Anat Sci Int (2015) 90:33–39 DOI 10.1007/s12565-014-0266-6

REVIEW ARTICLE

Organization of organelles and VAMP-associated vesicular transport systems in differentiating skeletal muscle cells Yuki Tajika • Maiko Takahashi • Hitoshi Ueno Tohru Murakami • Hiroshi Yorifuji



Received: 24 August 2014 / Accepted: 19 November 2014 / Published online: 5 December 2014 Ó Japanese Association of Anatomists 2014

Abstract Vesicular transport plays an important role in the regulation of cellular function and differentiation of the cell, and intracellular vesicles play a role in the delivery of membrane components and in sorting membrane proteins to appropriate domains in organelles and the plasma membrane. Research on vesicular transport in differentiating cells has mostly focused on neurons and epithelial cells, and few such studies have been carried out on skeletal muscle cells. Skeletal muscle cells have specialized organelles and plasma membrane domains, including T-tubules, sarcoplasmic reticulum, neuromuscular junctions, and myotendinous junctions. The differentiation of skeletal muscle cells is achieved by multiple steps, i.e., proliferation of myoblasts, formation of myotubes by cell–cell fusion, and maturation of myotubes into myofibers. Systematic vesicular transport is expected to play a role in the maintenance and development of skeletal muscle cells. Here, we review a map of the vesicular transport system during the differentiation of skeletal muscle cells. The characteristics of organelle arrangement in myotubes are described according to morphological studies. Vesicular transport in myotubes is explained by the expression profiles of soluble Nethylmaleimide-sensitive factor attachment protein receptor proteins. Keywords Muscle  Myoblast  Myotube  SNARE protein  VAMP

Y. Tajika (&)  M. Takahashi  H. Ueno  T. Murakami  H. Yorifuji Department of Anatomy, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan e-mail: [email protected] H. Yorifuji e-mail: [email protected]

Introduction Skeletal muscle cells are multinuclear cells with a very long cylindrical appearance, with the longest human skeletal muscle cells having a length of up to 14 cm (Paul 2001). Even the smallest human skeletal muscle cells found in the stapedius muscle are 2 mm in length, which is 100-fold longer than the average mononucleated cell (10–20 lm) (Grounds and Shavlakadze 2011). Skeletal muscle cells have contraction ability and are equipped with a specialized plasma membrane and intracellular structures, such as neuromuscular junctions, myotendinous or myo–myo junctions, myofibrils, T-system, and sarcoplasmic reticulum (SR). These specialized structures of skeletal muscle cells develop in a multiple-step process that consists of (1) the proliferation of mononuclear myoblasts, (2) the fusion of myoblasts to form multinuclear myotubes, and (3) the maturation of myotubes. Systematic vesicular transport is expected to play a role in the differentiation of skeletal muscle cells from myoblasts through to myotubes and for the maintenance of the mature skeletal muscle cells (Towler et al. 2004). However, most of the research on vesicular transport in differentiating cells has been performed on neurons and epithelial cells, and few such studies have focused on skeletal muscle cells. Here, we review recent information on the distribution of organelles and the vesicular transport system in myotubes.

Organelles in myoblasts and myotubes In the early 1960s satellite cells were identified as resident myoblasts in adult muscular tissue (Mauro 1961), and subsequent ultrastructural studies in the 1960s and 1970s described the morphological characteristics of these

