Abstract
Skeletal muscle plays a paramount role in physical activity, metabolism, and energy balance, while its homeostasis is being challenged by multiple unfavorable factors such as injury, aging, or obesity. Exosomes, a subset of extracellular vesicles, are now recognized as essential mediators of intercellular communication, holding great clinical potential in the treatment of skeletal muscle diseases. Herein, we outline the recent research progress in exosomal isolation, characterization, and mechanism of action, and emphatically discuss current advances in exosomes derived from multiple organs and tissues, and engineered exosomes regarding the regulation of physiological and pathological development of skeletal muscle. These remarkable advances expand our understanding of myogenesis and muscle diseases. Meanwhile, the engineered exosome, as an endogenous nanocarrier combined with advanced design methodologies of biomolecules, will help to open up innovative therapeutic perspectives for the treatment of muscle diseases.
Keywords: Exosome, Skeletal muscle, Muscle atrophy, Insulin resistance
1. Introduction
Skeletal muscle develops from muscle progenitor cells on the side of the paraxial mesoderm. Progenitor cells undergo terminal differentiation to form mononuclear myoblasts ( Zeschnigk et al., 1995), which further fuse into myotubes ( Steinacker et al., 2000) and ultimately generate biofunctional muscle tissue. Skeletal muscle is a highly dynamic and plastic organ that requires continuous surveillance to maintain biofunctional integrity in response to the demands of movement and metabolism. The biofunctional integrity of skeletal muscle is coordinated by complex regulatory circuits; however, multiple disadvantages, such as injury, aging, inactivity, and the current obesity epidemic, have disrupted this regulatory network and resulted in various skeletal muscle diseases ( Wu and Ballantyne, 2017; Larsson et al., 2019).
Early work on extracellular vesicles (EVs) has deepened our understanding of the interactions between organs and tissues. EVs are particles composed of lipid bilayers, and preliminarily classified into exosome, microvesicle, apoptotic body, and oncosome according to their biogenesis, release, and particle size. The diameters of EVs range from 20–30 nm (exomere, 35 nm) ( Zhang et al., 2018) to 10 μm (large oncosome, 1‒10 μm) ( Minciacchi et al., 2015), but most of them are less than 200 nm. Recent studies illustrated that almost all living cells transport specific combinations of proteins, nucleic acids, and lipids between donors and recipients via exosomes, thereby mediating the genetic exchanges and leading to the reprogramming of recipients ( Torralba et al., 2018; Asare-Werehene et al., 2020; Chen CH et al., 2021; He et al., 2021; Xu et al., 2022). Convincing evidence has indicated that exosomes play vital roles in the physiology and pathology of skeletal muscle, suggesting the possibility of a novel strategy to ameliorate skeletal muscle dysfunction.
Herein, we review the current progress in exosomal isolation, characterization and mechanism of action. We further comprehensively discuss the physiological and pathological regulatory mechanisms mediated by diverse organs- and tissues-derived exosomes, as well as engineered exosomes, in skeletal muscle. The remarkable advances made in recent years expand our understanding of myogenesis and muscle diseases. Moreover, they clearly help to elucidate exosome-mediated regulatory pathways in the physiology and pathology of skeletal muscle, and reveal innovative therapeutic perspectives in protecting against muscle diseases. Meanwhile, the engineering exosome, as an endogenous nanocarrier combined with advanced design methodology of biomolecules, further highlights the clinically translational potential of exosomes.
2. Overview of exosomes
The exosome is a pivotal subtype of EVs originating from endosome-derived multivesicular bodies ( Kalluri and LeBleu, 2020; Gurunathan et al., 2021). It was first reported by Pan and Johnstone (1983) and named as “exosome” by Johnstone et al. (1987). It is known to play essential roles in cellular communication by transferring bioactive cargos, while two major technical hindrances limit the relevant research. Firstly, these are challenges in simplifying the isolation method and improving the exosome yield, and secondly, it is difficult to effectively distinguish exosomes from other EV subtypes. In this section, we review the current research progress in exosomal isolation, characterization and mechanism of action.
2.1. Exosomal isolation
The isolation of exosomes is the first step in related research. Considering that a single isolation method applicable to different physicochemical and biochemical sources would be impractical, multiple methods have been established to extract exosomes from various sources. The commonly used methods include differential centrifugation, density gradient centrifugation, ultrafiltration, polymer precipitation, and immunoaffinity capture ( Li et al., 2017; Yang DB et al., 2020). In addition, efforts are still being pursued to improve existing methods and establish new ones. For example, the cushioned-density gradient ultracentrifugation (C-DGUC), a modification of DGUC, could purify exosomes from dense protein aggregates ( Li et al., 2018). In addition, size-exclusion chromatography is commonly used to isolate EVs from the serum because it could gain high-purity exosomes ( Monguió-Tortajada et al., 2019). Besides, asymmetric-flow field-flow fractionation (AF4) has been applied to isolate EVs and exosomes. Zhang et al. (2018) isolated and characterized two subpopulations of exosomes with AF4, and subsequently named them as large exosome vesicles (90‒120 nm) and small exosome vesicles (60‒80 nm). However, AF4 requires expertise to customize the protocol ( Zhang and Lyden, 2019). Furthermore, other exosomal isolation methods have also shown great potential, such as microfluidic technologies ( Hassanpour Tamrin et al., 2021), exosome detection method via the ultrafast-isolation system (EXODUS) ( Chen YC et al., 2021), nanostructures and microfluidic devices ( Le and Fan, 2021), rapid exosome isolation using ultrafiltration and size exclusion chromatography (REIUS) (la Shu et al., 2021). In short, as each isolation method has its advantages and drawbacks, it is highly practical to choose an appropriate method according to the samples and demands, rather than developing a universal method.
2.2. Exosomal characterization
The heterogeneity of exosomes and the differences between isolation methods, as well as residual non-exosomal substances, present certain requirements for the characterization of isolated exosomes. Traditionally, transmission electron microscopy (TEM), nanoparticle tracer analysis (NTA), and western blotting were used to characterize the isolated exosomes, and additional techniques have been developed for exosomal characterization. Firstly, scanning electron microscopy (SEM) ( Gao et al., 2018) and atomic force microscopy (AFM) ( Sharma et al., 2018) were adopted for characterizing cup-shaped morphology. With regards to exosome size characterization, single-particle interference reflection imaging sensor (SP-IRIS), microfluidic resistive pulse sensing (MRPS), and nanoflow cytometry measurement (NFCM) are alternative methods to NTA. By comparison among NTA, SP-IRIS, MRPS, and NFCM based on particle size, concentration, and fluorescence measurement accuracy, Arab et al. (2021) suggested that NFCM is the best one that can simultaneously provide particle size distribution, particle concentration, and fluorescence measurement accuracy at the single-particle level. Western blotting is generally applied to characterize exosome-membrane proteins, such as cluster of differentiation 9 (CD9) and CD63. Some researchers suggested excluding the possibility that some impurities mix in exosomes by testing negative biomarkers such as Golgi matrix protein 130 (GM130) and calnexin ( Samaeekia et al., 2018). However, given the complexity of exosomal cargos, there is no commonly recognized biomarker of impurities, and it requires judgement based on samples ( Théry et al., 2018). Clearly, it is of great value to develop multiple characterization methods to identify the collected subpopulations of EVs. The precise distinguishment of exosomes from other EVs will favor the development of isolation, function, and the ultimate theranostic therapy of exosome-mediated cell-free strategy.
