Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Carcinogenesis ; 36(9): 1071-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26153023

ABSTRACT

Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin ß1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.


Subject(s)
Apoptosis/drug effects , Artemisinins/pharmacology , Reactive Oxygen Species/metabolism , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Artemisinins/therapeutic use , Artesunate , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Damage/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Integrin beta1/biosynthesis , Mice , MicroRNAs/biosynthesis , Neoplasm Invasiveness , Neoplasm Transplantation , Neural Cell Adhesion Molecules/biosynthesis , PAX7 Transcription Factor/biosynthesis , Pyridines/pharmacology , Transplantation, Heterologous , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
2.
Sci Rep ; 7(1): 12537, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28970581

ABSTRACT

Regeneration of injured skeletal muscles relies on a tightly controlled chain of cellular and molecular events. We show that appropriate levels of S100B protein are required for timely muscle regeneration after acute injury. S100B released from damaged myofibers and infiltrating macrophages expands the myoblast population, attracts macrophages and promotes their polarization into M2 (pro-regenerative) phenotype, and modulates collagen deposition, by interacting with RAGE (receptor for advanced glycation end-products) or FGFR1 (fibroblast growth factor receptor 1) depending on the muscle repair phase and local conditions. However, persistence of high S100B levels compromises the regeneration process prolonging myoblast proliferation and macrophage infiltration, delaying M1/M2 macrophage transition, and promoting deposition of fibrotic tissue via RAGE engagement. Interestingly, S100B is released in high abundance from degenerating muscles of mdx mice, an animal model of Duchenne muscular dystrophy (DMD), and blocking S100B ameliorates histopathology. Thus, levels of S100B differentially affect skeletal muscle repair upon acute injury and in the context of muscular dystrophy, and S100B might be regarded as a potential molecular target in DMD.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophy, Duchenne/genetics , Regeneration/genetics , S100 Calcium Binding Protein beta Subunit/genetics , Animals , Disease Models, Animal , Humans , Macrophage Activation/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred mdx , Muscle Strength/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Receptor for Advanced Glycation End Products/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics
3.
Cell Death Differ ; 24(12): 2077-2088, 2017 12.
Article in English | MEDLINE | ID: mdl-28885620

ABSTRACT

Muscles of sarcopenic people show hypotrophic myofibers and infiltration with adipose and, at later stages, fibrotic tissue. The origin of infiltrating adipocytes resides in fibro-adipogenic precursors and nonmyogenic mesenchymal progenitor cells, and in satellite cells, the adult stem cells of skeletal muscles. Myoblasts and brown adipocytes share a common Myf5+ progenitor cell: the cell fate depends on levels of bone morphogenetic protein 7 (BMP-7), a TGF-ß family member. S100B, a Ca2+-binding protein of the EF-hand type, is expressed at relatively high levels in myoblasts from sarcopenic humans and exerts anti-myogenic effects via NF-κB-dependent inhibition of MyoD, a myogenic transcription factor acting upstream of the essential myogenic factor, myogenin. Adipogenesis requires high levels of ROS, and myoblasts of sarcopenic subjects show elevated ROS levels. Here we show that: (1) ROS overproduction in myoblasts results in upregulation of S100B levels via NF-κB activation; and (2) ROS/NF-κB-induced accumulation of S100B causes myoblast transition into brown adipocytes. S100B activates an NF-κB/Ying Yang 1 axis that negatively regulates the promyogenic and anti-adipogenic miR-133 with resultant accumulation of the brown adipogenic transcription regulator, PRDM-16. S100B also upregulates BMP-7 via NF-κB/Ying Yang 1 with resultant BMP-7 autocrine activity. Interestingly, myoblasts from sarcopenic humans show features of brown adipocytes. We also show that S100B levels and NF-κB activity are elevated in brown adipocytes obtained by culturing myoblasts in adipocyte differentiation medium and that S100B knockdown or NF-κB inhibition in myoblast-derived brown adipocytes reconverts them into fusion-competent myoblasts. At last, interstitial cells and, unexpectedly, a subpopulation of myofibers in muscles of geriatric but not young mice co-express S100B and the brown adipocyte marker, uncoupling protein-1. These results suggest that S100B is an important intracellular molecular signal regulating Myf5+ progenitor cell differentiation into fusion-competent myoblasts or brown adipocytes depending on its levels.


