Open Access

Extracellular vesicle microRNAs mediate skeletal muscle myogenesis and disease (Review)

  • Authors:
    • Haidong Wang
    • Bin Wang
  • View Affiliations

  • Published online on: July 27, 2016     https://doi.org/10.3892/br.2016.725
  • Pages: 296-300
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Skeletal muscle function is important for good health and independent living, and has been subject to numerous studies focused on skeletal muscle development, function and metabolism. However, progressive and degenerative changes in skeletal muscle function often occur following physiological and pathological stress, and these lead to the progression of diabetes, obesity, chronic kidney disease, and cardiovascular or respiratory diseases. Identifying the mechanisms that influence the processes regulating skeletal muscle function is a key priority. Recently, studies have demonstrated that microRNAs (miRNAs) play important roles in regulating biological processes. For instance, exosomes are key tools for communication between cells. Therefore, by determining how select miRNAs are transported to target organs and initiate their effects, these results will help explain muscle and organ crosstalk, improve our understanding and application of current therapeutic approaches and lead to the identification of new therapeutic strategies and targets aimed at maintaining and/or improving skeletal muscle health.

Introduction

Skeletal muscle is a major organ for all animals, including humans, and it comprises ~40% of the body's mass. Control of movement and posture are its primary functions, and the development and function of skeletal muscle is regulated by different factors. Recently, microRNA (miRNA) studies have provided an opportunity for improved understanding of the molecular processes of skeletal muscle diseases. Research suggests that miRNAs play important roles in skeletal muscle development, and several miRNAs have been identified as biomarkers for myogenesis, muscle mass, and nutrient metabolism in physiological and pathological states (1,2). Several miRNAs are specifically expressed in muscle (myomiRs). Myocyte proliferation and differentiation are influenced by miRNAs, and miRNAs may affect muscle fiber types by regulating several transcriptional repressors.

In addition, miRNAs have recently been identified in extracellular body fluids, such as serum, plasma, urine, milk, and spinal fluid (35). These circulating miRNAs (ci-miRs) are embedded in microvesicles (MVs) or exosomes, which transport proteins, lipids, mRNAs and miRNAs to regulate recipient cell functions (6). Multiple cell types have been demonstrated to release vesicles into the extracellular medium, including mesenchymal cells, adipocytes, fibroblasts, immune cells, platelets, myoblasts and tumor cells (716). Evidence suggests that exosomes carrying specific miRNAs, such as miR-1, miR-21, miR-133, miR-182, and miR-206, are targeted to myocytes and modulate the physiology and pathology status of myocytes by altering gene expression (5,17). To date, there is limited knowledge regarding miRNAs and exosome biology, therefore, further studies are required to clarify the molecular mechanisms and precise involvement of miRNAs in muscle development and regeneration.

miRNAs and microvesicles

miRNAs are small (~20–30 nucleotides in length), non-coding RNAs that are highly conserved between plants and mammals. miRNAs downregulate gene expression post-transcriptionally and fine-tune target genes in the organs of all animals, including humans. Organ-specific miRNAs may be important in controlling their development, function and disease. Furthermore, a single miRNA targets the expression of multiple genes, whereas one gene is regulated by multiple miRNAs (17,18). It has been predicted that miRNAs regulate ~60% of the protein-coding genes that could be involved in a wide range of biological processes (19). The miRNAs miR-1 and miR-133a are expressed in cardiac and skeletal muscle, whereas miR-206/miR-133b is expressed only in skeletal muscle (20,21).

Several biological processes, including muscle growth and differentiation, are mediated by a collection of specific miRNAs. Numerous miRNAs can be released from cells in the surrounding areas or into circulation, and appear resistant to harsh conditions, such as RNA enzyme degradation (22,23). Researchers hypothesize that miRNAs are also involved in cell-to-cell communication for the epigenetic regulation of recipient cells. miRNAs shuttled between cells appear to be preserved and mediated by extracellular vesicles (exosomes and MVs), which are emerging as potent genetic transfer agents (24,25).

