Skip to main content

MINI REVIEW article

Front. Physiol., 15 August 2023
Sec. Exercise Physiology

The role of exercise parameters on small extracellular vesicles and microRNAs cargo in preventing neurodegenerative diseases

  • 1Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Study of Bari, Bari, Italy
  • 2Applied Neurophysiology and Pain Unit, Department of Translational Biomedicine and Neuroscience (DiBraiN), Policlinico General Hospital, University of Study of Bari, Bari, Italy
  • 3Neurophysiology Operative Unit, Department of Translational Biomedicine and Neuroscience (DiBraiN), Policlinico General Hospital, University of Study of Bari, Bari, Italy

Physical activity (PA), which includes exercise, can reduce the risk of developing various non-communicable diseases, including neurodegenerative diseases (NDs), and mitigate their adverse effects. However, the mechanisms underlying this ability are not yet fully understood. Among several possible mechanisms proposed, such as the stimulation of brain-derived neurotrophic factor (BDNF), endothelial nitric oxide synthase (eNOS), insulin-like growth factor-1 (IGF-1), vascular endothelial growth factor (VEGF), and nerve growth factor (NGF), the possible involvement of particular vesicular structures enclosed in lipid membranes known as extracellular vesicles (EVs) has recently been investigated. These EVs would appear to exert a paracrine and systemic action through their ability to carry various molecules, particularly so-called microRNAs (miRNAs), performing a function as mediators of intercellular communication. Interestingly, EVs and miRNAs are differentially expressed following PA, but evidence on how different exercise parameters may differentially affect EVs and the miRNAs they carry is still scarce. In this review we summarized the current human findings on the effects of PA and different exercise parameters exerted on EVs and their cargo, focusing on miRNAs molecules, and discussing how this may represent one of the biological mechanisms through which exercise contributes to preventing and slowing NDs.

1 Introduction

Neurodegenerative diseases (NDs) are characterized by progressive nerve cell deterioration, leading to neurodegeneration and disability (Dugger and Dickson, 2017). Although NDs represent a serious health problem in communities, modifiable risk factors, such as physical activity (PA), have received relatively little attention. These disorders are becoming more prevalent, in part because the elderly population has increased in recent years (Heemels, 2016). The most frequent NDs are dementias such as Alzheimer’s disease (AD) and frontotemporal dementia (FTD), which account for 60%–70%. Other NDs include Parkinson’s disease (PD), which is the second most frequent ND, Huntington’s disease (HD), motor neuron diseases such as amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS) and spino-cerebellar ataxias. These diseases differ in their pathophysiology: some cause impairments in memory and cognition, while others affect the ability to move, speak, and breathe (Abeliovich and Gitler, 2016; Canter et al., 2016; Taylor et al., 2016; Wyss-Coray, 2016; Filippi et al., 2018). There is currently no cure for any of these diseases, though not for lack of trying (Heemels, 2016).

Epidemiological studies have shown that PA can reduce the risk of AD and dementia by 45% and 28%, respectively (Hamer and Chida, 2009). In recent years, PA has shown its potential as a therapeutic approach that can modify NDs (Frederiksen et al., 2018). PA has been described as a nondrug therapy against numerous diseases such as neurological, metabolic, psychiatric and cardiovascular diseases (Pedersen and Saltin, 2015).

With the great advancement of molecular techniques, researchers have identified several molecules whose production is found to be stimulated by PA, such as increased superoxide dismutase (SOD) (Feter et al., 2019), brain-derived neurotrophic factor (BDNF) (Erickson et al., 2011; Coelho et al., 2014), endothelial nitric oxide synthase (eNOS) (Lange-Asschenfeldt, and Kojda, 2008), insulin-like growth factor-1 (IGF-1), vascular endothelial growth factor (VEGF) (Maass et al., 2016), and nerve growth factor (NGF), and decreased production of damaging free radicals in the hippocampus region of the brain, which are mainly involved in memory (Ionescu-Tucker and Cotman, 2021). However, the exact mechanism by which PA exerts these effects is not yet fully understood. Understanding the molecular and cellular mechanisms of exercise-induced benefits is of great interest for preventive and therapeutic implications.

Recently, it has been observed that, during exercise, various organs and cells communicate through the secretion of biomolecules called exerkines. These molecules, including myokines, adipokines, hepatokines, act as signaling molecules that can have autocrine, paracrine, and endocrine effects. They are released into circulation and can influence multiple tissues and organs throughout the body. Exerkines play a crucial role in mediating the beneficial effects of exercise on various physiological processes, including muscle mass, metabolism, immune function, cardiovascular health, and neuroprotection (Magliulo et al., 2022; Rody et al., 2022). To preserve their integrity during transit, these molecules need to be transported by specialized structures such as extracellular vesicles (EVs), a wide family of membranous structures, including the most well-known exosomes (EXs) (Théry et al., 2018; van Niel et al., 2018), which facilitate intercellular communication and cross-talk between different organs (Magliulo et al., 2022). Consequently, EVs and specifically EXs have been proposed as an additional mechanism, probably underlying others already identified, through which PA may exert preventive and therapeutic effects on different kinds of pathologic conditions, including NDs (Whitham and Febbraio, 2019; Rigamonti et al., 2020). However, it is still unclear how exercise parameters like frequency, intensity, time, type, volume, and progression (FITT-VP) can affect the modulation of EVs and their cargo. There is still a gap between precisely characterizing the molecular pathways that exercise induces and the possible health advantages that have been reported. Developing tailored exercise interventions and creating molecular-level interventions that mimic exercise could benefit from a deeper understanding of these biological processes and pathways (Sanford et al., 2020). The study of EVs is still in its infancy, especially in terms of its relationship to PA. Therefore, we explore and summarize the currently available human evidence, regarding the effects that PA and related FITT-VP parameters may exert on EVs and their cargo, focusing on miRNAs molecules, aiming to examine how this process may represent one of the biological mechanisms by which exercise plays a role in the prevention and slowing of NDs.

2 Extracellular vesicles

EVs, defined as eukaryotic cell membrane fragments, consist of a phospholipid bilayer and serve as carriers of various molecules (DNA, mRNA, microRNA, cytokines, proteins and lipids), protecting them from degradative agents during transport in the bloodstream (Szatanek et al., 2017). Based on their origin and size, EVs are mainly divided into exosomes, microvesicles, and apoptotic bodies. Exosomes (50/150 nm ø) are EVs that originate from the late endosome, while microvesicles (100/1,000 nm ø) originate directly from the plasma membrane and apoptotic bodies (100/5,000 nm ø) are released following cell fragmentation during the late phase of apoptosis (Yuan et al., 2019).

