Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 626: 21-29, 2022 10 20.
Article in English | MEDLINE | ID: mdl-35970041

ABSTRACT

We previously showed that the adaptive response of BRAFV600-mutated melanoma cells to BRAF inhibition emerges from a subpopulation of cells expressing an intermittent lower level of the mRNA-binding protein HuR. In this study, following initial overexpression experiments in which we confirm our previous results, we use wild-type and mutants HuR full-length mRNA constructs and in vivo-interacting assays and demonstrate that a highly conserved interferon-γ-activated inhibitor of translation (GAIT)-like motif located upstream of the GU-rich elements of HuR major polyadenylation site (PAS2), interacts with constituents of the GAIT complex and affects HuR post-transcriptional expression regulation. Knockdown of the ribosomal protein L13a or the inhibition of the DAPK1-ZIPK axis involved in L13a phosphorylation, reduces the proportion of HuRLow cells at steady-state and attenuates the adaptive response of the whole melanoma-cell population to BRAF inhibition. These results have further potential therapeutic implications for disease conditions associated with HuR insufficient expression.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , 3' Untranslated Regions , ELAV Proteins/genetics , ELAV Proteins/metabolism , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Humans , Melanoma/drug therapy , Melanoma/genetics , Phosphorylation , Polyadenylation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA, Messenger/genetics
2.
Neoplasia ; 23(8): 775-782, 2021 08.
Article in English | MEDLINE | ID: mdl-34237504

ABSTRACT

The mechanisms of adaptive resistance to genetic-based targeted therapies of solid malignancies have been the subject of intense research. These studies hold great promise for finding co-targetable hub/pathways which in turn would control the downstream non-genetic mechanisms of adaptive resistance. Many such mechanisms have been described in the paradigmatic BRAF-mutated melanoma model of adaptive response to BRAF inhibition. Currently, a major challenge for these mechanistic studies is to confirm in vivo, at the single-cell proteomic level, the existence of dependencies between the co-targeted hub/pathways and their downstream effectors. Moreover, the drug-induced in vivo modulation of these dependencies needs to be demonstrated. Here, we implement such single-cell-based in vivo expression dependency quantification using immunohistochemistry (IHC)-based analyses of sequential biopsies in two xenograft models. These mimic phase 2 and 3 trials in our own therapeutic strategy to prevent the adaptive response to BRAF inhibition. In this mechanistic model, the dependencies between the targeted Li2CO3-inducible hub HuR and the resistance effectors are more likely time-shifted and transient since the minority of HuRLow cells, which act as a reservoir of adaptive plasticity, switch to a HuRHigh state as they paradoxically proliferate under BRAF inhibition. Nevertheless, we show that a copula/kernel density estimator (KDE)-based quantification of mutual information (MI) efficiently captures, at the individual level, the dependencies between HuR and two relevant resistance markers pERK and EGFR, and outperforms classic expression correlation coefficients. Ultimately, the validation of MI as a predictive IHC-based metric of response to our therapeutic strategy will be carried in clinical trials.


Subject(s)
Melanoma/drug therapy , Models, Theoretical , Molecular Targeted Therapy , Algorithms , Animals , Biomarkers, Tumor , Disease Management , Disease Models, Animal , Disease Susceptibility , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Melanoma/diagnosis , Melanoma/etiology , Melanoma/metabolism , Mice , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Single-Cell Analysis/methods , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Mol Ther Nucleic Acids ; 17: 63-77, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31226519