Cells secrete extracellular vesicles (EVs), MVs and exosomes, which are small, membrane-derived particles, usually 30–1,000 nm in diameter (26). Exosomes are defined as nanosized membrane vesicles with a diameter of 30–100 nm, originating from multivesicular endosomes that fuse with the plasma membrane and are released by cells into the extracellular environment. They differ from microvesicles, which have a diameter of 100–1,000 nm and originate from the plasma membrane (2729).

While the mechanisms of extracellular formation and secretion are not well-defined, evidence indicates that such vesicles possess the capability of ‘communicating’ with neighboring or distant cells by fusing with the plasma membrane and subsequently delivering their cargo, which consists of various molecules including proteins, mRNAs, and miRNAs (30,31). Moreover, transported miRNAs are capable of targeting mRNAs in recipient cells (30,32).

Exosomes are released from the plasma membrane and can be identified by specific markers, such as Hsp-60/70 in the lumen and CD9, CD63, CD81 and tissue-specific membrane proteins on the cell surface (22,33,34).

The data presented in the study by Yang et al (35) suggests that NF-κB may regulate exosomal protein expression at a remote site via circulation following ischemia-reperfusion injuries. Myoblasts and myotubes utilize exosome clustered miRNAs as endocrine signals to regulate important signaling pathways (e.g., the Wnt signaling pathway) for muscle homeostasis and regeneration. Furthermore, muscle behavior is influenced by the release of vesicles from multiple sources, such as mesenchymal stem cells. Recent evidence has demonstrated that miRNAs (e.g., miR-494 and myomiRs) released in exosomes from mesenchymal stem cells can promote muscle regeneration following injury by enhancing myogenesis and angiogenesis (24,36). Furthermore, stress may promote the release of exosomes that are carrying a variety of cargoes, inducing transfer to other cells in the local environment or farther away through systemic circulation (37).

miRNAs, microvesicles and muscle growth

Several miRNAs are highly enriched in skeletal muscle, and these can influence myocyte proliferation and differentiation. The importance of miRNAs in muscle development has been established in a previous study involving conditional transgenic mice lacking Dicer in myogenic progenitors. The study resulted in aberrant muscle differentiation accompanied by hyperplasia (38). Furthermore, miR-206 has been identified as the most abundant miRNA in adult vertebrate skeletal muscle and is known to promote skeletal muscle development and differentiation (22,39).

In fact, myotube-derived exosomes promoted the differentiation of target myoblasts by downregulating Cyclin-D1 and Myogenin (40). However, it was unclear whether miR-186, −329 and −362 were involved, since they were predicted binders for the 3′-UTRs of both genes. Intriguingly, using the same myoblast model, atrophic myotubes presented decreased intracellular levels, while showing increased exosomal fractions of miR-23a (41) and miR-182 (42). This result indicated that the exosome load was selectively choosing miRNAs under stressful conditions (43).

The upregulation of miR-1, miR-133, and miR-206 levels during myoblast differentiation is known to protect myocytes against atrophy (44). Interestingly, a mutation in the myostatin gene that causes a dramatic muscle increase in textile sheep, creates a target site for miR-206 and miR-1. In these sheep, myostatin downregulation determines the phenocopy of the double muscling Belgian Blue cattle (22).

Muroya et al (45) investigated the effects of grazing on the expression of miRNAs in cattle plasma with the hypothesis that the plasma miRNA profile reflects the physiological adaptation of different tissue types, such as skeletal muscle and adipose tissue. The miR-451 levels were elevated in the grazing cattle in comparison with the housed cattle. Synchronous miR-451 expression was also observed in the skeletal muscle, which may result in the secretion or intake of miRNAs between circulation and tissue cells in grazing cattle. Nakamura et al (36) investigated the role of mesenchymal stem cell (MSC) exosomes in skeletal muscle regeneration. MSC exosomes promoted myogenesis and angiogenesis in vitro, and muscle regeneration in an in vivo model of muscle injury. Although MSC exosomes had low concentrations of muscle-repair-related cytokines, several repair-related miRNAs were identified. The results of the study by Nakamura et al (36) suggest that the MSC-derived exosomes promoted muscle regeneration by enhancing myogenesis and angiogenesis, which is partially mediated by miRNAs, such as miR-494.

miRNAs, microvesicles and muscle wasting

Numerous previous studies have indicated that miRNA expression is involved in skeletal muscle diseases. In muscular dystrophy, inflammatory, myopathies, and congenital myopathies rhabdomyosarcomas (muscle tumor), individual miRNAs have been shown to cause or alleviate disease. It is known that ci-miRs are traceable in plasma or serum and appear resistant to harsh conditions, such as RNase activity. Among the vesicle-based carriers, exosomes are emerging as important regulators of long-range miRNA shuttling (43,46).

