Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Orphanet J Rare Dis ; 19(1): 180, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38685042

ABSTRACT

BACKGROUND: This cross-sectional controlled study aims to assess health-related quality of life (HRQoL) of children and adolescents with a molecular diagnosis of Marfan syndrome (MFS) or related disorders and to evaluate the factors associated with HRQoL in this population. Sixty-three children with MFS and 124 age- and sex-matched healthy children were recruited. HRQoL was assessed using the Pediatric Quality of Life Inventory (PedsQL™) generic questionnaire. The correlation between HRQoL scores and the different continuous parameters (age, body mass index, disease severity, systemic score, aortic sinus diameter, and aerobic physical capacity) was evaluated using Pearson's or Spearman's coefficient. A multiple linear regression analysis was performed on the two health summary self-reported PedsQL™ scores (physical and psychosocial) to identify the factors associated with HRQoL in the MFS group. RESULTS: Except for emotional functioning, all other domains of HRQoL (psychosocial and physical health, social and school functions) were significantly lower in children with MFS compared to matched healthy children. In the MFS group, the physical health summary score was significantly lower in female than in male patients (self-report: absolute difference [95%CI] = -8.7 [-17.0; -0.47], P = 0.04; proxy-report: absolute difference [95%CI] = -8.6 [-17.3; 0.02], P = 0.05) and also negatively correlated with the systemic score (self-report: R = -0.24, P = 0.06; proxy-report: R = -0.29, P = 0.03) and with the height Z-score (proxy-report: R = -0.29, P = 0.03). There was no significant difference in the physical health summary scores between the different genetic subgroups. In the subgroup of 27 patients who performed a cardiopulmonary exercise test, self- and proxy-reported physical health summary scores were highly correlated with their aerobic physical capacity assessed by peak oxygen consumption (VO2max) and ventilatory anaerobic threshold (VAT). In the multivariate analysis, the most important independent predictors of decreased physical health were increased height, decreased body mass index, decreased VAT and use of prophylactic therapy. CONCLUSIONS: This study reports an impaired HRQoL in children and adolescents with MFS or related conditions, in comparison with matched healthy children. Educational and rehabilitation programs must be developed and evaluated to improve exercise capacity and HRQoL in these patients. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03236571 . Registered 28 July 2017.


Subject(s)
Marfan Syndrome , Quality of Life , Humans , Marfan Syndrome/physiopathology , Male , Female , Cross-Sectional Studies , Child , Adolescent , Surveys and Questionnaires
2.
PLoS Biol ; 21(8): e3002261, 2023 08.
Article in English | MEDLINE | ID: mdl-37590318

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an early event in cell dissemination from epithelial tissues. EMT endows cells with migratory, and sometimes invasive, capabilities and is thus a key process in embryo morphogenesis and cancer progression. So far, matrix metalloproteinases (MMPs) have not been considered as key players in EMT but rather studied for their role in matrix remodelling in later events such as cell migration per se. Here, we used Xenopus neural crest cells to assess the role of MMP28 in EMT and migration in vivo. We show that a catalytically active MMP28, expressed by neighbouring placodal cells, is required for neural crest EMT and cell migration. We provide strong evidence indicating that MMP28 is imported in the nucleus of neural crest cells where it is required for normal Twist expression. Our data demonstrate that MMP28 can act as an upstream regulator of EMT in vivo raising the possibility that other MMPs might have similar early roles in various EMT-related contexts such as cancer, fibrosis, and wound healing.


Subject(s)
Epithelial-Mesenchymal Transition , Neural Crest , Cell Movement , Cell Nucleus , Epithelium
3.
Nat Commun ; 10(1): 1518, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944331

ABSTRACT

When migrating in vivo, cells are exposed to numerous conflicting signals: chemokines, repellents, extracellular matrix, growth factors. The roles of several of these molecules have been studied individually in vitro or in vivo, but we have yet to understand how cells integrate them. To start addressing this question, we used the cephalic neural crest as a model system and looked at the roles of its best examples of positive and negative signals: stromal-cell derived factor 1 (Sdf1/Cxcl12) and class3-Semaphorins. Here we show that Sdf1 and Sema3A antagonistically control cell-matrix adhesion via opposite effects on Rac1 activity at the single cell level. Directional migration at the population level emerges as a result of global Semaphorin-dependent confinement and broad activation of adhesion by Sdf1 in the context of a biased Fibronectin distribution. These results indicate that uneven in vivo topology renders the need for precise distribution of secreted signals mostly dispensable.