2.3. Exosomal mechanisms of action
It is well-established that exosomes exhibit regulatory roles through exosomal membrane proteins ( Gross et al., 2012; Gao et al., 2016) and cargos targeting the receptors located on the surface ( Wang et al., 2011) or inside the recipient ( Wei et al., 2020). Recently, Keller et al. (2020) reported a new exosomal mechanism of action: bacterial DNA and cytosine-guanine dinucleotide (CpG) DNA lead to an increased secretion of a disintegrin and metalloproteinase 10 (ADAM10)-bearing exosomes from both human and mice cells, and ADAM10-bearing exosomes act as decoys in response to bacterial infection to prevent damages to target tissues ( Fig. 1). This novel mechanism contributes to expanding our understanding of exosomes and exhibits great potential as endogenous decoys in the clinical treatment of virus-induced diseases, especially those of skeletal muscle.
Fig. 1. Exosomal biogenesis and mechanisms of action. Exosomes are described as vesicles of endosomal origin. Endocytosis is the first step to form early endosomes, and the early endosomes mature into late endosomes with specific cargos loaded, which further mature into multivesicular bodies. The latter are subject to two pathways: either lysosomal degradation, or secretion as exosomes with a size of 30‒150 nm in diameter through fusion with the plasma membrane and ultimate exocytosis. Cluster of differentiation 9 (CD9) and CD63 are membrane markers; tumor necrosis factor-α (TNF-α) and Wingless/Int-1 lipoglycoproteins (WNTs) are exosomal cargos on the surface. MiRNAs: microRNAs; ADAM10: a disintegrin and metalloproteinase 10.
3. Exosome-mediated regulatory mechanisms in skeletal muscle
Accumulating evidence shows that exosomes extensively participate in the physiology and pathology of skeletal muscle. Exosome-mediated cell-free strategy, resembling the beneficial effects of cell-based strategies without immune rejection, has been a popular field to explore the pathobiology and therapeutic strategy in multiple pathophysiologic processes. In this section, we focus our attention on exosomes, but not all EV subsets, derived from multiple tissues and organs (skeletal muscle, adipose, blood, urine, cancer, mesenchymal stem cells (MSCs), and other types of cells) in regulating the physiological and pathological development of skeletal muscle ( Table 1).
Table 1.
Biological functions and mechanism of exosomes in skeletal muscle
Origin | Factor | Function | Target | Reference |
---|---|---|---|---|
Myogenic progenitor cells | MiR-206 | Inhibits collagen biosynthesis to ensure optimal muscle remodeling | RRBP1 | Fry et al., 2017 |
Hindlimb muscle | Let-7-5p | Increases protein synthesis | IGF-1 | Huang et al., 2021 |
Adipocyte | MiR-27a | Induces IR | PPARγ | Yu et al., 2018; Wang et al., 2022 |
Gonadal white adipose tissue | MiR-222 | Promotes obesity-associated IR and glucose intolerance | IRS1 and p-AKT | Li et al., 2020 |
Adipose tissue macrophage (M1) | MiR-155 | Causes glucose intolerance and IR | PPARγ | Ying et al., 2017 |
Bone marrow-derived macrophages (M2) | MiR-690 | Improves glucose intolerance and insulin sensitivity | Nadk | Ying et al., 2021 |
Adipose tissue macrophage (M1) | MiR-29a | Causes IR | PPARδ | Liu et al., 2019 |
Serum | MiR-20b-5p | Impairs insulin action in human skeletal muscle | AKTIP and STAT3 | Katayama et al., 2019 |
Oral squamous cell carcinoma cells | MiR-181a-3p | Induces muscle cell atrophy and apoptosis | Unspecified | Qiu et al., 2020b |
C26 cell-induced colon cancer | MiR-195a-5p and miR-125b-1-3p | Induce skeletal muscle wasting | Bcl-2 | Miao et al., 2021 |
C26 cells | GDF-15 | Induces skeletal muscle atrophy | Bcl-2/ caspase-3 | Zhang et al., 2022 |
Umbilical cord MSCs | Unspecified | Ameliorates blood glucose levels and partially reverses IR | GLUT4 | Sun et al., 2018 |
Placenta-derived MSCs | MiR-29c | Promotes differentiation of myoblasts | Unspecified | Bier et al., 2018 |
Umbilical cord MSCs | CircHIPK3 | Prevents pyroptosis and repair ischemic muscle injury | MiR-421/ FoxO3a | Yan et al., 2020 |
Bone marrow MSCs | Unspecified | Facilitates recovery after injury | TGF-β | Iyer et al., 2020 |
Bone marrow MSCs | MiR-486-5p | Inhibits dexamethasone-induced muscle atrophy | FoxO1 | Li et al., 2021 |
Adipose tissue-derived MSCs | Unspecified | Attenuates the damage of biopsy punch to muscle bundles | MYOG/ MYOD | Byun et al., 2021 |
Muscle-derived fibroblasts | MiR-199a-5p | Promotes skeletal muscle fibrosis | Caveolin-1 | Zanotti et al., 2018 |
Engineered HEK293 cells | MiR-26a | Ameliorates muscle wasting | FoxO1 | Zhang et al., 2019 |
Engineered bone marrow MSCs | MiR-215 | Protects skeletal muscle against injury | FABP3 | Zhou Q et al., 2021 |
Engineered C2C12 cells | Hsp60 | Ameliorates muscle wasting and cachexia | PGC-1α isoform 1 | di Felice et al., 2022 |
Engineered human bone marrow mesenchymal stromal cells | IL6ST | Partially ameliorates IL6/IL6R complexes-induced anti-differentiation effects | STAT3 | Conceição et al., 2021 |
AKTIP: protein kinase B (AKT)-interacting protein; Bcl-2: B-cell lymphoma-2; CircHIPK3: circular RNA homeodomain-interacting protein kinase 3; FoxO3a: forkhead box O3a; FABP3: fatty acid-binding protein 3 gene; GDF-15: growth differentiation factor-15; GLUT4: glucose transporter 4; Hsp60: heat shock protein 60; IGF-1: insulin-like growth factor-1; IL6R: interleukin 6 (IL6) receptor; IL6ST: IL6 signal transducer; IRS1: insulin receptor substrate 1; IR: insulin resistance; Let-7-5p: lethal-7-5p; MiR: microRNA; MSCs: mesenchymal stem cells; MYOG: myogenin; MYOD: myoblast determination protein 1; Nadk: a gene encoding nicotinamide adenine dinucleotide (NAD) + kinase; p-AKT: phosphorylated protein kinase B; PGC-1α: peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α; RRBP1: ribosome-binding protein 1; STAT3: signal transducer and activator of transcription 3; TGF-β: transforming growth factor-β.
3.1. Regulatory mechanisms of skeletal muscle-derived exosomes in skeletal muscle
Skeletal muscle secretes exosomes, some of which enter the bloodstream and others are taken up by proximal muscle tissue ( Mytidou et al., 2021), to influence its own fate and biological activities, such as muscle remodeling. Fry et al. (2017) illustrated that myogenic progenitor cells (MPCs) contribute to optimal muscle remodeling in response to hypertrophic stimuli via the exosomal microRNA-206 (miR-206)/ribosome-binding protein 1 (RRBP1) axis. Skeletal muscle-derived exosomes are associated with muscle atrophy. Kim et al. (2018) reported that exosomes isolated from the conditioned media (CM) of inflammatory murine myoblast (C2C12) myotubes contribute to inflammation-induced muscle atrophy by causing myoblast inflammation and inhibiting the myogenic process. In addition to inflammatory models, myotube-derived exosomes induce functional improvements and alleviate muscle deterioration by improving muscle membrane integrity in dystrophic mice ( Leng et al., 2021), highlighting the therapeutic potential of exosomes for Duchenne muscular dystrophy (DMD) and other skeletal muscle disorders with compromised membranes. Furthermore, Huang et al. (2021) found that acupuncture with low-frequency electrical stimulation (Acu/LFES), a potential strategy to treat muscle atrophy ( Hu et al., 2015), promotes protein synthesis in the hindlimb and forelimb skeletal muscles by decreasing the expression of lethal-7-5p (let-7-5p), which inhibits insulin-like growth factor-1 (IGF-1), in the muscle of both hindlimbs and the circulating exosomes of C57/BL6 mice. Moreover, exosomes also exert a pro-fibrotic effect. Zanotti et al. (2018) reported that exosomal miR-199a-5p from muscle-derived fibroblasts in DMD patients targets caveolin-1, an anti-fibrotic signaling molecule ( Yi et al., 2014), and induces skeletal muscle fibrosis. Collectively, the above studies indicate that the exosomal cargos, such as miRNAs, are altered in response to physiological and pathological stimuli, and the altered cargos in turn influence the physiological and pathological fates of skeletal muscle, and thus could be applied for the treatment of skeletal muscle diseases.