Many miRNAs may potentially be used as biomarkers, since they circulate in the blood, are often tissue-specific and resistant to degradation due to circulation in protective exosomes (4749). Several miRNAs, including miR-1, −133, −206, and −499, involved in muscle homeostasis and metabolism were demonstrated to be associated with muscle wasting (5052). Furthermore, miR-30b and −181a are involved in the regulation of muscle regeneration and inflammation (50,53). Those muscle-specific miRNAs may be useful biomarkers for the early development of acute muscle wasting in critically ill patients (49).

A previous study demonstrated that tumor-derived microvesicles induced apoptosis in skeletal muscle cells. This proapoptotic activity was mediated by miRNA cargo, miR-21, which signals through the toll-like 7 receptor (TLR7) on murine myoblasts to promote cell death (5,54,55). Xu et al (56) verified that miRNA-486 decreases FoxO1 protein expression and promotes FoxO1 phosphorylation to suppress E3 ubiquitin ligases, and thus presents an excellent candidate for future studies on the mechanisms of regulation of muscle atrophy by miRNAs in cachexia. In addition, miR-206 and miR-21 were recently described as being important in muscle wasting during catabolic conditions (57).

miR-29a, a key regulator of tissue fibrosis, is highly expressed in the exosomes and marginal area of a remote ischemic conditioning (RIC) group. Even in the differentiated C2C12-derived exosomes, miR-29a expression is significantly increased under hypoxic conditions (58). Hu et al (59) confirmed that age-induced muscle senescence resulted from the activation of miR-29 by wnt-3a, which led to suppressed expression of the signaling proteins p85α, IGF-1 and B-myb, which coordinate to impair the proliferation of the MPCs and contribute to muscle atrophy.

In chronic kidney disease (CKD), muscle atrophy is a serious complication as it is associated with increased morbidity and mortality. Hu et al (60) confirmed that CKD suppresses miR-29 in the muscle, which leads to higher expression of the transcription factor YY-1, thereby suppressing myogenesis. However, further studies are required to identify whether miR-29 is transported by exosomes and microvesicles to affect muscle atrophy.

miRNAs, microvesicles and exercise

Exercise stimulates numerous structural, metabolic, and morphological adaptations in skeletal muscle. These adaptations are vital in order to maintain human health over a life span, and miRNAs constitute a new regulatory component that may be important in these adaptations (61). Since miRNAs are incorporated into exosomes, microvesicles, or protein complexes, they can be detected in human plasma (62). miRNAs in skeletal muscle are modified after physical exercise, especially after acute exercise. miR-1, −133a, and −206 are potential novel biomarkers for aerobic exercise capacity since they are highly correlated to standard performance parameters (62). Baggish et al (63) demonstrated altered expression of specific ci-miRs in response to both acute and chronic exercise interventions.

Circulating miRNA-126 increases in response to different forms of endurance exercise in healthy adults, however, there is no impact on the levels of miRNA-133, a marker for muscle damage (64). Nielsen et al (65) examined miRNAs in human plasma as a response to acute exercise and chronic endurance training with a novel methodological approach. Their data indicated that eight ci-miRs (miR-106a, −221, −30b, −151-5p, let-7i, miR-146a, −652,-151-3p) were downregulated immediately following acute exercise. Six ci-miRs (miR-338-3p, −330-3p, −223, −139-5p, miR-143, −1) were upregulated 1–3 h following acute endurance exercise. Basal ci-miRs levels were altered following 12 weeks of endurance training, and seven ci-miRs (miR-342-3p, let-7d, miR-766, −25, −148a, −185, −21) were decreased, while two ci-miRs (miR-185, −21) were increased following the training period (65).

