activity break”
AND
  • mental OR neuropsychological OR brain OR cogniti* OR neurocogni* OR executive OR attention OR memory OR “response time” OR “reaction time” OR accuracy OR error OR inhibition OR visual OR spatial OR visuospatial OR processing OR recall OR learning OR language OR oddball OR “task switching” OR “problem solving” OR Flanker OR Stroop OR Sternberg
AND
  • fMRI OR MRI OR “MR imaging” OR “magnetic resonance imaging”
Afterward, the results of the systematic search were loaded into a citation manager (Citavi 6.3), and the duplicates were removed (see Figure 2).

2.1. Inclusion and Exclusion Criteria

To screen for relevant studies, the established PICOS-principle was applied [76,77]. “PICOS” stands for participants (P), intervention (I), comparisons (C), outcomes (O), and study design (S) [76,77]. In the current systematic review, the following inclusion and exclusion criteria were used: (P) we applied no restrictions and included all age groups regardless of pathologies; (I) only acute physical exercise interventions were considered; (C) in this systematic literature search, no specific restrictions were used; (O) studies were considered relevant if they assessed changes in functional brain activation using fMRI (resting-state fMRI studies were included when they investigated the relationship between resting-state measures and cognitive performance). As cognition comprises a wide range of mental abilities that allow us, for example, to perceive, process, and retain information, as well as interact with our environment [78,79], it is necessary to classify the cognitive domains included in this systematic review. We included studies utilizing tests for cognitive domains such as attention, cognitive control, and memory [30,33] and excluded studies investigating solely perception (e.g., pain perception) or motor learning. (S) Only interventional studies that fulfill the previously stated criteria and have been published in a peer-reviewed journal were considered.
As shown in Figure 2, 16 studies were excluded after full-text screening because they did not meet our inclusion criteria. In particular, six studies did not perform neuroimaging using fMRI [80,81,82] or applied other neuroimaging techniques (e.g., functional near-infrared spectroscopy) [83,84,85]. The remaining 10 studies did not perform cognitive testing [86,87,88,89,90,91,92,93,94] or did not report data because they describe a study protocol [95].

2.2. Data Extraction

We extracted information about the first author, year of publication, population characteristics including health status, cardiorespiratory fitness level, age, gender, and anthropometric data (e.g., body weight, body height, and body-mass index [BMI]). Furthermore, details about exercise characteristics (e.g., type of physical exercise, exercise intensity, and exercise duration), cognitive testing (e.g., tested cognitive domain and administration after exercise cessation), and (pre-)processing of fMRI data (e.g., filter cut-off frequencies) were extracted. Moreover, information about the main behavioral findings and neuroimaging findings were gathered from the studies considered to be relevant. In addition, we extracted the x-y-z coordinates of significant exercise-related functional hemodynamic changes (if possible) and performed automatic anatomical labeling using AAL2 [96] to obtain uniform neuroanatomical labeling across the reviewed studies. In three studies, extraction of the exact x-y-z coordinates was not possible [38,72,73].

2.3. Risk of Bias Assessment

As recommended in the PRISMA statement, the methodological quality of the included studies and their risk of bias was assessed (see Figure 3) [76]. In this process, two independent evaluators used the Cochrane Collaboration’s Risk of Bias tool and rated the risk of bias as being “low”, “high”, or “unclear” [97]. Any discrepancies between the two evaluators regarding the ratings of the risk of bias in a particular study were resolved by a discussion among them or through consultation with a third author of the review [97].

3. Results

3.1. Risk of Bias

As shown in Figure 3, in the domains of sequence generation, allocation concealment, blinding of participants and personnel, and blinding of outcome assessments, all studies were rated as having an unclear risk of bias because the applied procedures were not described in sufficient detail. In the domains of incomplete outcome data and selective reporting, all studies were judged as having a low risk of bias. Furthermore, with regard to the domain of “other bias”, the majority of studies were judged as having an unclear bias because the study design did not meet recent recommendations [33] (e.g., it used only a posttest comparison [38,39,71,73,99,100,101,102,103,104] and/or the prescription of exercise intensity was not optimal because it was not based on a graded exercise test [39,71,72,73,103,104,105]).

3.2. Study Characteristics

In the reviewed studies, the acute effects of physical exercise were studied in healthy children [71,73], adolescents with and without bipolar disorder [105], younger adults with attention deficit hyperactivity disorder (ADHD) [100,102], healthy younger adults [38,39,72,99,101,102,106], and healthy older adults [103,104,107]. A detailed overview of the sample characteristics (e.g., age, gender, BMI, and cardiorespiratory fitness level) can be found in Table 1. The sample size in the reviewed studies varied between 9 [71,73,99] and 34 participants [107]. In particular, the following sample sizes were used: < 10 participants [71,73,99], between 10 to 20 participants [38,39,72,99,102,106], and > 20 participants [100,101,103,104,105,107].
Twelve of the studies were conducted in a within-subject design [38,39,71,72,73,100,101,102,103,104,106,107] and six studies used a between-subjects design [99,100,101,102,105,106]. Furthermore, in four studies, a cognitive test using fMRI was performed prior to and after cycling [72,105,106,107], whereas in 10 studies, testing was only performed after the cessation of the intervention conditions (e.g., cycling or seated rest/watching movie) [38,39,71,73,100,101,102,103,104,106]. In these 10 studies, the order of the separate testing sessions was counterbalanced [38,39,71,73,100,101,102,103,104,106]. Furthermore, the testing sessions were separated by a time interval of at least two days [38,100,101,102], seven days [39,71,73,106], or twelve days [103], and four studies reported that the testing sessions were performed at the same time of day [38,71,73,106]. After exercise cessation, the cognitive tests were administered based on the following criteria: (i) after 5 min [38], (ii) after 10 min [103], (iii) after approximately 15 min [72,104], (iv) within 15 min [39], (v) after 15.1 ± 4.3 min (moderate-intensity conditions) or 16.5 ± 5.1 min (light-intensity conditions) [107], (vi) after 20 min [106], (vii) 20.9 ± 1.8 min [100], (viii) after 25 min [105], (ix) approximately 63:38 min after exercise cessation [99], or (x) after reaching a level within 10% of the subject’s resting heart rate [71,73].
To test the cognitive performance of participants, in one study, a monetary incentive delay task [99], a mnemonic discrimination task [38], and a semantic memory task were implemented [104]. In two studies, a sustained attention task [72,105] or a checkerboard task [101,102] were applied. Four studies utilized the Flanker task [73,101,102,103] to test executive performance. In three studies a Go/No-Go task was used [100,101,102]. In four studies, working memory performance was examined by the N-Back paradigm [39,71,106,107]. In seven studies, cognitive testing was conducted using a block design [39,71,72,101,102,105,106], and in eight studies, an event-related design was applied [38,99,100,101,102,103,104,107].

3.3. Exercise Characteristics

All of the reviewed studies conducted an endurance exercise outside an fMRI scanner using a stationary cycling ergometer [38,39,71,72,73,100,101,102,103,104,105,106,107] or a treadmill [99]. In six studies, a graded exercise test was performed to determine the cardiorespiratory fitness level [38,99,100,101,102,107], and in one study, the cardiorespiratory fitness level was assessed via an indirect maximal oxygen uptake test [106]. The exercise intensity was set to HR 60–70% of the predicted maximal heart rate (HRmax) [39,71,72,73], 60–80% of the predicted HRmax [105], 50–70% of the measured HRmax [100,101,102], 65% of the measured HRmax [107], >70% of the measured HRmax [101], 30% of the subject’s VO2 peak [38], or 60–70% of the subject’s VO2 max [99]. In two studies, the Borg scale was used as a subjective rating of the perceived exertion, which ranges from 6 (no exertion) to 20 (maximal exertion) [108]; a rating of 15 (hard) was used to set the exercise intensity [103,104]. In five studies, HRmax was predicted using the formula ‘220 – age in years’ [39,71,72,73,105], and one study used the Karvonen formula to calculate the target HR for cycling [106].
The exercise duration was set to 10 min [38], 20 min [39,72,103,104,105,106,107], 21 min [101], or 30 min [71,73,99,100,101,102]. The durations of the warm-up were set to 2 min [71,73] or 5 min [39,100,101,103,104,105,106], whereas the period of cool-downs lasted 2 min [105], 3 min [71,73], 4 min [101], or 5 min [39,103,104,106,107].

3.4. fMRI Characteristics and Data Processing

The majority of the studies used a 3.0 T MR scanner [38,39,71,72,73,100,101,102,103,104,105,106,107] equipped with 8 channel head coils [72,105], 12 channel head coils [100,101,102,107], 32 channel head coils [38,39,103,104,106,107], or 64 channel head coils [100,101,102]. To process the data, SPM 8 (statistical parametric mapping) [39,99], SPM 12 [100,101,102,106], DPABI (a toolbox for Data Processing and Analysis for Brain Imaging) [71,73], FSL (Functional MRI of the Brain Software Library) [72,105,107], or AFNI (Analysis of Functional NeuroImages) were applied [38,103,104,107]. Using these software packages, the reviewed studies commonly conducted the following “basic” processing steps: (i) rejection of the first 3 [71,105], first 5 [39,104], first 6 [103], first 10 [73], or first 12 images [106]; (ii) slice-timing correction [38,39,71,73,99,103,106]; (iii) realignment (motion correction) [39,71,73,99,100,101,102,103,105,106]; (iv) intra-subject and inter-modal registration (e.g., to T1-weighted image) and spatial normalization (e.g., to MNI space) [38,39,71,72,73,99,100,101,102,103,104,105,106,107], and (v) spatial smoothing using a Gaussian kernel with 2 mm [38], 4 mm [104], 6 mm [105,107], 7 mm [99], or 8 mm at full width at half maximum (FWHM) [39,71,72,73,100,101,102,106]. In addition to the mentioned “basic” processing steps, in several of the reviewed studies, more sophisticated filter methods were applied in order to account for artifacts. In four studies, high-pass filters using cut-off frequencies of 1/128 Hz [99,100,101,102] or 1/60 Hz [105] were applied, whereas in one study, a bandpass filter with a range of 0.008–0.08 Hz was used [107] to reduce the influence of specific sources of noise (e.g., high-frequency physiological artifacts or non-physiological low-frequency artifacts introduced by scanner drift). In two studies and one study, respectively, an AR(1) model [100,101] and an AR(2) model [102] were used to account for temporal autocorrelation. Furthermore, to correct motion-related artifacts, sophisticated motion correction methods, such as the MCFLIRT algorithm [105], slice-oriented motion correction algorithm (SLOMOCO) [103], or an independent-component analysis (ICA)-based approach for automatic removal of the motion artifacts (i.e., ICA-AROMA) [107] were used. A nuisance regression was conducted in several of the reviewed studies to regress out signals from the motion-related artefacts (e.g., [six-rigid] body [head] movements) [38,73,99,100,101,102] or physiological artifacts arising from the white matter [38,73,107] or cerebrospinal fluid [38,73,107]. In addition, three studies monitored physiological parameters, such as heart rate [38,72,107] and blood pressure, during fMRI scans [72].
The majority of the studies analyzed task-based brain activation patterns using the general linear model approach [38,39,71,72,99,100,101,102,104,105,106]. Furthermore, in the reviewed studies, functional connectivity was assessed during resting-state utilizing a seed-based approach [73] or a cross-correlation approach [107]; during a cognitive task, a psychophysiological interaction approach (PPI) was used [38].

3.5. Findings

(i) Main findings with regard to behavioral changes
In comparison to the control condition, significant exercise-induced improvements were observed (i) in executive functioning and working memory performance in healthy children after 30 min of moderate-intensity cycling [71,73], (ii) in a mnemonic discrimination task in healthy younger adults after 10 min of moderate-intensity cycling [38], (iii) in executive functioning in younger adults with ADHD after 30 min of moderate-intensity cycling [102], (iv) in working memory tasks in younger adults with low cardiorespiratory fitness level after 20 min of moderate-intensity cycling [106], (v) in executive functioning in healthy younger adults after circa 30 min of moderate-intensity and vigorous-intensity cycling [101], (vi) in a working memory task in healthy older adults after 20 min of light-intensity and moderate-intensity cycling [107], and (vii) in executive functioning in healthy older adults after 20 min of vigorous-intensity cycling [103].
In contrast, compared to the control conditions, non-significant changes in performance were observed (i) in an attentional task in adolescents with and without bipolar disorder after 20 min of moderate-intensity cycling [105], (ii) in an attentional task in healthy younger adults after 20 min of moderate-intensity cycling [72], (iii) in a working memory task in healthy younger adults after 20 min of moderate-intensity cycling [39], (iv) in a Go/No-Go task in younger adults with and without ADHD after 30 min of moderate-intensity cycling [100], (v) in a reward processing task in healthy younger adults after 30 min of moderate-to-vigorous treadmill running [99], and (vi) in a memory task in healthy older adults after 20 min of vigorous-intensity cycling [104]. A more detailed overview of the main findings regarding exercise-induced changes in behavioral performance is provided in Table 1.
(ii) Main findings with regard to changes in functional brain hemodynamics
While the evidence regarding behavioral changes is mixed, significant changes in functional brain hemodynamics in response to acute physical exercise are routinely observed (for a detailed overview, see Table 1). These exercise-induced functional hemodynamic brain changes were primarily found in the frontal (e.g., inferior and superior frontal gyrus) and temporal lobes (e.g., temporal gyrus, fusiform gyrus, and hippocampus). In particular, a higher activation (compared to the control condition) was observed (i) in healthy younger adults in response to 20 min of moderate-intensity cycling in the right middle frontal gyrus, the right lingual gyrus, and the left fusiform gyrus during a 2-back task [39]; (ii) in healthy younger adults in response to 30 min of moderate-intensity cycling in the left superior frontal gyrus, in the right precentral gyrus, and in the triangular part of the left inferior frontal gyrus during a Go/No-Go task [101]; (iii) in healthy younger adults in response to 30 min of high-intensity interval exercise in the left lingual gyrus and in the precuneus during a visual task [101]; (iv) in younger adults with ADHD in response to 30 min of moderate-intensity cycling in left and right middle occipital gyrus, right supramarginal gyrus, and left inferior parietal gyrus during correct inhibitions in a Go/No-Go task [100]; and (v) in healthy older adults in response to 20 min of moderate-to-vigorous-intensity cycling in the orbital part of the left inferior frontal gyrus, in the left inferior temporal gyrus, in the right middle temporal gyrus, in the left fusiform gyrus, and in the bilateral hippocampus during a semantic memory task [104], as well as in the orbital part of the left inferior gyrus and in the left inferior parietal gyrus during a Flanker task [103]. In contrast, a lower activation of the structures of the frontal and temporal lobe was observed (i) in adolescents with bipolar disorder after 20 min of moderate-intensity cycling in the orbital part of the left inferior frontal gyrus, the right frontal pole extending to temporal pole, the bilateral hippocampus, and the right amygdala during a Go/No-Go task [105] and (ii) in younger adults with ADHD after 30 min of moderate-intensity cycling in the right precentral gyrus, the right middle temporal gyrus, the left superior frontal gyrus, the right middle frontal gyrus, and the paracentral lobule during a Flanker task, and in the right superior frontal gyrus and the right middle frontal gyrus during a visual task [102]. However, especially in the cingulate gyrus, exercise-related deactivations were also observed in healthy adults. In particular, a significantly lower activation was observed in healthy younger adults after 20 min of moderate-intensity cycling in the anterior cingulate gyrus [39,106], the right paracentral lobule, and the left inferior frontal gyrus during a working memory task [39], and in the left parietal operculum during an attentional task [72]. Furthermore, in healthy younger adults, 30 min of moderate-intensity cycling led to a decrease of the activity of the left anterior cingulate gyrus during a visual task [101], whereas 30 min of moderate-to-vigorous treadmill running decreased the activation of the caudate nucleus during a reward processing task [99]. In healthy older adults, 20 min of moderate-to-vigorous-intensity cycling led to exercise-related deactivations in the right anterior cingulate gyrus during a Flanker task [103].
Furthermore, we found evidence in the reviewed studies that different exercise protocols (continuous cycling at moderate intensity versus high-intensity interval cycling) [101], as well as the cardiorespiratory fitness level [106] and sex of the participants [101], influence exercise-related changes in functional brain hemodynamics (see Table 1 for a detailed overview).
With regard to the effects of different exercise protocols, it was observed that the left superior frontal gyrus and right insula showed higher activation in the moderate-intensity group during a Go/No-Go task, whereas in the high-intensity group, the left lingual gyrus, the right precuneus, and the left anterior cingulate cortex exhibited higher activation during a visual task [101].
Regarding the effects of cardiorespiratory fitness level (CRF), fitness level-dependent changes mainly manifested in the different activation patterns of the cerebellum. In particular, in the group with a high level of CRF, the activation of the right cerebellum was higher before cycling than after cycling (0-back and 1-back condition), whereas the activation of the left anterior cingulate gyrus and the left globus pallidus (a 2-back condition) was higher after cycling compared to before cycling [106]. In the group with a low level of CRF, the activation of the right cerebellum was higher after cycling compared to before cycling (0-back and 1-back condition), while the activation of the left anterior cingulate gyrus was lower after cycling compared to before cycling (2-back condition) [106]. The group comparison of the high-fit individuals and the low-fit individuals revealed that in the younger adults with a high level of CRF, the right cerebellum showed higher activation before cycling (0-back and 1-back condition), while the left anterior cingulate gyrus and the left globus pallidus exhibited lower activation before cycling (2-back condition) [106]. In comparison to the younger adults with a low level of CRF, the high-fit younger adults exhibited a lower activation of the right cerebellum (0-back and 1-back condition) and higher activations of the left anterior cingulate gyrus and the left globus pallidum (2-back condition) after cycling [106].
With respect to sex-specific differences in the functional brain activation patterns, male participants in the moderate-intensity group showed higher activation of the right precentral gyrus during a Go/No-Go task compared to female participants [101].
In addition, in children, an exercise-related increase after 30 min of moderate-intensity cycling was observed in the left superior/inferior parietal gyrus, the right superior parietal gyrus, the left hippocampus, and the bilateral cerebellum during a working memory task [71].
Suwabe et al. [38], who utilized the PPI approach to investigate the effects of 10 min of moderate-intensity cycling on task-based connectivity patterns, observed an increase in the functional connectivity between the hippocampal subfields dentate gyrus (DG)/CA3 and the cortical areas located in the temporal lobe (i.e., parahippocampal cortex and fusiform gyrus) and the parietal lobe (i.e., angular gyrus) [38]. In particular, positive associations between the hippocampus and the left angular gyrus, the left fusiform gyrus, the left parahippocampal cortex, and the left primary visual cortex were observed, while negative associations between the hippocampus and the left temporal pole were noted [38].
(iii) Neurobiobehavioral associations
In healthy children, it was observed that after 30 min of moderate-intensity cycling, the increment of the resting-state functional connectivity between the left cerebellum and the right inferior frontal gyrus was negatively correlated with better cognitive performance [73].
Furthermore, in a cohort of healthy younger adults, Mehren et al. [101] observed positive correlations between cardiorespiratory fitness levels and activation of the right insula (during Flanker task) and the left rolandic operculum (during a Go/No-Go task) in the moderate-intensity group. In the same study, a negative correlation between cardiorespiratory fitness level and the activation of the right postcentral gyrus (during a Go/No-Go task) in the high-intensity interval group was also observed [101]. In addition, in healthy younger adults, it was noticed that higher correlations between the hippocampal subfield DG/CA3 and the angular gyrus, the fusiform gyrus, and the parahippocampal cortex predicted higher performance in the mnemonic discrimination task [38].
In older adults, Voss et al. [107] observed that acute changes in behavioral performance and resting-state functional connectivity in the hippocampal-cortical connections in response to 20 min of moderate-intensity cycling can predict long-term training-related neurocognitive changes (i.e., neurocognitive changes after 12 weeks of aerobic training). For instance, the authors provide evidence that exercise-related changes of the right-lateralized fronto–parietal connections are related to acute and long-term gains in working memory performance [107]. A more detailed overview of the main findings is provided in Table 1.

