Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 15(12): e17405, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37927228

ABSTRACT

Fibrosis is associated with compromised muscle functionality in Duchenne muscular dystrophy (DMD). We report observations with tissues from dystrophic patients and mice supporting a model to explain fibrosis in DMD, which relies on the crosstalk between the complement and the WNT signaling pathways and the functional interactions of two cellular types. Fibro-adipogenic progenitors and macrophages, which populate the inflamed dystrophic muscles, act as a combinatorial source of WNT activity by secreting distinct subunits of the C1 complement complex. The resulting aberrant activation of the WNT signaling in responsive cells, such as fibro-adipogenic progenitors, contributes to fibrosis. Indeed, pharmacological inhibition of the C1r/s subunits in a murine model of DMD mitigated the activation of the WNT signaling pathway, reduced the fibrogenic characteristics of the fibro-adipogenic progenitors, and ameliorated the dystrophic phenotype. These studies shed new light on the molecular and cellular mechanisms responsible for fibrosis in muscular dystrophy and open to new therapeutic strategies.


Subject(s)
Muscle, Skeletal , Muscular Dystrophy, Duchenne , Humans , Mice , Animals , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Wnt Signaling Pathway , Fibrosis , Mice, Inbred mdx
5.
Front Physiol ; 13: 834705, 2022.
Article in English | MEDLINE | ID: mdl-35431987

ABSTRACT

Skeletal muscle is composed of syncytial muscle fibers, and by various mononucleated cellular types, such as muscle stem cells, immune cells, interstitial and stromal progenitors. These cell populations play a crucial role during muscle regeneration, and alterations of their phenotypic properties have been associated with defective repair and fibrosis in aging and dystrophic muscle. Studies involving in vivo gene modulation are valuable to investigate the mechanisms underlining cell function and dysfunction in complex pathophysiological settings. Electro-enhanced transfer of plasmids using square-wave generating devices represents a cost-effective approach that is widely used to transport DNA to muscle fibers efficiently. Still, it is not clear if this method can also be applied to mononuclear cells present in muscle. We demonstrate here that it is possible to efficiently deliver DNA into different muscle-resident cell populations in vivo. We evaluated the efficiency of this approach not only in healthy muscle but also in muscles of aging and dystrophic animal models. As an exemplificative application of this method, we used a strategy relying on a reporter gene-based plasmid containing regulatory sequences from the collagen 1 locus, and we determined collagen expression in various cell types reportedly involved in the production of fibrotic tissue in the dystrophic settings. The results enclosed in this manuscript reveal the suitability in applying electro-enhanced transfer of plasmid DNA to mononucleated muscle-resident cells to get insights into the molecular events governing diseased muscle physiology.

6.
Front Genet ; 13: 1056114, 2022.
Article in English | MEDLINE | ID: mdl-36685855

ABSTRACT

In 2002 we published an article describing a population of vessel-associated progenitors that we termed mesoangioblasts (MABs). During the past decade evidence had accumulated that during muscle development and regeneration things may be more complex than a simple sequence of binary choices (e.g., dorsal vs. ventral somite). LacZ expressing fibroblasts could fuse with unlabelled myoblasts but not among themselves or with other cell types. Bone marrow derived, circulating progenitors were able to participate in muscle regeneration, though in very small percentage. Searching for the embryonic origin of these progenitors, we identified them as originating at least in part from the embryonic aorta and, at later stages, from the microvasculature of skeletal muscle. While continuing to investigate origin and fate of MABs, the fact that they could be expanded in vitro (also from human muscle) and cross the vessel wall, suggested a protocol for the cell therapy of muscular dystrophies. We tested this protocol in mice and dogs before proceeding to the first clinical trial on Duchenne Muscular Dystrophy patients that showed safety but minimal efficacy. In the last years, we have worked to overcome the problem of low engraftment and tried to understand their role as auxiliary myogenic progenitors during development and regeneration.

