首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
The expression of fast and slow isoforms of the sarcoplasmic reticulum Ca2+-ATPase was studied in the developing chick embryo and in tissue-cultured myotubes. Monoclonal antibodies specific for each isoform were used as probes of protein expression. Analysis of expression of Ca2+-ATPase isoforms in chick thigh muscles by immunofluorescence microscopy revealed that all muscle fibers expressed both isoforms during their development. Primary generation muscle fibers expressed predominantly the slow isoform. Secondary generation fibers expressed both isoforms at comparable levels. Loss of the "inappropriate" isoforms occurred late in embryonic development. Immunoblot analysis of embryonic thigh muscle proteins indicated that the expression of the slow isoform varied little from embryonic Day 6 (ED6) to ED19, while expression of the fast isoform increased dramatically just prior to ED19. Tissue-cultured myotubes derived from ED12 chick thigh muscle myoblasts, plated at high density, expressed both isoforms of the Ca2+-ATPase at very similar levels. Clonal analysis of myoblasts taken from early (ED6) and late (ED12) chick thigh muscles showed that all muscle colonies expressed both forms, consistent with in vivo results. Fiber-type specific isoforms of the Ca2+-ATPase and myosin heavy chain are not coordinately expressed in developing chick skeletal muscle.  相似文献   

2.
We have identified three sarcolemma-associated antigens, including two antigens that are differentially distributed on skeletal muscle fibers of the fast, fast/slow, and slow types. Monoclonal antibodies were prepared using partially purified membranes of adult chicken skeletal muscles as immunogens and were used to characterize three antigens associated with the sarcolemma of muscle fibers. Immunofluorescence staining of cryosections of adult and embryonic chicken muscles showed that two of the three antigens differed in expression by fibers depending on developmental age and whether the fibers were of the fast, fast/slow, or slow type. Fiber type was assigned by determining the content of fast and slow myosin heavy chain. MSA-55 was expressed equally by fibers of all types. In contrast, MSA-slow and MSA-140 differed in their expression by muscle fibers depending on fiber type. MSA-slow was detected exclusively at the periphery of fast/slow and slow fibers, but was not detected on fast fibers. MSA-140 was detected on all fibers but fast/slow and slow fibers stained more intensely suggesting that these fiber types contain more MSA-140 than fast fibers. These sarcolemma-associated antigens were developmentally regulated in ovo and in vitro. MSA-55 and MSA-140 were detected on all primary muscle fibers by day 8 in ovo of embryonic development, whereas MSA-slow was first detected on muscle fibers just before hatching. Those antigens expressed by fast fibers (MSA-55 and MSA-140) were expressed only after myoblasts differentiated into myotubes, but were not expressed by fibroblasts in cell culture. Each antigen was also detected in one or more nonskeletal muscle cell types: MSA-55 and MSA-slow in cardiac myocytes and smooth muscle of gizzard (but not vascular structures) and MSA-140 in cardiac myocytes and smooth muscle of vascular structures. MSA-55 was identified as an Mr 55,000, nonglycosylated, detergent-soluble protein, and MSA-140 was an Mr 140,000, cell surface protein. The Mr of MSA-slow could not be determined by immunoblotting or immunoprecipitation techniques. These findings indicate that muscle fibers of different physiological function differ in the components associated with the sarcolemma. While the function of these sarcolemma-associated antigens is unknown, their regulated appearance during development in ovo and as myoblasts differentiate in culture suggests that they may be important in the formation, maturation, and function of fast, fast/slow, and slow muscle fibers.  相似文献   