4. Discussion

4.1. Risk of Bias

Overall, in most domains, the included studies were judged to have an unclear risk of bias (see Figure 3). In particular, in the domains of sequence generation, allocation concealment, blinding of participants and personnel, and blinding of outcome assessment, all included studies were rated as having an unclear risk of bias because they did not describe the used procedures in sufficient detail. Hence, we recommend that future studies should follow established guidelines (e.g., the CONSORT statement [112,113]) and report their study procedures in sufficient detail. Notably, the blinding of participants in exercise studies is exceedingly difficult because the participants notice if they are exercising or not. To minimize the risk of bias arising from not blinding participants (e.g., due to expectation effects), it is recommended that future studies should control appropriately for confounders, for instance, by assessing additional biopsychosocial variables (e.g., sleep, level of arousal, mood) [114,115,116,117,118] and/or a placebo group [116] (e.g., the placebo [sham] group performs the same exercise but without loading [119,120] or an inadequate dose to induce considerable effects [121]) when possible. Furthermore, with respect to the domain of “other bias”, we recommend that upcoming studies pay stronger attention to developing a more rigorous study design (see the next Section 4.2, Study characteristics, for a detailed discussion, as well as Figure 4) [33] and better exercise prescription (see Section 4.3, Exercise characteristics, for a detailed discussion).

4.2. Study Characteristics

An appropriate study design is crucial when the effects of acute physical exercise on cognition and their underlying neurobiological correlates are to be investigated (e.g., changes in functional brain activation). In the majority of the reviewed studies, a within-subject design with crossover posttest comparisons was used to contrast the brain activation patterns obtained after rest and after an acute bout of physical exercise. Studies using this type of within-subject design with crossover posttest comparisons, however, fail to account for day-to-day variations in factors such as sleep quality, which is known to influence cognitive performance [122,123,124,125] and brain activation [126,127,128]. Hence, using a within-subject design with crossover posttest comparisons makes it difficult to determine to what extent the observed changes were induced by the physical intervention itself or were a consequence of day-to-day variations [33]. To overcome these drawbacks, future investigations should make use of a within-subject crossover design with a pretest-posttest comparison (see Figure 4). This approach minimizes the influence of confounding factors and strengthens the validity of the observed effects [33].
Another crucial factor is the period between the cessation of the physical exercise and the administration of the cognitive tests (see Table 1). Behavioral results make it clear that, among other factors (e.g., exercise intensity), the length of this rest period affects cognitive performance substantially [30]. The neurobiological processes underlying this break effect (e.g., functional brain activation), however, have only been sparsely investigated [30,33]. Thus, studies investigating how brain activation is affected by the duration of the rest period (e.g., 10 min versus 60 min or 10 min versus the time needed to reach a level within 10% of the participant’s resting heart rate) are necessary to deepen our understanding of exercise–cognition interaction. Notably, fMRI scans require that every subject be prepared properly. This impedes immediate scans after the cessation of physical exercise. Fortunately, such delays between physical exercise and cognitive testing increase the effect sizes for cognitive improvements [30]
The experimental design of fMRI studies can be classified according to the following categories: (i) resting-state (e.g., analyzing fMRI data obtained while no explicit task is performed) or (ii) task-based (e.g., analyzing fMRI data obtained while a specific task is performed) [62]. While a resting-state fMRI commonly involves the assessment of functional connectivity (e.g., the temporal correlation between time series of different brain regions), in a task-based fMRI, either the magnitude of the cerebral hemodynamic response (e.g., using GLM approach) or functional connectivity (e.g., using PPI approach) are measured [62]. The majority of the reviewed studies conducted a task-based fMRI and used either a block design [39,71,72,101,102,105,106] or an event-related design [38,99,101,102,103,104,107]. Each design offers advantages and disadvantages [74,129]. The block design is a powerful paradigm to detect neuronal activity but (i) is limited with regard to its evaluation of temporal characteristics [129], (ii) can be influenced by canceling effects [130], and (iii) does not offer the possibility to perform a trial-to-trial analysis [74,129]. An event-related design provides valuable information about the temporal characteristics of neuronal activity, but many trials are needed for averaging to achieve an adequate signal-to-noise ratio [131] and appropriate statistical power [129,132]. A mixed design (consisting of a mixture of block design and event-related design) allows the investigation of ‘maintained’ versus ‘transient’ neural activity, but is challenging in terms of estimating the hemodynamic response function [74,129,133,134,135]. However, an in-depth discussion of all the advantages and disadvantages of different cognitive tests, fMRI experimental designs, and fMRI analysis methods is beyond the scope of this review; the reader may find valuable related information in the literature [62,74,129,133].
In summary, the advantages and disadvantages of the study design (e.g., within-subject crossover design with pretest-posttest comparisons), experimental fMRI design (e.g., block design), and fMRI analysis methods (e.g., PPI approach) should be carefully considered to address the specific research aim(s) appropriately.

4.3. Exercise Characteristics

The prescription of an acute bout of physical exercise is characterized by the following exercise variables: (i) exercise intensity, (ii) exercise duration and (iii) type of exercise (e.g., dancing or cycling) [33,136]. In this regard, it is important to acknowledge that exercise intensity can be specified and operationalized by using either parameters of external load (e.g., cycling while sustaining a distinct level of mean power output, i.e., in watts) or markers of internal load (e.g., cycling at a distinct percentage of the subject’s maximum heart rate) [137,138,139,140,141,142,143,144,145]. While the external load is characterized by the work completed by an individual independent of internal characteristics (e.g., the mean power output in watts over a distinct time duration and at a specific cadence), the internal load (e.g., heart rate, ratings of perceived exertion, or blood lactate response) is characterized by the psychophysiological response(s) to the external load and the influencing factors, such as personal characteristics (e.g., training status, sleep, nutrition, or genetics) and environmental factors (e.g., room temperature) [137,138,139,140,141,142,143,144,145]. In the literature, the product of exercise intensity, exercise duration, and type of physical exercise is commonly referred to as the dose [33,136], whereas a newer approach refines this definition and proposes that this dose can be objectified by using a specific marker(s) of the internal load as a proxy [29,138]. However, the dose–response relationship between acute physical exercise, cognition, and the underlying neurobiological processes (e.g., changes in cerebral hemodynamics) is currently poorly understood [26,33,35,146]. While one study provides clear evidence that brain activation patterns and cognitive performance are influenced by the exercise protocol [101], the remaining reviewed studies are too heterogeneous with regard to their studied exercise variables (e.g., exercise intensity and exercise duration) and too homogeneous with regard to the type of physical exercise they examine (only endurance exercises, such as stationary cycling or treadmill running) to derive solid conclusions in this direction. Cognizant of these limitations, further studies utilizing fMRI are required to investigate the effects of different exercise variables (e.g., different exercise intensities) or types of exercise (e.g., resistance exercise) on cognitive performance and the underlying functional brain activation changes. The investigation of the effects of acute resistance exercises [147,148] and/or acute coordinative/complex/motor-cognitive exercises [149,150,151] on cerebral hemodynamics (e.g., measured by fMRI) during a standardized cognitive test seem especially promising since these types of physical exercise have been demonstrated to lead to behavioral improvements in tests of attention, executive functions, and working memory. In addition, future studies should pay more attention to a more accurate prescription of exercise intensity because eight out of fourteen studies reviewed determined exercise intensity by using specific formulas to calculate the target heart rate [39,71,72,73,105,106] or used subjective ratings of the perceived exertion [102,103]. In particular, the use of specific formulas is less than optimal because such an approach cannot accurately predict exercise intensity [152,153,154,155,156,157,158]. Thus, in order to determine exercise intensity (or load) more accurately, it is recommended to conduct a graded exercise test for endurance exercise or to quantify the one-repetition maximum (1-RM) for resistance exercises in a separate testing session. Furthermore, it is recommended to pay stronger attention to an adequate exercise prescription, and we advocate for the reporting of both parameters of external load and markers of internal load in order to characterize the exercise protocol adequately [29,137,138,159]. By doing so, the comparability and reproducibility of the research results are very likely to be increased [137,138].

4.4. Data Processing

Measuring “true” brain activation is challenging because the BOLD signal can be influenced by non-neuronal confounders that arise from (i) instrumental noise, (ii) motion-related artifacts, and (iii) physiological artifacts [160,161,162,163,164,165,166,167]. Hence, in order to minimize the influence of artifacts and to achieve an optimal signal-to-noise and contrast-to-noise ratio, an appropriate (pre-)processing of the fMRI data is necessary. While there is currently no “gold standard” available for the processing of fMRI data [62,168], based on the analysis of the data processing procedures in the studies reviewed, it is recommended to incorporate the following “basic” data processing steps: (i) discard the first three to twelve images to avoid magnetization disequilibrium and allow the participants to habituate themselves to the scanner noise; (ii) employ slice-time correction to account for the time difference of the acquisition of each slice; (iii) utilize realignment to correct for subject motion; (iv) use intra-subject inter-modal registration (e.g., to a T1-weighted image) and spatial normalization (e.g., to MNI space); (v) apply spatial smoothing, which helps, for instance, to cope with interindividual anatomical variability and improve the signal-to-noise ratio; and (vi) apply denoising to remove the confounders and artifacts at known frequencies (e.g., scanner drifts ~< 0.01 Hz; cardiac artifacts ~0.15 Hz; respiratory artifacts ~0.3 Hz [62]). With regard to temporal filtering, only five out of the fourteen reviewed studies reported their detailed cut-off frequencies used for temporal filtering [99,101,102,105,107]. Thus, in order to enhance reproducibility, further studies should pay more attention to a detailed description of methodological details (e.g., used cut-off frequencies), thereby taking the published guidelines into account [75]. In addition to the mentioned “basic” processing steps, the correction of motion artifacts and physiological artifacts is necessary to achieve a relatively high signal quality. Artifacts arising from motion are serious confounders in fMRI studies and, thus, should be minimized [62,161,169,170,171]. The reviewed studies accounted for motion by using motion parameters as regressors or by using data-driven approaches (e.g., ICA-based methods). Indeed, the application of motion-correction techniques provides considerable improvements in data quality [169,172,173,174,175]. There seems to be no significant quality difference between the motion correction tools integrated into different popular software solutions [175,176]. However, it should be emphasized that the best approach to deal with motion artifacts is currently to reduce them as much as possible by, for instance, a comfortable padding and head fixation [62,177,178].
With regard to acute physical exercise, physiological artifacts that stem from changes in heart rate [179,180,181] and/or breathing [180,182,183,184,185,186,187,188,189] are a major challenge because acute physical exercise induces strong changes in systemic physiology (e.g., an increase in heart rate), which, in turn, may confound the BOLD signal substantially. To minimize the effects of such physiological artifacts on the BOLD signal, sophisticated cleanup techniques are needed. The available cleanup techniques can be classified as data-based approaches and approaches that rely on recordings of additional physiological parameters (e.g., cardiac or respiratory parameters) [160,164]. Since these cleanup procedures (e.g., using additional physiological measures, such as cardiac or respiratory signals as regressors) hold great potential to improve data quality [181,184], future studies should consider and incorporate them by default. It is strongly recommended to check the MR compatibility of the equipment used to quantify additional physiological parameters in order to avoid side effects [190,191]. More detailed information on how to account for different types of artifacts and noise can be found in the referenced literature [62,164,169,170,175,192,193,194,195,196,197,198]. All reviewed studies used software packages such as “Statistical parametric mapping” (SPM) or “Analysis of Functional NeuroImages” (AFNI) to analyze their fMRI data (see Table 1). A comprehensive overview of the main features of these common fMRI software packages is provided by Soares et al. [62].