Cells of the central nervous system (CNS), like neurons, oligodendrocytes and astrocytes, also secrete EVs, important in intracellular communication, neuronal plasticity and myelination, as well as in neuroprotective and regenerative processes. EXs appear capable of carrying enzymes with metabolic functions (e.g., catalase and superoxide dismutase-1) that are useful to neurons for increased resistance to oxidative stress (Yuan et al., 2019). However, the role played by EVs in the CNS remains a subject of conflicting evidence. Trotta et al. (2018) highlight their potential involvement in NDs like AD, PD, HD and ALS. These EVs are believed to facilitate the transport of misfolded proteins (tau, amyloid-β, and α-synuclein), which are implicated in the pathogenic process. The authors suggest that under normal conditions, glial cells secrete trophic and protective factors for neurons, but in pathological conditions, they undergo a phenotypic change, contributing to the spread of toxicity through EVs. Similarly, D’Anca et al. (2021) report that in MS, the first neurological disease in which EVs have been detected, it appears that these vesicles may play a role in myelin damage and inflammation but, on the other hand, they could also exert a neuroprotective role, being involved in the processes of modulating synaptic plasticity in the CNS and repairing damaged neurons.

Recent studies have suggested that EVs like EXs play a role in organ-to-organ communication, moreover, have shown that exercise can lead to an increase in the release of EVs, into the bloodstream. In addition, exercise can stimulate EXs production and microRNAs (miRNAs) processing in muscle, which may have implications for systemic metabolism. Furthermore, circulating exosomal miRNAs (exomiRs) have been found to be altered by acute exercise, with specific miRNAs showing changes in expression levels. These exercise-regulated exomiRs often target components of the IGF1 signaling pathway, which is involved in muscle growth and glucose homeostasis. Additionally, EVs secreted during exercise originate from various cell types in the circulatory system, suggesting a heterogeneous pool of cells involved in regulating systemic metabolism. The molecular mechanisms underlying EVs biogenesis and release are still not fully understood, but intracellular calcium release and signaling have been implicated. The release of EVs may be accelerated in muscle due to the large flux in calcium during muscle contractions (Sabaratnam et al., 2022).

EXs, currently, are the most studied EVs, although terminological confusion exists. Unless the EV is captured in the act of release with live imaging techniques, assigning an EV to a particular biogenesis pathway remains difficult, as a consensus on EV subtype-specific markers has not yet emerged (Théry et al., 2018). Because of the challenges to distinguishing between different classes of EVs with current isolation techniques, the term exosome-like EVs (ELVs) appears to be an appropriate term of use that encompasses the smallest EVs (exosomes and small micro-vesicles) (Warnier et al., 2022). The studies’ main characteristics and findings are shown in Table 1.

TABLE 1
www.frontiersin.org

TABLE 1. Summary characteristics of reviewed studies.

3 Exercise and exosome-like extracellular vesicles

ELVs have been shown to function as signal transducers that are secreted into the blood following PA. These ELVs connect with target cells through surface interaction or membrane fusion, which could mediate the release of the ELVs contents into the target cells and initiate downstream signalling (Safdar and Tarnopolsky, 2018).

Exercise-induced ELVs may improve cognitive performance indirectly by transporting molecules that stimulate neurogenesis in the hippocampus. Irisin and cathepsin B (CTSB) are both myokines released from EVLs during exercise (Safdar and Tarnopolsky, 2018) that have been shown to improve memory and induce adult neurogenesis in the hippocampus via induction of brain-derived neurotrophic factor (BDNF) (Moon et al., 2016), proving to be neuroprotective in AD patients by reducing β-Amyloid Peptide (the main component of amyloid plaques) levels, although this remains controversial (Morris et al., 2014; Selkoe and Hardy, 2016; Chen et al., 2017).

Only recently, it has been recognized that physical exercise has substantial benefits on executive and cognitive functions, influenced by exercise parameters. Furthermore, even acute exercise plays a vital role in priming cellular pathways, leading to structural and functional adaptations observed in regular exercisers. Although both aerobic and resistance exercise, for instance, acutely increases BDNF values, aerobic exercise shows greater variation. The effects of physical exercise on circulating BDNF values found in subjects with NDs seem to be comparable to those exerted on the general population (Lippi et al., 2020).

Many studies have shown that different types of training can modulate ELVs in different ways, for example, a single bout of exhaustive aerobic exercise triggers the release of EVLs, which are cleared from the circulation during the early recovery period after cycling but remain elevated after running exercise (Frühbeis et al., 2015). A few molecules from the periphery can cross the blood-brain barrier (BBB) and interact with the brain, including ELVs that can migrate through the BBB via the transcellular pathway (Chen et al., 2016). Pharmacological studies, aimed at the treatment of brain diseases such as PD (Haney et al., 2015), have exploited this inherent ability of the ELVs to deliver drugs into the brain, or simply confirmed how these can actually cross the BBB (Chen et al., 2016) or carry a genetic cargo such as small-interfering RNA (siRNA), small RNA molecules capable of inhibiting the expression of a given gene to prevent the production of harmful proteins (Alvarez-Erviti et al., 2011). The ability of ELVs to transport myokines released during exercise and their ability to penetrate the BBB, allows us to speculate that ELVs may play an important role in muscle-brain crosstalk and, as a result, in NDs.

Different studies suggest that ELVs are released by skeletal muscle (SkM) cells and can be found in the bloodstream. Guescini et al. (2015) found that muscle tissue releases ELVs carrying miRNAs into the bloodstream in response to physical exercise, similar findings were observed by Rome et al. (2019) and Trovato et al. (2019). Increased plasma levels of myokines and proteins highly enriched in ELVs were observed in humans after an hour of cycle exercise (Whitham et al., 2018), and ELVs packed with hundreds of peptides were also discovered to be released from skeletal muscles following running exercise (Frühbeis et al., 2015). Estrada et al. (2022), using targeted immunocapture of tetraspanin-expressing particles (a method for isolating specific ELVs subpopulations based on the presence of tetraspanin proteins), found that SkM myofiber ELVs account for 4%–5% of circulating ELVs under free-living conditions, confirming that SkM-ELVs, while accounting for a smaller percentage of all ELVs produced, can reach circulation in vivo and their potential contribution to systemic physiological functions in distant cells and tissues. Overall, this evidence suggests that ELVs produced by SkM cells are released into the bloodstream and may play a role in muscle communication and homeostasis. Additionally, up to 75% of reported myokines were found to exist in ELVs (Safdar et al., 2016), highlighting the possibility that ELVs mediate exercise-induced myokine release.