Guescini et al (66) investigated muscle tissue release of EVs carrying miRNAs in the bloodstream during physical exercise. A significant positive correlation was found between the aerobic fitness and muscle-specific miRNAs, and EV miR-133b and −181a-5p were significantly upregulated following acute exercise. Therefore, EVs could be a novel means for muscle communication involved in muscle remodeling and homeostasis.

Muscle and organ cross talk through microvesicles

Organ crosstalk may also be achieved by the release of miRNAs packaged in exosomes that are transported through circulation and delivery to other tissues (67,68). Accumulating evidence suggests that skeletal muscle is also involved in the crosstalk between other organs (69,70).

In the past year, multiple publications have introduced exciting details regarding cell-to-cell communication, and exosomes are quickly becoming biomarkers for disease progression and cancer recurrence. Research has shown that cell-to-cell communication using microvesicles and exosomes, produced by MSCs, can be transferred to damaged tissues to help repair skeletal muscle injuries (71).

The data confirmed that β-F1-ATPase translation was lower in obese individuals compared with healthy weight controls and was correlated with miR-127-5p expression. Moreover, studies demonstrated that miR-127-5p is present in muscle-derived blood exosomes, suggesting their putative involvement in intracellular cross-talk (72).

The field of direct cell-to-cell communication, especially myocyte to other neighboring cells, is an exciting one (73). miRNA enriched exosomes are highly regulated by various stressors and disease conditions, and have been implicated in skeletal muscle function (19). Therefore, it is possible that exercise leads to the release of miRNA-enriched exosomes into the circulatory system from working muscles, the heart, or adipose tissue to facilitate organ crosstalk and control gene expression (67).

Muscles and kidneys can crosstalk with the slow progression of CKD. The mechanisms for this interaction involve muscle secretomes, consisting of a variety of growth factors and cytokines that are expressed and secreted by skeletal muscle (69). Akt1-mediated fast/glycolytic skeletal muscle growth reversed muscle wasting and reduced renal damage in a UUO model (74). However, it is unclear if microvesicles and exosomes mediate muscle-kidney crosstalk.

Conclusion

The emergence of the exosome field provides an exciting opportunity to further understand cell-cell communication in skeletal muscle and muscle-organ crosstalk. miRNAs transported via extracellular vesicles may mediate skeletal muscle development, regeneration, function and diseases. miRNAs are potential biomarkers that may be powerful and exciting tools for the diagnosis and treatment of skeletal muscle diseases in the future.

Acknowledgements

The present study was supported by grants from National Natural Science Foundation of China (grant no. 31302049), Doctoral Program of Higher Education (grant no. 20131403120002), and Agro-scientific Research in the Public Interest (grant no. 201303119).

References

1 

Güller I and Russell AP: MicroRNAs in skeletal muscle: Their role and regulation in development, disease and function. J Physiol. 588:4075–4087. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Drummond MJ, Glynn EL, Fry CS, Dhanani S, Volpi E and Rasmussen BB: Essential amino acids increase microRNA-499, −208b, and −23a and downregulate myostatin and myocyte enhancer factor 2C mRNA expression in human skeletal muscle. J Nutr. 139:2279–2284. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Allegra A, Alonci A, Campo S, Penna G, Petrungaro A, Gerace D and Musolino C: Circulating microRNAs: New biomarkers in diagnosis, prognosis and treatment of cancer (Review). Int J Oncol. 41:1897–1912. 2012.PubMed/NCBI

4 

Alexandrov PN, Dua P, Hill JM, Bhattacharjee S, Zhao Y and Lukiw WJ: microRNA (miRNA) speciation in Alzheimer's disease (AD) cerebrospinal fluid (CSF) and extracellular fluid (ECF). Int J Biochem Mol Biol. 3:365–373. 2012.PubMed/NCBI

5 

He WA, Calore F, Londhe P, Canella A, Guttridge DC and Croce CM: Microvesicles containing miRNAs promote muscle cell death in cancer cachexia via TLR7. Proc Natl Acad Sci USA. 111:4525–4529. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Lo Cicero A, Delevoye C, Gilles-Marsens F, Loew D, Dingli F, Guéré C, André N, Vié K, van Niel G and Raposo G: Exosomes released by keratinocytes modulate melanocyte pigmentation. Nat Commun. 6:75062015. View Article : Google Scholar : PubMed/NCBI