4.5. Findings

There is growing evidence that acute bouts of aerobic exercise [30,199] or resistance exercise [147,148] improve cognitive performance. The fMRI studies reviewed here only partially support this notion because they report mixed findings (both improvements in behavioral performance [38,71,73,101,102,106,107] and non-significant changes in behavioral performance [39,72,99,100,104,105] were observed). Such heterogeneity may arise from the great variability in the applied research protocols, wherein it is known that behavioral improvements in response to acute physical exercise are dependent on different factors, such as the characteristics of participants (e.g., age), the cognitive domains tested (e.g., executive functions), the exercise characteristics (e.g., exercise intensity), and the time delays between the cessation of exercise and post-exercise cognitive testing [30,147,148,199].
However, independent of the degree of behavioral improvements, almost all reviewed studies reported profound changes in brain activation (especially in the frontal lobe, the cerebellum, and the hippocampus) in response to exercise (see Table 1). In general, the observations of a higher activation in distinct regions of the frontal lobe and cognitive improvements after exercise cessation are in line with the findings provided by other functional neuroimaging techniques (i.e., fNIRS) [34].
Furthermore, the results of Li et al. [106] and Mehren et al. [101] suggest that changes in functional brain activation patterns depend on (i) the cardiorespiratory fitness level, (ii) the exercise protocol, and (iii) sex. Although the number of fMRI studies in this area is limited, the moderating roles of (i) cardiorespiratory fitness levels [30], (ii) exercise intensity [30,200], and (iii) sex [30] are in accordance with the literature in summarizing the effects of acute physical exercise on behavioral performance. Thus, there is good reason to assume that investigations using fMRI can help to elucidate the effects of the mentioned moderators on neural correlates. The crucial role of using neuroimaging techniques (e.g., fMRI) to understand the neural processes involved in the interaction of acute exercise with cognition is underlined by the observed neurobehavioral associations. For instance, in three studies, correlations between changes in brain activation and behavioral performance were noticed [38,73,107], and in one study, correlations between cardiorespiratory fitness level and brain activation were observed [101]. However, based on the small number of studies using fMRI and the great variability between research protocols, it is too early to draw generalizable conclusions. Cognizant of these limitations, more comprehensive and well-designed trials are needed to utilize the possibilities provided by fMRI to understand exercise–cognition interaction.
Such upcoming studies could evaluate whether exercise-induced changes in neurotransmitter release and/or neurometabolic changes (e.g., by using positron emission tomography [PET]) [201,202,203,204] are related to exercise-induced improvements in cognitive performance. In this regard, PET studies suggest that during high-intensity exercise, lactate, which is released by the working muscles during physical exercise and can cross the blood–brain barrier via monocarboxylate transporters [205,206,207,208,209,210,211,212], is increasingly utilized by the brain to satisfy the increased energy demand of neural tissue [213]. Interestingly, after acute high-intensity interval exercises, changes in blood lactate levels have been linked to performance changes in executive functioning [214,215]. Based on these findings, further PET studies focusing on the associations between exercise-related changes in blood lactate and cognitive performance could provide neuroimaging evidence for the underlying neurobiological processes of acute exercise-related cognitive improvements. Furthermore, future studies could also assess whether acute physical exercise modulates hemodynamic changes in white matter during a cognitive task because there is growing evidence that acute hemodynamic changes occur in the white matter while solving cognitive tasks [216,217,218,219,220,221,222,223]. Another parameter worthy of examination in acute exercise-cognition studies is the (within-person) brain signal variability, which provides new insights into cognition-related processes of the human brain [224,225,226,227,228,229,230,231,232,233]. Furthermore, to derive valid and reliable conclusions in repeated measurements, a high level of reproducibility is necessary [234,235,236]. With regard to the assessment of cognitive performance prior to and after an acute bout of physical exercise, a considerable amount of test–retest errors for specific measures of cognition have been reported [237], suggesting that participants should be appropriately habituated to the whole testing procedure. Concerning the reproducibility of fMRI, it was observed that reproducibility is affected by experimental factors, such as the length of the scan [238] and the design of the cognitive task [239]. However, currently, there is no study available that investigates the reproducibility of exercise-induced functional brain activation changes (e.g., measured by fMRI). Such an evaluation of reproducibility is needed to improve our interpretations of the obtained results. In addition, there is some evidence that several biopsychosocial factors (e.g., circadian rhythms [240,241,242], level of sleepiness [243], and level of arousal [244]) can influence cognitive performance. In particular, it was observed (i) that measures of the cortical hemodynamic response (e.g., obtained during a cognitive test) are associated with total sleep duration [245,246] and sleepiness [247,248] and (ii) that total sleep duration is associated with acute exercise-related changes in behavioral performance (e.g., executive function performance) [82]. Interestingly, the relationship between total sleep duration and acute exercise-related changes in executive function performance are mediated by the volume of the caudate nuclei [82]. However, based on the evidence that cognitive performance is influenced by several biopsychosocial factors (e.g., sleep and arousal), future studies should consider these factors in order to elucidate their effects on acute exercise-related changes in cognitive performance and functional brain activation.

5. Conclusions

In summary, this review provides an overview of fMRI applications in the investigation of the effects of acute physical exercise on cognitive performance and their underlying neural correlates (e.g., changes in functional brain activation patterns). Although the small number of studies available and the great variability across study protocols limits the number of conclusions that can be drawn with certainty, our observations suggest that acute physical exercise (e.g., cycling) leads to profound changes in functional brain activation, while the evidence with regard to behavioral changes is equivocal. Thus, further well-designed trials are needed to strengthen our understanding of acute exercise-induced changes in functional brain activation, including an investigation of a possible dose–response relationship. Future studies should consider (i) a more rigorous study design (e.g., within-subject crossover design with a pretest-posttest comparison), (ii) the integration of sophisticated filter methods in fMRI data analysis (e.g., to account for physiological artifacts), (iii) a more detailed description of the applied processing steps in fMRI data analysis (e.g., the applied filter cut-off frequencies and the atlas used for neuroanatomical labeling), and (iv) a more precise exercise prescription (e.g., including the reporting of both parameters of external load and markers of internal load).

Author Contributions

Conceptualization: F.H.; methodology: F.H. and N.A.; software: F.H. and N.A.; validation: F.H. and N.A.; formal analysis: F.H. and N.A.; investigation (systematic literature search, data extraction, and risk of bias assessment): F.H. and N.A.; resources: M.T. and N.G.M.; data curation; F.H. and N.A.; writing—original draft: F.H.; writing—review and editing: F.H. and N.A.; N.L., M.T., and N.G.M.; visualization: F.H.; supervision: N.L., M.T., and N.G.M.; project administration: F.H. All authors have read and approved the final version. All authors have read and agreed to the published version of the manuscript.