3.2. Regulatory functions of adipose-derived exosomes in skeletal muscle
Both skeletal muscle and adipose tissue are of mesodermal origin ( Buckingham et al., 2003; Billon et al., 2008). Numerous studies, especially those related to exosomes, provided evidence for their close relationships with insulin resistance (IR). Yu et al. (2018) found that adipocyte-derived exosomal miR-27a induces IR in skeletal muscle by inhibiting the expression of peroxisome proliferator-activated receptor γ (PPARγ) and its downstream genes. Besides, Li et al. (2020) found that gonadal white adipose tissue-derived exosomal miR-222 is upregulated in obese patients with IR and in serum samples from type 2 diabetes mellitus (T2DM) patients. The elevated exosomal miR-222 is considered be associated with IR and glucose homeostasis/disposal in skeletal muscle, as it downregulates the insulin receptor substrate 1 (IRS1) and phosphorylated protein kinase B (p-AKT) levels. Macrophages residing within adipose tissue are associated with insulin sensitivity in skeletal muscle. Ying et al. (2017) found that miRNA-containing exosomes from adipose tissue macrophages (ATMs-Exos) of obese mice bring about glucose intolerance and IR in the skeletal muscle of lean mice, whereas ATMs-Exos from lean mice improve the glucose tolerance and insulin sensitivity of obese mice. ATMs-Exo-miR-155 is the key mediator that leads to glucose intolerance and IR in skeletal muscle by targeting PPARγ ( Ying et al., 2017), and exosomal miR-690 is the key mediator that improves glucose tolerance and insulin sensitivity in the skeletal muscle of obese mice by targeting Nadk (a gene encoding nicotinamide adenine dinucleotide (NAD) + kinase) ( Ying et al., 2021). It is worth noting that Liu et al. (2019) identified another mediator, miR-29a, in ATMs-Exos of obese mice, that causes IR in myocytes by targeting PPARδ. Beyond IR, adipose-derived exosomes play important roles in skeletal muscle regeneration. Wang CY et al. (2019) demonstrated that human adipose stem cells-derived exosomes (ASCs-Exos) significantly improve the regeneration and biomechanical properties of torn rotator cuff muscles by ameliorating atrophy and degeneration. What is more, exosomes from adipose-derived MSCs (AD-MSCs-Exos) promote the proliferation and expression of myocyte-related genes. Importantly, AD-MSCs-Exos attenuate the damage of biopsy punch to the shapes and sizes of the muscle bundles and increase the expression of myogenin (MYOG) and myoblast determination protein 1 (MYOD) ( Byun et al., 2021). To sum up, adipose-derived exosome is critical to skeletal muscle IR and regeneration, and exosomal miRNA is the most widely studied regulator in the current research ( Table 1). Given that adipose tissue is composed of multiple cell types, it will be an important study to distinguish the cell types/subtypes that exert beneficial effects through exosomes.
3.3. Regulatory roles of blood-derived exosomes in skeletal muscle
Blood is a circulating body fluid of vertebrates, which contains exosomes ( Li et al., 2014; Manterola et al., 2014; Zhao et al., 2017). Experimental studies indicated that serum-EV is a potential factor against muscle damage and aged skeletal muscle atrophy ( Cavallari et al., 2017; Sahu et al., 2021). In a rat model with tibialis anterior muscle injury, Iyer et al. (2020) reported that platelet-rich plasma-derived exosomes accelerated the recovery rhythm of contractile function by upregulating MYOG. In addition to muscle regeneration, serum exosome exerts a regulatory effect on T2DM. Katayama et al. (2019) documented that the overexpression of miR-20b-5p, a differentially expressed serum exosomal miRNA between normal subjects and T2DM patients, promotes basal glycogen synthesis in myocytes by targeting AKT-interacting protein (AKTIP) and signal transducer and activator of transcription 3 (STAT3). In older subjects doing long-term exercise, Nair et al. (2020) identified a subset of differentially expressed plasma-derived exosomal miRNAs that target genes in the insulin-like growth factor-1 (IGF-1) signaling pathway, a pathway involved in muscle growth and glucose homeostasis ( Vassilakos et al., 2019), highlighting the potential role of exercise-induced circulating exosomes in muscle growth and glucose homeostasis. Furthermore, Wang et al. (2022) reported the exercise-induced reduction of serum exosomal miR-27a level, which is positively associated with IR ( Yu et al., 2018) by targeting PPARγ. Briefly, blood exosomes are important mediators of both injury and metabolic disease in skeletal muscle. Considering the wide availability of blood, further studies are warranted to explore the unknown effects of blood exosomes on skeletal muscle, to advance their translational potential in the clinic.
3.4. Regulatory functions of urine-derived exosomes in skeletal muscle
Beyond being a metabolic waste, urine is considered as a promising non-invasive cell source for cell-based therapies in skeletal muscle regeneration ( Chen et al., 2017), while exosomal studies have shown that it might be related with cell-free therapies. Zhu et al. (2018) collected urine-derived stem cells-EV (USCs-EV) with a diameter of 30‒150 nm (within the range of exosomes) that expressed exosomal markers. The intramuscular injection of USCs-EV promoted muscle regeneration in a hindlimb ischemia mouse model. Furthermore, in vitro trials showed that USCs-EV promotes C2C12 cell proliferation in a dose-dependent manner ( Zhu et al., 2018). Using a rat stress urinary incontinence (SUI) model, Wu RY et al. (2019) found that USCs-exosomes (USCs-Exos) significantly accelerate the recovery of pubococcygeus muscle by promoting the activation, proliferation, and differentiation of muscle stem cells. In summary, urinary exosomes play significant roles in skeletal muscle regeneration. Given that urine is probably the most readily available patient-derived substance, it is likely to become a vital cell-source for exosome-mediated clinical treatments for skeletal muscle diseases.