ABSTRACT

Nucleic acid aptamers are often referred to as chemical antibodies. Because they possess several advantages, like their smaller size, temperature stability, ease of chemical modification, lack of immunogenicity and toxicity, and lower cost of production, aptamers are promising tools for clinical applications. Aptamers against cell surface protein biomarkers are of particular interest for cancer diagnosis and targeted therapy. In this study, we identified and characterized RNA aptamers targeting cells expressing integrin α5ß1. This αß heterodimeric cell surface receptor is implicated in tumor angiogenesis and solid tumor aggressiveness. In glioblastoma, integrin α5ß1 expression is associated with an aggressive phenotype and a decrease in patient survival. We used a complex and original hybrid SELEX (selective evolution of ligands by exponential enrichment) strategy combining protein-SELEX cycles on the recombinant α5ß1 protein, surrounded by cell-SELEX cycles using two different cell lines. We identified aptamer H02, able to differentiate, in cyto- and histofluorescence assays, glioblastoma cell lines, and tissues from patient-derived tumor xenografts according to their α5 expression levels. Aptamer H02 is therefore an interesting tool for glioblastoma tumor characterization.

4.
Oncotarget ; 7(38): 62194-62207, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27613837

ABSTRACT

The Wnt/beta catenin pathway has been highlighted as an important player of brain tumors aggressiveness and resistance to therapies. Increasing knowledges of the regulation of beta-catenin transactivation point out its hub position in different pathophysiological outcomes in glioma such as survival and migration. Crosstalks between integrins and beta-catenin pathways have been suggested in several tumor tissues. As we demonstrated earlier that α5ß1 integrin may be considered as a therapeutic target in high grade glioma through its contribution to glioma cell migration and resistance to chemotherapy, we addressed here the potential relationship between α5ß1 integrin and beta-catenin activation in glioma cells. We demonstrated that overexpression and activation by fibronectin of α5ß1 integrin allowed the transactivation of beta-catenin gene targets included in an EMT-like program that induced an increase in cell migration. Hampering of beta catenin activation and cell migration could be similarly achieved by a specific integrin antagonist. In addition we showed that α5ß1 integrin/AKT axis is mainly involved in these processes. However, blockade of beta-catenin by XAV939 (tankyrase inhibitor leading to beta-catenin degradation) did not synergize with p53 activation aiming to cell apoptosis as was the case with integrin antagonists. We therefore propose a dual implication of α5ß1 integrin/AKT axis in glioma cell resistance to therapies and migration each supported by different signaling pathways. Our data thus suggest that α5ß1 integrin may be added to the growing list of beta-catenin modulators and provide new evidences to assign this integrin as a valuable target to fight high grade glioma.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Glioma/pathology , Integrin alpha5beta1/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Apoptosis , Brain Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Fibronectins/metabolism , Glioma/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Immunohistochemistry , Integrin alpha5beta1/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Tankyrases/antagonists & inhibitors , Transcriptional Activation/drug effects , beta Catenin/antagonists & inhibitors
5.
Cancer Lett ; 376(2): 328-38, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27063097

ABSTRACT

Glioblastoma multiform (GBM) is the most common and most aggressive primary brain tumor. The fibronectin receptor, α5 integrin is a pertinent novel therapeutic target. Despite numerous data showing that α5 integrin support tumor cell migration and invasion, it has been reported that α5 integrin can also limit cell dispersion by increasing cell-cell interaction. In this study, we showed that α5 integrin was involved in cell-cell interaction and gliomasphere formation. α5-mediated cell-cell cohesion limited cell dispersion from spheroids in fibronectin-poor microenvironment. However, in fibronectin-rich microenvironment, α5 integrin promoted cell dispersion. Ligand-occupied α5 integrin and fibronectin were distributed in fibril-like pattern at cell-cell junction of evading cells, forming cell-cell fibrillar adhesions. Activated focal adhesion kinase was not present in these adhesions but was progressively relocalized with α5 integrin as cell migrates away from the spheroids. α5 integrin function in GBM appears to be more complex than previously suspected. As GBM overexpressed fibronectin, it is most likely that in vivo, α5-mediated dissemination from the tumor mass overrides α5-mediated tumor cell cohesion. In this respect, α5-integrin antagonists may be useful to limit GBM invasion in brain parenchyma.