However, a recent study (Watanabe et al., 2022), exploiting a proteomic approach used to identify potential tissue-specific ELVs marker proteins (i.e., ATP2A1, β-enolase, and desmin) that characterize ELVs derived from SkM, showed limited presence of SkM-ELVs in blood circulation, similar to Ismaeel et al. (2023), and they were hardly detected in the plasma, even after exercises, suggesting that SkM-ELVs do not appear to constitute a significant proportion of circulating ELVs, instead they were found to be highly accumulated within the SkM interstitium, indicating that they predominantly function within the tissue microenvironment. Furthermore, Watanabe et al. (2022) highlight that exercise-induced ELVs are more likely to be derived from leukocytes, platelets, and endothelial cells, while not ruling out the possibility that small but significant amounts of SkM-ELVs may enter the bloodstream and participate in inter-organ communication, as previously observed (Castano et al., 2022; Estrada et al., 2022).

These conflicting results may be due, at least in part, to the absence of specific SkM-ELVs markers defined for in vivo analysis, but they highlight the necessity of further research and, consequently, the current results should be viewed with caution.

4 How FITT-VP parameters modulate ELVs and microRNAs cargo

The potential benefits produced by exercise through EVLs are mainly due to their cargo. It is known that these vesicles can transport a variety of molecules including proteins, lipids, DNA, messenger RNAs (mRNA), siRNA and microRNA (miRNA); the latter has recently been the focus of different research (Karvinen et al., 2020; Doncheva et al., 2022; Garcia-Martin et al., 2022; Xu et al., 2022), showing how some miRNAs are taken into EVLs and released into the bloodstream without being degraded. Furthermore, these circulating miRNAs (c-miRNAs) can move to other cells and modulate their functions contributing to the pathogenesis of different diseases (Valadi et al., 2007).

miRNAs are small, single-stranded organized (∼19–22 nucleotide) sequences of RNAs that do not code for any protein but are functional in post-transcriptional regulation processes of coding genes and intercellular communication (Mohr and Mott, 2015). They primarily function by attaching to complementary mRNA sequences in recipient cells, interfering with the translational process, and stopping or changing the production of proteins.

Since miRNA species are known to play significant roles in a wide range of physiological and pathological processes and their dysregulation was observed in several NDs likewise ALS (Tasca et al., 2016; Pegoraro et al., 2017), AD, HD, and PD (Rajgor, 2018; Roser et al., 2018), they may also be engaged in the health benefits of exercise in terms of disease prevention; therefore, understanding the role of miRNAs carried by ELVs, produced during exercise, and their downstream targets would help us better understand how preventative lifestyle actually functions at the cellular level (Garai et al., 2021).

There is still a debate regarding the association of miRNAs with ELVs. Some studies suggest that miRNAs are predominantly associated with RNA-binding proteins rather than being contained within ELVs (Hoy and Buck, 2012). Whereas other studies indicate that miRNAs can be protected and released within ELVs (Wang et al., 2022). Moreover, the functional role of ELV-contained miRNAs in recipient cells is still uncertain. Some studies have questioned their functionality due to the low abundance of miRNAs in ELVs and the difficulty in evaluating their impact on recipient cells. An in vitro study (Albanese et al., 2021) found that, while ELVs showed interaction with various target cells, the fusion and transfer of ELV cargo into the cytoplasm of recipient cells were not detected, suggesting limited functional transfer of ELV-contained miRNAs. Conversely, other studies have reported the functional effects of miRNAs packed into the ELVs, suggesting that vesicular miRNAs can be internalized by a target cell regulating its gene expression (Rome et al., 2019; Castano et al., 2022).

However, altered levels of miRNAs are reported for numerous NDs and particularly type of miRNAs (miRs), defined as gatekeepers of both the nervous and immune system, seem capable of simultaneously modulating immune cell activation and neuronal function (Juźwik et al., 2019); interestingly miRNA expression changes after exercise (Guescini et al., 2015; D’Souza et al., 2018).

Guescini et al. (2015) reported that after acute aerobic exercise (40 min, vigorous intensity 80% of VO2max, treadmill exercise) muscle-specific miRNAs (miR-181a-5p, miR-133b, and miR-206), carried by ELVs, appear significantly increased or show an increasing trend. Differently, Aoi et al. (2013) observe that after acute and chronic aerobic exercise (a single bout of steady-state cycling exercise at 70% VO2max for 60 min cycling/exercise at 70% VO2max for 30 min, 3 times/week for 4 weeks) no significant changes in specific muscle miRNAs are detected in serum, except for miR-486 whose higher levels were observed in the early stage of HD (Packer et al., 2008; Karnati et al., 2015), which appears significantly decreased. After two consecutive bouts of mild muscle-damaging acute exercise (10 sets of 10 plyometric jumps at 90% of their maximum achievable jump height, with a 1 min interval between sets, 5 min rest followed by 5 sets of 4 min bouts of downhill running at a speed of 10 km/h, 10% decline, and 2 min standing interval between sets) the miR-31 cargo of circulating EVLs significantly decreased by 24 h post-performance of mild to moderate intensity (Lovett et al., 2018). Similarly, after a single bout of high-intensity interval exercise, was found that the increase in EVLs release returned to resting level after recovery, while the identification of 12 miRNAs as significantly altered in muscle and ELVs but not in plasma emphasizes the uniqueness of the signature of ELVs miRNAs as an independent component that may be an important mediator of cross-talk between tissues throughout the body, following exercise training (D’Souza et al., 2018).

Annibalini et al. (2019) report that after a single acute iso-inertial resistance exercise at the flywheel (5 sets of 10 maximal squats with 3 min rest between sets), there was an acute increase in the levels of circulating ELVs and the muscle-specific miRNAs (miR-206 and miR-146a) carried by them, that could be involved into the inflammatory response (Baggish et al., 2011). Some miRNAs, including miR-146a, are found to be involved in the regulation of inflammatory processes and are upregulated in NDs as AD (Lukiw et al., 2008), but the evidence is still conflicting (Ammal Kaidery et al., 2021). However, brain inflammation appears to have a dual function: to protect the brain from pathogens and neurotoxic agents during an acute phase response by promoting tissue repair (Yong et al., 2019), but to become detrimental when a chronic response sets in (Kinney et al., 2018).