Subject(s)
Cell Movement/physiology , Cell-Matrix Junctions/physiology , Neural Crest/cytology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cell Line , Cell Shape/drug effects , Cell Surface Extensions/drug effects , Cell-Matrix Junctions/drug effects , Cell-Matrix Junctions/metabolism , Chemokine CXCL12/metabolism , Female , Fibronectins/metabolism , Male , Manganese/metabolism , Mice , Nerve Tissue Proteins/physiology , Neural Crest/drug effects , Neural Crest/metabolism , Receptors, CXCR4/metabolism , Semaphorins/metabolism , Xenopus laevis/embryology , rac1 GTP-Binding Protein/metabolism
4.
PLoS Curr ; 92017 Dec 21.
Article in English | MEDLINE | ID: mdl-29399383

ABSTRACT

Histone acetyl transferases (HATs) and histone deacetylases (HDAC) control transcription during myogenesis. HDACs promote chromatin condensation, inhibiting gene transcription in muscle progenitor cells until myoblast differentiation is triggered and HDACs are released. HATs, namely CBP/p300, activate myogenic regulatory and elongation factors promoting myogenesis. HDAC inhibitors are known to improve regeneration in dystrophic muscles through follistatin upregulation. However, the potential of directly modulating HATs remains unexplored. We tested this possibility in a well-known zebrafish model of Duchenne muscular dystrophy. Interestingly, CBP/p300 transcripts were found downregulated in the absence of Dystrophin. While investigating CBP rescuing potential we observed that dystrophin-null embryos overexpressing CBP actually never show significant muscle damage, even before a first regeneration cycle could occur. We found that the pan-HDAC inhibitor trichostatin A (TSA) also prevents early muscle damage, however the single HAT CBP is as efficient even in low doses. The HAT domain of CBP is required for its full rescuing ability. Importantly, both CBP and TSA prevent early muscle damage without restoring endogenous CBP/p300 neither increasing follistatin transcripts. This suggests a new mechanism of action of epigenetic regulators protecting dystrophin-null muscle fibres from detaching, independent from the known improvement of regeneration upon damage of HDACs inhibitors. This study builds supporting evidence that epigenetic modulators may play a role in determining the severity of muscle dystrophy, controlling the ability to resist muscle damage. Determining the mode of action leading to muscle protection can potentially lead to new treatment options for muscular dystrophies in the future.

5.
Elife ; 42015 Oct 13.
Article in English | MEDLINE | ID: mdl-26459831

ABSTRACT

Dystrophin forms an essential link between sarcolemma and cytoskeleton, perturbation of which causes muscular dystrophy. We analysed Dystrophin binding dynamics in vivo for the first time. Within maturing fibres of host zebrafish embryos, our analysis reveals a pool of diffusible Dystrophin and complexes bound at the fibre membrane. Combining modelling, an improved FRAP methodology and direct semi-quantitative analysis of bleaching suggests the existence of two membrane-bound Dystrophin populations with widely differing bound lifetimes: a stable, tightly bound pool, and a dynamic bound pool with high turnover rate that exchanges with the cytoplasmic pool. The three populations were found consistently in human and zebrafish Dystrophins overexpressed in wild-type or dmd(ta222a/ta222a) zebrafish embryos, which lack Dystrophin, and in Gt(dmd-Citrine)(ct90a) that express endogenously-driven tagged zebrafish Dystrophin. These results lead to a new model for Dystrophin membrane association in developing muscle, and highlight our methodology as a valuable strategy for in vivo analysis of complex protein dynamics.