7 

Al-Nedawi K, Szemraj J and Cierniewski CS: Mast cell-derived exosomes activate endothelial cells to secrete plasminogen activator inhibitor type 1. Arterioscler Thromb Vasc Biol. 25:1744–1749. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Buschow SI, van Balkom BWM, Aalberts M, Heck AJR, Wauben M and Stoorvogel W: MHC class II-associated proteins in B-cell exosomes and potential functional implications for exosome biogenesis. Immunol Cell Biol. 88:851–856. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Forterre A, Jalabert A, Chikh K, Pesenti S, Euthine V, Granjon A, Errazuriz E, Lefai E, Vidal H and Rome S: Myotube-derived exosomal miRNAs downregulate Sirtuin1 in myoblasts during muscle cell differentiation. Cell Cycle. 13:78–89. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Guescini M, Guidolin D, Vallorani L, Casadei L, Gioacchini AM, Tibollo P, Battistelli M, Falcieri E, Battistin L, Agnati LF, et al: C2C12 myoblasts release micro-vesicles containing mtDNA and proteins involved in signal transduction. Exp Cell Res. 316:1977–1984. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Heijnen HFG, Schiel AE, Fijnheer R, Geuze HJ and Sixma JJ: Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 94:3791–3799. 1999.PubMed/NCBI

12 

Rabesandratana H, Toutant JP, Reggio H and Vidal M: Decay-accelerating factor (CD55) and membrane inhibitor of reactive lysis (CD59) are released within exosomes during In vitro maturation of reticulocytes. Blood. 91:2573–2580. 1998.PubMed/NCBI

13 

Romancino DP, Paterniti G, Campos Y, De Luca A, Di Felice V, d'Azzo A and Bongiovanni A: Identification and characterization of the nano-sized vesicles released by muscle cells. FEBS Lett. 587:1379–1384. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Wolfers J, Lozier A, Raposo G, Regnault A, Théry C, Masurier C, Flament C, Pouzieux S, Faure F, Tursz T, et al: Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 7:297–303. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Yang Y, Xiu F, Cai Z, Wang J, Wang Q, Fu Y and Cao X: Increased induction of antitumor response by exosomes derived from interleukin-2 gene-modified tumor cells. J Cancer Res Clin Oncol. 133:389–399. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Yu X, Huang C, Song B, Xiao Y, Fang M, Feng J and Wang P: CD4+CD25+ regulatory T cells-derived exosomes prolonged kidney allograft survival in a rat model. Cell Immunol. 285:62–68. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Matsuzaka Y and Hashido K: Roles of miR-1, miR-133a, and miR-206 in calcium, oxidative stress, and NO signaling involved in muscle diseases. RNA Dis. 2:558. 2015.

18 

Peter ME: Targeting of mRNAs by multiple miRNAs: the next step. Oncogene. 29:2161–2164. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Aoi W: Frontier impact of microRNAs in skeletal muscle research: a future perspective. Front Physiol. 5:4952015. View Article : Google Scholar : PubMed/NCBI

20 

Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM, Conlon FL and Wang DZ: The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet. 38:228–233. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Kim HK, Lee YS, Sivaprasad U, Malhotra A and Dutta A: Muscle-specific microRNA miR-206 promotes muscle differentiation. J Cell Biol. 174:677–687. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Berardi E and Sampaolesi M: Novel therapeutic approaches for skeletal muscle dystrophies. Muscle Cell and Tissue. Sakuma K: InTechOpen. (Rijeka). 393–412. 2015.