Funding

This systematic review was partially supported by the joint project “Autonomy in Old Age” (AiA) funded by the European Regional Development Fund (grant number: ZS/2019/07/99755). The authors are solely responsible for the work and the funders had no role in the preparation of the manuscript, the analysis of the studies, and the decision to publish.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hill, J.O.; Peters, J.C. Environmental contributions to the obesity epidemic. Science 1998, 280, 1371–1374. [Google Scholar] [CrossRef]
  2. Bell, A.C.; Ge, K.; Popkin, B.M. The road to obesity or the path to prevention: Motorized transportation and obesity in China. Obes. Res. 2002, 10, 277–283. [Google Scholar] [CrossRef] [PubMed]
  3. Fotheringham, M.J.; Wonnacott, R.L.; Owen, N. Computer use and physical inactivity in young adults: Public health perils and potentials of new information technologies. Ann. Behav. Med. 2000, 22, 269–275. [Google Scholar] [CrossRef] [PubMed]
  4. Johnson, R.W. Trends in Job Demands among Older Workers, 1992–2002. Mon. Labor Rev. 2004, 127, 48–56. [Google Scholar]
  5. Owen, N.; Sparling, P.B.; Healy, G.N.; Dunstan, D.W.; Matthews, C.E. Sedentary behavior: Emerging evidence for a new health risk. Mayo Clin. Proc. 2010, 85, 1138–1141. [Google Scholar] [CrossRef] [Green Version]
  6. Church, T.S.; Thomas, D.M.; Tudor-Locke, C.; Katzmarzyk, P.T.; Earnest, C.P.; Rodarte, R.Q.; Martin, C.K.; Blair, S.N.; Bouchard, C. Trends over 5 decades in U.S. occupation-related physical activity and their associations with obesity. PLoS ONE 2011, 6, e19657. [Google Scholar] [CrossRef] [Green Version]
  7. Copeland, J.L.; Clarke, J.; Dogra, S. Objectively measured and self-reported sedentary time in older Canadians. Prev. Med. Rep. 2015, 2, 90–95. [Google Scholar] [CrossRef] [Green Version]
  8. Blair, S.N. Physical inactivity: The biggest public health problem of the 21st century. Br. J. Sports Med. 2009, 43, 1–2. [Google Scholar]
  9. Falck, R.S.; Davis, J.C.; Liu-Ambrose, T. What is the association between sedentary behaviour and cognitive function? A systematic review. Br. J. Sports Med. 2016, 51, 800–811. [Google Scholar] [CrossRef] [Green Version]
  10. Ku, P.-W.; Liu, Y.-T.; Lo, M.-K.; Chen, L.-J.; Stubbs, B. Higher levels of objectively measured sedentary behavior is associated with worse cognitive ability: Two-year follow-up study in community-dwelling older adults. Exp. Gerontol. 2017, 99, 110–114. [Google Scholar] [CrossRef] [Green Version]
  11. 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. A Biol. Sci. Med. Sci. 2017, 72, 789–795. [Google Scholar] [CrossRef] [Green Version]
  12. Aichberger, M.C.; Busch, M.A.; Reischies, F.M.; Ströhle, A.; Heinz, A.; Rapp, M.A. Effect of Physical Inactivity on Cognitive Performance after 2.5 Years of Follow-Up. GeroPsych 2010, 23, 7–15. [Google Scholar] [CrossRef]
  13. Laurin, D.; Verreault, R.; Lindsay, J.; MacPherson, K.; Rockwood, K. Physical Activity and Risk of Cognitive Impairment and Dementia in Elderly Persons. Arch. Neurol. 2001, 58, 498–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bowen, M.E. A prospective examination of the relationship between physical activity and dementia risk in later life. Am. J. Health Promot. 2012, 26, 333–340. [Google Scholar] [CrossRef] [PubMed]
  15. Grande, G.; Vanacore, N.; Maggiore, L.; Cucumo, V.; Ghiretti, R.; Galimberti, D.; Scarpini, E.; Mariani, C.; Clerici, F. Physical activity reduces the risk of dementia in mild cognitive impairment subjects: A cohort study. J. Alzheimers Dis. 2014, 39, 833–839. [Google Scholar] [CrossRef] [PubMed]
  16. Paillard-Borg, S.; Fratiglioni, L.; Winblad, B.; Wang, H.-X. Leisure activities in late life in relation to dementia risk: Principal component analysis. Dement. Geriatr. Cogn. Disord. 2009, 28, 136–144. [Google Scholar] [CrossRef]
  17. Rovio, S.; Kåreholt, I.; Helkala, E.-L.; Viitanen, M.; Winblad, B.; Tuomilehto, J.; Soininen, H.; Nissinen, A.; Kivipelto, M. Leisure-time physical activity at midlife and the risk of dementia and Alzheimer’s disease. Lancet Neurol. 2005, 4, 705–711. [Google Scholar] [CrossRef]
  18. Paillard, T. Preventive effects of regular physical exercise against cognitive decline and the risk of dementia with age advancement. Sports Med. Open 2015, 1, 4. [Google Scholar] [CrossRef] [Green Version]
  19. Buchman, A.S.; Boyle, P.A.; Yu, L.; Shah, R.C.; Wilson, R.S.; Bennett, D.A. Total daily physical activity and the risk of AD and cognitive decline in older adults. Neurology 2012, 78, 1323–1329. [Google Scholar] [CrossRef] [Green Version]
  20. Bruijn, R.F.A.G.; Schrijvers, E.M.C.; Groot, K.A.; Witteman, J.C.M.; Hofman, A.; Franco, O.H.; Koudstaal, P.J.; Ikram, M.A. The association between physical activity and dementia in an elderly population: The Rotterdam Study. Eur. J. Epidemiol. 2013, 28, 277–283. [Google Scholar] [CrossRef]
  21. Hamer, M.; Chida, Y. Physical activity and risk of neurodegenerative disease: A systematic review of prospective evidence. Psychol. Med. 2009, 39, 3–11. [Google Scholar] [CrossRef] [PubMed]
  22. Ravaglia, G.; Forti, P.; Lucicesare, A.; Pisacane, N.; Rietti, E.; Bianchin, M.; Dalmonte, E. Physical activity and dementia risk in the elderly: Findings from a prospective Italian study. Neurology 2008, 70, 1786–1794. [Google Scholar] [CrossRef] [PubMed]
  23. Taylor, W.C. Transforming work breaks to promote health. Am. J. Prev. Med. 2005, 29, 461–465. [Google Scholar] [CrossRef] [PubMed]
  24. Bergouignan, A.; Legget, K.T.; Jong, N.; Kealey, E.; Nikolovski, J.; Groppel, J.L.; Jordan, C.; O’Day, R.; Hill, J.O.; Bessesen, D.H. Effect of frequent interruptions of prolonged sitting on self-perceived levels of energy, mood, food cravings and cognitive function. Int. J. Behav. Nutr. Phys. Act. 2016, 13, 113. [Google Scholar] [CrossRef] [Green Version]
  25. Panahi, S.; Tremblay, A. Sedentariness and Health: Is Sedentary Behavior More Than Just Physical Inactivity? Front. Public Health 2018, 6, 258. [Google Scholar] [CrossRef] [Green Version]
  26. Herold, F.; Hamacher, D.; Schega, L.; Müller, N.G. Thinking While Moving or Moving While Thinking-Concepts of Motor-Cognitive Training for Cognitive Performance Enhancement. Front. Aging Neurosci. 2018, 10, 228. [Google Scholar] [CrossRef] [Green Version]
  27. Budde, H.; Schwarz, R.; Velasques, B.; Ribeiro, P.; Holzweg, M.; Machado, S.; Brazaitis, M.; Staack, F.; Wegner, M. The need for differentiating between exercise, physical activity, and training. Autoimmun. Rev. 2016, 15, 110–111. [Google Scholar] [CrossRef]
  28. Caspersen, C.J.; Powell, K.E.; Christenson, G.M. Physical activity, exercise, and physical fitness: Definitions and distinctions for health-related research. Public Health Rep. 1985, 100, 126–131. [Google Scholar]
  29. Gronwald, T.; Budde, H. Commentary: Physical Exercise as Personalized Medicine for Dementia Prevention? Front. Physiol. 2019, 10, 726. [Google Scholar] [CrossRef] [Green Version]
  30. Chang, Y.K.; Labban, J.D.; Gapin, J.I.; Etnier, J.L. The effects of acute exercise on cognitive performance: A meta-analysis. Brain Res. 2012, 1453, 87–101. [Google Scholar] [CrossRef] [Green Version]
  31. Basso, J.C.; Suzuki, W.A. The Effects of Acute Exercise on Mood, Cognition, Neurophysiology, and Neurochemical Pathways: A Review. Brain Plast. 2017, 2, 127–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. McSween, M.-P.; Coombes, J.S.; MacKay, C.P.; Rodriguez, A.D.; Erickson, K.I.; Copland, D.A.; McMahon, K.L. The Immediate Effects of Acute Aerobic Exercise on Cognition in Healthy Older Adults: A Systematic Review. Sports Med. 2018, 49, 67–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Pontifex, M.B.; McGowan, A.L.; Chandler, M.C.; Gwizdala, K.L.; Parks, A.C.; Fenn, K.; Kamijo, K. A primer on investigating the after effects of acute bouts of physical activity on cognition. Psychol. Sport Exerc. 2019, 40, 1–22. [Google Scholar] [CrossRef]
  34. Herold, F.; Wiegel, P.; Scholkmann, F.; Müller, N.G. Applications of Functional Near-Infrared Spectroscopy (fNIRS) Neuroimaging in Exercise⁻Cognition Science: A Systematic, Methodology-Focused Review. J. Clin. Med. 2018, 7, 466. [Google Scholar] [CrossRef] [Green Version]
  35. Törpel, A.; Herold, F.; Hamacher, D.; Müller, N.G.; Schega, L. Strengthening the Brain—Is Resistance Training with Blood Flow Restriction an Effective Strategy for Cognitive Improvement? J. Clin. Med. 2018, 7, 377. [Google Scholar] [CrossRef] [Green Version]
  36. Hillman, C.H.; Kamijo, K.; Pontifex, M.B. The Relation of ERP Indices of Exercise to Brain Health and Cognition. In Functional Neuroimaging in Exercise and Sport Sciences; Boecker, H., Hillman, C.H., Scheef, L., Strüder, H.K., Eds.; Springer: New York, NY, USA, 2012; pp. 419–446. ISBN 978-1-4614-3292-0. [Google Scholar]
  37. Chang, Y.-K. Acute Exercise and Event-Related Potential. In Exercise-Cognition Interaction; Elsevier: Amsterdam, The Netherlands, 2016; pp. 105–130. ISBN 9780128007785. [Google Scholar]
  38. Suwabe, K.; Byun, K.; Hyodo, K.; Reagh, Z.M.; Roberts, J.M.; Matsushita, A.; Saotome, K.; Ochi, G.; Fukuie, T.; Suzuki, K.; et al. Rapid stimulation of human dentate gyrus function with acute mild exercise. Proc. Natl. Acad. Sci. USA 2018, 115, 10487–10492. [Google Scholar] [CrossRef] [Green Version]
  39. Li, L.; Men, W.W.; Chang, Y.K.; Fan, M.X.; Ji, L.; Wei, G.X. Acute aerobic exercise increases cortical activity during working memory: A functional MRI study in female college students. PLoS ONE 2014, 9, e99222. [Google Scholar] [CrossRef]
  40. Herold, F.; Wiegel, P.; Scholkmann, F.; Thiers, A.; Hamacher, D.; Schega, L. Functional near-infrared spectroscopy in movement science: A systematic review on cortical activity in postural and walking tasks. Neurophoton 2017, 4, 41403. [Google Scholar] [CrossRef] [Green Version]
  41. Pinti, P.; Tachtsidis, I.; Hamilton, A.; Hirsch, J.; Aichelburg, C.; Gilbert, S.; Burgess, P.W. The present and future use of functional near-infrared spectroscopy (fNIRS) for cognitive neuroscience. Ann. N. Y. Acad. Sci. 2018. [Google Scholar] [CrossRef]
  42. Scarapicchia, V.; Brown, C.; Mayo, C.; Gawryluk, J.R. Functional Magnetic Resonance Imaging and Functional Near-Infrared Spectroscopy: Insights from Combined Recording Studies. Front. Hum. Neurosci. 2017, 11, 419. [Google Scholar] [CrossRef]
  43. Soltanlou, M.; Sitnikova, M.A.; Nuerk, H.-C.; Dresler, T. Applications of Functional Near-Infrared Spectroscopy (fNIRS) in Studying Cognitive Development: The Case of Mathematics and Language. Front. Psychol. 2018, 9, 277. [Google Scholar] [CrossRef] [PubMed]
  44. Glover, G.H. Overview of functional magnetic resonance imaging. Neurosurg. Clin. N. Am. 2011, 22, 133–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Voss, J.L.; Bridge, D.J.; Cohen, N.J.; Walker, J.A. A Closer Look at the Hippocampus and Memory. Trends Cogn. Sci. 2017, 21, 577–588. [Google Scholar] [CrossRef] [PubMed]
  46. Knierim, J.J. The hippocampus. Curr. Biol. 2015, 25, R1116-21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Bird, C.M.; Burgess, N. The hippocampus and memory: Insights from spatial processing. Nat. Rev. Neurosci. 2008, 9, 182–194. [Google Scholar] [CrossRef] [PubMed]
  48. Maguire, E.A.; Frackowiak, R.S.J.; Frith, C.D. Recalling Routes around London: Activation of the Right Hippocampus in Taxi Drivers. J. Neurosci. 1997, 17, 7103–7110. [Google Scholar] [CrossRef] [Green Version]
  49. Squire, L.R.; Ojemann, J.G.; Miezin, F.M.; Petersen, S.E.; Videen, T.O.; Raichle, M.E. Activation of the hippocampus in normal humans: A functional anatomical study of memory. Proc. Natl. Acad. Sci. USA 1992, 89, 1837–1841. [Google Scholar] [CrossRef] [Green Version]
  50. O’Brien, J.L.; O’Keefe, K.M.; LaViolette, P.S.; DeLuca, A.N.; Blacker, D.; Dickerson, B.C.; Sperling, R.A. Longitudinal fMRI in elderly reveals loss of hippocampal activation with clinical decline. Neurology 2010, 74, 1969–1976. [Google Scholar] [CrossRef] [Green Version]
  51. Johnson, S.C.; Schmitz, T.W.; Moritz, C.H.; Meyerand, M.E.; Rowley, H.A.; Alexander, A.L.; Hansen, K.W.; Gleason, C.E.; Carlsson, C.M.; Ries, M.L.; et al. Activation of brain regions vulnerable to Alzheimer’s disease: The effect of mild cognitive impairment. Neurobiol. Aging 2006, 27, 1604–1612. [Google Scholar] [CrossRef] [Green Version]
  52. Small, S.A.; Perera, G.M.; DeLapaz, R.; Mayeux, R.; Stern, Y. Differential regional dysfunction of the hippocampal formation among elderly with memory decline and Alzheimer’s disease. Ann. Neurol. 1999, 45, 466–472. [Google Scholar] [CrossRef]
  53. Ogawa, S.; Lee, T.M.; Kay, A.R.; Tank, D.W. Brain magnetic resonance imaging with contrast dependent on blood oxygenation. Proc. Natl. Acad. Sci. USA 1990, 87, 9868–9872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ogawa, S.; Lee, T.M.; Nayak, A.S.; Glynn, P. Oxygenation-sensitive contrast in magnetic resonance image of rodent brain at high magnetic fields. Magn. Reson. Med. 1990, 14, 68–78. [Google Scholar] [CrossRef] [PubMed]
  55. Ogawa, S.; Tank, D.W.; Menon, R.; Ellermann, J.M.; Kim, S.G.; Merkle, H.; Ugurbil, K. Intrinsic signal changes accompanying sensory stimulation: Functional brain mapping with magnetic resonance imaging. Proc. Natl. Acad. Sci. USA 1992, 89, 5951–5955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ogawa, S. Finding the BOLD effect in brain images. NeuroImage 2012, 62, 608–609. [Google Scholar] [CrossRef]
  57. Gore, J.C. Principles and practice of functional MRI of the human brain. J. Clin. Investig. 2003, 112, 4–9. [Google Scholar] [CrossRef]
  58. Norris, D.G. Principles of magnetic resonance assessment of brain function. J. Magn. Reson. Imaging 2006, 23, 794–807. [Google Scholar] [CrossRef]
  59. Kim, S.-G.; Ogawa, S. Biophysical and physiological origins of blood oxygenation level-dependent fMRI signals. J. Cereb. Blood Flow Metab. 2012, 32, 1188–1206. [Google Scholar] [CrossRef]
  60. Logothetis, N.K. What we can do and what we cannot do with fMRI. Nature 2008, 453, 869–878. [Google Scholar] [CrossRef]
  61. Logothetis, N.K.; Pfeuffer, J. On the nature of the BOLD fMRI contrast mechanism. Magn. Reson. Imaging 2004, 22, 1517–1531. [Google Scholar] [CrossRef]
  62. Soares, J.M.; Magalhães, R.; Moreira, P.S.; Sousa, A.; Ganz, E.; Sampaio, A.; Alves, V.; Marques, P.; Sousa, N. A Hitchhiker’s Guide to Functional Magnetic Resonance Imaging. Front. Neurosci. 2016, 10, 515. [Google Scholar] [CrossRef] [Green Version]
  63. Francis, S.; Panchuelo, R.S. Physiological measurements using ultra-high field fMRI: A review. Physiol. Meas. 2014, 35, R167-85. [Google Scholar] [CrossRef] [PubMed]