3.
Vertebrate muscles are composed of an array of diverse fast and slow fiber types with different contractile properties. Differences among fibers in fast and slow MyHC expression could be due to extrinsic factors that act on the differentiated myofibers. Alternatively, the mononucleate myoblasts that fuse to form multinucleated muscle fibers could differ intrinsically due to lineage. To distinguish between these possibilities, we determined whether the changes in proportion of slow fibers were attributable to inherent differences in myoblasts. The proportion of fibers expressing slow myosin heavy chain (MyHC) was found to change markedly with time during embryonic and fetal human limb development. During the first trimester, a maximum of 75% of fibers expressed slow MyHC. Thereafter, new fibers formed which did not express this MyHC, so that the proportion of fibers expressing slow MyHC dropped to approximately 3% of the total by midgestation. Several weeks later, a subset of the new fibers began to express slow MyHC and from week 30 of gestation through adulthood, approximately 50% of fibers were slow. However, each myoblast clone (n = 2,119) derived from muscle tissues at six stages of human development (weeks 7, 9, 16, and 22 of gestation, 2 mo after birth and adult) expressed slow MyHC upon differentiation. We conclude from these results that the control of slow MyHC expression in vivo during muscle fiber formation in embryonic development is largely extrinsic to the myoblast. By contrast, human myoblast clones from the same samples differed in their expression of embryonic and neonatal MyHCs, in agreement with studies in other species, and this difference was shown to be stably heritable. Even after 25 population doublings in tissue culture, embryonic stage myoblasts did not give rise to myoblasts capable of expressing MyHCs typical of neonatal stages, indicating that stage-specific differences are not under the control of a division dependent mechanism, or intrinsic "clock." Taken together, these results suggest that, unlike embryonic and neonatal MyHCs, the expression of slow MyHC in vivo at different developmental stages during gestation is not the result of commitment to a distinct myoblast lineage, but is largely determined by the environment.  相似文献   

4.
In addition to its role as a strong antioxidant, vitamin C regulates the differentiation of several cell lineages. In vertebrate skeletal muscle, the vitamin C transporter SVCT2 is preferentially expressed in slow muscle fibers. To gain insights into the possible involvement of intracellular vitamin C on early myogenesis, we investigated the regulation of SVCT2 expression in cultures of chick fetal myoblasts. SVCT2 expression increases in cultures of both, slow and fast muscle-derived myoblasts, as they fuse to form mainly fast myotubes. Interestingly, we found that SVCT2 could be positively modulated by potassium-induced depolarization of myotubes. These findings suggest that SVCT2-mediated uptake of vitamin C could play diverse roles on skeletal muscle development and physiology.  相似文献   

5.
《The Journal of cell biology》1985,101(5):1643-1650
We prepared monoclonal antibodies specific for fast or slow classes of myosin heavy chain isoforms in the chicken and used them to probe myosin expression in cultures of myotubes derived from embryonic chicken myoblasts. Myosin heavy chain expression was assayed by gel electrophoresis and immunoblotting of extracted myosin and by immunostaining of cultures of myotubes. Myotubes that formed from embryonic day 5-6 pectoral myoblasts synthesized both a fast and a slow class of myosin heavy chain, which were electrophoretically and immunologically distinct, but only the fast class of myosin heavy chain was synthesized by myotubes that formed in cultures of embryonic day 8 or older myoblasts. Furthermore, three types of myotubes formed in cultures of embryonic day 5-6 myoblasts: one that contained only a fast myosin heavy chain, a second that contained only a slow myosin heavy chain, and a third that contained both a fast and a slow heavy chain. Myotubes that formed in cultures of embryonic day 8 or older myoblasts, however, were of a single type that synthesized only a fast class of myosin heavy chain. Regardless of whether myoblasts from embryonic day 6 pectoral muscle were cultured alone or mixed with an equal number of myoblasts from embryonic day 12 muscle, the number of myotubes that formed and contained a slow class of myosin was the same. These results demonstrate that the slow class of myosin heavy chain can be synthesized by myotubes formed in cell culture, and that three types of myotubes form in culture from pectoral muscle myoblasts that are isolated early in development, but only one type of myotube forms from older myoblasts; and they suggest that muscle fiber formation probably depends upon different populations of myoblasts that co-exist and remain distinct during myogenesis.  相似文献   

6.
7.
Postnatal myoblasts, the satellite cells, originating from slow and fast skeletal muscle fibres differentiate and fuse into myotubes expressing different phenotype of myosin heavy chain (MyHC) isoforms. Little is known, however, of factors which establish and maintain this phenotypic diversity. We used immunofluorescent labelling and Western blotting to examine the expression of slow and fast MyHC isoforms in myotubes formed in vitro from satellite cells isolated from mouse fast twitch extensor digitorum longus (EDL) and slow twitch soleus muscles. Satellite cells were cultured in serum-rich growth medium promoting myoblast proliferation until cross-striated and self-contracting myotubes were formed. We report that in both cultures myotubes expressed slow as well as fast MyHC isoforms, but the level of slow MyHC was higher in soleus culture than in EDL culture. Hence, the pattern of expression of slow and fast MyHC was characteristic of the muscle fibre type from which these cells derive. These results support the concept of phenotypic diversity among satellite cells in mature skeletal muscles and suggest that this diversity is generated in vitro irrespectively of serum mitogens.  相似文献   