7.
Antioxidants (Basel) ; 10(11)2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34829643

ABSTRACT

Aging is characterized by a progressive increase in oxidative stress, which favors lipid peroxidation and the formation of cholesterol oxide derivatives, including 7ß-hydroxycholesterol (7ß-OHC). This oxysterol, which is known to trigger oxidative stress, inflammation, and cell death, could contribute to the aging process and age-related diseases, such as sarcopenia. Identifying molecules or mixtures of molecules preventing the toxicity of 7ß-OHC is therefore an important issue. This study consists of determining the chemical composition of Tunisian Pistacia lentiscus L. seed oil (PLSO) used in the Tunisian diet and evaluating its ability to counteract the cytotoxic effects induced by 7ß-OHC in murine C2C12 myoblasts. The effects of 7ß-OHC (50 µM; 24 h), associated or not with PLSO, were studied on cell viability, oxidative stress, and on mitochondrial and peroxisomal damages induction. α-Tocopherol (400 µM) was used as the positive control for cytoprotection. Our data show that PLSO is rich in bioactive compounds; it contains polyunsaturated fatty acids, and several nutrients with antioxidant properties: phytosterols, α-tocopherol, carotenoids, flavonoids, and phenolic compounds. When associated with PLSO (100 µg/mL), the 7ß-OHC-induced cytotoxic effects were strongly attenuated. The cytoprotection was in the range of those observed with α-tocopherol. This cytoprotective effect was characterized by prevention of cell death and organelle dysfunction (restoration of cell adhesion, cell viability, and plasma membrane integrity; prevention of mitochondrial and peroxisomal damage) and attenuation of oxidative stress (reduction in reactive oxygen species overproduction in whole cells and at the mitochondrial level; decrease in lipid and protein oxidation products formation; and normalization of antioxidant enzyme activities: glutathione peroxidase (GPx) and superoxide dismutase (SOD)). These results provide evidence that PLSO has similar antioxidant properties than α-tocopherol used at high concentration and contains a mixture of molecules capable to attenuate 7ß-OHC-induced cytotoxic effects in C2C12 myoblasts. These data reinforce the interest in edible oils associated with the Mediterranean diet, such as PLSO, in the prevention of age-related diseases, such as sarcopenia.

8.
Oncogene ; 40(31): 4955-4966, 2021 08.
Article in English | MEDLINE | ID: mdl-34172934

ABSTRACT

A prototypic pediatric cancer that frequently shows activation of RAS signaling is embryonal rhabdomyosarcoma (ERMS). ERMS also show aberrant Hedgehog (HH)/GLI signaling activity and can be driven by germline mutations in this pathway. We show, that in ERMS cell lines derived from sporadic tumors i.e. from tumors not caused by an inherited genetic variant, HH/GLI signaling plays a subordinate role, because oncogenic mutations in HRAS, KRAS, or NRAS (collectively named oncRAS) inhibit the main HH target GLI1 via the MEK/ERK-axis, but simultaneously increase proliferation and tumorigenicity. oncRAS also modulate expression of stem cell markers in an isoform- and context-dependent manner. In Hh-driven murine ERMS that are caused by a Patched mutation, oncHRAS and mainly oncKRAS accelerate tumor development, whereas oncNRAS induces a more differentiated phenotype. These features occur when the oncRAS mutations are induced at the ERMS precursor stage, but not when induced in already established tumors. Moreover, in contrast to what is seen in human cell lines, oncRAS mutations do not alter Hh signaling activity and marginally affect expression of stem cell markers. Together, all three oncRAS mutations seem to be advantageous for ERMS cell lines despite inhibition of HH signaling and isoform-specific modulation of stem cell markers. In contrast, oncRAS mutations do not inhibit Hh-signaling in Hh-driven ERMS. In this model, oncRAS mutations seem to be advantageous for specific ERMS populations that occur within a specific time window during ERMS development. In addition, this window may be different for individual oncRAS isoforms, at least in the mouse.