123

34

satellite cells. Satellite cells in adult animals are less active mitotically and metabolically than those in juvenile animals based on their characteristics of heterochromatic nuclei, poorly developed Golgi apparatus, and low levels of glycogen (Snow 1977). The satellite cells of developing muscle (Ishikawa 1966) and young animals (Snow 1977) are, in contrast, more active based on the presence of euchromatic nuclei and a well-developed rough endoplasmic reticulum (ER). Satellite cells have caveolae and pinocytotic vesicles in their cytoplasm (Ishikawa 1966; Muir et al. 1965). The satellite cells are considered to be myogenic stem cells, and activated satellite cells, referred to as myoblasts, initiate the muscle differentiation process. Research on the differentiation of skeletal muscle cells shifted from morphological studies to molecular biology studies in the 1990s, with the latter revealing a number of transcription factors (Buckingham and Relaix 2007) and adhesion molecules (Krauss 2010) which regulate the differentiation of skeletal muscle cells. However, to date, no serious effort has been made to determine the intracellular features of myotubes. The myotubes, multinucleated muscle fibers that form from the fusion of myoblasts, are longer than typical mononuclear cells, usually growing to a length of 0.1–0.6 mm even in culture (Burattini et al. 2004). In a recent electron microscopy study, we examined the organelles in whole myotubes as a basis for studying membrane traffic in myotubes (Tajika et al. 2014). We found that the perinuclear region of the myotubes contained small-sized Golgi apparatuses and that autolysosomes and mitochondria were abundant in the perinuclear region. The rough ER extended from the perinuclear region toward the periphery of the myotube while the smooth ER was located further from the nuclei. A large number of vesicles were located near the tip of the myotubes. Vesicles were observed in both satellite cells (myoblasts) and myotubes. These vesicles are expected to play a role in the intracellular vesicular transport system for delivery of membrane components and including proteins to the appropriate organelles. To identify the nature of these vesicles in the vesicular transport system, in this review we focused on soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and describe their protein expression profiles in myoblasts and myotubes.

SNARE proteins Vesicular transport consists of multiple steps regulated by a number of molecules. In this multi-step process, coat proteins help to generate transport vesicles through vesicle

123

Y. Tajika et al.

budding from the membrane (Cai et al. 2007), motor proteins move vesicles along the cytoskeleton (Hirokawa et al. 2010), small GTPases tether the vesicles at the specific membrane compartment (Mizuno-Yamasaki et al. 2012), and SNARE proteins tether and fuse the vesicle to the target membrane (Hong 2005). Small GTPases are recruited from the cytoplasm to the target membrane depending on the GTPase cycle, whereas SNARE proteins are anchored to the membrane. Thus, we have focused on SNARE proteins to represent the nature of the vesicles in myoblasts and myotubes. More than 30 SNARE proteins have been identified in mammals. SNARE proteins are functionally categorized into vesicle-anchored SNAREs (v-SNAREs) and target membrane-anchored SNAREs (t-SNAREs). Each SNARE molecule has one or two coiled–coil domains (the SNARE motif). v-SNARE and t-SNARE assemble into a SNARE complex through the interaction of the SNARE motif, resulting in membrane fusion between vesicles and the target membrane. Restricted intracellular localization of each SNARE and specific combinations of SNAREs to form the SNARE complex ensure the direction of transport vesicles and specify the target membrane site. VAMP family proteins in skeletal muscle Seven v-SNAREs, i.e., VAMPs (vesicle-associated membrane proteins), are believed to reside on transport vesicles (Hong 2005). VAMPs are distributed in post-Golgi transport vesicles in a restricted manner (Table 1). Here, we describe the expression profiles of the VAMPs using data from mouse and rat models to compose a map of postGolgi transport systems within myotubes.

VAMP1/synaptobrevin VAMP1 was originally isolated from synaptic vesicles, leading to it also being called synaptobrevin. VAMP1 mediates exocytosis, i.e., neurotransmitter release (Baumert et al. 1989; Trimble et al. 1988). VAMP1-null mice die by postnatal day 15 due to neurological defects (Nystuen et al. 2006). Analyses using VAMP1-heterozygous mice revealed that VAMP1 plays an essential, non-redundant role in Ca2?-triggered vesicle exocytosis at mouse neuromuscular junctions (Liu et al. 2011). In addition to being present in the neuronal system, VAMP1 has been detected in the pancreas, kidney, and cardiac myocytes (Ferlito et al. 2010; Peters et al. 2006; Rossetto et al. 1996). VAMP1 has been found at the nerve terminals in skeletal muscles, but not in skeletal muscle cells (Tajika et al. 2007) or in developing myotubes (Tajika et al. 2014).

Vesicular transport in myotubes

35

Table 1 Expression of vesicle-associated membrane proteins during muscle differentiation VAMPsa

Intracellular localizations in non-muscle cells

VAMP expression during muscle differentiation Satellite cell (quiescent)

Myoblasts (proliferating)

Myotubes

Skeletal muscle cells (matured)

VAMP1

SV

-

-

-

-

VAMP2

SV, etc.