  64. Heeger, D.J.; Ress, D. What does fMRI tell us about neuronal activity? Nat. Rev. Neurosci. 2002, 3, 142–151. [Google Scholar] [CrossRef]
  65. Cinciute, S. Translating the hemodynamic response: Why focused interdisciplinary integration should matter for the future of functional neuroimaging. PeerJ 2019, 7, e6621. [Google Scholar] [CrossRef] [PubMed]
  66. Huneau, C.; Benali, H.; Chabriat, H. Investigating Human Neurovascular Coupling Using Functional Neuroimaging: A Critical Review of Dynamic Models. Front. Neurosci. 2015, 9, e1002435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Leybaert, L. Neurobarrier coupling in the brain: A partner of neurovascular and neurometabolic coupling? J. Cereb. Blood Flow Metab. 2005, 25, 2–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Liao, L.-D.; Tsytsarev, V.; Delgado-Martínez, I.; Li, M.-L.; Erzurumlu, R.; Vipin, A.; Orellana, J.; Lin, Y.-R.; Lai, H.-Y.; Chen, Y.-Y.; et al. Neurovascular coupling: In vivo optical techniques for functional brain imaging. BioMed. Eng. OnLine 2013, 12, 38. [Google Scholar] [CrossRef] [Green Version]
  69. Nippert, A.R.; Biesecker, K.R.; Newman, E.A. Mechanisms Mediating Functional Hyperemia in the Brain. Neuroscientist 2018, 24, 73–83. [Google Scholar] [CrossRef]
  70. Minzenberg, M.J. Pharmacological MRI approaches to understanding mechanisms of drug action. Curr. Top. Behav. Neurosci. 2012, 11, 365–388. [Google Scholar] [CrossRef]
  71. Chen, A.-G.; Zhu, L.-N.; Yan, J.; Yin, H.-C. Neural Basis of Working Memory Enhancement after Acute Aerobic Exercise: FMRI Study of Preadolescent Children. Front. Psychol. 2016, 7, 1804. [Google Scholar] [CrossRef] [Green Version]
  72. MacIntosh, B.J.; Crane, D.E.; Sage, M.D.; Rajab, A.S.; Donahue, M.J.; McIlroy, W.E.; Middleton, L.E. Impact of a single bout of aerobic exercise on regional brain perfusion and activation responses in healthy young adults. PLoS ONE 2014, 9, e85163. [Google Scholar] [CrossRef]
  73. Chen, A.-G.; Zhu, L.-N.; Xiong, X.; Li, Y. Acute aerobic exercise alters executive control network in preadolescent children. J. Sport Psychol. 2017, 26, 132–137. [Google Scholar]
  74. Amaro, E.; Barker, G.J. Study design in fMRI: Basic principles. Brain Cogn. 2006, 60, 220–232. [Google Scholar] [CrossRef] [PubMed]
  75. Poldrack, R.A.; Fletcher, P.C.; Henson, R.N.; Worsley, K.J.; Brett, M.; Nichols, T.E. Guidelines for reporting an fMRI study. Neuroimage 2008, 40, 409–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. J. Clin. Epidemiol. 2009, 62, 1006–1012. [Google Scholar] [CrossRef]
  77. Harris, J.D.; Quatman, C.E.; Manring, M.M.; Siston, R.A.; Flanigan, D.C. How to write a systematic review. Am. J. Sports Med. 2014, 42, 2761–2768. [Google Scholar] [CrossRef]
  78. Bostrom, N.; Sandberg, A. Cognitive enhancement: Methods, ethics, regulatory challenges. Sci. Eng. Ethics 2009, 15, 311–341. [Google Scholar] [CrossRef]
  79. Borson, S. Cognition, aging, and disabilities: Conceptual issues. Phys. Med. Rehabil. Clin. N. Am. 2010, 21, 375–382. [Google Scholar] [CrossRef] [Green Version]
  80. Gothe, N.; Pontifex, M.B.; Hillman, C.; McAuley, E. The acute effects of yoga on executive function. J. Phys. Act. Health 2013, 10, 488–495. [Google Scholar] [CrossRef]
  81. Chang, Y.-K.; Tsai, C.-L.; Hung, T.-M.; So, E.C.; Chen, F.-T.; Etnier, J.L. Effects of Acute Exercise on Executive Function: A Study with a Tower of London Task. J. Sport Exerc. Psychol. 2011, 33, 847–865. [Google Scholar] [CrossRef] [Green Version]
  82. Won, J.; Alfini, A.J.; Weiss, L.R.; Nyhuis, C.C.; Spira, A.P.; Callow, D.D.; Carson Smith, J. Caudate Volume Mediates the Interaction between Total Sleep Time and Executive Function after Acute Exercise in Healthy Older Adults. Brain Plast. 2019, 5, 69–82. [Google Scholar] [CrossRef] [Green Version]
  83. Faulkner, J.; Lambrick, D.; Kaufmann, S.; Stoner, L. Effects of Upright and Recumbent Cycling on Executive Function and Prefrontal Cortex Oxygenation in Young, Healthy, Men. J. Phys. Act. Health 2016, 13, 882–887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Faulkner, J.; Stoner, L.; Grigg, R.; Fryer, S.; Stone, K.; Lambrick, D. Acute effects of exercise posture on executive function in transient ischemic attack patients. Psychophysiology 2017, 54, 1239–1248. [Google Scholar] [CrossRef] [PubMed]
  85. Lambrick, D.; Stoner, L.; Grigg, R.; Faulkner, J. Effects of continuous and intermittent exercise on executive function in children aged 8–10 years. Psychophysiology 2016, 53, 1335–1342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Pontifex, M.B.; Gwizdala, K.L.; WENG, T.B.; Zhu, D.C.; Voss, M.W. Cerebral blood flow is not modulated following acute aerobic exercise in preadolescent children. Int. J. Psychophysiol. 2018, 134, 44–51. [Google Scholar] [CrossRef]
  87. Rajab, A.S.; Crane, D.E.; Middleton, L.E.; Robertson, A.D.; Hampson, M.; MacIntosh, B.J. A single session of exercise increases connectivity in sensorimotor-related brain networks: A resting-state fMRI study in young healthy adults. Front. Hum. Neurosci. 2014, 8, 625. [Google Scholar] [CrossRef] [Green Version]
  88. Scheef, L.; Jankowski, J.; Daamen, M.; Weyer, G.; Klingenberg, M.; Renner, J.; Mueckter, S.; Schürmann, B.; Musshoff, F.; Wagner, M.; et al. An fMRI study on the acute effects of exercise on pain processing in trained athletes. Pain 2012, 153, 1702–1714. [Google Scholar] [CrossRef]
  89. Schmitt, A.; Martin, J.A.; Rojas, S.; Vafa, R.; Scheef, L.; Strüder, H.K.; Boecker, H. Effects of low- and high-intensity exercise on emotional face processing: An fMRI face-matching study. Soc. Cogn. Affect. Neurosci. 2019, 14, 657–665. [Google Scholar] [CrossRef]
  90. Weng, T.B.; Pierce, G.L.; Darling, W.G.; Falk, D.; Magnotta, V.A.; Voss, M.W. The Acute Effects of Aerobic Exercise on the Functional Connectivity of Human Brain Networks. Brain Plast. 2017, 2, 171–190. [Google Scholar] [CrossRef] [Green Version]
  91. van Janse Rensburg, K.; Taylor, A.; Hodgson, T.; Benattayallah, A. Acute exercise modulates cigarette cravings and brain activation in response to smoking-related images: An fMRI study. Psychopharmacology 2009, 203, 589–598. [Google Scholar] [CrossRef]
  92. Masterson, T.D.; Kirwan, C.B.; Davidson, L.E.; Larson, M.J.; Keller, K.L.; Fearnbach, S.N.; Evans, A.; LeCheminant, J.D. Brain reactivity to visual food stimuli after moderate-intensity exercise in children. Brain Imaging Behav. 2018, 12, 1032–1041. [Google Scholar] [CrossRef]
  93. Chen, Y.-C.; Chen, C.; Martínez, R.M.; Etnier, J.L.; Cheng, Y. Habitual physical activity mediates the acute exercise-induced modulation of anxiety-related amygdala functional connectivity. Sci. Rep. 2019, 9, 19787. [Google Scholar] [CrossRef]
  94. Schmitt, A.; Upadhyay, N.; Martin, J.A.; Rojas, S.; Strüder, H.K.; Boecker, H. Modulation of Distinct Intrinsic Resting State Brain Networks by Acute Exercise Bouts of Differing Intensity. Brain Plast. 2019, 5, 39–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Himmelmeier, R.M.; Nouchi, R.; Saito, T.; Burin, D.; Wiltfang, J.; Kawashima, R. Study Protocol: Does an Acute Intervention of High-Intensity Physical Exercise Followed by a Brain Training Video Game Have Immediate Effects on Brain Activity of Older People During Stroop Task in fMRI?-A Randomized Controlled Trial With Crossover Design. Front. Aging Neurosci. 2019, 11, 260. [Google Scholar] [CrossRef] [Green Version]
  96. Tzourio-Mazoyer, N.; Landeau, B.; Papathanassiou, D.; Crivello, F.; Etard, O.; Delcroix, N.; Mazoyer, B.; Joliot, M. Automated anatomical labeling of activations in SPM using a macroscopic anatomical parcellation of the MNI MRI single-subject brain. NeuroImage 2002, 15, 273–289. [Google Scholar] [CrossRef] [PubMed]
  97. Higgins, J.P.T.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions. A Cochrane Pocketbook; John Wiley & Sons, Ltd.: Chichester, UK, 2008; ISBN 978-0470699515. [Google Scholar]
  98. The Cochrane Collaboration. Review Manager (RevMan 5.3) [Computer program]. Available online: https://community.cochrane.org/help/tools-and-software/revman-5 (accessed on 1 April 2019).
  99. Bothe, N.; Zschucke, E.; Dimeo, F.; Heinz, A.; Wüstenberg, T.; Ströhle, A. Acute exercise influences reward processing in highly trained and untrained men. Med. Sci. Sports Exerc. 2013, 45, 583–591. [Google Scholar] [CrossRef] [PubMed]
  100. Mehren, A.; Özyurt, J.; Thiel, C.M.; Brandes, M.; Lam, A.P.; Philipsen, A. Effects of Acute Aerobic Exercise on Response Inhibition in Adult Patients with ADHD. Sci. Rep. 2019, 9, 65. [Google Scholar] [CrossRef]
  101. Mehren, A.; Diaz Luque, C.; Brandes, M.; Lam, A.P.; Thiel, C.M.; Philipsen, A.; Özyurt, J. Intensity-Dependent Effects of Acute Exercise on Executive Function. Neural Plast. 2019, 2019, 8608317. [Google Scholar] [CrossRef] [Green Version]
  102. Mehren, A.; Özyurt, J.; Lam, A.P.; Brandes, M.; Müller, H.H.O.; Thiel, C.M.; Philipsen, A. Acute Effects of Aerobic Exercise on Executive Function and Attention in Adult Patients With ADHD. Front. Psychiatry 2019, 10, 132. [Google Scholar] [CrossRef] [Green Version]
  103. Won, J.; Alfini, A.J.; Weiss, L.R.; Callow, D.D.; Smith, J.C. Brain activation during executive control after acute exercise in older adults. Int. J. Psychophysiol. 2019, 146, 240–248. [Google Scholar] [CrossRef]
  104. Won, J.; Alfini, A.J.; Weiss, L.R.; Michelson, C.S.; Callow, D.D.; Ranadive, S.M.; Gentili, R.J.; Smith, J.C. Semantic Memory Activation After Acute Exercise in Healthy Older Adults. J. Int. Neuropsychol. Soc. 2019, 25, 557–568. [Google Scholar] [CrossRef] [Green Version]
  105. Metcalfe, A.W.S.; MacIntosh, B.J.; Scavone, A.; Ou, X.; Korczak, D.; Goldstein, B.I. Effects of acute aerobic exercise on neural correlates of attention and inhibition in adolescents with bipolar disorder. Transl. Psychiatry 2016, 6, e814. [Google Scholar] [CrossRef] [PubMed]
  106. Li, L.; Zhang, S.; Cui, J.; Chen, L.-Z.; Wang, X.; Fan, M.; Wei, G.-X. Fitness-Dependent Effect of Acute Aerobic Exercise on Executive Function. Front. Physiol. 2019, 10, 731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Voss, M.W.; Weng, T.B.; Narayana-Kumanan, K.; Cole, R.C.; Wharff, C.; Reist, L.; DuBose, L.; Sigurdsson, G.; Mills, J.A.; Long, J.D.; et al. Acute Exercise Effects Predict Training Change in Cognition and Connectivity. Med. Sci. Sports Exerc. 2019. [Google Scholar] [CrossRef] [PubMed]
  108. Borg, G.A. Psychophysical bases of perceived exertion. Med. Sci. Sports Exerc. 1982, 14, 377–381. [Google Scholar] [CrossRef] [PubMed]
  109. Day, J.R.; Rossiter, H.B.; Coats, E.M.; Skasick, A.; Whipp, B.J. The maximally attainable VO2 during exercise in humans: The peak vs. maximum issue. J. Appl. Physiol. 2003, 95, 1901–1907. [Google Scholar] [CrossRef] [Green Version]
  110. Poole, D.C.; Jones, A.M. Measurement of the maximum oxygen uptake V . o2max: V . o2peak is no longer acceptable. J. Appl. Physiol. 2017, 122, 997–1002. [Google Scholar] [CrossRef]
  111. Midgley, A.W.; McNaughton, L.R.; Polman, R.; Marchant, D. Criteria for determination of maximal oxygen uptake: A brief critique and recommendations for future research. Sports Med. 2007, 37, 1019–1028. [Google Scholar] [CrossRef]
  112. Schulz, K.F.; Altman, D.G.; Moher, D.; Group, C. CONSORT 2010 Statement: Updated guidelines for reporting parallel group randomised trials. BMC Med. 2010, 8, 18. [Google Scholar] [CrossRef] [Green Version]
  113. Dwan, K.; Li, T.; Altman, D.G.; Elbourne, D. CONSORT 2010 statement: Extension to randomised crossover trials. BMJ 2019, 366, l4378. [Google Scholar] [CrossRef] [Green Version]
  114. Lindheimer, J.B.; Szabo, A.; Raglin, J.S.; Beedie, C. Advancing the understanding of placebo effects in psychological outcomes of exercise: Lessons learned and future directions. Eur. J. Sport Sci. 2019, 1–28. [Google Scholar] [CrossRef]
  115. Lindheimer, J.B.; Szabo, A.; Raglin, J.S.; Beedie, C.; Carmichael, K.E.; O’Connor, P.J. Reconceptualizing the measurement of expectations to better understand placebo and nocebo effects in psychological responses to exercise. Eur. J. Sport Sci. 2019, 1–25. [Google Scholar] [CrossRef] [PubMed]
  116. Beedie, C.; Benedetti, F.; Barbiani, D.; Camerone, E.; Cohen, E.; Coleman, D.; Davis, A.; Elsworth-Edelsten, C.; Flowers, E.; Foad, A.; et al. Consensus statement on placebo effects in sports and exercise: The need for conceptual clarity, methodological rigour, and the elucidation of neurobiological mechanisms. Eur. J. Sport Sci. 2018, 18, 1383–1389. [Google Scholar] [CrossRef] [PubMed]
  117. Boot, W.R.; Simons, D.J.; Stothart, C.; Stutts, C. The Pervasive Problem With Placebos in Psychology: Why Active Control Groups Are Not Sufficient to Rule Out Placebo Effects. Perspect. Psychol. Sci. 2013, 8, 445–454. [Google Scholar] [CrossRef] [PubMed]
  118. Shawn Green, C.; Bavelier, D.; Kramer, A.F.; Vinogradov, S.; Ansorge, U.; Ball, K.K.; Bingel, U.; Chein, J.M.; Colzato, L.S.; Edwards, J.D.; et al. Improving Methodological Standards in Behavioral Interventions for Cognitive Enhancement. J. Cogn. Enhanc. 2019, 33, 316. [Google Scholar] [CrossRef] [Green Version]
  119. Lange, A.K.; Vanwanseele, B.; Foroughi, N.; Baker, M.K.; Shnier, R.; Smith, R.M.; Singh, M.A.F. Resistive Exercise for Arthritic Cartilage Health (REACH): A randomized double-blind, sham-exercise controlled trial. BMC Geriatr. 2009, 9, 1. [Google Scholar] [CrossRef] [Green Version]
  120. Vonk, M.; Wikkerink, S.; Regan, K.; Middleton, L.E. Similar changes in executive function after moderate resistance training and loadless movement. PLoS ONE 2019, 14, e0212122. [Google Scholar] [CrossRef] [Green Version]
  121. McCaskey, M.A.; Wirth, B.; Schuster-Amft, C.; Bruin, E.D. Postural sensorimotor training versus sham exercise in physiotherapy of patients with chronic non-specific low back pain: An exploratory randomised controlled trial. PLoS ONE 2018, 13, e0193358. [Google Scholar] [CrossRef]
  122. Alhola, P.; Polo-Kantola, P. Sleep deprivation: Impact on cognitive performance. Neuropsychiatr. Dis. Treat. 2007, 3, 553–567. [Google Scholar]
  123. Lo, J.C.; Groeger, J.A.; Santhi, N.; Arbon, E.L.; Lazar, A.S.; Hasan, S.; Schantz, M.; Archer, S.N.; Dijk, D.-J. Effects of partial and acute total sleep deprivation on performance across cognitive domains, individuals and circadian phase. PLoS ONE 2012, 7, e45987. [Google Scholar] [CrossRef] [Green Version]
  124. Nilsson, J.P.; Söderström, M.; Karlsson, A.U.; Lekander, M.; Akerstedt, T.; Lindroth, N.E.; Axelsson, J. Less effective executive functioning after one night’s sleep deprivation. J. Sleep Res. 2005, 14, 1–6. [Google Scholar] [CrossRef]