8.
Myosin heavy chain (MHC) is encoded by a multigene family containing members which are expressed in developmental and fiber type-specific patterns. In developing rats, primary (1°) and secondary (2°) myotjbes can be disfinguished by differences in MHC expression: 1° myotubes coexpress embryonic and slow MHC, while 2° myotubes initially express only embryonic MHC. We have used monoclonal antibodies which recognize the embryonic, slow, neonatal, and adult fast IIB/IIX MHCs to examine MHC accumulation in myoblasts obtained from hindlimbs of embryonic day (ED) 14 and ED 20 Sprague-Dawley rats during differentiation in vitro. Embryonic myoblasts (ED 14), which develop into 1° myotubes in vivo, differentiate as myocytes or small myotubes (i.e., 1–4 nuclei) which express both embryonic and slow MHC. They do not accumulate detectable levels of neonatal or adult fast IIB/IIX MHC. Fetal myoblasts, which develop into secondary myotubes in vivo, fuse to form large myotubes (i.e., 10–50 nuclei) and express predominantly embryonic MHC at 3 days in culture. These myotubes accumulate neonatal and adult fast IIB/IIX isoforms of MHC and eventually contract spontaneously. In contrast to embryonic myotubes, they do not accumulate slow MHC. Our results demonstrate that embryonic and fetal rat myoblasts express different phenotypes in vitro and suggest that they represent distinct myoblast lineages similar to those previously described in chickens and mice. These two lineages may be responsible for the generation of distinct populations of 1° and 2° myotubes in vivo. © 1993Wiley-Liss, Inc.  相似文献   

9.
We show that PTP1D, a protein tyrosine phosphatase that contains two SH2 domains, is preferentially expressed in slow skeletal muscle fibers. Immunohistochemical staining using polyclonal antibodies against PTP1D demonstrated that PTP1D was expressed in a subpopulation of rodent muscle fibers. These fibers were identified as slow Type I fibers based on histochemical ATPase assays and slow myosin heavy chain expression. Northern and Western analyses showed that PTP1D levels were higher in predominantly slow muscles than in predominantly fast muscles. This differential expression of PTP1D in slow muscle fibers appeared by birth. In cultures of mouse myogenic cells, PTP1D was expressed after MyoD and myogenin and appeared in myotubes derived from embryonic, fetal, and postnatal myoblasts. Remarkably, PTP1D was organized into sarcomeres in a pattern coincident with myosin heavy chain, suggesting that PTP1D associates with a component of the thick filament. These results show that PTP1D is preferentially expressed in slow muscle fibers. We speculate that PTP1D may play a role in slow muscle fiber function and differentiation.  相似文献   

10.
Laminin-2 is a component of skeletal and cardiac basal lamina expressed in normal mouse and human. Laminin alpha2 chain (LAMA2), however, is absent from muscles of some congenital muscular dystrophy patients and the dystrophia muscularis (dy/dy) mouse model. LAMA2 restoration was investigated following cell transplantation in vivo in dy/dy mouse. Allogeneic primary muscle cell cultures expressing the beta- galactosidase transgene under control of a muscular promoter, or histocompatible primary muscle cell cultures, were transplanted into dy/dy mouse muscles. FK506 immunosuppression was used in noncompatible models. All transplanted animals expressed LAMA2 in these immunologically-controlled models, and the degrees of LAMA2 restoration were shown to depend on the age of the animal at transplantation, on muscle pretreatment, and on duration time after transplantation in some cases. LAMA2 did not always colocalize with new or hybrid muscle fibers formed by the fusion of donor myoblasts. LAMA2 deposition around muscle fibers was often segmental and seemed to radiate from the center to the periphery of the injection site. Allogeneic conditionally immortalized pure myogenic cells expressing the beta-galactosidase transgene were characterized in vitro and in vivo. When injected into FK506- immunosuppressed dy/dy mice, these cells formed new or hybrid muscle fibers but essentially did not express LAMA2 in vivo. These data show that partial LAMA2 restoration is achieved in LAMA2-deficient dy/dy mouse by primary muscle cell culture transplantation. However, not all myoblasts, or myoblasts alone, or the muscle fibers they form are capable of LAMA2 secretion and deposition in vivo.  相似文献   