?

-

?

?

VAMP3 VAMP4

EE, RE TGN

NR

? ?

? ?

? NR

VAMP5

PM

-

?

?

?

VAMP7

LE, Ly, PM

NR

?

-

-

VAMP8

EE, LE

NR

?

?

NR

SV Synaptic vesicles, EE early endosome, RE recycling endosomes, TGN trans-Golgi network, PM plasma membrane, LE late endosome, Ly lysosome, NR not reported a

VAMPs, Family of vesicle-associated membrane proteins belonging to the category of v-SNAREs (N-ethylmaleimide-sensitive factor attachment protein receptors). For details on each VAMP described in Table 1, see section VAMP family proteins in skeletal muscle

VAMP2/synaptobrevin-2 VAMP2 was originally isolated from synaptic vesicles, leading to it being referred to as synaptobrevin-2 (Baumert et al. 1989; Trimble et al. 1988). In neurons, VAMP2 plays critical roles in Ca2?-triggered neurotransmitter release (Schoch et al. 2001). VAMP2 has also been detected in non-neuronal tissues, i.e., the kidney (Mendez et al. 2011; Procino et al. 2008), lung (Wang et al. 2012), pancreas (Regazzi et al. 1995; Weng et al. 2007), stomach (Karvar 2002), lymphocytes (Matti et al. 2013), adipose tissue (Martin et al. 1998; Zhao et al. 2009), and skeletal muscle (Rose et al. 2009). In the skeletal muscle, VAMP2 is preferentially expressed in the slow-twitch fibers and is localized mainly at the perinuclear cytoplasm (Tajika et al. 2008). It is also found in the cytoplasm of quiescent satellite cells (Fig. 1a) (Tajika et al. 2008), but disappears in the proliferating myoblasts during the muscle differentiation process (Tajika et al. 2014). In the myotubes, VAMP2 has been found to be distributed abundantly near the tip of the cell (Fig. 1b) (Tajika et al. 2010) . Knockdown of VAMP2 was performed using C2C12 culture myotubes, but it did not affect the fusion of myoblasts or the expression level of desmin, a muscle differentiation marker protein. Rudich and Klip (2003) proposed that VAMP2 is involved in the translocation of glucose transporter-4 (GLUT4) in mature skeletal muscle cells. However, to date, the functions of VAMP2 in satellite cells and myotubes remain to be elucidated. VAMP3/cellubrevin VAMP3/cellubrevin is expressed in a variety of tissues and plays a role in endocytosis in various types of cells (McMahon et al. 1993). VAMP3 protein was originally

detected in the skeletal muscle (Volchuk et al. 1994), and immunohistological analysis showed that VAMP3 is expressed in the capillaries. Skeletal muscle cells express VAMP3 at a low level (Tajika et al. 2007). During muscle differentiation, VAMP3 is found in both myoblasts and myotubes (Tajika et al. 2014). In myotubes VAMP3 is localized at the perinuclear region and the tips of protrusions in myoblasts, and in myotubes, it is localized mainly in the periphery of the cell and is colocalized with VAMP2. VAMP4 VAMP4 has been shown to regulate trafficking events in the trans-Golgi network (TGN) (Steegmaier et al. 1999). VAMP4 transcripts were detected in various tissues (Advani et al. 1998), but neither VAMP4 transcripts nor proteins have yet been reported in mature skeletal muscle. During the muscle differentiation process, VAMP4 is localized in the perinuclear region in both myoblasts and myotubes (Tajika et al. 2014). The Golgi apparatus in myotubes is fragmented and also localized in the perinuclear region (Lu et al. 2001; Tajika et al. 2014). The TGN in myotubes is localized in the vicinity of fragmented Golgi apparatus (Tajika et al. 2010). Thus, perinuclear VAMP4 is an indicator of this localization of the Golgi apparatus, especially the TGN, in myoblasts and myotubes. VAMP5 VAMP5 was originally cloned as a muscle-specific VAMP isoform and is believed to regulate the trafficking events between the cytoplasm and the sarcolemma (Takahashi et al. 2013; Zeng et al. 1998). VAMP5 protein has been found in various tissues except the brain (Schwenk et al. 2010; Takahashi et al. 2013). In the skeletal muscle, VAMP5 is

