Please use this identifier to cite or link to this item: https://doi.org/10.1007/s00418-015-1322-6
DC FieldValue
dc.titleInvolvement of unconventional myosin VI in myoblast function and myotube formation
dc.contributor.authorKarolczak, J
dc.contributor.authorPavlyk, I
dc.contributor.authorMajewski, L.
dc.contributor.authorSobczak, M
dc.contributor.authorNiewiadomski, P
dc.contributor.authorRzhepetskyy, Y
dc.contributor.authorSikorska, A
dc.contributor.authorNowak, N
dc.contributor.authorPomorski, P
dc.contributor.authorPrószynski, T
dc.contributor.authorEhler, E
dc.contributor.authorRedowicz, M.J
dc.date.accessioned2020-10-23T08:12:48Z
dc.date.available2020-10-23T08:12:48Z
dc.date.issued2015
dc.identifier.citationKarolczak, J, Pavlyk, I, Majewski, L., Sobczak, M, Niewiadomski, P, Rzhepetskyy, Y, Sikorska, A, Nowak, N, Pomorski, P, Prószynski, T, Ehler, E, Redowicz, M.J (2015). Involvement of unconventional myosin VI in myoblast function and myotube formation. Histochemistry and Cell Biology 144 (1) : 21-38. ScholarBank@NUS Repository. https://doi.org/10.1007/s00418-015-1322-6
dc.identifier.issn0948-6143
dc.identifier.urihttps://scholarbank.nus.edu.sg/handle/10635/179657
dc.description.abstractThe important role of unconventional myosin VI (MVI) in skeletal and cardiac muscle has been recently postulated (Karolczak et al. in Histochem Cell Biol 139:873–885, 2013). Here, we addressed for the first time a role for this unique myosin motor in myogenic cells as well as during their differentiation into myotubes. During myoblast differentiation, the isoform expression pattern of MVI and its subcellular localization underwent changes. In undifferentiated myoblasts, MVI-stained puncti were seen throughout the cytoplasm and were in close proximity to actin filaments, Golgi apparatus, vinculin-, and talin-rich focal adhesion as well as endoplasmic reticulum. Colocalization of MVI with endoplasmic reticulum was enhanced during myotube formation, and differentiation-dependent association was also seen in sarcoplasmic reticulum of neonatal rat cardiomyocytes (NRCs). Moreover, we observed enrichment of MVI in myotube regions containing acetylcholine receptor-rich clusters, suggesting its involvement in the organization of the muscle postsynaptic machinery. Overexpression of the H246R MVI mutant (associated with hypertrophic cardiomyopathy) in myoblasts and NRCs caused the formation of abnormally large intracellular vesicles. MVI knockdown caused changes in myoblast morphology and inhibition of their migration. On the subcellular level, MVI-depleted myoblasts exhibited aberrations in the organization of actin cytoskeleton and adhesive structures as well as in integrity of Golgi apparatus and endoplasmic reticulum. Also, MVI depletion or overexpression of H246R mutant caused the formation of significantly wider or aberrant myotubes, respectively, indicative of involvement of MVI in myoblast differentiation. The presented results suggest an important role for MVI in myogenic cells and possibly in myoblast differentiation. © 2015, The Author(s).
dc.publisherSpringer Verlag
dc.rightsAttribution 4.0 International
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/
dc.sourceUnpaywall 20201031
dc.subjectactin
dc.subjectbeta actin
dc.subjectcholinergic receptor
dc.subjectgamma actin
dc.subjectmutant protein
dc.subjectmyosin VI
dc.subjecttalin
dc.subjectunclassified drug
dc.subjectvinculin
dc.subjectmyosin heavy chain
dc.subjectmyosin VI
dc.subjectactin filament
dc.subjectamino acid sequence
dc.subjectanimal cell
dc.subjectanimal tissue
dc.subjectArticle
dc.subjectcell adhesion
dc.subjectcell differentiation
dc.subjectcell function
dc.subjectcell growth
dc.subjectcell migration
dc.subjectcell structure
dc.subjectcellular distribution
dc.subjectcomplex formation
dc.subjectcontrolled study
dc.subjectdown regulation
dc.subjectendoplasmic reticulum
dc.subjectfocal adhesion
dc.subjectgene overexpression
dc.subjectgene silencing
dc.subjectgenetic transfection
dc.subjectGolgi complex
dc.subjectgrowth inhibition
dc.subjecthypertrophic cardiomyopathy
dc.subjectintracellular space
dc.subjectloss of function mutation
dc.subjectmouse
dc.subjectmyoblast
dc.subjectmyotube
dc.subjectnewborn
dc.subjectnonhuman
dc.subjectphenotype
dc.subjectpriority journal
dc.subjectprotein depletion
dc.subjectprotein expression
dc.subjectprotein localization
dc.subjectprotein synthesis
dc.subjectrat
dc.subjectregulatory mechanism
dc.subjectsarcoplasmic reticulum
dc.subjectupregulation
dc.subjectanimal
dc.subjectcardiac muscle cell
dc.subjectcell differentiation
dc.subjectcell line
dc.subjectcell motion
dc.subjectcell shape
dc.subjectchemistry
dc.subjectcytology
dc.subjectcytoplasm
dc.subjectmetabolism
dc.subjectmuscle development
dc.subjectmyoblast
dc.subjectphysiology
dc.subjectskeletal muscle cell
dc.subjectultrastructure
dc.subjectRattus
dc.subjectActin Cytoskeleton
dc.subjectAnimals
dc.subjectCell Adhesion
dc.subjectCell Differentiation
dc.subjectCell Line
dc.subjectCell Movement
dc.subjectCell Shape
dc.subjectCytoplasm
dc.subjectEndoplasmic Reticulum
dc.subjectGolgi Apparatus
dc.subjectMice
dc.subjectMuscle Development
dc.subjectMuscle Fibers, Skeletal
dc.subjectMyoblasts
dc.subjectMyocytes, Cardiac
dc.subjectMyosin Heavy Chains
dc.subjectRats
dc.subjectSarcoplasmic Reticulum
dc.typeArticle
dc.contributor.departmentBIOMEDICAL ENGINEERING
dc.description.doi10.1007/s00418-015-1322-6
dc.description.sourcetitleHistochemistry and Cell Biology
dc.description.volume144
dc.description.issue1
dc.description.page21-38
dc.published.statePublished
Appears in Collections:Staff Publications
Elements

Show simple item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
10_1007_s00418-015-1322-6.pdf5.67 MBAdobe PDF

OPEN

NoneView/Download

Google ScholarTM

Check

Altmetric


This item is licensed under a Creative Commons License Creative Commons