11.
《The Journal of cell biology》1988,107(6):2191-2197
The developmental pattern of slow myosin expression has been studied in mouse embryos from the somitic stage to the period of secondary fiber formation and in myogenic cells, cultured from the same developmental stages. The results obtained, using a combination of different polyclonal and monoclonal antibodies, indicate that slow myosin is coexpressed in virtually all the cells that express embryonic (fast) myosin in somites and limb buds in vivo as well as in culture. On the contrary fetal or late myoblasts (from 15-d-old embryos) express in culture only embryonic (fast) myosin. At this stage, muscle cells in vivo, as already shown (Crow, M.T., and F.A. Stockdale. 1986. Dev. Biol. 113:238-254; Dhoot, G.K. 1986. Muscle & Nerve. 9:155-164; Draeger, A., A.G. Weeds, and R.B. Fitzsimons. 1987. J. Neurol. Sci. 81:19-43; Miller, J.B., and F.A. Stockdale. 1986. J. Cell Biol. 103:2197-2208), consist of primary myotubes, which express both myosins, and secondary myotubes, which express preferentially embryonic (fast) myosin. Under no circumstance neonatal or adult fast myosins were detected. Western blot analysis confirmed the immunocytochemical data. These results suggest that embryonic myoblasts in mammals are all committed to the mixed embryonic-(fast) slow lineage and, accordingly, all primary fibers express both myosins, whereas fetal myoblasts mostly belong to the embryonic (fast) lineage and likely generate fibers containing only embryonic (fast) myosin. The relationship with current models of avian myogenesis are discussed.  相似文献   

12.
We have created new mouse muscle cell lines of an immortalized type, expressing normal differentiation at the myotube stage: sarcomeric organization, functional excitation-contraction coupling, and triadic differentiation. The DNA immortalizing recombinant utilizes a deletion mutant of the regulatory region of the human vimentin promoter controlling the expression of a SV40 thermosensitive large T antigen, in which the small t sequence has been deleted. Skeletal mouse replicative myoblasts synthesized predominantly vimentin. After myoblast fusion the vimentin gene is strongly repressed in multinucleated syncytia. Furthermore, the normal activity of the vimentin promoter in myoblasts is increased in the large T antigen-expressing cells. We observed that continuous and rapid division of myoblasts occurs at permissive temperature, suggesting that immortalization is achieved even though the small t antigen is absent. When fusion is induced by changing media conditions, large T antigen expression is totally repressed by the vimentin promoter. When the temperature is elevated to 39 degrees C, the preexisting large T antigen is inactivated. The resulting myotubes from normal mouse differentiate totally normally as indicated by their morphology, ultrastructure, and electrophysiological properties. Mutant (muscular dysgenesis) immortalized cells express the same properties as mutant primary counterparts with no contraction, no slow Ca2+ current, and no triadic differentiation. These immortalized cell lines are potentially very useful for further pharmacology, transplantation, and cell biology studies. The vimentin promoter control of immortalizing recombinant DNA can be used for any mammalian normal and mutant muscle cell lines.  相似文献   

13.
Skeletal muscle has the ability to regenerate new muscle fibers after injury. The process of new muscle formation requires that quiescent mononuclear muscle precursor cells (myoblasts) become activated, proliferate, differentiate, and fuse into multinucleated myotubes which, in turn, undergo further differentiation and mature to form functional muscle fibers. Previous data demonstrated the crucial role played by 3-hydroxy 3-methylglutaryl coenzyme A reductase (HMGR), the rate-limiting enzyme of cholesterol biosynthetic pathway, in fetal rat myoblast (L6) differentiation. This finding, along with epidemiological studies assessing the myotoxic effect of statins, HMGR inhibitors, allowed us to speculate that HMGR could be strongly involved in skeletal muscle repair. Thus, our research was aimed at evaluating such involvement: in vitro and in vivo experiments were performed on both mouse adult satellite cell derived myoblasts (SCDM) and mouse muscles injured with cardiotoxin. Results demonstrate that HMGR inhibition by the statin Simvastatin reduces SCDM fusion index, fast MHC protein levels by 60% and slow MHC by 40%. Most importantly, HMGR inhibition delays skeletal muscle regeneration in vivo. Thus, besides complaining of myopathies, patients given Simvastatin could also undergo an impairment in muscle repair.  相似文献   