  125. Randazzo, A.C.; Muehlbach, M.J.; Schweitzer, P.K.; Walsh, J.K. Cognitive function following acute sleep restriction in children ages 10–14. Sleep 1998, 21, 861–868. [Google Scholar] [PubMed]
  126. Chee, M.W.L.; Chuah, L.Y.M. Functional neuroimaging insights into how sleep and sleep deprivation affect memory and cognition. Curr. Opin. Neurol. 2008, 21, 417–423. [Google Scholar] [CrossRef] [PubMed]
  127. Drummond, S.P.; Gillin, J.C.; Brown, G.G. Increased cerebral response during a divided attention task following sleep deprivation. J. Sleep Res. 2001, 10, 85–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Killgore, W.D.S.; Weber, M. Sleep Deprivation and Cognitive Performance. In Sleep Deprivation and Disease; Bianchi, M.T., Ed.; Springer: New York, NY, USA, 2014; pp. 209–229. ISBN 978-1-4614-9086-9. [Google Scholar]
  129. Petersen, S.E.; Dubis, J.W. The mixed block/event-related design. NeuroImage 2012, 62, 1177–1184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Meltzer, J.A.; Negishi, M.; Constable, R.T. Biphasic hemodynamic responses influence deactivation and may mask activation in block-design fMRI paradigms. Hum. Brain Mapp. 2008, 29, 385–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Dale, A.M.; Buckner, R.L. Selective averaging of rapidly presented individual trials using fMRI. Hum. Brain Mapp. 1997, 5, 329–340. [Google Scholar] [CrossRef]
  132. Miezin, F.M.; Maccotta, L.; Ollinger, J.M.; Petersen, S.E.; Buckner, R.L. Characterizing the hemodynamic response: Effects of presentation rate, sampling procedure, and the possibility of ordering brain activity based on relative timing. Neuroimage 2000, 11, 735–759. [Google Scholar] [CrossRef] [Green Version]
  133. Donaldson, D.I. Parsing brain activity with fMRI and mixed designs: What kind of a state is neuroimaging in? Trends Neurosci. 2004, 27, 442–444. [Google Scholar] [CrossRef]
  134. Behroozi, M.; Reza Daliri, M.; Boyaci, H. Statistical Analysis Methods for the fMRI Data. Basic Clin. Neurosci. 2011, 2, 67–74. [Google Scholar]
  135. Otten, L.J.; Henson, R.N.A.; Rugg, M.D. State-related and item-related neural correlates of successful memory encoding. Nat. Neurosci. 2002, 5, 1339–1344. [Google Scholar] [CrossRef]
  136. Wasfy, M.M.; Baggish, A.L. Exercise Dose in Clinical Practice. Circulation 2016, 133, 2297–2313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Gronwald, T.; Bem Alves, A.C.; Murillo-Rodríguez, E.; Latini, A.; Schuette, J.; Budde, H. Standardization of exercise intensity and consideration of a dose-response is essential. Commentary on “Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models”, by Lourenco et al., published 2019 in Nature Medicine. J. Sport Health Sci. 2019, 8, 353–354. [Google Scholar] [CrossRef] [PubMed]
  138. Herold, F.; Müller, P.; Gronwald, T.; Müller, N.G. Dose-response matters–A perspective on the exercise prescription in exercise-cognition research. Front. Psychol. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Impellizzeri, F.M.; Marcora, S.M.; Coutts, A.J. Internal and External Training Load: 15 Years On. Int. J. Sports Physiol. Perform. 2019, 14, 270–273. [Google Scholar] [CrossRef] [PubMed]
  140. Vanrenterghem, J.; Nedergaard, N.J.; Robinson, M.A.; Drust, B. Training Load Monitoring in Team Sports: A Novel Framework Separating Physiological and Biomechanical Load-Adaptation Pathways. Sports Med. 2017, 47, 2135–2142. [Google Scholar] [CrossRef] [PubMed]
  141. Wallace, L.K.; Slattery, K.M.; Coutts, A.J. The ecological validity and application of the session-RPE method for quantifying training loads in swimming. J. Strength Cond. Res. 2009, 23, 33–38. [Google Scholar] [CrossRef] [Green Version]
  142. Halson, S.L. Monitoring training load to understand fatigue in athletes. Sports Med. 2014, 44 (Suppl. 2), S139–S147. [Google Scholar] [CrossRef] [Green Version]
  143. Bourdon, P.C.; Cardinale, M.; Murray, A.; Gastin, P.; Kellmann, M.; Varley, M.C.; Gabbett, T.J.; Coutts, A.J.; Burgess, D.J.; Gregson, W.; et al. Monitoring Athlete Training Loads: Consensus Statement. Int. J. Sports Physiol. Perform. 2017, 12, S2-161–S2-170. [Google Scholar] [CrossRef]
  144. Burgess, D.J. The Research Doesn’t Always Apply: Practical Solutions to Evidence-Based Training-Load Monitoring in Elite Team Sports. Int. J. Sports Physiol. Perform. 2017, 12, S2136–S2141. [Google Scholar] [CrossRef]
  145. McLaren, S.J.; Macpherson, T.W.; Coutts, A.J.; Hurst, C.; Spears, I.R.; Weston, M. The Relationships between Internal and External Measures of Training Load and Intensity in Team Sports: A Meta-Analysis. Sports Med. 2018, 48, 641–658. [Google Scholar] [CrossRef] [Green Version]
  146. Hillman, C.H.; Erickson, K.I.; Kramer, A.F. Be smart, exercise your heart: Exercise effects on brain and cognition. Nat. Rev. Neurosci. 2008, 9, 58–65. [Google Scholar] [CrossRef] [PubMed]
  147. Soga, K.; Masaki, H.; Gerber, M.; Ludyga, S. Acute and Long-term Effects of Resistance Training on Executive Function. J. Cogn. Enhanc. 2018, 56, 729. [Google Scholar] [CrossRef]
  148. Wilke, J.; Giesche, F.; Klier, K.; Vogt, L.; Herrmann, E.; Banzer, W. Acute Effects of Resistance Exercise on Cognitive Function in Healthy Adults: A Systematic Review with Multilevel Meta-Analysis. Sports Med. 2019, 49, 905–916. [Google Scholar] [CrossRef] [PubMed]
  149. Budde, H.; Voelcker-Rehage, C.; Pietrabyk-Kendziorra, S.; Ribeiro, P.; Tidow, G. Acute coordinative exercise improves attentional performance in adolescents. Neurosci. Lett. 2008, 441, 219–223. [Google Scholar] [CrossRef] [PubMed]
  150. Takahashi, S.; Grove, P.M. Comparison of the effects of running and badminton on executive function: A within-subjects design. PLoS ONE 2019, 14, e0216842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Zach, S.; Shalom, E. The Influence of Acute Physical Activity on Working Memory. Percept. Mot. Ski. 2016, 122, 365–374. [Google Scholar] [CrossRef] [PubMed]
  152. Arena, R.; Myers, J.; Kaminsky, L.A. Revisiting age-predicted maximal heart rate: Can it be used as a valid measure of effort? Am. Heart J. 2016, 173, 49–56. [Google Scholar] [CrossRef] [Green Version]
  153. Correa Mesa, J.F.; Cruz Martinez, L.E.; Correa Morales, J.C.; Rojas Valencia, J.T. Maximum Heart Rate during exercise: Reliability of the 220-age and Tanaka formulas in healthy young people at a moderate altitude. Rev. Fac. Med. 2015, 62, 579–585. [Google Scholar] [CrossRef]
  154. Esco, M.R.; Chamberlain, N.; Flatt, A.A.; Snarr, R.L.; Bishop, P.A.; Williford, H.N. Cross-Validation of Age-Predicted Maximal Heart Rate Equations Among Female Collegiate Athletes. J. Strength Cond. Res. 2015, 29, 3053–3059. [Google Scholar] [CrossRef]
  155. Robergs, R.A.; Landwehr, R. The surprising history of the “HRmax = 220-age” equiation. J. Exerc. Physiol. Online 2002, 5, 1–10. [Google Scholar]
  156. Sarzynski, M.A.; Rankinen, T.; Earnest, C.P.; Leon, A.S.; Rao, D.C.; Skinner, J.S.; Bouchard, C. Measured maximal heart rates compared to commonly used age-based prediction equations in the Heritage Family Study. Am. J. Hum. Biol. 2013, 25, 695–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Silva, V.A.P.; Bottaro, M.; Justino, M.A.; Ribeiro, M.M.; Lima, R.M.; Oliveira, R.J. Maximum Heart Rate in Brazilian Elderly Women: Comparing Measured and Predicted Values. Arq. Bras. Cardiol. 2007, 88, 147–162. [Google Scholar] [CrossRef]
  158. Strzelczyk, T.A.; Quigg, R.J.; Pfeifer, P.B.; Parker, M.A.; Greenland, P. Accuracy of estimating exercise prescription intensity in patients with left ventricular systolic dysfunction. J. Cardiopulm. Rehabil. 2001, 21, 158–163. [Google Scholar] [CrossRef] [PubMed]
  159. Gronwald, T.; Velasques, B.; Ribeiro, P.; Machado, S.; Murillo-Rodríguez, E.; Ludyga, S.; Yamamoto, T.; Budde, H. Increasing exercise’s effect on mental health: Exercise intensity does matter. Proc. Natl. Acad. Sci. USA 2018, 115, E11890–E11891. [Google Scholar] [CrossRef] [Green Version]
  160. Murphy, K.; Birn, R.M.; Bandettini, P.A. Resting-state fMRI confounds and cleanup. Neuroimage 2013, 80, 349–359. [Google Scholar] [CrossRef] [Green Version]
  161. Liu, T.T. Noise contributions to the fMRI signal: An overview. Neuroimage 2016, 143, 141–151. [Google Scholar] [CrossRef]
  162. Peeters, R.; Sunaert, S. Clinical BOLD fMRI: Artifacts, Tips and Tricks. In Clinical Functional MRI; Springer: Berlin/Heidelberg, Germany, 2007; Volume 23, pp. 227–249. [Google Scholar] [CrossRef]
  163. Peeters, R.; Sunaert, S. Clinical BOLD fMRI and DTI: Artifacts, Tips, and Tricks. In Clinical Functional MRI; Stippich, C., Ed.; Springer: Berlin/Heidelberg, Germany, 2015; pp. 313–336. ISBN 978-3-662-45122-9. [Google Scholar]
  164. Caballero-Gaudes, C.; Reynolds, R.C. Methods for cleaning the BOLD fMRI signal. Neuroimage 2017, 154, 128–149. [Google Scholar] [CrossRef] [Green Version]
  165. Hutton, C.; Josephs, O.; Stadler, J.; Featherstone, E.; Reid, A.; Speck, O.; Bernarding, J.; Weiskopf, N. The impact of physiological noise correction on fMRI at 7 T. Neuroimage 2011, 57, 101–112. [Google Scholar] [CrossRef] [Green Version]
  166. Bulte, D.; Wartolowska, K. Monitoring cardiac and respiratory physiology during FMRI. Neuroimage 2017, 154, 81–91. [Google Scholar] [CrossRef]
  167. Özbay, P.S.; Chang, C.; Picchioni, D.; Mandelkow, H.; Chappel-Farley, M.G.; van Gelderen, P.; Zwart, J.A.; Duyn, J. Sympathetic activity contributes to the fMRI signal. Commun. Biol. 2019, 2, 421. [Google Scholar] [CrossRef]
  168. Strother, S.C. Evaluating fMRI preprocessing pipelines. IEEE Eng. Med. Biol. Mag. 2006, 25, 27–41. [Google Scholar] [CrossRef] [PubMed]
  169. Power, J.D.; Schlaggar, B.L.; Petersen, S.E. Recent progress and outstanding issues in motion correction in resting state fMRI. Neuroimage 2015, 105, 536–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Power, J.D.; Mitra, A.; Laumann, T.O.; Snyder, A.Z.; Schlaggar, B.L.; Petersen, S.E. Methods to detect, characterize, and remove motion artifact in resting state fMRI. Neuroimage 2014, 84, 320–341. [Google Scholar] [CrossRef]
  171. Gorgolewski, K.J.; Storkey, A.J.; Bastin, M.E.; Whittle, I.; Pernet, C. Single subject fMRI test-retest reliability metrics and confounding factors. Neuroimage 2013, 69, 231–243. [Google Scholar] [CrossRef] [PubMed]
  172. Patriat, R.; Molloy, E.K.; Meier, T.B.; Kirk, G.R.; Nair, V.A.; Meyerand, M.E.; Prabhakaran, V.; Birn, R.M. The effect of resting condition on resting-state fMRI reliability and consistency: A comparison between resting with eyes open, closed, and fixated. Neuroimage 2013, 78, 463–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Pruim, R.H.R.; Mennes, M.; Buitelaar, J.K.; Beckmann, C.F. Evaluation of ICA-AROMA and alternative strategies for motion artifact removal in resting state fMRI. Neuroimage 2015, 112, 278–287. [Google Scholar] [CrossRef] [PubMed]
  174. Pruim, R.H.R.; Mennes, M.; van Rooij, D.; Llera, A.; Buitelaar, J.K.; Beckmann, C.F. ICA-AROMA: A robust ICA-based strategy for removing motion artifacts from fMRI data. Neuroimage 2015, 112, 267–277. [Google Scholar] [CrossRef]
  175. Oakes, T.R.; Johnstone, T.; Ores Walsh, K.S.; Greischar, L.L.; Alexander, A.L.; Fox, A.S.; Davidson, R.J. Comparison of fMRI motion correction software tools. Neuroimage 2005, 28, 529–543. [Google Scholar] [CrossRef] [Green Version]
  176. Morgan, V.L.; Dawant, B.M.; Li, Y.; Pickens, D.R. Comparison of fMRI statistical software packages and strategies for analysis of images containing random and stimulus-correlated motion. Comput. Med. Imaging Graph. 2007, 31, 436–446. [Google Scholar] [CrossRef] [Green Version]
  177. Edward, V.; Windischberger, C.; Cunnington, R.; Erdler, M.; Lanzenberger, R.; Mayer, D.; Endl, W.; Beisteiner, R. Quantification of fMRI artifact reduction by a novel plaster cast head holder. Hum. Brain Mapp. 2000, 11, 207–213. [Google Scholar] [CrossRef]
  178. Heim, S.; Amunts, K.; Mohlberg, H.; Wilms, M.; Friederici, A.D. Head motion during overt language production in functional magnetic resonance imaging. Neuroreport 2006, 17, 579–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Chang, C.; Cunningham, J.P.; Glover, G.H. Influence of heart rate on the BOLD signal: The cardiac response function. Neuroimage 2009, 44, 857–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Frank, L.R.; Buxton, R.B.; Wong, E.C. Estimation of respiration-induced noise fluctuations from undersampled multislice fMRI data. Magn. Reson. Med. 2001, 45, 635–644. [Google Scholar] [CrossRef] [PubMed]
  181. Shmueli, K.; van Gelderen, P.; Zwart, J.A.; Horovitz, S.G.; Fukunaga, M.; Jansma, J.M.; Duyn, J.H. Low-frequency fluctuations in the cardiac rate as a source of variance in the resting-state fMRI BOLD signal. Neuroimage 2007, 38, 306–320. [Google Scholar] [CrossRef] [Green Version]
  182. Weiss, F.; Zamoscik, V.; Schmidt, S.N.L.; Halli, P.; Kirsch, P.; Gerchen, M.F. Just a very expensive breathing training? Risk of respiratory artefacts in functional connectivity-based real-time fMRI neurofeedback. Neuroimage 2020, 210, 116580. [Google Scholar] [CrossRef] [PubMed]
  183. Abbott, D.F.; Opdam, H.I.; Briellmann, R.S.; Jackson, G.D. Brief breath holding may confound functional magnetic resonance imaging studies. Hum. Brain Mapp. 2005, 24, 284–290. [Google Scholar] [CrossRef]
  184. Birn, R.M.; Diamond, J.B.; Smith, M.A.; Bandettini, P.A. Separating respiratory-variation-related fluctuations from neuronal-activity-related fluctuations in fMRI. Neuroimage 2006, 31, 1536–1548. [Google Scholar] [CrossRef]
  185. Birn, R.M.; Murphy, K.; Handwerker, D.A.; Bandettini, P.A. fMRI in the presence of task-correlated breathing variations. Neuroimage 2009, 47, 1092–1104. [Google Scholar] [CrossRef] [Green Version]
  186. Gretton, A.; Belitski, A.; Murayama, Y.; Schölkopf, B.; Logothetis, N. The effect of artifacts on dependence measurement in fMRI. Magn. Reson. Imaging 2006, 24, 401–409. [Google Scholar] [CrossRef]
  187. Kemna, L.J.; Posse, S. Effect of respiratory CO(2) changes on the temporal dynamics of the hemodynamic response in functional MR imaging. Neuroimage 2001, 14, 642–649. [Google Scholar] [CrossRef] [Green Version]
  188. Magon, S.; Basso, G.; Farace, P.; Ricciardi, G.K.; Beltramello, A.; Sbarbati, A. Reproducibility of BOLD signal change induced by breath holding. Neuroimage 2009, 45, 702–712. [Google Scholar] [CrossRef]
  189. Raj, D.; Anderson, A.W.; Gore, J.C. Respiratory effects in human functional magnetic resonance imaging due to bulk susceptibility changes. Phys. Med. Biol. 2001, 46, 3331–3340. [Google Scholar] [CrossRef] [PubMed]