Subject(s)
Dystrophin/analysis , Muscle, Skeletal/chemistry , Muscle, Skeletal/growth & development , Zebrafish/embryology , Animals , Fluorescence Recovery After Photobleaching , Humans
7.
Dev Dyn ; 241(8): 1365-73, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700438

ABSTRACT

BACKGROUND: The circadian clock is a well-described temporal organizer in adult organisms. Despite the particularly evident need for temporal control during embryo development, the effect of environmental cues is still greatly neglected. Few studies have reported circadian clock gene expression in early embryonic stages. However, nothing is known about circadian clock gene expression and function in the first stages of avian embryogenesis. RESULTS/CONCLUSIONS: In this work, the presence and spatial distribution of core circadian clock Bmal1 and Clock transcripts were thoroughly characterized during the first 50 hr of chick development using reverse transcriptase-polymerase chain reaction (RT-PCR), single and double whole-mount in situ hybridization and subsequent cross-section histology analysis. RT-PCR detected both Bmal1 and Clock transcripts since the egg is laid and until the embryo reaches the 22-somite stage. Whole-mount in situ hybridization showed that Bmal1 and Clock are expressed in the Hensen's node and primitive streak at early gastrula stage. Later, both mRNAs are present in the developing nervous system, optic vesicle, notochord, foregut, and somites. Clock was further identified in the developing heart. Noticeably, Bmal1 and Clock are expressed with a "salt and pepper" pattern, suggesting the existence of nonentrained oscillatory transcription which could play a nondependent dark/light function during chick embryo development.


Subject(s)
ARNTL Transcription Factors/metabolism , CLOCK Proteins/metabolism , ARNTL Transcription Factors/genetics , Animals , CLOCK Proteins/genetics , Chick Embryo , Chickens , Circadian Clocks/genetics , Circadian Clocks/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , In Situ Hybridization , Reverse Transcriptase Polymerase Chain Reaction
8.
Dev Biol ; 354(2): 191-207, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21420400

ABSTRACT

Cells anchor to substrates by binding to extracellular matrix (ECM). In addition to this anchoring function however, cell-ECM binding is a mechanism for cells to sense their surroundings and to communicate and coordinate behaviour amongst themselves. Several ECM molecules and their receptors play essential roles in muscle development and maintenance. Defects in these proteins are responsible for some of the most severe muscle dystrophies at every stage of life from neonates to adults. However, recent studies have also revealed a role of cell-ECM interactions at much earlier stages of development as skeletal muscle forms. Here we review which ECM molecules are present during the early phases of myogenesis, how myogenic cells interact with the ECM that surrounds them and the potential consequences of those interactions. We conclude that cell-ECM interactions play significant roles during all stages of skeletal muscle development in the embryo and suggest that this "extracellular matrix dimension" should be added to our conceptual network of factors contributing to skeletal myogenesis.


Subject(s)
Extracellular Matrix/metabolism , Muscle Development , Muscle, Skeletal/embryology , Animals , Cell Communication , Extracellular Matrix Proteins/metabolism , Humans , Mice , Rats , Signal Transduction
9.
PLoS One ; 5(7): e11728, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20668528

ABSTRACT

BACKGROUND: During myogenesis several transcription factors and regulators of protein synthesis and assembly are rapidly degraded by the ubiquitin-proteasome system (UPS). Given the potential role of the deubiquitinating enzyme (DUB) ataxin-3 in the UPS, and the high expression of the murine ataxin-3 homolog in muscle during embryogenesis, we sought to define its role in muscle differentiation. METHODOLOGY/PRINCIPAL FINDINGS: Using immunofluorescence analysis, we found murine ataxin-3 (mATX3) to be highly expressed in the differentiated myotome of E9.5 mouse embryos. C2C12 myoblasts depleted of mATX3 by RNA interference exhibited a round morphology, cell misalignment, and a delay in differentiation following myogenesis induction. Interestingly, these cells showed a down-regulation of alpha5 and alpha7 integrin subunit levels both by immunoblotting and immunofluorescence. Mouse ATX3 was found to interact with alpha5 integrin subunit and to stabilize this protein by repressing its degradation through the UPS. Proteomic analysis of mATX3-depleted C2C12 cells revealed alteration of the levels of several proteins related to integrin signaling. CONCLUSIONS: Ataxin-3 is important for myogenesis through regulation of integrin subunit levels.