3.5. Regulatory mechanisms of cancer-derived exosomes in skeletal muscle
Cancer cachexia is a cancer-induced systemic multifactorial disease characterized by specific losses of skeletal muscle, muscle strength, and adipose tissue that deteriorates the quality of life and increases the mortality of patients ( Baracos et al., 2018; Schmidt et al., 2018). Recent studies on exosomes may accelerate the development of therapeutic interventions to prevent or reverse this muscle wasting syndrome ( Li et al., 2016; Hu et al., 2019). EVs play a role in endoplasmic reticulum stress (ERS)-induced deterioration by promoting the malignant transformation of predisposed cells ( Wu CH et al., 2019) and cancer cell apoptosis ( Qiu et al., 2020a). Qiu et al. (2020b) further reported that exosomes from the CM of oral squamous cell carcinoma cells treated by tunicamycin lead to muscle atrophy and apoptosis by mediating transmissible ERS signaling. Exosomal miR-181a-3p was subsequently identified as a key factor. Circulating exosome intensity is relevant to cancer cachexia. Miao et al. (2021) showed that C26 colon carcinoma cell-derived exosomal miR-195a-5p and miR-125b-1-3p decrease the sizes of C2C12 myotubes and diminish muscle strength and muscle weight in C26 tumor-bearing mice by reducing the expression of B-cell lymphoma-2 (Bcl-2), an anti-apoptosis protein ( Schöneich et al., 2014), at both the messenger RNA (mRNA) and protein levels. In a subsequent study, Zhang et al. (2022) elucidated that growth differentiation factor-15 (GDF-15) is enriched in C26 colon carcinoma cell-derived exosomes (C26-Exos). The overexpression of GDF-15 in non-cachexic MC38-derived exosomes increases the potency of inducing muscle atrophy, whereas the knockdown of GDF-15 weakens the ability of C26-Exos to trigger muscle atrophy. Mechanistically, GDF-15 contributes to muscle atrophy via targeting Bcl-2/caspase-3 pathways ( Zhang et al., 2022). Zhou L et al. (2021) generated colon cancer cachexia and lung cancer cachexia mice models by the subcutaneous injection of CT26 and Lewis lung carcinoma (LLC) tumor cells, respectively. Importantly, both models showed increased circulating levels of exosomes that were largely derived from tumors. Meanwhile, amiloride, a potassium-sparing diuretic with anticancer effect ( Rojas et al., 2017), markedly alleviated muscle atrophy, and inhibited the secretion of tumor-derived exosomes without inducing systemic toxicities in healthy mice, highlighting the role of tumor-derived exosome in cancer cachexia ( Zhou L et al., 2021). In short, these studies suggest that cancer-derived exosomal cargos (such as miRNAs and proteins) and their circulating levels change with pathological development, providing a significant clinical value for muscle atrophy in cancer cachexia by inhibiting the production of tumor exosomes.
3.6. Regulatory roles of mesenchymal stem cell-derived exosomes in skeletal muscle
MSCs are multipotent—that is, they possess great potential to differentiate into multiple types of cells. Because MSCs are easily obtained and cultured in vitro, there is a remarkable desire to test whether they exert beneficial effects on specific diseases ( Wu et al., 2018; Fu et al., 2019). Sun et al. (2018) reported that the exosomes of human umbilical cord-derived MSC (hucMSC-Exos) ameliorate blood glucose levels and partially reverse IR in T2DM, that is, at least in part, to be mediated by upregulating the expression of membrane translocation of glucose transporter 4 (GLUT4) in muscle. Apart from IR, MSCs-derived exosomes play a role in muscle injury. Bier et al. (2018) found that CM and exosomes isolated from placenta-derived MSCs (PL-MSCs) promote the differentiation of myoblasts from Duchenne patients and mdx mice, and decrease the expression of fibrogenic genes in myoblasts of patients with DMD; the miR-29c at least partially mediates the effects of CM and exosome. Furthermore, Yan et al. (2020) showed that hucMSC-Exo treatment significantly promotes running distance and muscle force in a hindlimb ischemia mice model, which are blocked by the injection of human umbilical cord MSC-derived exosomes (UMSC-Exos) and small interfering RNA (siRNA)-targeting circular RNA homeodomain-interacting protein kinase 3 (si-circHIPK3). Besides, Iyer et al. (2020) revealed that muscles treated with exosomes of bone marrow-derived MSC (BMSC-Exos) reduce the expression of transforming growth factor-β (TGF-β) in a muscle injury rat model, while Li et al. (2021) reported that BMSC-Exos blunt dexamethasone-induced muscle atrophy both in vitro and in vivo via the miR-486-5p/forkhead box O1 (FoxO1) axis. In conclusion, the above studies indicate that MSCs-derived exosomes exert beneficial roles against IR and skeletal muscle injury; therefore, it is valuable to search for other types of stem cells that exert beneficial effects on skeletal muscle diseases through exosomes.
3.7. Regulatory activity of exosomes derived from other tissues and cells in skeletal muscle
Exosomes are secreted by almost all living cells and can be detected in various body fluids. Tavakoli Dargani et al. (2018) showed that embryonic stem cell exosomes (ES-Exos) inhibit doxorubicin- and inflammation-induced pyroptosis in mouse soleus muscle cells (Sol 8), while the exosome inhibitor GW4869 abolishes the beneficial effect of ES-Exos.
Circulating placental exosomes are increased in pregnancies with gestational diabetes mellitus (GDM), which are associated with skeletal muscle IR. Nair et al. (2018) reported that GDM pregnancy-derived placental exosomes attenuate the migration and glucose uptake of primary skeletal muscle from normal glucose tolerant (NGT) subjects in response to insulin stimulation, whereas placental exosomes from NGT enhance the migration and glucose uptake of skeletal muscle from diabetic patients.
Furthermore, Aminzadeh et al. (2018) reported that the absence of dystrophin deteriorates exercise ability and increases mortality in DMD patients, while cardiosphere-derived cells (CDCs) and CDC-derived exosomes (CDC-Exos) improve both cardiac and skeletal muscle structure and exercise ability in DMD patients. In particular, CDC-Exos markedly promote myofiber proliferation, myoblast determination and differentiation three weeks after intrasoleus injection, indicative of enhanced muscle regeneration. Meanwhile, the intrasoleus injection of CDC-Exos decreases inflammation and fibrosis, highlighting the therapeutic potential of CDC-Exos for DMD patients ( Aminzadeh et al., 2018).
In brief, a massive number of cell types communicate with skeletal muscle via exosomes, so more attention should be paid to exploring additional types of cells that communicate with skeletal muscle via exosomes.
4. Engineered exosomes in skeletal muscle
Exosomes are widely involved in the physiology and pathology of skeletal muscle and hold great clinical potential in the treatment of skeletal muscle diseases. However, a large number of donors are necessary to induce sufficient effects in vivo due to the low purity and insufficient yield of exosomes ( Ying et al., 2017, 2021). Artificially engineered exosomes possess tissue-specific targeting capability and modified cargos, and thus may be a donor-saving approach for in vivo studies. MiR-26a is required for skeletal muscle differentiation and regeneration ( Dey et al., 2012). Zhang et al. (2019) showed that the intramuscular injection of miR-26a-containing exosomes generated by transfecting the Lamp2b-miR-26a vector into HEK293 cells, ameliorates the muscle wasting of unilateral ureteral obstruction mice by targeting FoxO1. Wang B et al. (2019) generated miR-26a-overexpressing exosomes (Exo/miR-26a) containing a muscle-specific targeting peptide that helps them to be selectively delivered to muscle cells. The intramuscular injection of Exo/miR-26a prevents chronic kidney disease (CKD)-induced muscle wasting in a 5/6 nephrectomized CKD mice model. MiR-215, a tumor inhibitor ( Ge et al., 2016), exhibits protective effects against cancer cachexia-induced muscle atrophy. Zhou Q et al. (2021) developed exosomes overexpressing miR-215 (miR-215-modified exosomes) by transfecting miR-215 mimics into rat bone marrow MSCs (rBMSCs), and the miR-215-modified exosomes reversed the H2O2-induced inhibition of cell viability and increased the apoptosis of L6 cells. In addition to miRNAs, exosomal proteins also exert bioactive effects. Heat shock protein 60 (Hsp60)-bearing exosomes are reported to induce PPARγ coactivator-α1 (PGC-1α) expression ( Barone et al., 2016). di Felice et al. (2022) generated Hsp60-bearing exosomes (with a diameter of 50–140 nm) by genetically modifying C2C12 cell lines with an Hsp60-overexpressing plasmid. The obtained exosomes activated the expression of PGC-1α isoform 1, which is directly involved in mitochondrial biogenesis and muscle atrophy suppression, in C2C12 cell lines. In addition to delivering biomolecules, engineered exosomes could regulate skeletal muscle development as a decoy receptor. Conceição et al. (2021) generated engineered EVs (average diameter of 144 nm, also known as exosomes) to express interleukin 6 (IL6) signal transducer (IL6ST) decoy receptors that selectively inhibit the IL6 trans-signaling pathway in C2C12 myoblasts and myotubes, thereby partially ameliorating the IL6/IL6 receptor (IL6R) complexes-induced anti-differentiation effects. On the whole, the engineered exosomes with modified contents and tissue-specific targeting capability present a tremendous application prospect in the treatment of muscle diseases.