  190. Wong, S.W.H.; Xue, G.; Bechara, A. Integrating fMRI with psychophysiological measurements in the study of decision-making. J. Neurosci. Psychol. Econ. 2011, 4, 85–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Kugel, H.; Bremer, C.; Püschel, M.; Fischbach, R.; Lenzen, H.; Tombach, B.; van Aken, H.; Heindel, W. Hazardous situation in the MR bore: Induction in ECG leads causes fire. Eur. Radiol. 2003, 13, 690–694. [Google Scholar] [CrossRef] [PubMed]
  192. Bright, M.G.; Tench, C.R.; Murphy, K. Potential pitfalls when denoising resting state fMRI data using nuisance regression. Neuroimage 2017, 154, 159–168. [Google Scholar] [CrossRef] [PubMed]
  193. Churchill, N.W.; Strother, S.C. PHYCAA+: An optimized, adaptive procedure for measuring and controlling physiological noise in BOLD fMRI. Neuroimage 2013, 82, 306–325. [Google Scholar] [CrossRef]
  194. Churchill, N.W.; Yourganov, G.; Spring, R.; Rasmussen, P.M.; Lee, W.; Ween, J.E.; Strother, S.C. PHYCAA: Data-driven measurement and removal of physiological noise in BOLD fMRI. Neuroimage 2012, 59, 1299–1314. [Google Scholar] [CrossRef]
  195. Kasper, L.; Bollmann, S.; Diaconescu, A.O.; Hutton, C.; Heinzle, J.; Iglesias, S.; Hauser, T.U.; Sebold, M.; Manjaly, Z.-M.; Pruessmann, K.P.; et al. The PhysIO Toolbox for Modeling Physiological Noise in fMRI Data. J. Neurosci. Methods 2017, 276, 56–72. [Google Scholar] [CrossRef] [Green Version]
  196. Murphy, K.; Fox, M.D. Towards a consensus regarding global signal regression for resting state functional connectivity MRI. Neuroimage 2017, 154, 169–173. [Google Scholar] [CrossRef] [Green Version]
  197. Power, J.D.; Plitt, M.; Laumann, T.O.; Martin, A. Sources and implications of whole-brain fMRI signals in humans. Neuroimage 2017, 146, 609–625. [Google Scholar] [CrossRef] [Green Version]
  198. Parkes, L.; Fulcher, B.; Yücel, M.; Fornito, A. An evaluation of the efficacy, reliability, and sensitivity of motion correction strategies for resting-state functional MRI. Neuroimage 2018, 171, 415–436. [Google Scholar] [CrossRef] [PubMed]
  199. Ludyga, S.; Gerber, M.; Brand, S.; Holsboer-Trachsler, E.; Puhse, U. Acute effects of moderate aerobic exercise on specific aspects of executive function in different age and fitness groups: A meta-analysis. Psychophysiology 2016, 11, 1611–1626. [Google Scholar] [CrossRef] [PubMed]
  200. Oberste, M.; Javelle, F.; Sharma, S.; Joisten, N.; Walzik, D.; Bloch, W.; Zimmer, P. Effects and Moderators of Acute Aerobic Exercise on Subsequent Interference Control: A Systematic Review and Meta-Analysis. Front. Psychol. 2019, 10, 609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Boecker, H.; Henriksen, G.; Sprenger, T.; Miederer, I.; Willoch, F.; Valet, M.; Berthele, A.; Tölle, T.R. Positron emission tomography ligand activation studies in the sports sciences: Measuring neurochemistry in vivo. Methods 2008, 45, 307–318. [Google Scholar] [CrossRef] [PubMed]
  202. Boecker, H.; Othman, A.; Mueckter, S.; Scheef, L.; Pensel, M.; Daamen, M.; Jankowski, J.; Schild, H.; Tölle, T.; Schreckenberger, M. Advocating neuroimaging studies of transmitter release in human physical exercise challenges studies. Open Access J. Sports Med. 2010, 1, 167–175. [Google Scholar] [CrossRef] [Green Version]
  203. Boecker, H.; Sprenger, T.; Spilker, M.E.; Henriksen, G.; Koppenhoefer, M.; Wagner, K.J.; Valet, M.; Berthele, A.; Tolle, T.R. The runner’s high: Opioidergic mechanisms in the human brain. Cereb. Cortex 2008, 18, 2523–2531. [Google Scholar] [CrossRef] [Green Version]
  204. Boecker, H.; Tölle, T.R.; Valet, M.; Sprenger, T. Effects of Aerobic Exercise on Mood and Human Opioidergic Activation Measured by Positron Emission Tomography. In Functional Neuroimaging in Exercise and Sport Sciences; Boecker, H., Hillman, C.H., Scheef, L., Strüder, H.K., Eds.; Springer: New York, NY, USA, 2012; pp. 499–510. ISBN 9781461432920. [Google Scholar]
  205. van Hall, G.; Stromstad, M.; Rasmussen, P.; Jans, O.; Zaar, M.; Gam, C.; Quistorff, B.; Secher, N.H.; Nielsen, H.B. Blood lactate is an important energy source for the human brain. J. Cereb. Blood Flow Metab. 2009, 29, 1121–1129. [Google Scholar] [CrossRef]
  206. Quistorff, B.; Secher, N.H.; van Lieshout, J.J. Lactate fuels the human brain during exercise. FASEB J. 2008, 22, 3443–3449. [Google Scholar] [CrossRef]
  207. Proia, P.; Di Liegro, C.M.; Schiera, G.; Fricano, A.; Di Liegro, I. Lactate as a Metabolite and a Regulator in the Central Nervous System. Int. J. Mol. Sci. 2016, 17, 1450. [Google Scholar] [CrossRef] [Green Version]
  208. Dennis, A.; Thomas, A.G.; Rawlings, N.B.; Near, J.; Nichols, T.E.; Clare, S.; Johansen-Berg, H.; Stagg, C.J. An Ultra-High Field Magnetic Resonance Spectroscopy Study of Post Exercise Lactate, Glutamate and Glutamine Change in the Human Brain. Front. Physiol. 2015, 6, 351. [Google Scholar] [CrossRef]
  209. Taher, M.; Leen, W.G.; Wevers, R.A.; Willemsen, M.A. Lactate and its many faces. Eur. J. Paediatr. Neurol. 2016, 20, 3–10. [Google Scholar] [CrossRef] [PubMed]
  210. Riske, L.; Thomas, R.K.; Baker, G.B.; Dursun, S.M. Lactate in the brain: An update on its relevance to brain energy, neurons, glia and panic disorder. Ther. Adv. Psychopharmacol. 2017, 7, 85–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Brooks, G.A. The Science and Translation of Lactate Shuttle Theory. Cell Metab. 2018, 27, 757–785. [Google Scholar] [CrossRef] [PubMed]
  212. Sobral-Monteiro-Junior, R.; Maillot, P.; Gatica-Rojas, V.; Ávila, W.R.M.; Paula, A.M.B.; Guimarães, A.L.S.; Santos, S.H.S.; Pupe, C.C.B.; Deslandes, A.C. Is the “lactormone” a key-factor for exercise-related neuroplasticity? A hypothesis based on an alternative lactate neurobiological pathway. Med. Hypotheses 2019, 123, 63–66. [Google Scholar] [CrossRef]
  213. Kemppainen, J.; Aalto, S.; Fujimoto, T.; Kalliokoski, K.K.; Långsjö, J.; Oikonen, V.; Rinne, J.; Nuutila, P.; Knuuti, J. High intensity exercise decreases global brain glucose uptake in humans. J. Physiol. 2005, 568, 323–332. [Google Scholar] [CrossRef] [Green Version]
  214. Tsukamoto, H.; Suga, T.; Takenaka, S.; Tanaka, D.; Takeuchi, T.; Hamaoka, T.; Isaka, T.; Ogoh, S.; Hashimoto, T. Repeated high-intensity interval exercise shortens the positive effect on executive function during post-exercise recovery in healthy young males. Physiol. Behav. 2016, 160, 26–34. [Google Scholar] [CrossRef] [Green Version]
  215. Hashimoto, T.; Tsukamoto, H.; Takenaka, S.; Olesen, N.D.; Petersen, L.G.; Sørensen, H.; Nielsen, H.B.; Secher, N.H.; Ogoh, S. Maintained exercise-enhanced brain executive function related to cerebral lactate metabolism in men. FASEB J. 2017, 32, 1417–1427. [Google Scholar] [CrossRef] [Green Version]
  216. Li, M.; Newton, A.T.; Anderson, A.W.; Ding, Z.; Gore, J.C. Characterization of the hemodynamic response function in white matter tracts for event-related fMRI. Nat. Commun. 2019, 10, 1140. [Google Scholar] [CrossRef] [Green Version]
  217. Ding, Z.; Huang, Y.; Bailey, S.K.; Gao, Y.; Cutting, L.E.; Rogers, B.P.; Newton, A.T.; Gore, J.C. Detection of synchronous brain activity in white matter tracts at rest and under functional loading. Proc. Natl. Acad. Sci. USA 2018, 115, 595–600. [Google Scholar] [CrossRef] [Green Version]
  218. Fraser, L.M.; Stevens, M.T.; Beyea, S.D.; D’Arcy, R.C.N. White versus gray matter: fMRI hemodynamic responses show similar characteristics, but differ in peak amplitude. BMC Neurosci. 2012, 13, 91. [Google Scholar] [CrossRef] [Green Version]
  219. Gawryluk, J.R.; D’Arcy, R.C.N.; Mazerolle, E.L.; Brewer, K.D.; Beyea, S.D. Functional mapping in the corpus callosum: A 4T fMRI study of white matter. Neuroimage 2011, 54, 10–15. [Google Scholar] [CrossRef]
  220. Gawryluk, J.R.; Mazerolle, E.L.; D’Arcy, R.C.N. Does functional MRI detect activation in white matter? A review of emerging evidence, issues, and future directions. Front. Neurosci. 2014, 8, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Mazerolle, E.L.; Beyea, S.D.; Gawryluk, J.R.; Brewer, K.D.; Bowen, C.V.; D’Arcy, R.C.N. Confirming white matter fMRI activation in the corpus callosum: Co-Localization with DTI tractography. Neuroimage 2010, 50, 616–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Tae, W.S.; Yakunina, N.; Kim, T.S.; Kim, S.S.; Nam, E.-C. Activation of auditory white matter tracts as revealed by functional magnetic resonance imaging. Neuroradiology 2014, 56, 597–605. [Google Scholar] [CrossRef] [PubMed]
  223. Yarkoni, T.; Barch, D.M.; Gray, J.R.; Conturo, T.E.; Braver, T.S. BOLD correlates of trial-by-trial reaction time variability in gray and white matter: A multi-study fMRI analysis. PLoS ONE 2009, 4, e4257. [Google Scholar] [CrossRef] [Green Version]
  224. Garrett, D.D.; Samanez-Larkin, G.R.; MacDonald, S.W.S.; Lindenberger, U.; McIntosh, A.R.; Grady, C.L. Moment-to-moment brain signal variability: A next frontier in human brain mapping? Neurosci. Biobehav. Rev. 2013, 37, 610–624. [Google Scholar] [CrossRef] [Green Version]
  225. Grady, C.L.; Garrett, D.D. Brain signal variability is modulated as a function of internal and external demand in younger and older adults. Neuroimage 2017, 169, 510–523. [Google Scholar] [CrossRef]
  226. Garrett, D.D.; Lindenberger, U.; Hoge, R.D.; Gauthier, C.J. Age differences in brain signal variability are robust to multiple vascular controls. Sci. Rep. 2017, 7, 10149. [Google Scholar] [CrossRef]
  227. Armbruster-Genç, D.J.N.; Ueltzhöffer, K.; Fiebach, C.J. Brain Signal Variability Differentially Affects Cognitive Flexibility and Cognitive Stability. J. Neurosci. 2016, 36, 3978–3987. [Google Scholar] [CrossRef] [Green Version]
  228. van Horn, J.D.; Grafton, S.T.; Miller, M.B. Individual Variability in Brain Activity: A Nuisance or an Opportunity? Brain Imaging Behav. 2008, 2, 327–334. [Google Scholar] [CrossRef] [Green Version]
  229. Dinstein, I.; Heeger, D.J.; Behrmann, M. Neural variability: Friend or foe? Trends Cogn. Sci. 2015, 19, 322–328. [Google Scholar] [CrossRef]
  230. Garrett, D.D.; Kovacevic, N.; McIntosh, A.R.; Grady, C.L. Blood oxygen level-dependent signal variability is more than just noise. J. Neurosci. 2010, 30, 4914–4921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Garrett, D.D.; Nagel, I.E.; Preuschhof, C.; Burzynska, A.Z.; Marchner, J.; Wiegert, S.; Jungehülsing, G.J.; Nyberg, L.; Villringer, A.; Li, S.-C.; et al. Amphetamine modulates brain signal variability and working memory in younger and older adults. Proc. Natl. Acad. Sci. USA 2015, 112, 7593–7598. [Google Scholar] [CrossRef] [Green Version]
  232. Nomi, J.S.; Bolt, T.S.; Ezie, C.E.C.; Uddin, L.Q.; Heller, A.S. Moment-to-Moment BOLD Signal Variability Reflects Regional Changes in Neural Flexibility across the Lifespan. J. Neurosci. 2017, 37, 5539–5548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Protzner, A.B.; Kovacevic, N.; Cohn, M.; McAndrews, M.P. Characterizing functional integrity: Intraindividual brain signal variability predicts memory performance in patients with medial temporal lobe epilepsy. J. Neurosci. 2013, 33, 9855–9865. [Google Scholar] [CrossRef] [PubMed]
  234. Button, K.S.; Ioannidis, J.P.; Mokrysz, C.; Nosek, B.A.; Flint, J.; Robinson, E.S.J.; Munafò, M.R. Power failure: Why small sample size undermines the reliability of neuroscience. Nat. Rev. Neurosci. 2013, 14, 365–376. [Google Scholar] [CrossRef] [Green Version]
  235. de Vet, H.C.; Terwee, C.B.; Knol, D.L.; Bouter, L.M. When to use agreement versus reliability measures. J. Clin. Epidemiol. 2006, 59, 1033–1039. [Google Scholar] [CrossRef] [Green Version]
  236. Atkinson, G.; Nevill, A.M. Statistical methods for assessing measurement error (reliability) in variables relevant to sports medicine. Sports Med. 1998, 26, 217–238. [Google Scholar] [CrossRef]
  237. Donath, L.; Ludyga, S.; Hammes, D.; Rossmeissl, A.; Andergassen, N.; Zahner, L.; Faude, O. Absolute and relative reliability of acute effects of aerobic exercise on executive function in seniors. BMC Geriatr. 2017, 17, 247. [Google Scholar] [CrossRef] [Green Version]
  238. Birn, R.M.; Molloy, E.K.; Patriat, R.; Parker, T.; Meier, T.B.; Kirk, G.R.; Nair, V.A.; Meyerand, M.E.; Prabhakaran, V. The effect of scan length on the reliability of resting-state fMRI connectivity estimates. Neuroimage 2013, 83, 550–558. [Google Scholar] [CrossRef] [Green Version]
  239. Bennett, C.M.; Miller, M.B. fMRI reliability: Influences of task and experimental design. Cogn. Affect. Behav. Neurosci. 2013, 13, 690–702. [Google Scholar] [CrossRef] [PubMed]
  240. Burke, T.M.; Scheer, F.A.J.L.; Ronda, J.M.; Czeisler, C.A.; Wright, K.P. Sleep inertia, sleep homeostatic and circadian influences on higher-order cognitive functions. J. Sleep Res. 2015, 24, 364–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Anderson, J.A.E.; Campbell, K.L.; Amer, T.; Grady, C.L.; Hasher, L. Timing is everything: Age differences in the cognitive control network are modulated by time of day. Psychol. Aging 2014, 29, 648–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Hodyl, N.A.; Schneider, L.; Vallence, A.-M.; Clow, A.; Ridding, M.C.; Pitcher, J.B. The cortisol awakening response is associated with performance of a serial sequence reaction time task. Int. J. Psychophysiol. 2016, 100, 12–18. [Google Scholar] [CrossRef]
  243. Kojima, S.; Abe, T.; Morishita, S.; Inagaki, Y.; Qin, W.; Hotta, K.; Tsubaki, A. Acute moderate-intensity exercise improves 24-h sleep deprivation-induced cognitive decline and cerebral oxygenation: A near-infrared spectroscopy study. Respir. Physiol. Neurobiol. 2019, 274, 103354. [Google Scholar] [CrossRef]
  244. Byun, K.; Hyodo, K.; Suwabe, K.; Ochi, G.; Sakairi, Y.; Kato, M.; Dan, I.; Soya, H. Positive effect of acute mild exercise on executive function via arousal-related prefrontal activations: An fNIRS study. Neuroimage 2014, 98, 336–345. [Google Scholar] [CrossRef]
  245. Kato, K.; Miyata, S.; Ando, M.; Matsuoka, H.; Yasuma, F.; Iwamoto, K.; Kawano, N.; Banno, M.; Ozaki, N.; Noda, A. Influence of sleep duration on cortical oxygenation in elderly individuals. Psychiatry Clin. Neurosci. 2017, 71, 44–51. [Google Scholar] [CrossRef]
  246. Kato, K.; Iwamoto, K.; Kawano, N.; Noda, Y.; Ozaki, N.; Noda, A. Differential effects of physical activity and sleep duration on cognitive function in young adults. J. Sport Health Sci. 2017, 7, 227–236. [Google Scholar] [CrossRef]
  247. Nishida, M.; Kikuchi, S.; Miwakeichi, F.; Suda, S. Night duty and decreased brain activity of medical residents: A wearable optical topography study. Med. Educ. Online 2017, 22, 1379345. [Google Scholar] [CrossRef] [Green Version]