In contrast, similarly to Lovett et al. (2018), another study (Estébanez et al., 2021) does not observe changes in plasma levels of ELVs and miRNAs (miR-146a), after 8 weeks of mild to moderate intensity resistance training (2 sessions/week, 3 sets/12-8-12 repetitions) in elderly subjects. Similarly, Just et al. (2020) after a single bout of low-intensity ischemic resistance exercise (BFRE) (5 sets of knee extension to volitional failure at 30% of 1RM) do not observe an increase in muscle-specific miRNAs packed into the ELVs and, likewise, Silver et al. (2020) did not observe significant modifications in the expression of miRNAs (miR-1, miR-16, miR-23b and miR-133a/b) carried by ELVs after a single acute bout of endurance exercise (cycling for 60 min at 70% of VO2peak).

Maggio et al. (2023) studied how miRNAs carried in ELVs are modulated following different protocols of aerobic exercises: acute aerobic activity (AAE) (single steady-state moderate-intensity workout on a treadmill) and aerobic training (AT) (moderate to vigorous intensity 40 min, 3 days/week for 8 weeks, 55% of their VO2max), maximal aerobic exercise (AMAE) (graded exercise test to exhaustion on a treadmill) and 2 weeks of high altitude aerobic training (AAT) (15 days of training camp at 2,000 m altitude, 23 sessions). After AAE they observe only an increasing trend in EVLs, but no significant upregulation of miRNAs carried by ELVs except for miR-146a; in contrast, these stimuli do not appear sufficient to generate a similar response, after AT. Following AMAE no change in the number of ELVs is detected, while there is an immediate post-exercise increase in miRNAs (miR-206, miR-133b, miR-486-5p, miR-181a-5p, miR-16) carried by ELVs that return to basal levels faster than at AAE, suggesting that it may be related to exercise intensity. Finally, no increase in the number of ELVs is observed following AAT, but rather a decrease, with no significant change in miRNA expression levels.

Few studies have investigated the effects of exercise on the modulation of ELVs and the miRNAs they contain and carry, over the long term. Garai et al. (2021) do so by evaluating the effects of a concomitant training protocol (60 min, 3 times/week of endurance and strength training) performed for 6 months, on the expression of specific ELV-transported miRNAs, in previously sedentary healthy young adults (YG) and comparing this profile with that present in healthy senior subjects who have been engaged in regular exercise (endurance and strength training) for at least 25 years (SG). They thus identified 12 similarly regulated miRNAs in both the YG and SG groups. They observe a reduction in some specific miRNAs (miR-23a, miR-451a, miR-223-3p, miR-150-5p, miR-15a, miR-142-3p) among which the expression of miR-142-3p was found to be significantly reduced compared to sedentary subjects.

While the expression levels of these miRNAs, whether carried by ELVs or present freely in the bloodstream, have been observed to be significantly higher in the serum of individuals affected by various NDs (Barbagallo et al., 2020; Da Silva et al., 2021), the current evidence does not conclusively establish a causal relationship between regular physical activity and the reduction of the risk of developing neurodegenerative diseases or the improvement of their symptoms through the modulation of ELVs and specific miRNAs carried by them. These, however, may provide a non-negligible biological plausibility in support of a possible role played by exercise and may allow hypotheses to be generated about which clusters of integrated pathways may be involved. Further studies are needed to move beyond speculative inferences and establish a link of causality.

5 Limitations and future directions

Although this area of research is promising, several limitations arise in structuring studies. The first of these is the difficulty of studying different EVs subpopulations; assigning an EV to a particular biogenesis pathway is incredibly difficult unless, for example, the EV is captured in the act of release with live imaging techniques. This is because a consensus on specific markers of EV subtypes, such as endosomal-derived “exosomes” and “ectosomes” (microparticles/microvesicles) derived from the plasma membrane, has not yet developed (Théry et al., 2018).

Furthermore, although it can be considered a marker of the onset and progression of some NDs, challenges and limitations are emerging in understanding the functional role of miRNAs contained within ELVs and their impact on recipient cells.

Despite growing evidence, there are still few studies about the effects exerted by exercise on ELVs and their cargo, even fewer that specifically assess FITT-VP parameters and have been conducted on subjects with NDs. Even in the latter case, the studies are limited by the absence of control of dosages and types of treatments and by only man samples (Da Silva et al., 2021). Furthermore, in some works, training parameters such as volume and progression are not always controlled and almost all studies use small samples.

Finally, it should be considered that many studies perform assessments immediately after a single exercise, focusing on the acute and transient aspects of exercise, however serum-level changes in the number and content of ELVs may be influenced by stress-related factors (Beninson and Fleshner, 2014; Hou et al., 2019), while only a few studies analyze long-term effects (Aoi et al., 2013; Estébanez et al., 2021; Garai et al., 2021; Da Silva et al., 2021; Maggio et al., 2023).

New studies should focus on larger samples of subjects affected or at risk of developing NDs, evaluating the long-term effects of exercise on modulation of ELVs and their cargo, with specific attention to the packed miRNAs in ELVs as several evidences support their ability to be assimilated by target tissue cells, contrary to free miRNAs, and regulate gene expression (Valadi et al., 2007; Rome et al., 2019; Xu et al., 2022; Liu and Wang, 2023). Special attention should also be paid to controlling the FITT-VP parameters of the intervention protocol, as these aspects could positively or negatively affect the outcomes.

6 Conclusion

Exercise has been associated with potential benefits in terms of neuroprotection and the prevention of NDs. The effects of exercise on ELVs and their cargo, specifically miRNAs, may be one of the mechanisms underlying these observed benefits. Different exercise parameters can lead to distinct changes in ELVs levels and miRNAs expression profiles; however, given the variety of exercise protocols, the optimal dose-response connection of exercise, and related FITT-VP parameters, remain uncertain.

It seems that acute aerobic exercise practised at vigorous intensity, can result in increased levels of ELVs and muscle-specific miRNAs (miR-181a-5p and miR-133b) carried by EVLs. This suggests that acute aerobic exercise may have an impact on miRNA expression in EVLs, differently from endurance training and chronic exercise. Acute resistance exercise at mild to moderate intensity, differently from low-intensity ischemic resistance exercise, can induce acute increases in ELVs levels and muscle-specific miRNAs (miR-206 and miR-146a) that may be involved in the inflammatory response, although the evidence for their role in inflammation and NDs is still conflicting. Long-term concomitant exercise training (endurance/strength) can result in the downregulation of specific ELV-transported miRNAs (miR-142-3p, miR-23a) potentially contributing to the reduction of the risk of developing NDs.

Overall, exercise parameters have been shown to influence the release of ELVs and the modulation of miRNAs carried by these vesicles. Further research is needed to fully understand the specific mechanisms and implications of exercise-induced ELVs release and miRNAs modulation in the NDs context, as well as their potential therapeutic applications.