123

36

Y. Tajika et al.

Fig. 1 a Localization of vesicle-associated membrane protein 2 (VAMP2) in isolated skeletal muscle cells. Skeletal muscle cells were isolated from extensor digitorum longus muscles of 4-week-old mice by collagenase digestion for 2 h. Fixed skeletal muscle cells were immunolabeled for VAMP2 (green) and Pax3/7 (red). Nuclei were stained with DAPI (blue). VAMP2 was detected mainly around the nuclei of skeletal muscle cells and Pax3/7positive satellite cells (arrow). b Localization of VAMP2 and VAMP5 in differentiating myotubes. C2C12 myoblasts were differentiated into multinuclear myotubes and labeled for VAMP2 (green), VAMP5 (red), and nuclei (blue). Both VAMP2 and VAMP5 were detected in the cytoplasm of myotubes, localized mainly near the tips of the myotubes. Images were obtained by confocal microscopy. Bars 20 lm. (Color figure online)

preferentially expressed in fast-twitch (type IIa) fibers (Takahashi et al. 2013). During the muscle differentiation process, VAMP5 is found in myotubes (Fig. 1b) and is localized mainly at the periphery of the cell. VAMP2 and VAMP3 show similar localization at the periphery of myotubes, but their localization is different from that of VAMP5, which is closer to the plasma membrane (Tajika et al. 2014). The precise roles of VAMP5 remain to be elucidated. Knockdown of VAMP5 using C2C12 myotubes has been found to have no effect on the fusion of myoblasts or the expression level of desmin, a muscle differentiation marker protein, nor on muscle differentiation (Takahashi et al. 2013). VAMP7/TI-VAMP VAMP7/TI-VAMP is generally known to regulate vesicular transport to late endosomes and lysosomes (Advani

123

et al. 1999). VAMP7 is also distributed in various organelles, such as the Golgi apparatus, TGN, and early endosomes, depending on the cell type (Chaineau et al. 2009). VAMP7 protein is not expressed in mature skeletal muscle (Advani et al. 1999; Sato et al. 2011), but it is expressed in myoblasts, with its expression decreasing as muscle differentiation progresses (Tajika et al. 2014). VAMP8/endobrevin VAMP8/endobrevin has been shown to be distributed in the endosomal systems (Wang et al. 2010; Wong et al. 1998), but its transcripts or proteins have not been reported in mature skeletal muscle. During the process of muscle differentiation, VAMP8 is found in myoblasts as endosome-like dots (Tajika et al. 2014), with subsequent diffuse localization in myotubes. VAMP8 and VAMP7 are both

Vesicular transport in myotubes

37

Fig. 2 Organelles and VAMPs in myotubes. To show the organelles and VAMPs clearly, we omitted differentiating myofibrils in this figure. Mitochondria and autolysosomes are abundant around the center of the cell. The Golgi apparatus and trans-Golgi network labeled with VAMP4 are localized in the perinuclear region. The

endoplasmic reticulum (ER) expands from the perinuclear region toward the periphery. VAMP2, 3, and 5 are localized at the periphery of the myotubes, with VAMP2 and 3 localized similarly and VAMP5 localized closer to the sarcolemma. Portions of VAMP3 and 5 are localized at the developing T-tubules

involved in the fusion of lysosomes to autophagosomes (Furuta et al. 2010). Autolysosomes are abundant in myotubes, but they are not associated with VAMP7 or VAMP8 (Tajika et al. 2014).