Subject(s)
Brain Neoplasms/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Cell-Matrix Junctions/metabolism , Extracellular Matrix/metabolism , Integrin alphaV/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Humans , Integrin alphaV/genetics , Neoplasm Invasiveness , RNA Interference , Signal Transduction , Spheroids, Cellular , Time Factors , Transfection
6.
Oncotarget ; 6(39): 41884-901, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26474461

ABSTRACT

Distant metastases arise in 20-30% of patients with squamous cell carcinoma of the head and neck (HNSCC) in the 2 years following treatment. Therapeutic options are limited and the outcome of the patients is poor. The identification of predictive biomarkers of patient at risk for distant metastasis and therapies are urgently needed. We previously identified a clinical subgroup, called "R1" characterized by high propensity for rapid distant metastasis. Here, we showed that "R1" patients do not or at very low level express caveolin-1 (Cav1). Low or no expression of Cav1 is of bad prognosis. Disappearance of Cav1 enables cells to undergo epithelial-mesenchymal transition (EMT). EMT is associated with enhanced migration and invasion. Our study uncovered a new target, α5ß1 integrin. Targeting α5ß1 integrins might not only prevent metastasis of HNSCC but also delay the development of the primary tumor by reducing tumor cell viability. Cav1 detection might be taken into consideration in the future in the clinic not only to identify patients at high risk of metastasis but also to select patient who might benefit from an anti-integrin therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Caveolin 1/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Head and Neck Neoplasms/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/secondary , Carcinoma, Squamous Cell/therapy , Caveolin 1/genetics , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , Integrin alpha5beta1/metabolism , Kaplan-Meier Estimate , Prognosis , RNA Interference , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Time Factors , Transfection
7.
Biochim Biophys Acta ; 1840(9): 2978-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24810979

ABSTRACT

BACKGROUND: Integrins are extracellular matrix receptors involved in several pathologies. Despite homologies between the RGD-binding α5ß1 and αvß3 integrins, selective small antagonists for each heterodimer have been proposed. Herein, we evaluated the effects of such small antagonists in a cellular context, the U87MG cell line, which express both integrins. The aim of the study was to determine if fibronectin-binding integrin antagonists are able to impact on cell adhesion and migration in relationships with their defined affinity and selectivity for α5ß1 and αvß3/ß5 purified integrins. METHODS: Small antagonists were either selective for α5ß1 integrin, for αvß3/ß5 integrin or non-selective. U87MG cell adhesion was evaluated on fibronectin or vitronectin. Migration assays included wound healing recovery and single cell tracking experiments. U87MG cells stably manipulated for the expression of α5 integrin subunit were used to explore the impact of α5ß1 integrin in the biological assays. RESULTS: U87MG cell adhesion on fibronectin or vitronectin was respectively dependent on α5ß1 or αvß3/ß5 integrin. Wound healing migration was dependent on both integrins. However U87MG single cell migration was highly dependent on α5ß1 integrin and was inhibited selectively by α5ß1 integrin antagonists but increased by αvß3/ß5 integrin antagonists. CONCLUSIONS: We provide a rationale for testing new integrin ligands in a cell-based assay to characterize more directly their potential inhibitory effects on integrin cellular functions. GENERAL SIGNIFICANCE: Our data highlight a single cell tracking assay as a powerful cell-based test which may help to characterize true functional integrin antagonists that block α5ß1 integrin-dependent cell migration.


Subject(s)
Antineoplastic Agents , Glioma/drug therapy , Integrin alpha5beta1/antagonists & inhibitors , Integrin alphaVbeta3/antagonists & inhibitors , Integrin beta Chains , Neoplasm Proteins/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Drug Screening Assays, Antitumor , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Integrin alpha5beta1/biosynthesis , Integrin alpha5beta1/genetics , Integrin alphaVbeta3/biosynthesis , Integrin alphaVbeta3/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics
8.
PLoS One ; 9(3): e92059, 2014.
Article in English | MEDLINE | ID: mdl-24642576