Subject(s)
Antigens, CD/metabolism , Cell Differentiation/physiology , Integrin alpha Chains/metabolism , Integrin alpha5/metabolism , Muscle Development/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Antigens, CD/genetics , Ataxin-3 , Cell Differentiation/genetics , Cell Line , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fluorescent Antibody Technique , Immunoblotting , Immunoprecipitation , In Vitro Techniques , Integrin alpha Chains/genetics , Integrin alpha5/genetics , Isoelectric Focusing , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Myoblasts/cytology , Myoblasts/metabolism , Nuclear Proteins/genetics , Pregnancy , Protein Binding , Proteomics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Transcription Factors/genetics
10.
Biochim Biophys Acta ; 1803(10): 1154-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20637808

ABSTRACT

Ataxin-3 (ATXN3) is a widely expressed protein that binds to ubiquitylated proteins, has deubiquitylating activity in vitro and is thought to modulate substrate degradation through the ubiquitin-proteasome pathway. Expansion of a polyglutamine tract in ATXN3 causes Machado-Joseph disease, a late-onset neurodegenerative disorder characterized by ubiquitin-positive aggregate formation and specific neuronal death. Although ATXN3 has been involved in transcriptional repression and in the ubiquitin-proteasome pathway, its biological function is still unknown. In this work, we show that depletion of ATXN3 using small-interference RNA (siRNA) causes a prominent phenotype in both human and mouse cell lines. A mild increase in ubiquitylation occurs and cells exhibit ubiquitin-positive foci, which is consistent with ATXN3 putative function as a deubiquitylating enzyme. In addition, siATXN3-silenced cells exhibit marked morphological changes such as rounder shape and loss of adhesion protrusions. At a structural level, the microtubule, microfilament and intermediate filament networks are severely compromised and disorganized. This cytoskeletal phenotype is reversible and dependent on ATXN3 levels. Cell-extracellular matrix connection is also affected in ATXN3-depleted cells as talin expression is reduced in the focal adhesions and lower levels of alpha-1 integrin subunit are expressed at their surface. Although the cytoskeletal and adhesion problems do not originate any major change in the cell cycle of siATXN3-depleted cells, cell death is increased in siATXN3 cultures compared to controls. In summary, in this work we show that the absence of ATXN3 leads to an overt cytoskeletal/adhesion defect raising the possibility that this protein may play a role in the cytoskeleton.


Subject(s)
Apoptosis/physiology , Cytoskeleton/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , 3T3 Cells , Animals , Ataxin-3 , Blotting, Western , Cell Cycle/physiology , Focal Adhesions/physiology , HeLa Cells , Humans , In Situ Nick-End Labeling , Integrin alpha1/metabolism , Mice , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
11.
J Neurosci ; 29(40): 12506-20, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19812326

ABSTRACT

In vertebrates, the embryonic nervous system is shaped and patterned by a series of temporally and spatially regulated cell divisions, cell specifications, and cell adhesions and movements. Morphogens of the Hedgehog, Wnt, and bone morphogenetic protein families have been shown to play a crucial role in the control of cell division and specification in the trunk neural tube, but their possible implication in the regulation of adhesive events has been poorly documented. In the present study, we demonstrate that Sonic hedgehog regulates neural epithelial cell adhesion and polarity through regulation of integrin activity, cadherin cell-cell contact, and cell polarity genes in immature neural epithelial cells before the specification of neuronal cells. We propose that Sonic hedgehog orchestrates neural tube morphogenesis by coordinating adhesive and motility events with cell proliferation and differentiation.