Author contributions

FF, LP, and GG contributed to conception of the review. LP and SC organized the database. FF and LP wrote the first draft of the manuscript. MD, DP, GG, and SC wrote sections of the manuscript. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abeliovich, A., and Gitler, A. D. (2016). Defects in trafficking bridge Parkinson's disease pathology and genetics. Nature 539 (7628), 207–216. doi:10.1038/nature20414

PubMed Abstract | CrossRef Full Text | Google Scholar

Albanese, M., Chen, Y.-F. A., Hüls, C., Gärtner, K., Tagawa, T., Mejias-Perez, E., et al. (2021). MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells. PLoS Genet. 17 (12), e1009951. doi:10.1371/journal.pgen.1009951

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., and Wood, M. J. A. (2011). Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29, 341–345. doi:10.1038/nbt.1807

PubMed Abstract | CrossRef Full Text | Google Scholar

Ammal Kaidery, N., Ahuja, M., Sharma, S. M., and Thomas, B. (2021). An emerging role of miRNAs in neurodegenerative diseases: mechanisms and perspectives on miR146a. Antioxidants Redox Signal. 35 (7), 580–594. doi:10.1089/ars.2020.8256

CrossRef Full Text | Google Scholar

Annibalini, G., Contarelli, S., Lucertini, F., Guescini, M., Maggio, S., Ceccaroli, P., et al. (2019). Muscle and systemic molecular responses to a single flywheel based iso-inertial training session in resistance-trained men. Front. Physiology 10, 554. doi:10.3389/fphys.2019.00554

CrossRef Full Text | Google Scholar

Aoi, W., Ichikawa, H., Mune, K., Tanimura, Y., Mizushima, K., Naito, Y., et al. (2013). Muscle-enriched microRNA miR-486 decreases in circulation in response to exercise in young men. Front. Physiology 4, 80. doi:10.3389/fphys.2013.00080

PubMed Abstract | CrossRef Full Text | Google Scholar

Baggish, A. L., Hale, A., Weiner, R. B., Lewis, G. D., Systrom, D., Wang, F., et al. (2011). Dynamic regulation of circulating microRNA during acute exhaustive exercise and sustained aerobic exercise training. J. Physiology 589 (16), 3983–3994. doi:10.1113/jphysiol.2011.213363

PubMed Abstract | CrossRef Full Text | Google Scholar

Barbagallo, C., Mostile, G., Baglieri, G., Giunta, F., Luca, A., Raciti, L., et al. (2020). Specific signatures of serum miRNAs as potential biomarkers to discriminate clinically similar neurodegenerative and vascular-related diseases. Cell. Mol. Neurobiol. 40 (4), 531–546. doi:10.1007/s10571-019-00751-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Beninson, L. A., and Fleshner, M. (2014). Exosomes: an emerging factor in stress-induced immunomodulation. Seminars Immunol. 26 (5), 394–401. doi:10.1016/j.smim.2013.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Canter, R. G., Penney, J., and Tsai, L. H. (2016). The road to restoring neural circuits for the treatment of Alzheimer's disease. Nature 539 (7628), 187–196. doi:10.1038/nature20412

PubMed Abstract | CrossRef Full Text | Google Scholar

Castaño, C., Meza-Ramos, A., Batlle, M., Guasch, E., Novials, A., and Párrizas, M. (2022). Treatment with EV-miRNAs alleviates obesity-associated metabolic dysfunction in mice. Int. J. Mol. Sci. 23 (23), 14920. doi:10.3390/ijms232314920

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, C. C., Liu, L., Ma, F., Wong, C. W., Guo, X. E., Chacko, J. V., et al. (2016). Elucidation of exosome migration across the blood-brain barrier model in vitro. Cell. Mol. Bioeng. 9 (4), 509–529. doi:10.1007/s12195-016-0458-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, G. F., Xu, T. H., Yan, Y., Zhou, Y. R., Jiang, Y., Melcher, K., et al. (2017). Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 38 (9), 1205–1235. doi:10.1038/aps.2017.28

PubMed Abstract | CrossRef Full Text | Google Scholar

Coelho, F. G., Vital, T. M., Stein, A. M., Arantes, F. J., Rueda, A. V., Camarini, R., et al. (2014). Acute aerobic exercise increases brain-derived neurotrophic factor levels in elderly with Alzheimer's disease. J. Alzheimer's Dis. JAD 39 (2), 401–408. doi:10.3233/JAD-131073

PubMed Abstract | CrossRef Full Text | Google Scholar

Da Silva, F. C., Rode, M. P., Vietta, G. G., Iop, R. D. R., Creczynski-Pasa, T. B., Martin, A. S., et al. (2021). Expression levels of specific microRNAs are increased after exercise and are associated with cognitive improvement in Parkinson's disease. Mol. Med. Rep. 24 (2), 618. doi:10.3892/mmr.2021.12257

PubMed Abstract | CrossRef Full Text | Google Scholar

D'Anca, M., Fenoglio, C., Buccellato, F. R., Visconte, C., Galimberti, D., and Scarpini, E. (2021). Extracellular vesicles in multiple sclerosis: role in the pathogenesis and potential usefulness as biomarkers and therapeutic tools. Cells 10 (7), 1733. doi:10.3390/cells10071733

PubMed Abstract | CrossRef Full Text | Google Scholar

Doncheva, A. I., Romero, S., Ramirez-Garrastacho, M., Lee, S., Kolnes, K. J., Tangen, D. S., et al. (2022). Extracellular vesicles and microRNAs are altered in response to exercise, insulin sensitivity and overweight. Acta Physiol. Oxf. Engl. 236 (4), e13862. doi:10.1111/apha.13862

CrossRef Full Text | Google Scholar

D'Souza, R. F., Woodhead, J. S. T., Zeng, N., Blenkiron, C., Merry, T. L., Cameron-Smith, D., et al. (2018). Circulatory exosomal miRNA following intense exercise is unrelated to muscle and plasma miRNA abundances. Am. J. Physiology. Endocrinol. Metabolism 315 (4), E723–E733. doi:10.1152/ajpendo.00138.2018

CrossRef Full Text | Google Scholar

Dugger, B. N., and Dickson, D. W. (2017). Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 9 (7), a028035. doi:10.1101/cshperspect.a028035

PubMed Abstract | CrossRef Full Text | Google Scholar

Erickson, K. I., Voss, M. W., Prakash, R. S., Basak, C., Szabo, A., Chaddock, L., et al. (2011). Exercise training increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci. U. S. A. 108 (7), 3017–3022. doi:10.1073/pnas.1015950108