14.
Regenerating areas of adult chicken fast muscle (pectoralis major) and slow muscle (anterior latissimus dorsi) were examined in order to determine synthesis patterns of myosin light chains, heavy chains and tropomyosin. In addition, these patterns were also examined in muscle cultures derived from satellite cells of adult fast and slow muscle. One week after cold-injury the regenerating fast muscle showed a pattern of synthesis that was predominately embryonic. These muscles synthesized the embryonic myosin heavy chain, beta-tropomyosin and reduced amounts of myosin fast light chain-3 which are characteristic of embryonic fast muscle but synthesized very little myosin slow light chains. The regenerating slow muscle, however, showed a nearly complete array of embryonic peptides including embryonic myosin heavy chain, fast and slow myosin light chains and both alpha-fast and slow tropomyosins. Peptide map analysis of the embryonic myosin heavy chains synthesized by regenerating fast and slow muscles showed them to be identical. Thus, in both muscles there is a return to embryonic patterns during regeneration but this return appears to be incomplete in the pectoralis major. By 4 weeks postinjury both regenerating fast and slow muscles had stopped synthesizing embryonic isoforms of myosin and tropomyosin and had returned to a normal adult pattern of synthesis. Adult fast and slow muscles yielded a satellite cell population that formed muscle fibers in culture. Fibers derived from either population synthesized the embryonic myosin heavy chain in addition to alpha-fast and beta-tropomyosin. Thus, muscle fibers derived in culture from satellite cells of fast and slow muscles synthesized a predominately embryonic pattern of myosin heavy chains and tropomyosin. In addition, however, the satellite cell-derived myotubes from fast muscle synthesized only fast myosin light chains while the myotubes derived from slow muscle satellite cells synthesized both fast and slow myosin light chains. Thus, while both kinds of satellite cells produced embryonic type myotubes in culture the overall patterns were not identical. Satellite cells of fast and slow muscle appear therefore to have diverged from each other in their commitment during maturation in vivo.  相似文献   

15.
Myogenesis involves changes in both gene expression and cellular architecture. Little is known of the organization, in muscle in vivo, of the subcellular organelles involved in protein synthesis despite the potential importance of targeted protein synthesis for formation and maintenance of functional domains such as the neuromuscular junction. A panel of antibodies to markers of the ER, the Golgi complex, and the centrosome were used to localize these organelles by immunofluorescence in myoblasts and myotubes of the mouse muscle cell line C2 in vitro, and in intact single muscle fibers from the rat flexor digitorum brevis. Antibodies to the ER stained structures throughout the cytoplasm of both C2 myoblasts and myotubes. In contrast, the spatial relationship between nucleus, centrosome, and Golgi complex was dramatically altered. These changes could also be observed in a low- calcium medium that allowed differentiation while preventing myoblast fusion. Muscle fibers in vivo resembled myotubes except that the ER occupied a smaller volume of cytoplasm and no staining was found for one of the Golgi complex markers, the enzyme alpha-mannosidase II. Electron microscopy, however, clearly showed the presence of stacks of Golgi cisternae in both junctional and extrajunctional regions of muscle fibers. The perinuclear distribution of the Golgi complex was also observed in live muscle fibers stained with a fluorescent lipid. Thus, the distribution of subcellular organelles of the secretory pathway was found to be similar in myotubes and muscle fibers, and all organelles were found in both junctional and extrajunctional areas of muscle.  相似文献   

16.
S M Hughes  H M Blau 《Cell》1992,68(4):659-671
Muscle fibers specialized for fast or slow contraction are arrayed in characteristic patterns within developing limbs. Clones of myoblasts analyzed in vitro express fast and slow myosin isoforms typical of the muscle from which they derive. As a result, it has been suggested that distinct myoblast lineages generate and maintain muscle fiber pattern. We tested this hypothesis in vivo by using a retrovirus to label myoblasts genetically so that the fate of individual clones could be monitored. Both myoblast clones labeled in muscle in situ and clones labeled in tissue culture and then injected into various muscles contribute progeny to all fiber types encountered. Thus, extrinsic signals override the intrinsic commitment of myoblast nuclei to particular programs of gene expression. We conclude that in postnatal development, pattern is not dictated by myoblast lineage.  相似文献   