Subject(s)
Disease Susceptibility , Genes, ras , Neoplasms/etiology , Neoplasms/metabolism , Protein Isoforms/genetics , Age Factors , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Mice , Mice, Knockout , Mutation , Neoplasms/pathology , Neoplastic Stem Cells , Oncogenes , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
9.
Biomedicines ; 9(1)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445754

ABSTRACT

Bone and muscle have been recognized as endocrine organs since they produce and secrete "hormone-like factors" that can mutually influence each other and other tissues, giving rise to a "bone-muscle crosstalk". In our study, we made use of myogenic (C2C12 cells) and osteogenic (2T3 cells) cell lines to investigate the effects of muscle cell-produced factors on the maturation process of osteoblasts. We found that the myogenic medium has inhibitory effects on bone cell differentiation and we identified sclerostin as one of the myokines produced by muscle cells. Sclerostin is a secreted glycoprotein reportedly expressed by bone/cartilage cells and is considered a negative regulator of bone growth due to its role as an antagonist of the Wnt/ß-catenin pathway. Given the inhibitory role of sclerostin in bone, we analyzed its expression by muscle cells and how it affects bone formation and homeostasis. Firstly, we characterized and quantified sclerostin synthesis by a myoblast cell line (C2C12) and by murine primary muscle cells by Western blotting, real-time PCR, immunofluorescence, and ELISA assay. Next, we investigated in vivo production of sclerostin in distinct muscle groups with different metabolic and mechanical loading characteristics. This analysis was done in mice of different ages (6 weeks, 5 and 18 months after birth) and revealed that sclerostin expression is dynamically modulated in a muscle-specific way during the lifespan. Finally, we transiently expressed sclerostin in the hind limb muscles of young mice (2 weeks of age) via in vivo electro-transfer of a plasmid containing the SOST gene in order to investigate the effects of muscle-specific overproduction of the protein. Our data disclosed an inhibitory role of the muscular sclerostin on the bones adjacent to the electroporated muscles. This observation suggests that sclerostin released by skeletal muscle might synergistically interact with osseous sclerostin and potentiate negative regulation of osteogenesis possibly by acting in a paracrine/local fashion. Our data point out a role for muscle as a new source of sclerostin.

10.
Commun Biol ; 4(1): 62, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33437023

ABSTRACT

Recent computational advancements in the simulation of biochemical processes allow investigating the mechanisms involved in protein regulation with realistic physics-based models, at an atomistic level of resolution. These techniques allowed us to design a drug discovery approach, named Pharmacological Protein Inactivation by Folding Intermediate Targeting (PPI-FIT), based on the rationale of negatively regulating protein levels by targeting folding intermediates. Here, PPI-FIT was tested for the first time on the cellular prion protein (PrP), a cell surface glycoprotein playing a key role in fatal and transmissible neurodegenerative pathologies known as prion diseases. We predicted the all-atom structure of an intermediate appearing along the folding pathway of PrP and identified four different small molecule ligands for this conformer, all capable of selectively lowering the load of the protein by promoting its degradation. Our data support the notion that the level of target proteins could be modulated by acting on their folding pathways, implying a previously unappreciated role for folding intermediates in the biological regulation of protein expression.


Subject(s)
Drug Evaluation, Preclinical/methods , Prion Diseases/drug therapy , Prion Proteins/chemistry , Prion Proteins/metabolism , Protein Folding , Animals , Binding Sites , Computer Simulation , Endoplasmic Reticulum/metabolism , Fibroblasts , HEK293 Cells , Humans , Ligands , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Reproducibility of Results
11.
J Clin Invest ; 130(11): 5652-5664, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32946430

ABSTRACT

Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.


Subject(s)
Cell Differentiation , Muscular Dystrophies , Myoblasts , Pluripotent Stem Cells , Stem Cell Transplantation , Humans , Muscle, Skeletal/physiology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/therapy , Myoblasts/metabolism , Myoblasts/pathology , Myoblasts/transplantation , Myocardium/metabolism , Myocardium/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Pluripotent Stem Cells/transplantation , Regeneration
13.
Front Physiol ; 9: 352, 2018.
Article in English | MEDLINE | ID: mdl-29674978

ABSTRACT

The formation and activity of mammalian tissues entail finely regulated processes, involving the concerted organization and interaction of multiple cell types. In recent years the prospective isolation of distinct progenitor and stem cell populations has become a powerful tool in the hands of developmental biologists and has rendered the investigation of their intrinsic properties possible. In this protocol, we describe how to purify progenitors with different lineage history and degree of differentiation from embryonic and fetal skeletal muscle by fluorescence-activated cell sorting (FACS). The approach takes advantage of a panel of murine strains expressing fluorescent reporter genes specifically in the myogenic progenitors. We provide a detailed description of the dissection procedures and of the enzymatic dissociation required to maximize the yield of mononucleated cells for subsequent FACS-based purification. The procedure takes ~6-7 h to complete and allows for the isolation and the subsequent molecular and phenotypic characterization of developmental myogenic progenitors.