23 

Cheng G: Circulating miRNAs: Roles in cancer diagnosis, prognosis and therapy. Adv Drug Deliv Rev. 81:75–93. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Consalvi S, Sandoná M and Saccone V: Epigenetic reprogramming of muscle progenitors: inspiration for clinical therapies. Stem Cells Int. 2016:60936012016. View Article : Google Scholar : PubMed/NCBI

25 

Lee Y, El Andaloussi S and Wood MJA: Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum Mol Genet. 21(R1): R125–R134. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Suzuki T, Yamashita K, Jomen W, Ueki S, Aoyagi T, Fukai M, Furukawa H, Umezawa K, Ozaki M and Todo S: The novel NF-kappaB inhibitor, dehydroxymethylepoxyquinomicin, prevents local and remote organ injury following intestinal ischemia/reperfusion in rats. J Surg Res. 149:69–75. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Lai RC, Chen TS and Lim SK: Mesenchymal stem cell exosome: a novel stem cell-based therapy for cardiovascular disease. Regen Med. 6:481–492. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Jenjaroenpun P, Kremenska Y, Nair VM, Kremenskoy M, Joseph B and Kurochkin IV: Characterization of RNA in exosomes secreted by human breast cancer cell lines using next-generation sequencing. PeerJ. 1:e2012013. View Article : Google Scholar : PubMed/NCBI

29 

Huang L, Ma W, Ma Y, Feng D, Chen H and Cai B: Exosomes in mesenchymal stem cells, a new therapeutic strategy for cardiovascular diseases? Int J Biol Sci. 11:238–245. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Burke M, Choksawangkarn W, Edwards N, Ostrand-Rosenberg S and Fenselau C: Exosomes from myeloid-derived suppressor cells carry biologically active proteins. J Proteome Res. 13:836–843. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MAJ, Hopmans ES, Lindenberg JL, de Gruijl TD, Würdinger T and Middeldorp JM: Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci USA. 107:6328–6333. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Yoon YJ, Kim OY and Gho YS: Extracellular vesicles as emerging intercellular communicasomes. BMB Rep. 47:531–539. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Yellon DM and Davidson SM: Exosomes: Nanoparticles involved in cardioprotection? Circ Res. 114:325–332. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Ban JJ, Lee M, Im W and Kim M: Low pH increases the yield of exosome isolation. Biochem Biophys Res Commun. 461:76–79. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Yang JC, Lin MW, Rau CS, Jeng SF, Lu TH, Wu YC, Chen YC, Tzeng SL, Wu CJ and Hsieh CH: Altered exosomal protein expression in the serum of NF-κB knockout mice following skeletal muscle ischemia-reperfusion injury. J Biomed Sci. 22:402015. View Article : Google Scholar : PubMed/NCBI

36 

Nakamura Y, Miyaki S, Ishitobi H, Matsuyama S, Nakasa T, Kamei N, Akimoto T, Higashi Y and Ochi M: Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration. FEBS Lett. 589:1257–1265. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Zablocki D and Sadoshima J: Inside-Out Signaling: moving the AT1 Receptor in to Get the Message Out. Circulation. 131:2097–2100. 2015. View Article : Google Scholar : PubMed/NCBI

38 

O'Rourke JR, Georges SA, Seay HR, Tapscott SJ, McManus MT, Goldhamer DJ, Swanson MS and Harfe BD: Essential role for Dicer during skeletal muscle development. Dev Biol. 311:359–368. 2007. View Article : Google Scholar : PubMed/NCBI

39 

McCarthy JJ, Esser KA and Andrade FH: MicroRNA-206 is overexpressed in the diaphragm but not the hindlimb muscle of mdx mouse. Am J Physiol Cell Physiol. 293:C451–C457. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Forterre A, Jalabert A, Berger E, Baudet M, Chikh K, Errazuriz E, De Larichaudy J, Chanon S, Weiss-Gayet M, Hesse AM, et al: Proteomic analysis of C2C12 myoblast and myotube exosome-like vesicles: a new paradigm for myoblast-myotube cross talk? PLoS One. 9:e841532014. View Article : Google Scholar : PubMed/NCBI