Rab13 are involved in insulin-dependent GLUT4 translocation in skeletal muscle cells (Klip et al. 2014). However, the interactions between VAMPs and other molecules have not been identified in myoblasts or myotubes. Future studies on the molecular interactions and molecular dynamics of VAMPs will clarify the route and target of the vesicles in myotubes. Further studies are required to address how the VAMPbearing vesicles contribute to the differentiation of skeletal muscle cells. We observed that VAMP3 and VAMP5 are colocalized with caveolin-3, a marker of developing T-tubules, leading us to suggest that VAMP3 and VAMP5 may be involved in T-tubule development (Tajika et al. 2014). In addition to T-tubules, characteristic structures, such as neuromuscular junctions (Sanes and Lichtman 2001) and myotendinous junctions (Burkin and Kaufman 1999), should be investigated in future studies, although many difficulties are encountered when attempting to reproduce a whole differentiation process of the skeletal muscle in experimental model systems. Myoblasts in culture are available as cell lines (C2C12, BC3H1, and MM14 from mouse and L6 from rat) or can be isolated from rodent and avian muscle tissues (Neville et al. 1997). These cells do not differentiate to show the full contractile function and the generation of different fiber types. Animal models can be used to study muscle differentiation and do show the terminal differentiation, but it is hard to analyze the time-dependent changes of a single skeletal muscle cell. Research on vesicular transport in myotubes has just marked its beginning. Further expansion of this field will yield insight into not only the nature of skeletal muscle cells but also muscular diseases.

Discussion Expression profiles of the VAMPs during muscle differentiation are summarized in Table 1. Proliferating myoblasts (C2C12 mouse myoblast and/or primary culture myoblasts) express VAMP3, 4, and 8. VAMP3 and 8 may play roles in endosomal trafficking, and VAMP4 may be involved in TGN trafficking. Myotubes express VAMP2, 3, 4, 5, and 8 (Fig. 2). VAMP2, 3, and 5 may regulate endosomal trafficking at the periphery of the myotubes, with VAMP2 and VAMP3 showing similar localization and VAMP5 localized solely near the plasma membrane. These observations may reflect the multiple steps of endosomal trafficking between the cytoplasm and plasma membrane. VAMP4 in myotubes is involved in TGN trafficking at the perinuclear region, while VAMP8 in myotubes shows diffuse signals and therefore may not play significant roles in myotubes. These data were obtained by static morphological analyses and, therefore, are not supported by information on time-dependent changes in the localization of VAMPs or the molecular interactions of VAMPs with other molecules. VAMPs interact with t-SNAREs to form the SNARE complex and function to tether and fuse vesicles to the target membrane (Hong 2005). The formation of the SNARE complex is also regulated by other molecules, such as small GTPases (Grosshans et al. 2006). Only a few studies have reported the expression and the distribution of small GTPases in differentiating skeletal muscle cells. ADP ribosylation factor (ARF) 1 regulates vesicle budding from the ER to the SR (Nori et al. 2004). ARF6 is distributed at the plasma membrane and influences myoblast fusion (Bach et al. 2010; Chen et al. 2003). Rab8A and

Acknowledgments We thank Ms. Harumi Matsuda and Mr. Yoshihiro Morimura (Department of Anatomy, Gunma University Graduate School of Medicine) for secretarial assistance. This work was supported by MEXT KAKENHI Grant Number 25860138. Conflict of interest

None.