Subject(s)
Cadherins/metabolism , Cell Adhesion/genetics , Cell Polarity/genetics , Hedgehog Proteins/metabolism , Integrins/metabolism , Neural Tube/embryology , Neurulation/genetics , Animals , Anoikis/genetics , Body Patterning/genetics , Chick Embryo , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression , Mice , Neural Tube/cytology , Neural Tube Defects/genetics , Quail , Signal Transduction/genetics , Transcriptional Activation/genetics
12.
Birth Defects Res C Embryo Today ; 81(2): 65-83, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17600780

ABSTRACT

Segmentation of the vertebrate embryo body is a fundamental developmental process that occurs with strict temporal precision. Temporal control of this process is achieved through molecular segmentation clocks, evidenced by oscillations of gene expression in the unsegmented presomitic mesoderm (PSM, precursor tissue of the axial skeleton) and in the distal limb mesenchyme (limb chondrogenic precursor cells). The first segmentation clock gene, hairy1, was identified in the chick embryo PSM in 1997. Ten years later, chick hairy2 expression unveils a molecular clock operating during limb development. This review revisits vertebrate embryo segmentation with special emphasis on the current knowledge on somitogenesis and limb molecular clocks. A compilation of human congenital disorders that may arise from deregulated embryo clock mechanisms is presented here, in an attempt to reconcile different sources of information regarding vertebrate embryo development. Challenging open questions concerning the somitogenesis clock are presented and discussed, such as When?, Where?, How?, and What for? Hopefully the next decade will be equally rich in answers.


Subject(s)
Biological Clocks , Congenital Abnormalities , Embryonic Development , Gene Expression Regulation, Developmental , Transcription Factors/genetics , Animals , Birds , Cell Differentiation , Humans , Somites/cytology , Transcription Factors/metabolism
13.
Biochem Biophys Res Commun ; 352(1): 153-7, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17112470

ABSTRACT

The apical ectodermal ridge (AER) controls limb outgrowth and patterning, such that its removal causes changes in mesodermal gene expression, cell death and limb truncation. Fibroblast growth factor (FGF) family members are expressed in the AER and can rescue limb bud outgrowth after AER removal. Cells localized underneath the AER are maintained in an undifferentiated state by the FGFs produced by the AER. MAPK phosphatase 3 (mkp3) is a downstream effector of FGF8 signalling during limb bud development and is expressed in the distal limb mesenchyme. The present work evidences a gradient of mkp3 transcripts along the chick limb bud, in a distal to proximal direction. mkp3 transcription occurs only in the most distal limb bud cells and its mRNA gradient throughout the limb results from progressive mRNA decay. We show that FGF8-soaked beads induce ectopic mkp3 expression, indicating that AER-derived FGF8 protein may activate mkp3 in the distal mesenchyme.


Subject(s)
Gene Expression Regulation, Developmental , Limb Buds/metabolism , Protein Tyrosine Phosphatases/genetics , RNA Stability/physiology , Animals , Chick Embryo , Exons/genetics , Fibroblast Growth Factor 8/metabolism , Limb Buds/embryology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
14.
Development ; 133(9): 1635-44, 2006 May.
Article in English | MEDLINE | ID: mdl-16554364

ABSTRACT

We addressed the potential role of cell-laminin interactions during epaxial myotome formation in the mouse embryo. Assembly of the myotomal laminin matrix occurs as epaxial myogenic precursor cells enter the myotome. Most Myf5-positive and myogenin-negative myogenic precursor cells localise near assembled laminin, while myogenin-expressing cells are located either away from this matrix or in areas where it is being assembled. In Myf5(nlacZ/nlacZ) (Myf5-null) embryos, laminin, collagen type IV and perlecan are present extracellularly near myogenic precursor cells, but do not form a basement membrane and cells are not contained in the myotomal compartment. Unlike wild-type myogenic precursor cells, Myf5-null cells do not express the alpha6beta1 integrin, a laminin receptor, suggesting that integrin alpha6beta1-laminin interactions are required for myotomal laminin matrix assembly. Blocking alpha6beta1-laminin binding in cultured wild-type mouse embryo explants resulted in dispersion of Myf5-positive cells, a phenotype also seen in Myf5(nlacZ/nlacZ) embryos. Furthermore, inhibition of alpha6beta1 resulted in an increase in Myf5 protein and ectopic myogenin expression in dermomyotomal cells, suggesting that alpha6beta1-laminin interactions normally repress myogenesis in the dermomyotome. We conclude that Myf5 is required for maintaining alpha6beta1 expression on myogenic precursor cells, and that alpha6beta1 is necessary for myotomal laminin matrix assembly and cell guidance into the myotome. Engagement of laminin by alpha6beta1 also plays a role in maintaining the undifferentiated state of cells in the dermomyotome prior to their entry into the myotome.