5. Conclusions
Exosomes comprise the most widely investigated subset among the four main subsets of EVs (exosomes, microvesicles, apoptotic body, and oncosome), and their functions are beyond the initial expectations. However, in addition to their poor yield and insufficient purity, there are still a number of concerns regarding exosomes. Firstly, several tissues/organs deserve attention regarding the exosome-mediated regulation of skeletal muscle. For example, the nervous system plays an important role in skeletal muscle development ( Yang XF et al., 2020), while little is known about the exosome-mediated regulatory mechanisms of the nervous system in skeletal muscle. Secondly, gut microbes are critical for skeletal muscle. Choi et al. (2015) reported that stool EVs from high-fat diet (HFD)-fed mice impair glucose metabolism in skeletal muscle, which revealed a subject of intense study by elaborating the unknown effects of gut microbe-derived exosomes on skeletal muscle. Thirdly, the source of exosomes is limited, which indirectly burdens the investigation and clinical application of exosomes. Since many organisms, including archaea ( Evguenieva-Hackenberg et al., 2014), bacteria ( Deng et al., 2015), fungi ( Oliveira et al., 2013), and plant cells ( Xu et al., 2021), secrete exosomes, the comparison results between different sources of exosomes from cells and animals to milk, bacteria, fungi, and plants warrant further investigations. At last, due to its relatively low immune rejection rate compared with cell-based therapy, trials for exosome-based translational medicine could become a hot topic, while the clinical application of engineered exosomes is undeveloped and insufficient. Native exosomes contain a specific combination of proteins, nucleic acids, and lipids, while the engineered exosomes are typically modified for a single bio-molecule, which is considered insufficient and could be a time-wasting operation compared with mimics and recombinant proteins for clinical treatment. The combination of proteins, nucleic acids, lipids, and drugs may be the future of clinical applications. Notably, decoy receptors may be another application strategy.
As comprehensively discussed in this work, advances in the research on exosome-mediated cellular bio-information exchange and reprogramming of skeletal muscle cells have expanded our knowledge. On the one hand, the cargos and circulating levels of exosomes change in response to physiological and pathological stimuli, and these changes in turn affect the physiological and pathological fates of skeletal muscle. On the other hand, engineered exosomes with modified contents and tissue-specific targeting ligands hold tremendous promise for targeted therapy in treating skeletal muscle diseases ( Fig. 2). We believe that those marked advances provide extraordinary opportunities to deepen the understanding and boost the clinical applications of exosomes to tackle skeletal muscle diseases.
Fig. 2. Graphical abstract of potential clinical applications of engineered exosomes. A vector containing a muscle-specific targeting peptide and plasmid/adenovirus containing a precursor gene are co-transfected into donor cells, and then the donor cells are anticipated to secrete engineered exosomes with muscle-specific targeting capability and combined cargos such as proteins, nucleic acids, lipids, and drugs. These engineered exosomes can provide targeted therapy for the treatment of skeletal muscle diseases.
Review criteria
We searched PubMed for original, full-length, English-language articles published within the last five years (2017–2022). The search terms (individually or in combination) included: “extracellular vesicle,” “exosome,” “exosome isolation,” “muscle,” “skeletal muscle,” “myotube,” “C2C12,” “myogenesis,” “muscle atrophy,” and “muscle loss.” We preferred those original articles that report novel discovery in the exosome field and the exosomal function and/or mechanism relevant to skeletal muscle. The professional opinions of each of the authors were considered in the selection of articles relevant to this review, and all listed references were evaluated by at least one of the authors.
Acknowledgments
This work was supported by the National Key Research and Development Program of China (No. 2021YFF1000602). We thank members of the SHI laboratory (Northwest A&F University) for helpful discussions. We thank Dr. Jiangwei WU (Northwest A&F University) for critical reading of the manuscript. We thank Xiuxiu LI (Shandong Agricultural University) and Baojian WU (Shihezi University) for suggestions of the manuscript.
Author contributions
Gongshe YANG and Xin’e SHI outlined the content of the review and made the final corrections for the manuscript. Zhaolu WANG, Jinjin YANG, Xiaohui SUN, and Xi SUN collected the literature and wrote the manuscript. Zhaolu WANG drew the figures. All authors have read and agreed the final manuscript.
Compliance with ethics guidelines
Zhaolu WANG, Jinjin YANG, Xiaohui SUN, Xi SUN, Gongshe YANG, and Xin'e SHI declare that they have no conflict of interest.
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
- Aminzadeh MA, Rogers RG, Fournier M, et al. , 2018. Exosome-mediated benefits of cell therapy in mouse and human models of Duchenne muscular dystrophy. Stem Cell Rep, 10( 3): 942- 955. 10.1016/j.stemcr.2018.01.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arab T, Mallick ER, Huang YY, et al. , 2021. Characterization of extracellular vesicles and synthetic nanoparticles with four orthogonal single-particle analysis platforms. J Extracell Vesicles, 10( 6): e12079. 10.1002/jev2.12079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asare-Werehene M, Nakka K, Reunov A, et al. , 2020. The exosome-mediated autocrine and paracrine actions of plasma gelsolin in ovarian cancer chemoresistance. Oncogene, 39( 7): 1600- 1616. 10.1038/s41388-019-1087-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baracos VE, Martin L, Korc M, et al. , 2018. Cancer-associated cachexia. Nat Rev Dis Primers, 4: 17105. 10.1038/nrdp.2017.105 [DOI] [PubMed] [Google Scholar]
- Barone R, Macaluso F, Sangiorgi C, et al. , 2016. Skeletal muscle Heat shock protein 60 increases after endurance training and induces peroxisome proliferator-activated receptor gamma coactivator 1 α1 expression. Sci Rep, 6: 19781. 10.1038/srep19781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bier A, Berenstein P, Kronfeld N, et al. , 2018. Placenta-derived mesenchymal stromal cells and their exosomes exert therapeutic effects in Duchenne muscular dystrophy. Biomaterials, 174: 67- 78. 10.1016/j.biomaterials.2018.04.055 [DOI] [PubMed] [Google Scholar]
- Billon N, Monteiro MC, Dani C, 2008. Developmental origin of adipocytes: new insights into a pending question. Biol Cell, 100( 10): 563- 575. 10.1042/bc20080011 [DOI] [PubMed] [Google Scholar]