41 

Hudson MB, Woodworth-Hobbs ME, Zheng B, Rahnert JA, Blount MA, Gooch JL, Searles CD and Price SR: miR-23a is decreased during muscle atrophy by a mechanism that includes calcineurin signaling and exosome-mediated export. Am J Physiol Cell Physiol. 306:C551–C558. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Hudson MB, Rahnert JA, Zheng B, Woodworth-Hobbs ME, Franch HA and Price SR: miR-182 attenuates atrophy-related gene expression by targeting FoxO3 in skeletal muscle. Am J Physiol Cell Physiol. 307:C314–C319. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Quattrocelli M and Sampaolesi M: The mesmiRizing complexity of microRNAs for striated muscle tissue engineering. Adv Drug Deliv Rev. 88:37–52. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Safdar A, Abadi A, Akhtar M, Hettinga BP and Tarnopolsky MA: miRNA in the regulation of skeletal muscle adaptation to acute endurance exercise in C57Bl/6J male mice. PLoS One. 4:e56102009. View Article : Google Scholar : PubMed/NCBI

45 

Muroya S, Ogasawara H and Hojito M: Grazing affects exosomal circulating microRNAs in cattle. PLoS One. 10:e01364752015. View Article : Google Scholar : PubMed/NCBI

46 

Atay S and Godwin AK: Tumor-derived exosomes: A message delivery system for tumor progression. Commun Integr Biol. 7:e282312014. View Article : Google Scholar : PubMed/NCBI

47 

De Guire V, Robitaille R, Tétreault N, Guérin R, Ménard C, Bambace N and Sapieha P: Circulating miRNAs as sensitive and specific biomarkers for the diagnosis and monitoring of human diseases: promises and challenges. Clin Biochem. 46:846–860. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Van Roosbroeck K, Pollet J and Calin GA: miRNAs and long noncoding RNAs as biomarkers in human diseases. Expert Rev Mol Diagn. 13:183–204. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Bloch SA, Donaldson AV, Lewis A, Banya WA, Polkey MI, Griffiths MJ and Kemp PR: miR-181a: a potential biomarker of acute muscle wasting following elective high-risk cardiothoracic surgery. Crit Care. 19:1472015. View Article : Google Scholar : PubMed/NCBI

50 

Chen JF, Callis TE and Wang DZ: MicroRNAs and muscle disorders. J Cell Sci. 122:13–20. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Donaldson A, Natanek SA, Lewis A, Man WDC, Hopkinson NS, Polkey MI and Kemp PR: Increased skeletal muscle-specific microRNA in the blood of patients with COPD. Thorax. 68:1140–1149. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Lewis A, Riddoch-Contreras J, Natanek SA, Donaldson A, Man WDC, Moxham J, Hopkinson NS, Polkey MI and Kemp PR: Downregulation of the serum response factor/miR-1 axis in the quadriceps of patients with COPD. Thorax. 67:26–34. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Naguibneva I, Ameyar-Zazoua M, Polesskaya A, Ait-Si-Ali S, Groisman R, Souidi M, Cuvellier S and Harel-Bellan A: The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nat Cell Biol. 8:278–284. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Croce CM: Novel function of microRNAs. Clin Cancer Res. 21:2015. View Article : Google Scholar

55 

Camargo RG, Quintas Teixeira Ribeiro H, Geraldo MV, Matos-Neto E, Neves RX, Carnevali LC Jr, Donatto FF, Alcântara PS, Ottoch JP and Seelaender M: Cancer Cachexia and MicroRNAs. Mediators Inflamm. 2015:3675612015. View Article : Google Scholar : PubMed/NCBI