Subject(s)
Embryo, Mammalian , Gene Expression Regulation, Developmental , Integrin alpha6beta1/metabolism , Laminin/metabolism , Muscle Development , Muscle, Skeletal/embryology , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Integrin alpha6beta1/genetics , Mice , Mice, Knockout , Models, Biological , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenin/metabolism , Organ Culture Techniques
15.
Dev Dyn ; 232(4): 1069-78, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15739233

ABSTRACT

Cells interact with the extracellular matrix through receptors, most commonly of the integrin family. We (Cachaco et al. [2003] Development 130:1659-1671) and others (Schwander et al. [2003] Dev. Cell 4:673-685) have demonstrated a role for beta1 integrins in mouse primary myogenesis. However, it is unclear what alpha subunits pair with beta1 during this process in vivo. Here, we determined alpha subunit expression patterns at embryonic day (E) 11.5-E14.5. Differentiated myotomal myocytes express all alpha subunits studied. As the muscle masses form both in trunk (E12.5) and limbs (E11.5-E12.5), laminin receptors alpha6beta1 and alpha7beta1 are undetectable, and an assembled laminin matrix is absent. Instead alpha1beta1, alpha4beta1, alpha5beta1, and an alpha v-containing integrin are expressed and unassembled laminin and fibronectin are abundant around myogenic cells. At E13.5-E14.5, alpha6beta1 and alpha7beta1 are expressed, and a laminin matrix forms around individual myotubes. Thus, myogenic cells change their integrin expression pattern during the course of primary myogenesis in the mouse, suggesting different roles for fibronectin- and laminin-containing matrices in this process.


Subject(s)
Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental/physiology , Integrins/biosynthesis , Muscle Development/physiology , Myoblasts/physiology , Animals , Mice , Myoblasts/cytology
16.
Dev Dyn ; 231(2): 402-15, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15366018

ABSTRACT

Integrins are cellular adhesion receptors that mediate signaling and play key roles in the development of multicellular organisms. However, their role in the cellular events leading to myotome formation is completely unknown. Here, we describe the expression patterns of the alpha1, alpha4, alpha5, alpha6, and alpha7 integrin subunits in the mouse myotome and correlate them with the expression of several differentiation markers. Our results indicate that these integrin subunits may be differentially involved in the various phases of myogenic determination and differentiation. A detailed characterization of the myogenic cell types expressing the alpha4 and alpha6 subunits showed a regionalization of the myotome and dermomyotome based on cell-adhesion properties. We conclude that alpha6beta1 may be an early marker of epaxial myogenic progenitor cells. In contrast, alpha4beta1 is up-regulated in the intercalated myotome after myocyte differentiation. Furthermore, alpha4beta1 is expressed in the hypaxial dermomyotome and is maintained by early hypaxial myogenic progenitor cells colonizing the myotome.