PubMed Abstract | CrossRef Full Text | Google Scholar

Estébanez, B., Visavadiya, N. P., de Paz, J. A., Whitehurst, M., Cuevas, M. J., González-Gallego, J., et al. (2021). Resistance training diminishes the expression of exosome CD63 protein without modification of plasma miR-146a-5p and cfDNA in the elderly. Nutrients 13 (2), 665. doi:10.3390/nu13020665

PubMed Abstract | CrossRef Full Text | Google Scholar

Estrada, A. L., Valenti, Z. J., Hehn, G., Amorese, A. J., Williams, N. S., Balestrieri, N. P., et al. (2022). Extracellular vesicle secretion is tissue-dependent ex vivo and skeletal muscle myofiber extracellular vesicles reach the circulation in vivo. Am. J. Physiology. Cell. Physiology 322 (2), C246–C259. doi:10.1152/ajpcell.00580.2020

CrossRef Full Text | Google Scholar

Feter, N., Spanevello, R. M., Soares, M. S. P., Spohr, L., Pedra, N. S., Bona, N. P., et al. (2019). How does physical activity and different models of exercise training affect oxidative parameters and memory? Physiology Behav. 201, 42–52. doi:10.1016/j.physbeh.2018.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Filippi, M., Bar-Or, A., Piehl, F., Preziosa, P., Solari, A., Vukusic, S., et al. (2018). Multiple sclerosis. Nat. Rev. Dis. Prim. 4 (1), 43. doi:10.1038/s41572-018-0041-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Frederiksen, K. S., Gjerum, L., Waldemar, G., and Hasselbalch, S. G. (2018). Effects of physical exercise on Alzheimer’s disease biomarkers: a systematic review of intervention studies. J. Alzheimers Dis. 61 (1), 359–372. doi:10.3233/JAD-170567

PubMed Abstract | CrossRef Full Text | Google Scholar

Frühbeis, C., Helmig, S., Tug, S., Simon, P., and Krämer-Albers, E. M. (2015). Physical exercise induces rapid release of small extracellular vesicles into the circulation. J. Extracell. Vesicles 4, 28239. doi:10.3402/jev.v4.28239

PubMed Abstract | CrossRef Full Text | Google Scholar

Garai, K., Adam, Z., Herczeg, R., Banfai, K., Gyebrovszki, A., Gyenesei, A., et al. (2021). Physical activity as a preventive lifestyle intervention acts through specific exosomal miRNA species-evidence from human short- and long-term pilot studies. Front. Physiology 12, 658218. doi:10.3389/fphys.2021.658218

CrossRef Full Text | Google Scholar

Garcia-Martin, R., Wang, G., Brandão, B. B., Zanotto, T. M., Shah, S., Kumar Patel, S., et al. (2022). MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature 601 (7893), 446–451. doi:10.1038/s41586-021-04234-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Guescini, M., Canonico, B., Lucertini, F., Maggio, S., Annibalini, G., Barbieri, E., et al. (2015). Muscle releases alpha-sarcoglycan positive extracellular vesicles carrying miRNAs in the bloodstream. PloS One 10 (5), e0125094. doi:10.1371/journal.pone.0125094

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamer, M., and Chida, Y. (2009). Physical activity and risk of neurodegenerative disease: a systematic review of prospective evidence. Psychol. Med. 39 (1), 3–11. doi:10.1017/S0033291708003681

PubMed Abstract | CrossRef Full Text | Google Scholar

Haney, M. J., Klyachko, N. L., Zhao, Y., Gupta, R., Plotnikova, E. G., He, Z., et al. (2015). Exosomes as drug delivery vehicles for Parkinson's disease therapy. J. Control. Release Official J. Control. Release Soc. 207, 18–30. doi:10.1016/j.jconrel.2015.03.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Heemels, M. T. (2016). Neurodegenerative diseases. Nature 539, 179. doi:10.1038/539179a

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, Z., Qin, X., Hu, Y., Zhang, X., Li, G., Wu, J., et al. (2019). Longterm exercise-derived exosomal miR-342-5p: A novel exerkine for cardioprotection. Circulation Res. 124 (9), 1386–1400. doi:10.1161/CIRCRESAHA.118.314635

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoy, A. M., and Buck, A. H. (2012). Extracellular small RNAs: what, where, why? Biochem. Soc. Trans. 40 (4), 886–890. doi:10.1042/BST20120019

PubMed Abstract | CrossRef Full Text | Google Scholar

Ionescu-Tucker, A., and Cotman, C. W. (2021). Emerging roles of oxidative stress in brain aging and Alzheimer's disease. Neurobiol. Aging 107, 86–95. doi:10.1016/j.neurobiolaging.2021.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Ismaeel, A., Van Pelt, D. W., Hettinger, Z. R., Fu, X., Richards, C. I., Butterfield, T. A., et al. (2023). Extracellular vesicle distribution and localization in skeletal muscle at rest and following disuse atrophy. Skelet. Muscle 13 (1), 6. doi:10.1186/s13395-023-00315-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Just, J., Yan, Y., Farup, J., Sieljacks, P., Sloth, M., Venø, M., et al. (2020). Blood flow-restricted resistance exercise alters the surface profile, miRNA cargo and functional impact of circulating extracellular vesicles. Sci. Rep. 10 (1), 5835. doi:10.1038/s41598-020-62456-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Juźwik, C. A., S Drake, S., Zhang, Y., Paradis-Isler, N., Sylvester, A., Amar-Zifkin, A., et al. (2019). microRNA dysregulation in neurodegenerative diseases: A systematic review. Prog. Neurobiol. 182, 101664. doi:10.1016/j.pneurobio.2019.101664

PubMed Abstract | CrossRef Full Text | Google Scholar

Karnati, H. K., Panigrahi, M. K., Gutti, R. K., Greig, N. H., and Tamargo, I. A. (2015). miRNAs: key players in neurodegenerative disorders and epilepsy. J. Alzheimer's Dis. JAD 48 (3), 563–580. doi:10.3233/JAD-150395

CrossRef Full Text | Google Scholar

Karvinen, S., Sievänen, T., Karppinen, J. E., Hautasaari, P., Bart, G., Samoylenko, A., et al. (2020). MicroRNAs in extracellular vesicles in sweat change in response to endurance exercise. Front. Physiology 11, 676. doi:10.3389/fphys.2020.00676