ABSTRACT

Focal adhesion kinase (FAK) plays an important role in signal transduction pathways initiated at sites of integrin-mediated cell adhesion to the extracellular matrix. Thus, FAK is involved in many aspects of the metastatic process including adhesion, migration and invasion. Recently, several small molecule inhibitors which target FAK catalytic activity have been developed by pharmaceutical companies. The current study was aimed at addressing whether inhibiting FAK targeting to focal adhesions (FA) represents an efficient alternative strategy to inhibit FAK downstream pathways. Using a mutagenesis approach to alter the targeting domain of FAK, we constructed a FAK mutant that fails to bind paxillin. Inhibiting FAK-paxillin interactions led to a complete loss of FAK localization at FAs together with reduced phosphorylation of FAK and FAK targets such as paxillin and p130Cas. This in turn resulted in altered FA dynamics and inhibition of cell adhesion, migration and invasion. Moreover, the migration properties of cells expressing the FAK mutant were reduced as compared to FAK-/- cells. This was correlated with a decrease in both phospho-Src and phospho-p130Cas levels at FAs. We conclude that targeting FAK-paxillin interactions is an efficient strategy to reduce FAK signalling and thus may represent a target for the development of new FAK inhibitors.


Subject(s)
Crk-Associated Substrate Protein/genetics , Fibroblasts/drug effects , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation , Paxillin/genetics , src-Family Kinases/genetics , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Crk-Associated Substrate Protein/metabolism , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Mice , Mutagenesis, Site-Directed , Paxillin/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Recombinant Fusion Proteins , Signal Transduction , src-Family Kinases/metabolism
9.
Cancer Lett ; 336(2): 307-18, 2013 Aug 19.
Article in English | MEDLINE | ID: mdl-23523610

ABSTRACT

Integrins emerge nowadays as crucial actors of tumor aggressiveness and resistance to therapies. Integrin α5ß1, the fibronectin receptor, determines malignant properties of colon carcinoma which is one of the most important causes of cancer-related deaths in the world. Here we show that inhibition of α5 integrin subunit expression by siRNA or α5ß1 integrin function by specific antagonist affects the survival of HCT116 colon cancer cells. We also evidence that pharmacological reactivation of the tumor suppressor p53 by Nutlin-3a inhibits specifically the expression of the α5 integrin subunit both at the transcriptional and protein level. Inversely repression of α5 integrin modulates p53 activity. A clear relationship between p53 activation by Nutlin-3a, α5 repression and cell survival is shown. No such effects are obtained in cells lacking p53 or when another non-genotoxic activator of p53, RITA, is used. Our results emphasize the crucial role of α5ß1 integrin in colon tumors. Data also suggest that interfering with the integrin α5ß1 through the reactivation of p53 by Nutlin-3a may be of valuable interest as a new therapeutic option for colon tumors expressing high level of the integrin and a wild type p53.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Imidazoles/pharmacology , Integrin alpha5/biosynthesis , Piperazines/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Integrin alpha5/genetics , Molecular Targeted Therapy , Signal Transduction/drug effects , Transcription, Genetic , Transfection
10.
Cancer Res ; 72(14): 3463-70, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22593187

ABSTRACT

Integrins play a role in the resistance of advanced cancers to radiotherapy and chemotherapy. In this study, we show that high expression of the α5 integrin subunit compromises temozolomide-induced tumor suppressor p53 activity in human glioblastoma cells. We found that depletion of the α5 integrin subunit increased p53 activity and temozolomide sensitivity. However, when cells were treated with the p53 activator nutlin-3a, the protective effect of α5 integrin on p53 activation and cell survival was lost. In a functional p53 background, nutlin-3a downregulated the α5 integrin subunit, thereby increasing the cytotoxic effect of temozolomide. Clinically, α5ß1 integrin expression was associated with a more aggressive phenotype in brain tumors, and high α5 integrin gene expression was associated with decreased survival of patients with high-grade glioma. Taken together, our findings indicate that negative cross-talk between α5ß1 integrin and p53 supports glioma resistance to temozolomide, providing preclinical proof-of-concept that α5ß1 integrin represents a therapeutic target for high-grade brain tumors. Direct activation of p53 may remain a therapeutic option in the subset of patients with high-grade gliomas that express both functional p53 and a high level of α5ß1 integrin.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Glioblastoma/metabolism , Integrin alpha5beta1/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Dacarbazine/therapeutic use , Humans , Imidazoles/pharmacology , Integrin alpha5beta1/metabolism , Mice , Piperazines/pharmacology , Temozolomide , Treatment Outcome
11.
Mol Biol Cell ; 22(7): 964-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21289086