- Buckingham M, Bajard L, Chang T, et al. , 2003. The formation of skeletal muscle: from somite to limb. J Anat, 202( 1): 59- 68. 10.1046/j.1469-7580.2003.00139.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Byun SE, Sim C, Chung Y, et al. , 2021. Skeletal muscle regeneration by the exosomes of adipose tissue-derived mesenchymal stem cells. Curr Issues Mol Biol, 43( 3): 1473- 1488. 10.3390/cimb43030104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavallari C, Ranghino A, Tapparo M, et al. , 2017. Serum-derived extracellular vesicles (EVs) impact on vascular remodeling and prevent muscle damage in acute hind limb ischemia. Sci Rep, 7: 8180. 10.1038/s41598-017-08250-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen CH, Zheng HH, Luo YM, et al. , 2021. SUMOylation promotes extracellular vesicle-mediated transmission of lncRNA ELNAT1 and lymph node metastasis in bladder cancer. J Clin Invest, 131( 8): e146431. 10.1172/jci146431 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen W, Xie MK, Yang B, et al. , 2017. Skeletal myogenic differentiation of human urine-derived cells as a potential source for skeletal muscle regeneration. J Tissue Eng Regen Med, 11( 2): 334- 341. 10.1002/term.1914 [DOI] [PubMed] [Google Scholar]
- Chen YC, Zhu QF, Cheng LM, et al. , 2021. Exosome detection via the ultrafast-isolation system: EXODUS. Nat Methods, 18( 2): 212- 218. 10.1038/s41592-020-01034-x [DOI] [PubMed] [Google Scholar]
- Choi Y, Kwon Y, Kim DK, et al. , 2015. Gut microbe-derived extracellular vesicles induce insulin resistance, thereby impairing glucose metabolism in skeletal muscle. Sci Rep, 5: 15878. 10.1038/srep15878 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conceição M, Forcina L, Wiklander OPB, et al. , 2021. Engineered extracellular vesicle decoy receptor-mediated modulation of the IL6 trans-signalling pathway in muscle. Biomaterials, 266: 120435. 10.1016/j.biomaterials.2020.120435 [DOI] [PubMed] [Google Scholar]
- Deng ZB, Mu JY, Tseng M, et al. , 2015. Enterobacteria-secreted particles induce production of exosome-like S1P-containing particles by intestinal epithelium to drive Th17-mediated tumorigenesis. Nat Commun, 6: 6956. 10.1038/ncomms7956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dey BK, Gagan J, Yan Z, et al. , 2012. miR-26a is required for skeletal muscle differentiation and regeneration in mice. Genes Dev, 26( 19): 2180- 2191. 10.1101/gad.198085.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- di Felice V, Barone R, Trovato E, et al. , 2022. Physiactisome: a new nanovesicle drug containing heat shock protein 60 for treating muscle wasting and cachexia. Cells, 11( 9): 1406 10.3390/cells11091406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evguenieva-Hackenberg E, Hou LL, Glaeser S, et al. , 2014. Structure and function of the archaeal exosome. WIREs RNA, 5( 5): 623- 635. 10.1002/wrna.1234 [DOI] [PubMed] [Google Scholar]
- Fry CS, Kirby TJ, Kosmac K, et al. , 2017. Myogenic progenitor cells control extracellular matrix production by fibroblasts during skeletal muscle hypertrophy. Cell Stem Cell, 20( 1): 56- 69. 10.1016/j.stem.2016.09.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fu XR, Liu G, Halim A, et al. , 2019. Mesenchymal stem cell migration and tissue repair. Cells, 8( 8): 784. 10.3390/cells8080784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao HH, Gao Y, Pang WJ, et al. , 2018. Iridoviral infection can be reduced by UCHL1-loaded exosomes from the testis of Chinese giant salamanders ( Andrias davidianus). Vet Microbiol, 224: 50- 57. 10.1016/j.vetmic.2018.08.025 [DOI] [PubMed] [Google Scholar]
- Gao W, Liu HB, Yuan J, et al. , 2016. Exosomes derived from mature dendritic cells increase endothelial inflammation and atherosclerosis via membrane TNF-α mediated NF-κB pathway. J Cell Mol Med, 20( 12): 2318- 2327. 10.1111/jcmm.12923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge GQ, Zhang W, Niu LG, et al. , 2016. miR-215 functions as a tumor suppressor in epithelial ovarian cancer through regulation of the X-chromosome-linked inhibitor of apoptosis. Oncol Rep, 35( 3): 1816- 1822. 10.3892/or.2015.4482 [DOI] [PubMed] [Google Scholar]
- Gross JC, Chaudhary V, Bartscherer K, et al. , 2012. Active Wnt proteins are secreted on exosomes. Nat Cell Biol, 14( 10): 1036- 1045. 10.1038/ncb2574 [DOI] [PubMed] [Google Scholar]
- Gurunathan S, Kang MH, Kim JH, 2021. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomedicine, 16: 1281- 1312. 10.2147/ijn.S291956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hassanpour Tamrin S, Sanati Nezhad A, Sen A, 2021. Label-free isolation of exosomes using microfluidic technologies. ACS Nano, 15( 11): 17047- 17079. 10.1021/acsnano.1c03469 [DOI] [PubMed] [Google Scholar]
- He Y, Rodrigues RM, Wang XL, et al. , 2021. Neutrophil-to-hepatocyte communication via LDLR-dependent miR-223-enriched extracellular vesicle transfer ameliorates nonalcoholic steatohepatitis. J Clin Invest, 131( 3): e141513. 10.1172/jci141513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu L, Klein JD, Hassounah F, et al. , 2015. Low-frequency electrical stimulation attenuates muscle atrophy in CKD—a potential treatment strategy. J Am Soc Nephrol, 26( 3): 626- 635. 10.1681/asn.2014020144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu WJ, Ru ZY, Zhou YL, et al. , 2019. Lung cancer-derived extracellular vesicles induced myotube atrophy and adipocyte lipolysis via the extracellular IL-6-mediated STAT3 pathway. Biochim Biophys Acta (BBA)-Mol Cell Biol Lipids, 1864( 8): 1091- 1102. 10.1016/j.bbalip.2019.04.006 [DOI] [PubMed] [Google Scholar]
- Huang Y, Yu MS, Kuma A, et al. , 2021. Downregulation of let-7 by electrical acupuncture increases protein synthesis in mice. Front Physiol, 12: 697139. 10.3389/fphys.2021.697139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iyer SR, Scheiber AL, Yarowsky P, et al. , 2020. Exosomes isolated from platelet-rich plasma and mesenchymal stem cells promote recovery of function after muscle injury. Am J Sports Med, 48( 9): 2277- 2286. 10.1177/0363546520926462 [DOI] [PubMed] [Google Scholar]
- Johnstone RM, Adam M, Hammond JR, et al. , 1987. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem, 262( 19): 9412- 9420. 10.1016/S0021-9258(18)48095-7 [DOI] [PubMed] [Google Scholar]
- Kalluri R, LeBleu VS, 2020. The biology, function, and biomedical applications of exosomes. Science, 367( 6478): eaau6977. 10.1126/science.aau6977 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katayama M, Wiklander OPB, Fritz T, et al. , 2019. Circulating exosomal miR-20b-5p is elevated in type 2 diabetes and could impair insulin action in human skeletal muscle. Diabetes, 68( 3): 515- 526. 10.2337/db18-0470 [DOI] [PubMed] [Google Scholar]
- Keller MD, Ching KL, Liang FX, et al. , 2020. Decoy exosomes provide protection against bacterial toxins. Nature, 579( 7798): 260- 264. 10.1038/s41586-020-2066-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S, Lee MJ, Choi JY, et al. , 2018. Roles of exosome-like vesicles released from inflammatory C2C12 myotubes: regulation of myocyte differentiation and myokine expression. Cell Physiol Biochem, 48( 5): 1829- 1842. 10.1159/000492505 [DOI] [PubMed] [Google Scholar]