CrossRef Full Text | Google Scholar

Kinney, J. W., Bemiller, S. M., Murtishaw, A. S., Leisgang, A. M., Salazar, A. M., and Lamb, B. T. (2018). Inflammation as a central mechanism in Alzheimer's disease. Alzheimer's dementia (New York, N. Y.) 4, 575–590. doi:10.1016/j.trci.2018.06.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Lange-Asschenfeldt, C., and Kojda, G. (2008). Alzheimer's disease, cerebrovascular dysfunction and the benefits of exercise: from vessels to neurons. Exp. Gerontol. 43 (6), 499–504. doi:10.1016/j.exger.2008.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Lippi, G., Mattiuzzi, C., and Sanchis-Gomar, F. (2020). Updated overview on interplay between physical exercise, neurotrophins, and cognitive function in humans. J. Sport Health Sci. 9 (1), 74–81. doi:10.1016/j.jshs.2019.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y. J., and Wang, C. (2023). A review of the regulatory mechanisms of extracellular vesicles-mediated intercellular communication. Cell. Commun. Signal. CCS 21 (1), 77. doi:10.1186/s12964-023-01103-6

CrossRef Full Text | Google Scholar

Lovett, J. A. C., Durcan, P. J., and Myburgh, K. H. (2018). Investigation of circulating extracellular vesicle MicroRNA following two consecutive bouts of muscle-damaging exercise. Front. Physiology 9, 1149. doi:10.3389/fphys.2018.01149

PubMed Abstract | CrossRef Full Text | Google Scholar

Lukiw, W. J., Zhao, Y., and Cui, J. G. (2008). An NF-kappaB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J. Biol. Chem. 283 (46), 31315–31322. doi:10.1074/jbc.M805371200

PubMed Abstract | CrossRef Full Text | Google Scholar

Maass, A., Düzel, S., Brigadski, T., Goerke, M., Becke, A., Sobieray, U., et al. (2016). Relationships of peripheral IGF-1, VEGF and BDNF levels to exercise-related changes in memory, hippocampal perfusion and volumes in older adults. NeuroImage 131, 142–154. doi:10.1016/j.neuroimage.2015.10.084

PubMed Abstract | CrossRef Full Text | Google Scholar

Maggio, S., Canonico, B., Ceccaroli, P., Polidori, E., Cioccoloni, A., Giacomelli, L., et al. (2023). Modulation of the circulating extracellular vesicles in response to different exercise regimens and study of their inflammatory effects. Int. J. Mol. Sci. 24 (3), 3039. doi:10.3390/ijms24033039

PubMed Abstract | CrossRef Full Text | Google Scholar

Magliulo, L., Bondi, D., Pini, N., Marramiero, L., and Di Filippo, E. S. (2022). The wonder exerkines-novel insights: a critical state-of-the-art review. Mol. Cell. Biochem. 477 (1), 105–113. doi:10.1007/s11010-021-04264-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Mohr, A. M., and Mott, J. L. (2015). Overview of microRNA biology. Seminars Liver Dis. 35 (1), 3–11. doi:10.1055/s-0034-1397344

PubMed Abstract | CrossRef Full Text | Google Scholar

Moon, H. Y., Becke, A., Berron, D., Becker, B., Sah, N., Benoni, G., et al. (2016). Running-induced systemic cathepsin B secretion is associated with memory function. Cell. Metab. 24 (2), 332–340. doi:10.1016/j.cmet.2016.05.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Morris, G. P., Clark, I. A., and Vissel, B. (2014). Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer's disease. Acta Neuropathol. Commun. 2, 135. doi:10.1186/s40478-014-0135-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Packer, A. N., Xing, Y., Harper, S. Q., Jones, L., and Davidson, B. L. (2008). The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington's disease. J. Neurosci. Official J. Soc. Neurosci. 28 (53), 14341–14346. doi:10.1523/JNEUROSCI.2390-08.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Pedersen, B. K., and Saltin, B. (2015). Exercise as medicine - evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand. J. Med. Sci. Sports 25 (3), 1–72. doi:10.1111/sms.12581

PubMed Abstract | CrossRef Full Text | Google Scholar

Pegoraro, V., Merico, A., and Angelini, C. (2017). Micro-RNAs in ALS muscle: differences in gender, age at onset and disease duration. J. Neurological Sci. 380, 58–63. doi:10.1016/j.jns.2017.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajgor, D. (2018). Macro roles for microRNAs in neurodegenerative diseases. Non-coding RNA Res. 3 (3), 154–159. doi:10.1016/j.ncrna.2018.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Rigamonti, A. E., Bollati, V., Pergoli, L., Iodice, S., De Col, A., Tamini, S., et al. (2020). Effects of an acute bout of exercise on circulating extracellular vesicles: tissue-sex-and BMI-related differences. Int. J. Obes. 44 (5), 1108–1118. (2005). doi:10.1038/s41366-019-0460-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Rody, T., De Amorim, J. A., and De Felice, F. G. (2022). The emerging neuroprotective roles of exerkines in Alzheimer's disease. Front. Aging Neurosci. 14, 965190. doi:10.3389/fnagi.2022.965190

PubMed Abstract | CrossRef Full Text | Google Scholar

Rome, S., Forterre, A., Mizgier, M. L., and Bouzakri, K. (2019). Skeletal muscle-released extracellular vesicles: state of the art. Front. Physiology 10, 929. doi:10.3389/fphys.2019.00929

PubMed Abstract | CrossRef Full Text | Google Scholar

Roser, A. E., Caldi Gomes, L., Schünemann, J., Maass, F., and Lingor, P. (2018). Circulating miRNAs as diagnostic biomarkers for Parkinson's disease. Front. Neurosci. 12, 625. doi:10.3389/fnins.2018.00625

PubMed Abstract | CrossRef Full Text | Google Scholar

Sabaratnam, R., Wojtaszewski, J. F. P., and Højlund, K. (2022). Factors mediating exercise-induced organ crosstalk. Acta Physiol. Oxf. Engl. 234 (2), e13766. doi:10.1111/apha.13766

CrossRef Full Text | Google Scholar

Safdar, A., and Tarnopolsky, M. A. (2018). Exosomes as mediators of the systemic adaptations to endurance exercise. Cold Spring Harb. Perspect. Med. 8 (3), a029827. doi:10.1101/cshperspect.a029827

PubMed Abstract | CrossRef Full Text | Google Scholar

Safdar, A., Saleem, A., and Tarnopolsky, M. A. (2016). The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat. Rev. Endocrinol. 12 (9), 504–517. doi:10.1038/nrendo.2016.76

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanford, J. A., Nogiec, C. D., Lindholm, M. E., Adkins, J. N., Amar, D., Dasari, S., et al. (2020). molecular transducers of physical activity consortium (MoTrPAC): mapping the dynamic responses to exercise. Cell. 181 (7), 1464–1474. doi:10.1016/j.cell.2020.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Selkoe, D. J., and Hardy, J. (2016). The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol. Med. 8 (6), 595–608. doi:10.15252/emmm.201606210