17.
The fiber-type composition of postnatal chicken leg muscle spindles with from one to four intrafusal fibers was examined in sections incubated with monoclonal antibodies against fast and slow myosin heavy chains. In monofibral spindles the lone intrafusal fiber was almost always fast. In duofibral spindles usually one slow and one fast fiber were present. Trifibral spindles most often displayed two fast and one slow fiber, whereas quadrofibral receptors characteristically contained two slow and two fast fibers. Earlier results showed that the primary intrafusal myotube in nascent spindles has almost always a fast myosin heavy chain profile and that the proportion of slow myotubes and fibers increases as intrafusal fiber bundles grow in size. Data from postnatal chicken leg muscles collected here suggest that up to the first four fibers this proportional increase can be largely accounted for if consecutive intrafusal fibers arise in a fast-slow-fast-slow sequence. The late recognition during myogenesis of primary intrafusal myotubes and their fast myosin heavy chain profiles warrant exploring if nascent chicken muscles spindles are first seeded by fast fetal myoblasts. © 1995 Wiley-Liss, Inc.  相似文献   

18.
Bilateral asymmetry of the paired snapper/pincer claws may be reversed in adult snapping shrimps (Alpheus heterochelis). Removal of the snapper claw triggers transformation of the contralateral pincer claw into a snapper and the regeneration of a new pincer claw at the old snapper site. During this process the pincer closer muscle is remodeled to a snapper-type, and these alterations have been examined with the electron microscope. There is selective death of the central band of fast fibers, accompanied by an accumulation of electron-dense crysttaline bodies in the degenerating fibers. Two principal types of hemocytes (amebocytes and coagulocytes) invade the area and the degenerating muscle fibers. New myotubes also appear in this central site. The myotubes are characterized by a prolific network of presumptive sarcoplasmic reticulum and transverse tubules, nascent myofibrils, and crystalline bodies. The myotubes are innervated by many motor nerve terminals, and they subsequently differentiate into long-sarcomere (8–12 m), slow muscle fibers. Remodeling of the central band, therefore, occurs by degeneration of the fast fibers and their replacement by new slow fibers. Remnants of the degenerating fast fibers act as scaffolding for the myotubes which originate from adjacent satellite cells. The crystalline bodies may represent protein stores from the degeneration of the fast fibers, recycled for use in the genesis of new fibers. The invading hemocytes appear to play several roles, initially phagocytosing the fast muscle fibers, transporting the crystalline bodies into the new myotubes, and acting as stem cells for the new muscle fibers. Apart from the central band of fibers, the remaining pincer-type slow fibers with sarcomere lengths of 5–7 m are transformed via sarcomere lengthening into snapper-type slow fibers with sarcomere lengths of 7–12 m. Thus, during claw transformation in adult snapping shrimps, the pincer closer muscle is remodeled into a snapper closer muscle by selective death of the fast-fiber band, replacement of the fast-fiber band by new slow fibers, and transformation of the existing slow fibers to an even-slower variety. Note. This paper is dedicated to the fond memory of Professor M.S. Laverack whose enjoyment of biological research and gentle encouragement of such endeavours touched all those who knew him.  相似文献   

19.
20.
Roles for the integrin VLA-4 and its counter receptor VCAM-1 in myogenesis.   总被引:47,自引:0,他引:47  
Mammalian myogenesis is biphasic: primary myoblasts fuse to form primary myotubes, then secondary myoblasts align along the primary myotubes and form secondary myotubes, which comprise most of adult muscle. We provide evidence that an integrin (VLA-4) and its counter receptor (VCAM-1) have a role in secondary myogenesis. Both receptors are synthesized by cultured muscle cells: VLA-4 is induced as myotubes form, whereas VCAM-1 is present on myoblasts and myotubes. In vivo, both molecules are expressed at sites of secondary myogenesis, VLA-4 on primary and secondary myotubes, and VCAM-1 on secondary myoblasts and on regions of secondary myotubes apposed to primary myotubes. These patterns suggest that VLA-4-VCAM-1 interactions influence alignment of secondary myoblasts along primary myotubes and/or the fusion of secondary myoblasts. In support of the latter possibility, antibodies to VLA-4 or VCAM-1 inhibit myotube formation in culture.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号