Subject(s)
Body Patterning , Embryo, Mammalian/physiology , Integrins/metabolism , Muscles/embryology , Protein Subunits/metabolism , Animals , Biomarkers , Cell Adhesion/physiology , Cell Differentiation/physiology , Embryo, Mammalian/anatomy & histology , Female , Gestational Age , In Situ Hybridization , Integrins/genetics , Mice , Muscles/cytology , Muscles/metabolism , MyoD Protein/metabolism , Pregnancy , Protein Subunits/genetics , RNA, Messenger/metabolism , Receptors, Laminin/metabolism , Stem Cells/cytology , Stem Cells/physiology
17.
Development ; 130(8): 1659-71, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12620989

ABSTRACT

Integrins are extracellular matrix receptors composed of alpha and beta subunits involved in cell adhesion, migration and signal transduction. The beta1 subunit has two isoforms, beta 1A ubiquitously expressed and beta 1D restricted to striated muscle. They are not functionally equivalent. Replacement of beta 1A by beta 1D (beta 1D knock-in) in the mouse leads to midgestation lethality on a 50% Ola/50% FVB background [Baudoin, C., Goumans, M. J., Mummery, C. and Sonnenberg, A. (1998). Genes Dev. 12, 1202-1216]. We crossed the beta 1D knock-in line into a less penetrant genetic background. This led to an attenuation of the midgestation lethality and revealed a second period of lethality around birth. Midgestation death was apparently not caused by failure in cell migration, but rather by abnormal placentation. The beta 1D knock-in embryos that survived midgestation developed until birth, but exhibited severely reduced skeletal muscle mass. Quantification of myotube numbers showed that substitution of beta 1A with beta 1D impairs primary myogenesis with no direct effect on secondary myogenesis. Furthermore, long-term primary myotube survival was affected in beta 1D knock-in embryos. Finally, overexpression of beta 1D in C2C12 cells impaired myotube formation while overexpression of beta 1A primarily affected myotube maturation. Together these results demonstrate for the first time distinct roles for beta1 integrins in primary versus secondary myogenesis and that the beta 1A and beta 1D variants are not functionally equivalent in this process.


Subject(s)
Cell Differentiation/physiology , Integrin beta1/genetics , Integrin beta1/metabolism , Muscle Development/physiology , Animals , Apoptosis/physiology , Cell Line , Cell Movement/physiology , Cell Survival , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , In Situ Hybridization , Mice , Mice, Transgenic
18.
Gene Expr Patterns ; 2(1-2): 133-6, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12617851

ABSTRACT

Cell-extracellular matrix interactions play crucial roles in limb muscle development but practically nothing is known on what integrins are involved before the differentiation of muscle precursor cells (MPCs) in the limb muscle masses. In this study we determine the expression patterns of integrins during early forelimb muscle development in the mouse. alpha6beta1 integrin is downregulated in the lateral dermomyotome when delamination of MPCs occurs. In late E9.5 embryos, alpha1beta1 and alpha5beta1 are expressed in a pattern very similar to pax3, which marks MPCs migrating to the limb bud. After myf5 upregulation in the limb bud, alpha1beta1 and alpha5beta1 expression is maintained and the alpha4beta1 integrin starts being expressed.


Subject(s)
Gene Expression Regulation, Developmental , Integrin beta1/genetics , Muscle, Skeletal/metabolism , Animals , Extremities/embryology , Extremities/physiology , Gene Expression Profiling , Immunohistochemistry , In Situ Hybridization , Integrin beta1/biosynthesis , Mice/embryology , Muscle, Skeletal/embryology
19.
Mech Dev ; 119 Suppl 1: S131-4, 2002 Dec.
Article in English | MEDLINE | ID: mdl-14516674

ABSTRACT

Cell-extracellular matrix interactions play crucial roles in limb muscle development but practically nothing is known on what integrins are involved before the differentiation of muscle precursor cells (MPCs) in the limb muscle masses. In this study we determine the expression patterns of integrins during early forelimb muscle development in the mouse. alpha6beta1 integrin is downregulated in the lateral dermomyotome when delamination of MPCs occurs. In late E9.5 embryos, alpha1beta1 and alpha5beta1 are expressed in a pattern very similar to pax3, which marks MPCs migrating to the limb bud. After myf5 upregulation in the limb bud, alpha1beta1 and alpha5beta1 expression is maintained and the alpha4beta1 integrin starts being expressed.


Subject(s)
Gene Expression Regulation, Developmental , Muscle Development , Animals , Extremities , Integrins/metabolism , Limb Buds/cytology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...