ABSTRACT

Cell migration is a highly complex process that requires the coordinated formation of membrane protrusion and focal adhesions (FAs). Focal adhesion kinase (FAK), a major signaling component of FAs, is involved in the disassembly process of FAs through phosphorylation and dephosphorylation of its tyrosine residues, but the role of such phosphorylations in nascent FA formation and turnover near the cell front and in cell protrusion is less well understood. In the present study, we demonstrate that, depending on the phosphorylation status of Tyr-925 residue, FAK modulates cell migration via two specific mechanisms. FAK⁻/⁻ mouse embryonic fibroblasts (MEFs) expressing nonphosphorylatable Y925F-FAK show increased interactions between FAK and unphosphorylated paxillin, which lead to FA stabilization and thus decreased FA turnover and reduced cell migration. Conversely, MEFs expressing phosphomimetic Y925E-FAK display unchanged FA disassembly rates, show increase in phosphorylated paxillin in FAs, and exhibit increased formation of nascent FAs at the cell leading edges. Moreover, Y925E-FAK cells present enhanced cell protrusion together with activation of the p130(CAS)/Dock180/Rac1 signaling pathway. Together, our results demonstrate that phosphorylation of FAK at Tyr-925 is required for FAK-mediated cell migration and cell protrusion.


Subject(s)
Cell Surface Extensions/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , Cells, Cultured , Crk-Associated Substrate Protein/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Mice , Mice, Knockout , Paxillin/metabolism , Phosphorylation , rac1 GTP-Binding Protein/metabolism
12.
Cell Signal ; 23(1): 228-38, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20837140

ABSTRACT

The human NPY Y(1) receptor undergoes fast agonist-induced internalization via clathrin-coated pits then recycles back to the cell membrane. In an attempt to identify the molecular determinants involved in this process, we studied several C-terminal truncation mutants tagged with EFGP. In the absence of agonist, Y(1) receptors lacking the last 32 C-terminal amino acids (Y(1)Δ32) are constitutively internalized, unlike full-length Y(1) receptors. At steady state, internalized Y(1)Δ32 receptors co-localize with transferrin, a marker of early and recycling endosomes. Inhibition of constitutive internalization of Y(1)Δ32 receptors by hypertonic sucrose or by co-expression of Rab5aS34N, a dominant negative form of the small GTPase Rab5a or depletion of all three isoforms of Rab5 indicates the involvement of clathrin-coated pits. In contrast, a truncated receptor lacking the last 42 C-terminal amino acids (Y(1)Δ42) does not constitutively internalize, consistent with the possibility that there is a molecular determinant responsible for constitutive internalization located in the last 10 amino acids of Y(1)Δ32 receptors. We show that the agonist-independent internalization of Y(1)Δ32 receptors involves a tyrosine-based motif YXXΦ. The potential role of this motif in the behaviour of full-length Y(1) receptors has also been explored. Our results indicate that a C-terminal tyrosine-based motif is critical for the constitutive internalization of truncated Y(1)Δ32 receptors. We suggest that this motif is masked in full-length Y(1) receptors which do not constitutively internalize in the absence of agonist.