PubMed Abstract | CrossRef Full Text | Google Scholar

Silver, J. L., Alexander, S. E., Dillon, H. T., Lamon, S., and Wadley, G. D. (2020). Extracellular vesicular miRNA expression is not a proxy for skeletal muscle miRNA expression in males and females following acute, moderate intensity exercise. Physiol. Rep. 8 (16), e14520. doi:10.14814/phy2.14520

PubMed Abstract | CrossRef Full Text | Google Scholar

Szatanek, R., Baj-Krzyworzeka, M., Zimoch, J., Lekka, M., Siedlar, M., and Baran, J. (2017). The methods of choice for extracellular vesicles (EVs) characterization. Int. J. Mol. Sci. 18 (6), 1153. doi:10.3390/ijms18061153

PubMed Abstract | CrossRef Full Text | Google Scholar

Tasca, E., Pegoraro, V., Merico, A., and Angelini, C. (2016). Circulating microRNAs as biomarkers of muscle differentiation and atrophy in ALS. Clin. Neuropathol. 35 (1), 22–30. doi:10.5414/NP300889

PubMed Abstract | CrossRef Full Text | Google Scholar

Taylor, J. P., Brown, R. H., and Cleveland, D. W. (2016). Decoding ALS: from genes to mechanism. Nature 539 (7628), 197–206. doi:10.1038/nature20413

PubMed Abstract | CrossRef Full Text | Google Scholar

Théry, C., Witwer, K. W., Aikawa, E., Alcaraz, M. J., Anderson, J. D., Andriantsitohaina, R., et al. (2018). Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 7 (1), 1535750. doi:10.1080/20013078.2018.1535750

PubMed Abstract | CrossRef Full Text | Google Scholar

Trotta, T., Panaro, M. A., Cianciulli, A., Mori, G., Di Benedetto, A., and Porro, C. (2018). Microglia-derived extracellular vesicles in Alzheimer's disease: a double-edged sword. Biochem. Pharmacol. 148, 184–192. doi:10.1016/j.bcp.2017.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Trovato, E., Di Felice, V., and Barone, R. (2019). Extracellular vesicles: delivery vehicles of myokines. Front. Physiology 10, 522. doi:10.3389/fphys.2019.00522

PubMed Abstract | CrossRef Full Text | Google Scholar

Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., and Lötvall, J. O. (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell. Biol. 9 (6), 654–659. doi:10.1038/ncb1596

PubMed Abstract | CrossRef Full Text | Google Scholar

van Niel, G., D'Angelo, G., and Raposo, G. (2018). Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell. Biol. 19 (4), 213–228. doi:10.1038/nrm.2017.125

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, W., Hao, L. P., Song, H., Chu, X. Y., and Wang, R. (2022). The potential roles of exosomal non-coding RNAs in hepatocellular carcinoma. Front. Oncol. 12, 790916. doi:10.3389/fonc.2022.790916

PubMed Abstract | CrossRef Full Text | Google Scholar

Warnier, G., De Groote, E., Britto, F. A., Delcorte, O., Nederveen, J. P., Nilsson, M. I., et al. (2022). Effects of an acute exercise bout in hypoxia on extracellular vesicle release in healthy and prediabetic subjects. Am. J. Physiology. Regul. Integr. Comp. Physiology 322 (2), R112–R122. doi:10.1152/ajpregu.00220.2021

CrossRef Full Text | Google Scholar

Watanabe, S., Sudo, Y., Makino, T., Kimura, S., Tomita, K., Noguchi, M., et al. (2022). Skeletal muscle releases extracellular vesicles with distinct protein and microRNA signatures that function in the muscle microenvironment. PNAS Nexus 1 (4), pgac173. doi:10.1093/pnasnexus/pgac173

PubMed Abstract | CrossRef Full Text | Google Scholar

Whitham, M., and Febbraio, M. A. (2019). Redefining tissue crosstalk via shotgun proteomic analyses of plasma extracellular vesicles. Proteomics 19 (1-2), e1800154. doi:10.1002/pmic.201800154

PubMed Abstract | CrossRef Full Text | Google Scholar

Whitham, M., Parker, B. L., Friedrichsen, M., Hingst, J. R., Hjorth, M., Hughes, W. E., et al. (2018). Extracellular vesicles provide a means for tissue crosstalk during exercise. Cell. Metab. 27 (1), 237–251.e4. doi:10.1016/j.cmet.2017.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Wyss-Coray, T. (2016). Ageing, neurodegeneration and brain rejuvenation. Nature 539 (7628), 180–186. doi:10.1038/nature20411

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, D., Di, K., Fan, B., Wu, J., Gu, X., Sun, Y., et al. (2022). MicroRNAs in extracellular vesicles: sorting mechanisms, diagnostic value, isolation, and detection technology. Front. Bioeng. Biotechnol. 10, 948959. doi:10.3389/fbioe.2022.948959

PubMed Abstract | CrossRef Full Text | Google Scholar

Yong, H. Y. F., Rawji, K. S., Ghorbani, S., Xue, M., and Yong, V. W. (2019). The benefits of neuroinflammation for the repair of the injured central nervous system. Cell. Mol. Immunol. 16 (6), 540–546. doi:10.1038/s41423-019-0223-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, Q., Li, X. D., Zhang, S. M., Wang, H. W., and Wang, Y. L. (2019). Extracellular vesicles in neurodegenerative diseases: insights and new perspectives. Genes. & Dis. 8 (2), 124–132. doi:10.1016/j.gendis.2019.12.001

CrossRef Full Text | Google Scholar

Keywords: physical activity, sport, exosome, microvesicles, Parkinson, Alzheimer, ALS, neuroprotection

Citation: Fischetti F, Poli L, De Tommaso M, Paolicelli D, Greco G and Cataldi S (2023) The role of exercise parameters on small extracellular vesicles and microRNAs cargo in preventing neurodegenerative diseases. Front. Physiol. 14:1241010. doi: 10.3389/fphys.2023.1241010

Received: 15 June 2023; Accepted: 04 August 2023;
Published: 15 August 2023.

Edited by:

José Antonio de Paz, University of León, Spain

Reviewed by:

Danilo Bondi, University of Studies G.d’Annunzio Chieti and Pescara, Italy
Jinju Wang, Marshall University, United States

Copyright © 2023 Fischetti, Poli, De Tommaso, Paolicelli, Greco and Cataldi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Gianpiero Greco, gianpiero.greco@uniba.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.