- la Shu S, Allen CL, Benjamin-Davalos S, et al. , 2021. A rapid exosome isolation using ultrafiltration and size exclusion chromatography (REIUS) method for exosome isolation from melanoma cell lines. In: Hargadon KM (Ed.), Melanoma: Methods and Protocols. Humana, New York, p. 289- 304. 10.1007/978-1-0716-1205-7_22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larsson L, Degens H, Li MS, et al. , 2019. Sarcopenia: aging-related loss of muscle mass and function. Physiol Rev, 99( 1): 427- 511. 10.1152/physrev.00061.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le MN, Fan ZH, 2021. Exosome isolation using nanostructures and microfluidic devices. Biomed Mater, 16( 2): 022005. 10.1088/1748-605X/abde70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leng L, Dong X, Gao XJ, et al. , 2021. Exosome-mediated improvement in membrane integrity and muscle function in dystrophic mice. Mol Ther, 29( 4): 1459- 1470. 10.1016/j.ymthe.2020.12.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li DM, Song HC, Shuo LH, et al. , 2020. Gonadal white adipose tissue-derived exosomal MiR-222 promotes obesity-associated insulin resistance. Aging (Albany NY), 12( 22): 22719- 22743. 10.18632/aging.103891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li K, Wong DK, Hong KY, et al. , 2018. Cushioned-density gradient ultracentrifugation (C-DGUC): a refined and high performance method for the isolation, characterization, and use of exosomes. In: Patel T (Ed.), Extracellular RNA: Methods and Protocols. Humana Press, New York, p. 69- 83. 10.1007/978-1-4939-7652-2_7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M, Zeringer E, Barta T, et al. , 2014. Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos Trans Roy Soc B Biol Sci, 369( 1652): 20130502. 10.1098/rstb.2013.0502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li P, Kaslan M, Lee SH, et al. , 2017. Progress in exosome isolation techniques. Theranostics, 7( 3): 789- 804. 10.7150/thno.18133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li XX, Wang SH, Zhu RJ, et al. , 2016. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J Hematol Oncol, 9: 42. 10.1186/s13045-016-0269-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li ZY, Liu C, Li SL, et al. , 2021. BMSC-derived exosomes inhibit dexamethasone-induced muscle atrophy via the miR-486-5p/FoxO1 axis. Front Endocrinol (Lausanne), 12: 681267. https://doi.org/ 10.3389/fendo.2021.681267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu T, Sun YC, Chen Pg et al. , 2019. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochem Biophys Res Commun, 515( 2): 352- 358. 10.1016/j.bbrc.2019.05.113 [DOI] [PubMed] [Google Scholar]
- Manterola L, Guruceaga E, Pérez-Larraya JG, et al. , 2014. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro-Oncology, 16( 4): 520- 527. 10.1093/neuonc/not218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miao CX, Zhang WL, Feng LX, et al. , 2021. Cancer-derived exosome miRNAs induce skeletal muscle wasting by Bcl-2-mediated apoptosis in colon cancer cachexia. Mol Ther Nucleic Acids, 24: 923- 938. 10.1016/j.omtn.2021.04.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minciacchi VR, You S, Spinelli C, et al. , 2015. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget, 6( 13): 11327- 11341. 10.18632/oncotarget.3598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monguió-Tortajada M, Morón-Font M, Gámez-Valero A, et al. , 2019. Extracellular-vesicle isolation from different biological fluids by size-exclusion chromatography. Curr Protoc Stem Cell Biol, 49: e82. 10.1002/cpsc.82 [DOI] [PubMed] [Google Scholar]
- Mytidou C, Koutsoulidou A, Katsioloudi A, et al. , 2021. Muscle-derived exosomes encapsulate myomiRs and are involved in local skeletal muscle tissue communication. FASEB J, 35( 2): e21279. 10.1096/fj.201902468RR [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair S, Jayabalan N, Guanzon D, et al. , 2018. Human placental exosomes in gestational diabetes mellitus carry a specific set of miRNAs associated with skeletal muscle insulin sensitivity. Clin Sci (Lond), 132( 22): 2451- 2467. 10.1042/cs20180487 [DOI] [PubMed] [Google Scholar]
- Nair VD, Ge YC, Li SD, et al. , 2020. Sedentary and trained older men have distinct circulating exosomal microRNA profiles at baseline and in response to acute exercise. Front Physiol, 11: 605. 10.3389/fphys.2020.00605 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliveira DL, Rizzo J, Joffe LS, et al. , 2013. Where do they come from and where do they go: candidates for regulating extracellular vesicle formation in fungi. Int J Mol Sci, 14( 5): 9581- 9603. 10.3390/ijms14059581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan BT, Johnstone RM, 1983. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell, 33( 3): 967- 978. 10.1016/0092-8674(83)90040-5 [DOI] [PubMed] [Google Scholar]
- Qiu L, Liu Z, Wu CZ, et al. , 2020a. C6-ceramide induces salivary adenoid cystic carcinoma cell apoptosis via IP3R-activated UPR and UPR-independent pathways. Biochem Biophys Res Commun, 525( 4): 997- 1003. 10.1016/j.bbrc.2020.02.164 [DOI] [PubMed] [Google Scholar]
- Qiu L, Chen W, Wu CZ, et al. , 2020b. Exosomes of oral squamous cell carcinoma cells containing miR-181a-3p induce muscle cell atrophy and apoptosis by transmissible endoplasmic reticulum stress signaling. Biochem Biophys Res Commun, 533( 4): 831- 837. 10.1016/j.bbrc.2020.09.066 [DOI] [PubMed] [Google Scholar]
- Rojas EA, Corchete LA, San-Segundo L, et al. , 2017. Amiloride, an old diuretic drug, is a potential therapeutic agent for multiple myeloma. Clin Cancer Res, 23( 21): 6602- 6615. 10.1158/1078-0432.Ccr-17-0678 [DOI] [PubMed] [Google Scholar]
- Sahu A, Clemens ZJ, Shinde SN, et al. , 2021. Regulation of aged skeletal muscle regeneration by circulating extracellular vesicles. Nat Aging, 1( 12): 1148- 1161. 10.1038/s43587-021-00143-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samaeekia R, Rabiee B, Putra I, et al. , 2018. Effect of human corneal mesenchymal stromal cell-derived exosomes on corneal epithelial wound healing. Invest Ophthalmol Vis Sci, 59( 12): 5194- 5200. 10.1167/iovs.18-24803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt SF, Rohm M, Herzig S, et al. , 2018. Cancer cachexia: more than skeletal muscle wasting. Trends Cancer, 4( 12): 849- 860. 10.1016/j.trecan.2018.10.001 [DOI] [PubMed] [Google Scholar]
- Schöneich C, Dremina E, Galeva N, et al. , 2014. Apoptosis in differentiating C2C12 muscle cells selectively targets Bcl-2-deficient myotubes. Apoptosis, 19( 1): 42- 57. 10.1007/s10495-013-0922-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma S, LeClaire M, Gimzewski JK, 2018. Ascent of atomic force microscopy as a nanoanalytical tool for exosomes and other extracellular vesicles. Nanotechnology, 29( 13): 132001. 10.1088/1361-6528/aaab06 [DOI] [PubMed] [Google Scholar]
- Steinacker JM, Opitz-Gress A, Baur S, et al. , 2000. Expression of myosin heavy chain isoforms in skeletal muscle of patients with peripheral arterial occlusive disease. J Vasc Surg, 31( 3): 443- 449. 10.1067/mva.2000.102848 [DOI] [PubMed] [Google Scholar]
- Sun YX, Shi H, Yin SQ, et al. , 2018. Human mesenchymal stem cell derived exosomes alleviate type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano, 12( 8): 7613- 7628. 10.1021/acsnano.7b07643 [DOI] [PubMed] [Google Scholar]
- Tavakoli Dargani Z, Singla R, Johnson T, et al. , 2018. Exosomes derived from embryonic stem cells inhibit doxorubicin and inflammation-induced pyroptosis in muscle cells. Can J Physiol Pharmacol, 96( 3): 304- 307. 10.1139/cjpp-2017-0340 [DOI] [PubMed] [Google Scholar]