14.
Proc Natl Acad Sci U S A ; 114(43): E8996-E9005, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073096

ABSTRACT

Tissue regeneration depends on the timely activation of adult stem cells. In skeletal muscle, the adult stem cells maintain a quiescent state and proliferate upon injury. We show that muscle stem cells (MuSCs) use direct translational repression to maintain the quiescent state. High-resolution single-molecule and single-cell analyses demonstrate that quiescent MuSCs express high levels of Myogenic Differentiation 1 (MyoD) transcript in vivo, whereas MyoD protein is absent. RNA pulldowns and costainings show that MyoD mRNA interacts with Staufen1, a potent regulator of mRNA localization, translation, and stability. Staufen1 prevents MyoD translation through its interaction with the MyoD 3'-UTR. MuSCs from Staufen1 heterozygous (Staufen1+/-) mice have increased MyoD protein expression, exit quiescence, and begin proliferating. Conversely, blocking MyoD translation maintains the quiescent phenotype. Collectively, our data show that MuSCs express MyoD mRNA and actively repress its translation to remain quiescent yet primed for activation.


Subject(s)
Gene Expression Regulation/physiology , MyoD Protein/metabolism , RNA-Binding Proteins/metabolism , Stem Cells/physiology , Animals , Cell Differentiation , Mice , Muscle Cells/physiology , MyoD Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
15.
Front Aging Neurosci ; 7: 140, 2015.
Article in English | MEDLINE | ID: mdl-26257645

ABSTRACT

Skeletal muscle atrophy or wasting accompanies various chronic illnesses and the aging process, thereby reducing muscle function. One of the most important components contributing to effective muscle repair in postnatal organisms, the satellite cells (SCs), have recently become the focus of several studies examining factors participating in the atrophic process. We critically examine here the experimental evidence linking SC function with muscle loss in connection with various diseases as well as aging, and in the subsequent recovery process. Several recent reports have investigated the changes in SCs in terms of their differentiation and proliferative capacity in response to various atrophic stimuli. In this regard, we review the molecular changes within SCs that contribute to their dysfunctional status in atrophy, with the intention of shedding light on novel potential pharmacological targets to counteract the loss of muscle mass.

16.
Sci Transl Med ; 6(267): 267ra176, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25520397

ABSTRACT

We have previously observed that Wnt signaling activates a fibrogenic program in adult muscle stem cells, called satellite cells, during aging. We genetically labeled satellite cells in a mouse model of Duchenne muscular dystrophy to follow their fate during the progression of the disease. We observed that a fraction of satellite cells had a reduced myogenic potential and showed enhanced expression of profibrotic genes compared to age-matched controls. By combining in vitro and in vivo results, we found that expression of transforming growth factor-ß2 (TGFß2) was induced in response to elevated canonical Wnt signaling in dystrophic muscles and that the resulting increase in TGFß activity affected the behavior of satellite cells in an autocrine or paracrine fashion. Indeed, pharmacological inhibition of the TGFß pathway in vivo reduced the fibrogenic characteristics of satellite cells. These studies shed new light on the cellular and molecular mechanisms responsible for stem cell dysfunction in dystrophic muscle and may contribute to the development of more effective and specific therapeutic approaches for the prevention of muscle fibrosis.