123

38

References Advani RJ, Bae HR, Bock JB et al (1998) Seven novel mammalian SNARE proteins localize to distinct membrane compartments. J Biol Chem 273:10317–10324 Advani RJ, Yang B, Prekeris R et al (1999) VAMP-7 mediates vesicular transport from endosomes to lysosomes. J Cell Biol 146:765–776 Bach A-S, Enjalbert S, Comunale F et al (2010) ADP-ribosylation factor 6 regulates mammalian myoblast fusion through phospholipase D1 and phosphatidylinositol 4,5-bisphosphate signaling pathways. Mol Biol Cell 21:2412–2424. doi:10.1091/mbc. E09-12-1063 Baumert M, Maycox PR, Navone F et al (1989) Synaptobrevin: an integral membrane protein of 18,000 Daltons present in small synaptic vesicles of rat brain. EMBO J 8:379–384 Buckingham M, Relaix F (2007) The role of Pax genes in the development of tissues and organs: pax3 and Pax7 regulate muscle progenitor cell functions. Annu Rev Cell Dev Biol 23:645–673. doi:10.1146/annurev.cellbio.23.090506.123438 Burattini S, Ferri P, Battistelli M et al (2004) C2C12 murine myoblasts as a model of skeletal muscle development: morphofunctional characterization. Eur J Histochem 48:223–233 Burkin DJ, Kaufman SJ (1999) The alpha7beta1 integrin in muscle development and disease. Cell Tissue Res 296:183–190 Cai H, Reinisch K, Ferro-Novick S (2007) Coats, tethers, Rabs, and SNAREs work together to mediate the intracellular destination of a transport vesicle. Dev Cell 12:671–682. doi:10.1016/j. devcel.2007.04.005 Chaineau M, Danglot L, Galli T (2009) Multiple roles of the vesicular-SNARE TI-VAMP in post-Golgi and endosomal trafficking. FEBS Lett 583:3817–3826. doi:10.1016/j.febslet. 2009.10.026 Chen EH, Pryce BA, Tzeng JA et al (2003) Control of myoblast fusion by a guanine nucleotide exchange factor, loner, and its effector ARF6. Cell 114:751–762 Ferlito M, Fulton WB, Zauher MA et al (2010) VAMP-1, VAMP-2, and syntaxin-4 regulate ANP release from cardiac myocytes. J Mol Cell Cardiol 49:791–800. doi:10.1016/j.yjmcc.2010.08. 020 Furuta N, Fujita N, Noda T et al (2010) Combinational soluble Nethylmaleimide-sensitive factor attachment protein receptor proteins VAMP8 and Vti1b mediate fusion of antimicrobial and canonical autophagosomes with lysosomes. Mol Biol Cell 21:1001–1010. doi:10.1091/mbc.E09-08-0693 Grosshans BL, Ortiz D, Novick P (2006) Rabs and their effectors: achieving specificity in membrane traffic. Proc Natl Acad Sci USA 103:11821–11827. doi:10.1073/pnas.0601617103 Grounds MD, Shavlakadze T (2011) Growing muscle has different sarcolemmal properties from adult muscle: a proposal with scientific and clinical implications. BioEssays 33:458–468. doi:10.1002/bies.201000136 Hirokawa N, Niwa S, Tanaka Y (2010) Molecular motors in neurons: transport mechanisms and roles in brain function, development, and disease. Neuron 68:610–638. doi:10.1016/j.neuron.2010.09. 039 Hong W (2005) SNAREs and traffic. Biochim Biophys Acta 1744:120–144. doi:10.1016/j.bbamcr.2005.03.014 Ishikawa H (1966) Electron microscopic observations of satellite cells with special reference to the development of mammalian skeletal muscles. Z Anat Entwicklungsgesch 125:43–63 Karvar S (2002) Localization and function of soluble N-ethylmaleimide-sensitive factor attachment protein-25 and vesicle-associated membrane protein-2 in functioning gastric parietal cells. J Biol Chem 277:50030–50035. doi:10.1074/jbc.M207694200