- Théry C, Witwer KW, Aikawa E, 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. 10.1080/20013078.2018.1535750 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torralba D, Baixauli F, Villarroya-Beltri C, et al. , 2018. Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts. Nat Commun, 9: 2658. 10.1038/s41467-018-05077-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vassilakos G, Lei HQ, Yang Y, et al. , 2019. Deletion of muscle IGF-I transiently impairs growth and progressively disrupts glucose homeostasis in male mice. FASEB J, 33( 1): 181- 194. 10.1096/fj.201800459R [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang B, Zhang AQ, Wang HD, et al. , 2019. miR- 26a limits muscle wasting and cardiac fibrosis through exosome-mediated microRNA transfer in chronic kidney disease. Theranostics, 9( 7): 1864- 1877. 10.7150/thno.29579 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang CY, Song W, Chen B, et al. , 2019. Exosomes isolated from adipose-derived stem cells: a new cell-free approach to prevent the muscle degeneration associated with torn rotator cuffs. Am J Sports Med, 47( 13): 3247- 3255. 10.1177/0363546519876323 [DOI] [PubMed] [Google Scholar]
- Wang DX, Zhang XH, Li YB, et al. , 2022. Exercise-induced browning of white adipose tissue and improving skeletal muscle insulin sensitivity in obese/non-obese growing mice: do not neglect exosomal miR-27a. Front Nutr, 9: 940673. 10.3389/fnut.2022.940673 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang SW, Cesca F, Loers G, et al. , 2011. Synapsin I is an oligomannose-carrying glycoprotein, acts as an oligomannose-binding lectin, and promotes neurite outgrowth and neuronal survival when released via glia-derived exosomes. J Neurosci, 31( 20): 7275- 7290. 10.1523/jneurosci.6476-10.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei MY, Gao XT, Liu LJ, et al. , 2020. Visceral adipose tissue derived exosomes exacerbate colitis severity via pro-inflammatory miRNAs in high fat diet fed mice. ACS Nano, 14( 4): 5099- 5110. 10.1021/acsnano.0c01860 [DOI] [PubMed] [Google Scholar]
- Wu CH, Silvers CR, Messing EM, et al. , 2019. Bladder cancer extracellular vesicles drive tumorigenesis by inducing the unfolded protein response in endoplasmic reticulum of nonmalignant cells. J Biol Chem, 294( 9): 3207- 3218. 10.1074/jbc.RA118.006682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu HZ, Ballantyne CM, 2017. Skeletal muscle inflammation and insulin resistance in obesity. J Clin Invest, 127( 1): 43- 54. 10.1172/jci88880 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu PP, Zhang B, Shi H, et al. , 2018. MSC-exosome: a novel cell-free therapy for cutaneous regeneration. Cytotherapy, 20( 3): 291- 301. 10.1016/j.jcyt.2017.11.002 [DOI] [PubMed] [Google Scholar]
- Wu RY, Huang CS, Wu QK, et al. , 2019. Exosomes secreted by urine-derived stem cells improve stress urinary incontinence by promoting repair of pubococcygeus muscle injury in rats. Stem Cell Res Ther, 10: 80. 10.1186/s13287-019-1182-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu XH, Yuan TJ, Dad HA, et al. , 2021. Plant exosomes as novel nanoplatforms for microRNA transfer stimulate neural differentiation of stem cells in vitro and in vivo. Nano Lett, 21( 19): 8151- 8159. 10.1021/acs.nanolett.1c02530 [DOI] [PubMed] [Google Scholar]
- Xu YT, Zhang L, Ocansey DKW, et al. , 2022. HucMSC-Ex alleviates inflammatory bowel disease via the lnc78583-mediated miR3202/HOXB13 pathway. J Zhejiang Univ-Sci B (Biomed & Biotechnol), 23( 5): 423- 431. 10.1631/jzus.B2100793 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan B, Zhang Y, Liang C, et al. , 2020. Stem cell-derived exosomes prevent pyroptosis and repair ischemic muscle injury through a novel exosome/circHIPK3/FOXO3a pathway. Theranostics, 10( 15): 6728- 6742. 10.7150/thno.42259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang DB, Zhang WH, Zhang HY, et al. , 2020. Progress, opportunity, and perspective on exosome isolation-efforts for efficient exosome-based theranostics. Theranostics, 10( 8): 3684- 3707. 10.7150/thno.41580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang XF, Xue PP, Chen HR, et al. , 2020. Denervation drives skeletal muscle atrophy and induces mitochondrial dysfunction, mitophagy and apoptosis via miR-142a-5p/MFN1 axis. Theranostics, 10( 3): 1415- 1432. 10.7150/thno.40857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yi SL, Liu XJ, Zhong JQ, et al. , 2014. Role of caveolin-1 in atrial fibrillation as an anti-fibrotic signaling molecule in human atrial fibroblasts. PLoS ONE, 9( 1): e85144. 10.1371/journal.pone.0085144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ying W, Riopel M, Bandyopadhyay G, et al. , 2017. Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell, 171( 2): 372- 384.e12. 10.1016/j.cell.2017.08.035 [DOI] [PubMed] [Google Scholar]
- Ying W, Gao H, Dos Reis FCG, et al. , 2021. MiR-690, an exosomal-derived miRNA from M2-polarized macrophages, improves insulin sensitivity in obese mice. Cell Metab, 33( 4): 781- 790.e5. 10.1016/j.cmet.2020.12.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu Y, Du HW, Wei SN, et al. , 2018. Adipocyte-derived exosomal miR-27a induces insulin resistance in skeletal muscle through repression of PPARγ. Theranostics, 8( 8): 2171- 2188. 10.7150/thno.22565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zanotti S, Gibertini S, Blasevich F, et al. , 2018. Exosomes and exosomal miRNAs from muscle-derived fibroblasts promote skeletal muscle fibrosis. Matrix Biol, 74: 77- 100. 10.1016/j.matbio.2018.07.003 [DOI] [PubMed] [Google Scholar]
- Zeschnigk M, Kozian D, Kuch C, et al. , 1995. Involvement of M-cadherin in terminal differentiation of skeletal muscle cells. J Cell Sci, 108( 9): 2973- 2981. 10.1242/jcs.108.9.2973 [DOI] [PubMed] [Google Scholar]
- Zhang AQ, Wang HD, Wang B, et al. , 2019. Exogenous miR-26a suppresses muscle wasting and renal fibrosis in obstructive kidney disease. FASEB J, 33( 12): 13590- 13601. 10.1096/fj.201900884R [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang HY, Lyden D, 2019. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat Protoc, 14( 4): 1027- 1053. 10.1038/s41596-019-0126-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang HY, Freitas D, Kim HS, et al. , 2018. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat Cell Biol, 20( 3): 332- 343. 10.1038/s41556-018-0040-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang WL, Sun WK, Gu XF, et al. , 2022. GDF-15 in tumor-derived exosomes promotes muscle atrophy via Bcl-2/caspase-3 pathway. Cell Death Discov, 8: 162. 10.1038/s41420-022-00972-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao L, Yu J, Wang J, et al. , 2017. Isolation and identification of miRNAs in exosomes derived from serum of colon cancer patients. J Cancer, 8( 7): 1145- 1152. 10.7150/jca.18026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou L, Zhang T, Shao W, et al. , 2021. Amiloride ameliorates muscle wasting in cancer cachexia through inhibiting tumor-derived exosome release. Skelet Muscle, 11: 17. 10.1186/s13395-021-00274-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Q, Yang HC, Pan HC, et al. , 2021. Exosomes isolated from the miR-215-modified bone marrow mesenchymal stem cells protect H2O2-induced rat myoblasts via the miR-215/FABP3 pathway. Exp Mol Pathol, 119: 104608. 10.1016/j.yexmp.2021.104608 [DOI] [PubMed] [Google Scholar]
- Zhu QW, Li Q, Niu X, et al. , 2018. Extracellular vesicles secreted by human urine-derived stem cells promote ischemia repair in a mouse model of hind-limb ischemia. Cell Physiol Biochem, 47( 3): 1181- 1192. 10.1159/000490214 [DOI] [PubMed] [Google Scholar]