56 

Xu J, Li R, Workeneh B, Dong Y, Wang X and Hu Z: Transcription factor FoxO1, the dominant mediator of muscle wasting in chronic kidney disease, is inhibited by microRNA-486. Kidney Int. 82:401–411. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Soares RJ, Cagnin S, Chemello F, Silvestrin M, Musaro A, De Pitta C, Lanfranchi G and Sandri M: Involvement of microRNAs in the regulation of muscle wasting during catabolic conditions. J Biol Chem. 289:21909–21925. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Yamaguchi T, Izumi Y, Nakamura Y, Yamazaki T, Shiota M, Sano S, Tanaka M, Osada-Oka M, Shimada K, Miura K, et al: Repeated remote ischemic conditioning attenuates left ventricular remodeling via exosome-mediated intercellular communication on chronic heart failure after myocardial infarction. Int J Cardiol. 178:239–246. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Hu Z, Klein JD, Mitch WE, Zhang L, Martinez I and Wang XH: MicroRNA-29 induces cellular senescence in aging muscle through multiple signaling pathways. Aging (Albany NY). 6:160–175. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Hu J, Du J, Zhang L, Price SR, Klein JD and Wang XH: XIAP reduces muscle proteolysis induced by CKD. J Am Soc Nephrol. 21:1174–1183. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Russell AP and Lamon S: Exercise, skeletal muscle and circulating microRNAs. Prog Mol Biol Transl Sci. 135:471–496. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Mooren FC, Viereck J, Krüger K and Thum T: Circulating microRNAs as potential biomarkers of aerobic exercise capacity. Am J Physiol Heart Circ Physiol. 306:H557–H563. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Baggish AL, Hale A, Weiner RB, Lewis GD, Systrom D, Wang F, Wang TJ and Chan SY: Dynamic regulation of circulating microRNA during acute exhaustive exercise and sustained aerobic exercise training. J Physiol. 589:3983–3994. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Uhlemann M, Möbius-Winkler S, Fikenzer S, Adam J, Redlich M, Möhlenkamp S, Hilberg T, Schuler GC and Adams V: Circulating microRNA-126 increases after different forms of endurance exercise in healthy adults. Eur J Prev Cardiol. 21:484–491. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Nielsen S, Åkerström T, Rinnov A, Yfanti C, Scheele C, Pedersen BK and Laye MJ: The miRNA plasma signature in response to acute aerobic exercise and endurance training. PLoS One. 9:e873082014. View Article : Google Scholar : PubMed/NCBI

66 

Guescini M, Canonico B, Lucertini F, Maggio S, Annibalini G, Barbieri E, Luchetti F, Papa S and Stocchi V: Muscle releases alpha-sarcoglycan positive extracellular vesicles carrying miRNAs in the bloodstream. PLoS One. 10:e01250942015. View Article : Google Scholar : PubMed/NCBI

67 

Zierath JR and Wallberg-Henriksson H: Looking ahead perspective: where will the future of exercise biology take us? Cell Metab. 22:25–30. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Hamrick MW: The skeletal muscle secretome: an emerging player in muscle-bone crosstalk. Bonekey Rep. 1:602012. View Article : Google Scholar : PubMed/NCBI

70 

Rondon-Berrios H, Wang Y and Mitch WE: Can muscle-kidney crosstalk slow progression of CKD? J Am Soc Nephrol. 25:2681–2683. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Zhu YG, Feng XM, Abbott J, Fang XH, Hao Q, Monsel A, Qu JM, Matthay MA and Lee JW: Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells. 32:116–125. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Tran L, Campbell L, Coletta D, Mandarino L and Katsanos C: Skeletal muscle β-F1-ATPase translation is inhibited by hyperlipidemia-induced miR-127-5p expression in human obesity. FASEB J. 29(Suppl 1): 974–976. 2015.

73 

Nolta JA: New advances in understanding stem cell fate and function. Stem Cells. 33:313–315. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Hanatani S, Izumiya Y, Araki S, Rokutanda T, Kimura Y, Walsh K and Ogawa H: Akt1-mediated fast/glycolytic skeletal muscle growth attenuates renal damage in experimental kidney disease. J Am Soc Nephrol. 25:2800–2811. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 5 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang H and Wang H: Extracellular vesicle microRNAs mediate skeletal muscle myogenesis and disease (Review). Biomed Rep 5: 296-300, 2016
APA
Wang, H., & Wang, H. (2016). Extracellular vesicle microRNAs mediate skeletal muscle myogenesis and disease (Review). Biomedical Reports, 5, 296-300. https://doi.org/10.3892/br.2016.725
MLA
Wang, H., Wang, B."Extracellular vesicle microRNAs mediate skeletal muscle myogenesis and disease (Review)". Biomedical Reports 5.3 (2016): 296-300.
Chicago
Wang, H., Wang, B."Extracellular vesicle microRNAs mediate skeletal muscle myogenesis and disease (Review)". Biomedical Reports 5, no. 3 (2016): 296-300. https://doi.org/10.3892/br.2016.725