123

Y. Tajika et al. Klip A, Sun Y, Chiu TT, Foley KP (2014) Signal transduction meets vesicle traffic: the software and hardware of GLUT4 translocation. AJP Cell Physiol 306:C879–C886. doi:10.1152/ajpcell. 00069.2014 Krauss RS (2010) Regulation of promyogenic signal transduction by cell–cell contact and adhesion. Exp Cell Res 316:3042–3049. doi:10.1016/j.yexcr.2010.05.008 Liu Y, Sugiura Y, Lin W (2011) The role of synaptobrevin1/VAMP1 in Ca2?-triggered neurotransmitter release at the mouse neuromuscular junction. J Physiol 589:1603–1618. doi:10.1113/ jphysiol.2010.201939 Lu Z, Joseph D, Bugnard E et al (2001) Golgi complex reorganization during muscle differentiation: visualization in living cells and mechanism. Mol Biol Cell 12:795–808 Martin LB, Shewan A, Millar CA et al (1998) Vesicle-associated membrane protein 2 plays a specific role in the insulin-dependent trafficking of the facilitative glucose transporter GLUT4 in 3T3L1 adipocytes. J Biol Chem 273:1444–1452. doi:10.1074/jbc. 273.3.1444 Matti U, Pattu V, Halimani M et al (2013) Synaptobrevin2 is the v-SNARE required for cytotoxic T-lymphocyte lytic granule fusion. Nat Commun 1:1439. doi:10.1038/ncomms2467 Mauro A (1961) Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol 9:493–495 McMahon HT, Ushkaryov YA, Edelmann L et al (1993) Cellubrevin is a ubiquitous tetanus-toxin substrate homologous to a putative synaptic vesicle fusion protein. Nature 364:346–349. doi:10. 1038/364346a0 Mendez M, Gross KW, Glenn ST et al (2011) Vesicle-associated membrane protein-2 (VAMP2) mediates cAMP-stimulated renin release in mouse juxtaglomerular cells. J Biol Chem 286:28608–28618. doi:10.1074/jbc.M111.225839 Mizuno-Yamasaki E, Rivera-Molina F, Novick P (2012) GTPase networks in membrane traffic. Annu Rev Biochem 81:637–659. doi:10.1146/annurev-biochem-052810-093700 Muir AR, Kanji AH, Allbrook D (1965) The structure of the satellite cells in skeletal muscle. J Anat 99:435–444 Neville C, Rosenthal N, McGrew M et al (1997) Skeletal muscle cultures. Methods Cell Biol 52:85–116 Nori A, Bortoloso E, Frasson F et al (2004) Vesicle budding from endoplasmic reticulum is involved in calsequestrin routing to sarcoplasmic reticulum of skeletal muscles. Biochem J 379:505–512. doi:10.1042/BJ20031875 Nystuen AM, Schwendinger JK, Sachs AJ et al (2006) A null mutation in VAMP1/synaptobrevin is associated with neurological defects and prewean mortality in the lethal-wasting mouse mutant. Neurogenetics 8:1–10. doi:10.1007/s10048-006-0068-7 Paul AC (2001) Muscle length affects the architecture and pattern of innervation differently in leg muscles of mouse, guinea pig, and rabbit compared to those of human and monkey muscles. Anat Rec 262:301–309 Peters C, Miller D, Giovannucci D (2006) Identification, localization and interaction of SNARE proteins in atrial cardiac myocytes. J Mol Cell Cardiol 40:361–374. doi:10.1016/j.yjmcc.2005.12. 007 Procino G, Barbieri C, Tamma G et al (2008) AQP2 exocytosis in the renal collecting duct—involvement of SNARE isoforms and the regulatory role of Munc18b. J Cell Sci 121:2097–2106. doi:10. 1242/jcs.022210 Regazzi R, Wollheim CB, Lang J et al (1995) VAMP-2 and cellubrevin are expressed in pancreatic beta-cells and are essential for Ca(2 ?)-but not for GTP gamma S-induced insulin secretion. EMBO J 14:2723–2730 Rose AJ, Jeppesen J, Kiens B, Richter EA (2009) Effects of contraction on localization of GLUT4 and v-SNARE isoforms in