  248. Suda, M.; Sato, T.; Kameyama, M.; Ito, M.; Suto, T.; Yamagishi, Y.; Uehara, T.; Fukuda, M.; Mikuni, M. Decreased cortical reactivity underlies subjective daytime light sleepiness in healthy subjects: A multichannel near-infrared spectroscopy study. Neurosci. Res. 2008, 60, 319–326. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of the neurovascular unit and changes in cerebral hemodynamics and oxygenation induced by neural activity (e.g., as a consequence of solving a cognitive task). Furthermore, in the lower left part of the illustration, the basic physical principles of magnetic resonance imaging using the BOLD contrast (Blood Oxygenation Level Depend) are displayed.
Figure 1. Schematic illustration of the neurovascular unit and changes in cerebral hemodynamics and oxygenation induced by neural activity (e.g., as a consequence of solving a cognitive task). Furthermore, in the lower left part of the illustration, the basic physical principles of magnetic resonance imaging using the BOLD contrast (Blood Oxygenation Level Depend) are displayed.
Brainsci 10 00175 g001
Figure 2. Flow chart with information about the search, screening, and selection processes that led to the identification of the relevant articles for inclusion in this systematic review.
Figure 2. Flow chart with information about the search, screening, and selection processes that led to the identification of the relevant articles for inclusion in this systematic review.
Brainsci 10 00175 g002
Figure 3. Illustration of the analysis of the risk of bias assessment of the included studies. This figure was created by using Review Manager [98] and follows the recommendations outlined in the Cochrane Collaboration guidelines [97]. A “green plus” indicates a low risk of bias and a “yellow question mark” indicates an unclear risk of bias.
Figure 3. Illustration of the analysis of the risk of bias assessment of the included studies. This figure was created by using Review Manager [98] and follows the recommendations outlined in the Cochrane Collaboration guidelines [97]. A “green plus” indicates a low risk of bias and a “yellow question mark” indicates an unclear risk of bias.
Brainsci 10 00175 g003
Figure 4. Schematic illustration of the recommended study design (a within-subject crossover design with a pretest-posttest comparison).
Figure 4. Schematic illustration of the recommended study design (a within-subject crossover design with a pretest-posttest comparison).
Brainsci 10 00175 g004
Table 1. Overview of the selected population characteristics and main findings of the reviewed studies.
Table 1. Overview of the selected population characteristics and main findings of the reviewed studies.
First Author (year)Sample Characteristics and Study Design
(1.) Health Status
(2.) Cardiorespiratory Fitness Level (mean ± SD in ml/kg/min)
(3.) Number of Participants (gender)
(4.) Age (mean ± SD or range in years)
(5.) (i) Body Height (mean ± SD or range in cm)/ (ii) Body Mass (mean ± SD or range in kg)/ (iii) BMI (mean ± SD)
Main Findings
(1.) Behavioral Level
(2.) Functional Level
Bothe et al. (2013) [99](1.) Young healthy trained and inactive adults
(2.) HT (T) 56.0 ± 5.2; HT (P) 55.1 ± 6.2/ PIA (T) 48.4 ± 6.8; PIA (P) 50.2 ± 2.9
(3.) HT (T) N = 9 (9 m); HT (P) N = 10 (10 m)/ PIA (T) N = 11; PIA (P) N = 13 (13 m)
(4.) HT (T) 24.0 ± 4.0; HT (P) 26.1 ± 3.4/ PIA (T) 25.1 ± 3.0; PIA (P) 23.9 ± 2.7
(5.) (i) N.A./ (ii) N.A./ (iii) HT (T) 22.8 ± 2.0; HT (P) 23.0 ± 1.9, PIA (T) 22.4 ± 1.5; PIA (P) 21.6 ± 1.5
Treadmill running vs. placebo (stretching):
(1.) - no significant differences
(2.) ↓ lt. caudate nucleus in gain anticipation
 ↓ rt. caudate nucleus in gain feedback
Chen et al. (2016) [71](1.) Healthy children
(2.) N.A.
(3.) N = 9 (4 f/ 5 m)
(4.) 10.0 ± 0.0
(5.) (i) N.A./ (ii) N.A./ (iii) N.A.
Cycling vs. seated rest:
(1.) ↓ mean RT in the 2-back condition
(2.) ↑ lt. superior/inferior parietal gyrus, rt. superior parietal gyrus, lt. hippocampus, lt. cerebellum, rt. cerebellum
Chen et al. (2017) [73](1.) Healthy children
(2.) N.A.
(3.) N = 9 (4 f/ 5 m)
(4.) 10.0 ± 0.0
(5.) (i) N.A./ (ii) N.A./ (iii) N.A.
Cycling vs. seated rest:
(1.) ↓ mean RT in Eriksen flanker task
(2.) ↑ resting-state functional connectivity in the lt. cerebellum-rt. inferior gyrus is significantly negatively correlated with better performance (e.g., shorter reaction time) in an Eriksen flanker task
Li et al. (2014) [39](1.) Healthy younger adults
(2.) N.A.
(3.) N = 15 (15 f)
(4.) 19-22
(5.) (i) 161.6 ± 3.1/ (ii) 50.8 ± 3.4/ (iii) 19.5 ± 1.4
Cycling vs. seated rest:
(1.) - no significant differences
(2.) ↑ rt. middle frontal gyrus, rt. lingual gyrus, lt. fusiform gyrus/↓ anterior cingulate gyrus, rt. paracentral lobule, and lt. inferior frontal gyrus in the 2-back condition
Li et al. (2019) [106](1.) Healthy high-fit and low-fit younger adults
(2.) HF 26.5 ± 1.9/ LF 19.9 ± 1.0
(3.) HF N = 12 (12 f)/ LF N = 12 (12 f)
(4.) HF 25.5 ± 0.7/ LF 25.8 ± 0.6
(5.) (i) HF 164.8 ± 5.9; LF 161.1 ± 4.9/ (ii) HF 55.7 ± 5.1; LF 52.0 ± 5.0/ (iii) HF 20.5 ± 1.1; LF 20.0 ± 1.5
After cycling vs. prior cycling:
(1.) ↑ accuracy in LF in 0-back
 ↓ accuracy in HF in 1-back
(2.) ↓ rt. cerebellum in HF in 0-back and 1-back
 ↑ rt. cerebellum in LF in 0-back and 1-back
 ↑ lt. and rt. cerebellum, rt. paracentral lobule, and rt. medial temporal pole in HF and LF in 1-back
 ↑ lt. anterior cingulate gyrus and in lt. pallidum in HF in 2-back
 ↓ lt. anterior cingulate cortex in LF in 2-back
HF vs. LF:
(1.) ↑ accuracy in 1-back and 2-back (before cycling)
(2.) ↑ rt. cerebellum in 0-back (before cycling)
 ↓ rt. cerebellum in 0-back and 1-back (after cycling)
 ↑ lt. inferior parietal lobule in 0-back and 1-back
 ↓ lt. anterior cingulate gyrus and in lt. globus pallidus in 2-back (before cycling)
 ↑ lt. anterior cingulate gyrus and in lt. globus pallidus in 2-back (after cycling)
 ↑ rt. superior frontal gyrus in 2-back
MacIntosh et al. (2014) [72](1.) Healthy younger adults
(2.) N.A.
(3.) N = 16 (10 f/ 6 m)
(4.) 26.7 ± 4.1 (20-35)
(5.) (i) 171.0 ± 10.5/ (ii) 67.9 ± 13.9/ (iii) N.A.
After vs. prior cycling:
(1.) - no significant change
(2.) ↓ lt. parietal operculum
Mehren et al. (2019) [100](1.) Younger adults with and without ADHD
(2.) ADHD 36.6 ± 7.5/ HC 42.0 ± 7.3
(3.) ADHD N = 23 (3 f/ 20 m)/ HC N = 23 (4 f/ 20 m)
(4.) ADHD 31.4 ± 9.6/HC 29.5 ± 7.0
(5.) (i) N.A./ (ii) N.A./ (iii) ADHD 25.6 ± 4.3; HC 24.1 ± 2.6
Cycling vs. rest (watching movie):
(1.) – no significant differences
(2.) ↑ lt. middle occipital gyrus, rt. middle occipital gyrus, rt. supramarginal gyrus, and lt. inferior parietal gyrus during correct inhibitions in Go/No-Go task in ADHD
- negative correlation between exercise-related changes in brain activation in lt. insula, lt. precentral gyrus, and rt. postcentral gyrus during correct inhibitions in Go/No-Go task and task performance in the control condition in ADHD
ADHD vs. HC
(1.) – no significant differences
(2.) ↑ lt. superior occipital gyrus, rt. precuneus, and lt. supramarginal gyrus
Mehren et al. (2019) [101](1.) Younger healthy adults
(2.) MIE 39.6 ± 7.1/ HIE 37.0 ± 8.3
(3.) MIE N = 32 (16 f/ 16 m)/ HIE N = 31 (16 f/ 15 m)
(4.) MIE 29.3 ± 8.5/ HIE 28.6 ± 7.7
(5.) (i) N.A./ (ii) N.A./ (iii) MIE 23.8 ± 2.3; HIE 24.5 ± 4.8
Cycling vs. rest (watching movie):
(1.) ↓ reaction times in a congruent and incongruent condition (Flanker task) in HIIE
 ↑ sensitivity index (Go/No-Go task) in MICE
(2.) ↑ lt. superior frontal gyrus, rt. precentral gyrus and the triangular part of the lt. inferior frontal gyrus in Go/No-Go task (contrast “hits”) in MICE
 ↓ lt. anterior cingulate gyrus in a visual task in MICE
 ↑ lt. lingual gyrus and precuneus in a visual task in HIIE
 ↑ rt. precentral gyrus in Go/No-Go task (contrast “hits”) in MICE (males vs. females)
MICE vs. HIIE:
(1.) - no significant differences
(2.) ↑ lt. superior frontal gyrus and rt. insula in Go/No-Go task (contrast “hits”)
 ↓ lt. lingual gyrus, rt. precuneus, and lt. anterior cingulate gyrus in a visual task
Associations between brain activation and CRF:
- positive association between activation of rt. insula and VO2 peak in MICE (Flanker task contrast “incongruent – congruent”)
- negative association between activation of rt. postcentral gyrus VO2 peak in HIIE (Go/No-Go task – contrast “hits”)
- positive association between activation of lt. rolandic operculum VO2 peak in MICE (Go/No-Go task – contrast “correct inhibitions - hits”)
Mehren et al. (2019) [102](1.) Younger adults with and without ADHD
(2.) ADHD 37.1 ± 7.2/ HC 41.5 ± 7.3
(3.) ADHD N = 20 (4 f/ 16 m)/ HC N = 20 (5 f/ 15 m)
(4.) ADHD 29.9 ± 9.5/ HC 29.0 ± 7.4
(5.) (i) N.A./ (ii) N.A./ (iii) ADHD 25.0 ± 3.8; HC 24.3 ± 2.7
Cycling vs. rest (watching movie):
(1.) ↓ interference score (incongruent-congruent) in HC
 ↓ mean RT in a congruent and incongruent condition in ADHD
 ↓ RT variability in a congruent condition in ADHD
- significant positive correlation between RT differences (i.e., differences between exercise and control condition) in incongruent trials and VO2 peak in ADHD
(2.) ↓ rt. precentral gyrus and rt. middle temporal gyrus in a congruent condition in ADHD
 ↓ rt. superior frontal gyrus, rt. middle frontal gyrus and paracentral lobule in incongruent condition in ADHD
 ↓ rt. superior frontal gyrus and rt. middle frontal gyrus in a visual task in ADHD
Metcalfe et al. (2016) [105](1.) Adolescents with and without BD
(2.) N.A.
(3.) BD N = 30 (17 f/13 m)/ HC N = 20 (11 f/9 m)
(4.) BD 16.8 ± 1.4/ HC 16.1 ± 1.4
(5.) (i) N.A./ (ii) N.A./ (iii) N.A.
BD vs. HC:
(1.) – no significant differences
(2.) ↓ orbital part of the lt. inferior frontal gyrus, rt. frontal pole extending to temporal pole, rt. and lt. hippocampus and rt. amygdala in Go trials
Suwabe et al. (2018) [38](1.) Healthy younger adults
(2.) 37.9 ± 8.2
(3.) N = 16 (12 f/4 m)
(4.) 21.1 ± 2.0
(5.) (i) 164.2 ± 9.1/ (ii) 55.4 ± 7.7/ (iii) 20.5 ± 1.6
Cycling vs. seated rest:
(1.) ↑ better performance in mnemonic discrimination task (in high- and medium-similarity lures)
(2.) - significant positive associations between dentate gyrus/CA3 subfield bilaterally and lt. angular gyrus, lt. fusiform gyrus, lt. parahippocampal cortex, and lt. primary visual cortex
 - significant negative association between dentate gyrus/CA3 subfield bilaterally and lt. temporal pole
 - significant association between the whole hippocampus (CA1, subiculum and dentate gyrus/CA3) and bilateral parahippocampal cortex
 - higher correlations between dentate gyrus/CA3 and angular gyrus, fusiform gyrus as well as parahippocampal cortex predicted improvements in cognitive performance
Voss et al. (2019) [107](1.) Healthy older adults
(2.) 20.1 ± 5.0
(3.) N = 34 (20 f/ 14 m)
(4.) 67.1 ± 4.3 (60-80)
(5.) (i) N.A./ (ii) N.A./ (iii) 29.1 ± 5.3
After vs. prior cycling:
(1.) ↑ accuracy in 1-back and 2-back condition in LIE and MIE and ↑ accuracy in 1-back and 2-back condition in MIE vs. LIE
- acute improvements in working memory performance predict long-term improvements in working memory performance (after 12-weeks of training)
(2.) - acute changes in the following connections predict long-term changes in the same connections: (i) lt. middle occipital lobule and rt. angular gyrus, (ii) lt. superior frontal gyrus and lt. inferior temporal gyrus, (iii) lt. superior frontal gyrus and rt. inferior parietal gyrus, (iv) rt inferior parietal gyrus and rt. triangular part of the inferior frontal gyrus, (v) rt. inferior parietal gyrus and opercular part of the lt. inferior frontal gyrus, (vi) lt. lingual gyrus and rt. fusiform gyrus, (vii) rt. superior frontal gyrus and rt. middle temporal gyrus, (viii) rt. middle temporal gyrus and rt. inferior parietal gyrus, (ix) lt. and rt. precuneus, (x) lt. superior frontal gyrus and lt. medial superior frontal gyrus, (xi) rt. middle temporal gyrus and opercular part of the lt. inferior frontal gyrus, and (xii) rt. superior frontal gyrus and rt. inferior parietal gyrus
- acute and long-term changes in (i) connections between rt. middle temporal gyrus and rt. superior frontal gyrus, and (ii) connections between rt. inferior parietal gyrus and opercular part of the rt. inferior frontal gyrus are associated with performance in the 2-back condition
Won et al. (2019) [103](1.) Healthy older adults
(2.) N.A.
(3.) N = 32 (24 f/ 8 m)
(4.) 66.2 ± 7.3 (55-85)
(5.) (i) 166.6 ± 9.0/ (ii) 71.3 ± 14.2/ (iii) 25.5 ± 4.1
Cycling vs. seated rest:
(1.) ↑ accuracy in a congruent and incongruent condition
(2.) ↑ orbital part of the lt. inferior frontal gyrus and lt. inferior parietal gyrus in incongruent and incongruent-congruent condition
 ↓ rt. cingulate gyrus in incongruent and incongruent-congruent condition
Won et al. (2019) [104](1.) Healthy older adults
(2.) N.A.
(3.) N = 26 (20 f/ 6 m)
(4.) 65.9 ± 7.2 (55-85)
(5.) (i) 166.5 ± 8.5/ (ii) 73.6 ± 14.1/ (iii) 26.1 ± 4.3
Cycling vs. seated rest:
(1.) - no significant differences
(2.) ↑ orbital part of lt. inferior frontal gyrus, lt. inferior temporal gyrus, rt. middle temporal gyrus, lt. fusiform gyrus, and rt. and lt. hippocampus
ADHD: attention deficit hyperactivity disorder; BD: bipolar disorder; BMI: body mass index; cm: centimeter; f: female; HC: healthy controls; HF: high-fit group; HIIE: high-intensity interval exercise group; HT: highly trained individuals; kg: kilogram; LF: low-fit group; lt.: left; m: male; MICE: moderate-intensity continuous exercise group; min: minutes; N: number; N.A.: not applicable; P: placebo exercise (stretching and gymnastic tasks); PIA: physically inactive individuals; rt.: right; RT: reaction time; SD: standard deviation; T: treadmill exercise; VO2 max: highest value of maximal oxygen uptake attainable by a subject [109,110,111]; during cardiorespiratory fitness test; VO2 peak: highest “system-limited” oxygen uptake attained during the cardiorespiratory fitness test [109,110,111]; vs.: versus; ↑: significant increase; ↓: significant decrease.

Share and Cite

MDPI and ACS Style

Herold, F.; Aye, N.; Lehmann, N.; Taubert, M.; Müller, N.G. The Contribution of Functional Magnetic Resonance Imaging to the Understanding of the Effects of Acute Physical Exercise on Cognition. Brain Sci. 2020, 10, 175. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci10030175

AMA Style

Herold F, Aye N, Lehmann N, Taubert M, Müller NG. The Contribution of Functional Magnetic Resonance Imaging to the Understanding of the Effects of Acute Physical Exercise on Cognition. Brain Sciences. 2020; 10(3):175. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci10030175

Chicago/Turabian Style

Herold, Fabian, Norman Aye, Nico Lehmann, Marco Taubert, and Notger G. Müller. 2020. "The Contribution of Functional Magnetic Resonance Imaging to the Understanding of the Effects of Acute Physical Exercise on Cognition" Brain Sciences 10, no. 3: 175. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci10030175

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