Subject(s)
Receptors, Neuropeptide Y/metabolism , Tyrosine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Clathrin/chemistry , Clathrin/metabolism , HEK293 Cells , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuropeptide Y/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics , Signal Transduction , Transferrin/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
13.
Dev Cell ; 18(4): 643-54, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20412778

ABSTRACT

Muscle progenitors, labeled by the transcription factor Pax7, are responsible for muscle growth during development. The signals that regulate the muscle progenitor number during myogenesis are unknown. We show, through in vivo analysis, that Bmp signaling is involved in regulating fetal skeletal muscle growth. Ectopic activation of Bmp signaling in chick limbs increases the number of fetal muscle progenitors and fibers, while blocking Bmp signaling reduces their numbers, ultimately leading to small muscles. The Bmp effect that we observed during fetal myogenesis is diametrically opposed to that previously observed during embryonic myogenesis and that deduced from in vitro work. We also show that Bmp signaling regulates the number of satellite cells during development. Finally, we demonstrate that Bmp signaling is active in a subpopulation of fetal progenitors and satellite cells at the extremities of muscles. Overall, our results show that Bmp signaling plays differential roles in embryonic and fetal myogenesis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , Muscles/embryology , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction , Stem Cells/cytology , Animals , Cell Differentiation , Chick Embryo , In Situ Hybridization , Mice , Models, Biological , Muscle, Skeletal/metabolism , PAX7 Transcription Factor/metabolism , Tendons/pathology
14.
Neuromuscul Disord ; 17(2): 148-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17236769

ABSTRACT

Limb-girdle muscular dystrophy type 2A (LGMD2A) is an autosomal recessive muscular disorder caused by mutations in the gene coding for calpain 3, a calcium-dependent protease. We developed an in vitro assay that can detect the proteolytic activity of calpain 3 in a muscle sample. This assay is based on the use of an inactive calpain 3 as a substrate for active calpain 3 molecules. A total of 79 human biopsies have been analysed using an unbiased single blind method. Results were confronted with the molecular diagnosis for confirmation. Proteolytic activity was either reduced or absent in 68% of LGMD2A biopsies. In the remaining 32%, normal proteolytic activity was found despite the presence of calpain 3 mutation(s), suggesting that other calpain 3 properties might be impaired to give rise to the LGMD2A phenotype. Our assay is easily adaptable to routine and appears to be more sensitive than common analysis by immunodetection.


Subject(s)
Calpain/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscular Dystrophies, Limb-Girdle/enzymology , Animals , Blotting, Western , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Heterozygote , Humans , Mice , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , NIH 3T3 Cells , Phenotype , Reproducibility of Results , Tissue Banks , Transfection
15.
Mol Ther ; 13(2): 250-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16290124

ABSTRACT

Calpainopathy (limb-girdle muscular dystrophy type 2A, LGMD2A) is a recessive muscular disorder caused by deficiency in the calcium-dependent cysteine protease calpain 3. To date, no treatment exists for this disease. We evaluated the potential of recombinant adeno-associated virus (rAAV) vectors for gene therapy in a murine model for LGMD2A. To drive the expression of calpain 3, we used rAAV2/1 pseudotyped vectors and muscle-specific promoters to avoid calpain 3 cell toxicity. We report efficient and stable transgene expression in muscle with restoration of the proteolytic activity and without evident toxicity. In addition, calpain 3 was correctly targeted to the sarcomere. Moreover, its presence resulted in improvement of the histological features and in therapeutic efficacy at the physiological levels, including correction of atrophy and full rescue of the contractile force deficits. Our results establish the feasibility of AAV-mediated calpain 3 gene transfer as a therapeutic approach.


Subject(s)
Calpain/genetics , Calpain/therapeutic use , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Muscle Proteins/genetics , Muscle Proteins/therapeutic use , Muscular Dystrophies, Limb-Girdle/therapy , Animals , Calpain/biosynthesis , Calpain/deficiency , Disease Models, Animal , Enzyme Activation/genetics , Enzyme Stability/genetics , Genetic Vectors/therapeutic use , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/biosynthesis , Muscle Proteins/deficiency , Muscle, Skeletal/enzymology , Muscular Dystrophies, Limb-Girdle/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...