Vesicular transport in myotubes rat skeletal muscle. Am J Physiol Regul Integr Comp Physiol 297:R1228–R1237. doi:10.1152/ajpregu.00258.2009 Rossetto O, Gorza L, Schiavo G et al (1996) VAMP/synaptobrevin isoforms 1 and 2 are widely and differentially expressed in nonneuronal tissues. J Cell Biol 132:167–179 Rudich A, Klip A (2003) Push/pull mechanisms of GLUT4 traffic in muscle cells. Acta Physiol Scand 178:297–308 Sanes JR, Lichtman JW (2001) Induction, assembly, maturation and maintenance of a postsynaptic apparatus. Nat Rev Neurosci 2:791–805. doi:10.1038/35097557 Sato M, Yoshimura S, Hirai R et al (2011) The role of VAMP7/TIVAMP in cell polarity and lysosomal exocytosis in vivo. Traffic 12:1383–1393. doi:10.1111/j.1600-0854.2011.01247.x Schoch S, Deak F, Konigstorfer A et al (2001) SNARE function analyzed in synaptobrevin/VAMP knockout mice. Science 294:1117–1122. doi:10.1126/science.1064335 Schwenk RW, Dirkx E, Coumans WA et al (2010) Requirement for distinct vesicle-associated membrane proteins in insulin- and AMP-activated protein kinase (AMPK)-induced translocation of GLUT4 and CD36 in cultured cardiomyocytes. Diabetologia 53:2209–2219. doi:10.1007/s00125-010-1832-7 Snow MH (1977) The effects of aging on satellite cells in skeletal muscles of mice and rats. Cell Tissue Res 185:399–408 Steegmaier M, Klumperman J, Foletti DL et al (1999) Vesicleassociated membrane protein 4 is implicated in trans-Golgi network vesicle trafficking. Mol Biol Cell 10:1957–1972 Tajika Y, Sato M, Murakami T et al (2007) VAMP2 is expressed in muscle satellite cells and up-regulated during muscle regeneration. Cell Tissue Res 328:573–581. doi:10.1007/s00441-0060376-0 Tajika Y, Murakami T, Sato M et al (2008) VAMP2 is expressed in myogenic cells during rat development. Dev Dyn 237:1886–1892. doi:10.1002/dvdy.21596 Tajika Y, Takahashi M, Hino M et al (2010) VAMP2 marks quiescent satellite cells and myotubes, but not activated myoblasts. Acta Histochem Cytochem 43:107–114. doi:10.1267/ahc.10010 Tajika Y, Takahashi M, Khairani AF et al (2014) Vesicular transport system in myotubes: ultrastructural study and signposting with

39 vesicle-associated membrane proteins. Histochem Cell Biol 141:441–454. doi:10.1007/s00418-013-1164-z Takahashi M, Tajika Y, Khairani AF et al (2013) The localization of VAMP5 in skeletal and cardiac muscle. Histochem Cell Biol. 139(4):573–582. doi:10.1007/s00418-012-1050-0 Towler MC, Kaufman SJ, Brodsky FM (2004) Membrane traffic in skeletal muscle. Traffic 5:129–139. doi:10.1111/j.1600-0854. 2003.00164.x Trimble WS, Cowan DM, Scheller RH (1988) VAMP-1: a synaptic vesicle-associated integral membrane protein. Proc Natl Acad Sci USA 85:4538–4542 Volchuk A, Mitsumoto Y, He L et al (1994) Expression of vesicleassociated membrane protein 2 (VAMP-2)/synaptobrevin II and cellubrevin in rat skeletal muscle and in a muscle cell line. Biochem J 304(Pt 1):139–145 Wang C-C, Ng CP, Shi H et al (2010) A role for VAMP8/endobrevin in surface deployment of the water channel aquaporin 2. Mol Cell Biol 30:333–343. doi:10.1128/MCB.00814-09 Wang P, Howard MD, Zhang H et al (2012) Characterization of VAMP-2 in the lung: implication in lung surfactant secretion. Cell Biol Int 36:785–791. doi:10.1042/CBI20110146 Weng N, Thomas DDH, Groblewski GE (2007) Pancreatic acinar cells express vesicle-associated membrane protein 2- and 8-specific populations of zymogen granules with distinct and overlapping roles in secretion. J Biol Chem 282:9635–9645. doi:10.1074/jbc.M611108200 Wong SH, Zhang T, Xu Y et al (1998) Endobrevin, a novel synaptobrevin/VAMP-like protein preferentially associated with the early endosome. Mol Biol Cell 9:1549–1563 Zeng Q, Subramaniam VN, Wong SH et al (1998) A novel synaptobrevin/VAMP homologous protein (VAMP5) is increased during in vitro myogenesis and present in the plasma membrane. Mol Biol Cell 9:2423–2437 Zhao P, Yang L, Lopez JA et al (2009) Variations in the requirement for v-SNAREs in GLUT4 trafficking in adipocytes. J Cell Sci 122:3472–3480. doi:10.1242/jcs.047449

123

Organization of organelles and VAMP-associated vesicular transport systems in differentiating skeletal muscle cells.

Vesicular transport plays an important role in the regulation of cellular function and differentiation of the cell, and intracellular vesicles play a ...
3MB Sizes 0 Downloads 6 Views