Subject(s)
Muscular Dystrophy, Duchenne/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Transforming Growth Factor beta2/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Autocrine Communication , Cell Differentiation , Cell Line , Cell Lineage , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Male , Mice, Inbred mdx , Mice, Transgenic , Muscle Development , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Paracrine Communication , Satellite Cells, Skeletal Muscle/pathology , Transfection , Up-Regulation , Wnt Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 110(46): 18549-54, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24170859

ABSTRACT

The adaptor protein Numb has been implicated in the switch between cell proliferation and differentiation made by satellite cells during muscle repair. Using two genetic approaches to ablate Numb, we determined that, in its absence, muscle regeneration in response to injury was impaired. Single myofiber cultures demonstrated a lack of satellite cell proliferation in the absence of Numb, and the proliferation defect was confirmed in satellite cell cultures. Quantitative RT-PCR from Numb-deficient satellite cells demonstrated highly up-regulated expression of p21 and Myostatin, both inhibitors of myoblast proliferation. Transfection with Myostatin-specific siRNA rescued the proliferation defect of Numb-deficient satellite cells. Furthermore, overexpression of Numb in satellite cells inhibited Myostatin expression. These data indicate a unique function for Numb during the initial activation and proliferation of satellite cells in response to muscle injury.


Subject(s)
Cell Proliferation , Membrane Proteins/deficiency , Muscle Development/physiology , Nerve Tissue Proteins/deficiency , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Analysis of Variance , Animals , Barium Compounds , Blotting, Western , Body Weights and Measures , Chlorides , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Knockout , Muscle Development/genetics , RNA Interference , RNA, Small Interfering/genetics , Regeneration/genetics , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/metabolism
18.
Dev Biol ; 379(2): 195-207, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23639729

ABSTRACT

Myf5 is a member of the muscle-specific determination genes and plays a critical role in skeletal muscle development. Whereas the expression of Myf5 during embryonic and fetal myogenesis has been extensively studied, its expression in progenitors that will ultimately give rise to adult satellite cells, the stem cells responsible for muscle repair, is still largely unexplored. To investigate this aspect, we have generated a mouse strain carrying a CreER coding sequence in the Myf5 locus. In this strain, Tamoxifen-inducible Cre activity parallels endogenous Myf5 expression. Combining Myf5(CreER) and Cre reporter alleles, we were able to evaluate the contribution of cells expressing Myf5 at distinct developmental stages to the pool of satellite cells in adult hindlimb muscles. Although it was possible to trace back the origin of some rare satellite cells to a subpopulation of Myf5(+ve) progenitors in the limb buds at the late embryonic stage (∼E12), a significant number of satellite cells arise from cells which expressed Myf5 for the first time at the fetal stage (∼E15). These studies provide direct evidence that adult satellite cells derive from progenitors that first express the myogenic determination gene Myf5 during fetal stages of myogenesis.


Subject(s)
Cell Lineage/physiology , Fetus/physiology , Muscle Development/physiology , Muscle, Skeletal/cytology , Myogenic Regulatory Factor 5/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/metabolism , Animals , Blotting, Southern , DNA Primers/genetics , Flow Cytometry , Galactosides , Indoles , Integrases , Mice , Microscopy, Fluorescence , Satellite Cells, Skeletal Muscle/cytology , Tamoxifen
20.
Mol Cell ; 40(5): 749-61, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21145483

ABSTRACT

Pax3 plays critical roles during developmental and postnatal myogenesis. We have previously shown that levels of Pax3 protein are regulated by monoubiquitination and proteasomal degradation during postnatal myogenesis, but none of the key regulators of the monoubiquitination process were known. Here we show that Pax3 monoubiquitination is mediated by the ubiquitin-activating/conjugating activity of Taf1, a component of the core transcriptional machinery that was recently reported to be downregulated during myogenic differentiation. We show that Taf1 binds directly to Pax3 and overexpression of Taf1 increases the level of monoubiquitinated Pax3 and its degradation by the proteasome. A decrease of Taf1 results in a decrease in Pax3 monoubiquitination, an increase in the levels of Pax3 protein, and a concomitant increase in Pax3-mediated inhibition of myogenic differentiation and myoblast migration. These results suggest that Taf1 regulates Pax3 protein levels through its ability to mediate monoubiquitination, revealing a critical interaction between two proteins that are involved in distinct aspects of myogenic differentiation. Finally, these results suggest that the components of the core transcriptional are integrally involved in the process of myogenic differentiation, acting as nodal regulators of the differentiation program.


Subject(s)
Paired Box Transcription Factors/metabolism , Satellite Cells, Skeletal Muscle/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Ubiquitin/metabolism , Animals , Cells, Cultured , Histone Acetyltransferases , Mice , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...