Subject(s)
Adipocytes, Brown/metabolism , MicroRNAs/metabolism , Myoblasts/metabolism , NF-kappa B/metabolism , Oxidative Stress/physiology , S100 Calcium Binding Protein beta Subunit/metabolism , Adipocytes, Brown/cytology , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Humans , Male , Mice , MicroRNAs/genetics , Myoblasts/cytology , Reactive Oxygen Species/metabolism , S100 Calcium Binding Protein beta Subunit/genetics , Transfection , YY1 Transcription Factor/metabolism
4.
Exp Gerontol ; 40(3): 189-97, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15763396

ABSTRACT

The mature myofibres of human skeletal muscle are surrounded by a type of adult stem cell, known as the satellite cell, which lies outside the sarcolemma but within the basal lamina. These cells remain quiescent until external stimuli trigger their re-entry into the cell cycle. In humans, ageing is characterised by a progressive loss of muscle mass and strength (sarcopenia) associated with a decline in functional ability. One of the possible causes of this decline in muscle performance is a decrease in the antioxidative capacity of skeletal muscle, resulting in an abnormal accumulation of the reactive oxygen species (ROS) critical for cell life. The present study shows that: (i) the antioxidant activity of Catalase and Gluthatione transferase in satellite cells derived from the elderly is drastically reduced compared to that in cells isolated from young individuals; (ii) cell membrane fluidity is considerably different between the two age groups; and (iii) basal [Ca(2+)](i) levels in satellite cells increase significantly in an age-dependent manner. In view of the data obtained, we hypothesise that the destabilising oxidative damage that occurs during ageing in skeletal muscle also affects quiescent satellite cells, which spend their life in close anatomic and functional contact with adult fibres. This status is derived from a decrease in the antioxidative capacity, and may negatively affect the ageing satellite cells ability to repair muscle.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Satellite Cells, Skeletal Muscle/enzymology , Adult , Aged , Aged, 80 and over , Biopsy , Calcium/metabolism , Catalase/metabolism , Cytosol/enzymology , Enzyme Activation/physiology , Female , Glutathione Transferase/metabolism , Humans , Infant, Newborn , Male , Membrane Fluidity
5.
PLoS One ; 7(1): e28700, 2012.
Article in English | MEDLINE | ID: mdl-22276098

ABSTRACT

In high-density myoblast cultures S100B enhances basic fibroblast growth factor (bFGF) receptor 1 (FGFR1) signaling via binding to bFGF and blocks its canonical receptor, receptor for advanced glycation end-products (RAGE), thereby stimulating proliferation and inhibiting differentiation. Here we show that upon skeletal muscle injury S100B is released from myofibers with maximum release at day 1 post-injury in coincidence with satellite cell activation and the beginning of the myoblast proliferation phase, and declining release thereafter in coincidence with reduced myoblast proliferation and enhanced differentiation. By contrast, levels of released bFGF are remarkably low at day 1 post-injury, peak around day 5 and decline thereafter. We also show that in low-density myoblast cultures S100B binds RAGE, but not bFGF/FGFR1 thereby simultaneously stimulating proliferation via ERK1/2 and activating the myogenic program via p38 MAPK. Clearance of S100B after a 24-h treatment of low-density myoblasts results in enhanced myotube formation compared with controls as a result of increased cell numbers and activated myogenic program, whereas chronic treatment with S100B results in stimulation of proliferation and inhibition of differentiation due to a switch of the initial low-density culture to a high-density culture. However, at relatively high doses, S100B stimulates the mitogenic bFGF/FGFR1 signaling in low-density myoblasts, provided bFGF is present. We propose that S100B is a danger signal released from injured muscles that participates in skeletal muscle regeneration by activating the promyogenic RAGE or the mitogenic bFGF/FGFR1 depending on its own concentration, the absence or presence of bFGF, and myoblast density.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Immunologic/metabolism , Regeneration/physiology , S100 Proteins/metabolism , Animals , Blotting, Western , Cattle , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Cells, Cultured , Fibroblast Growth Factor 2/genetics , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Skeletal/cytology , Myoblasts/cytology , Protein Binding , Receptor for Advanced Glycation End Products , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , S100 Proteins/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Age (Dordr) ; 33(4): 523-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21140295

ABSTRACT

During aging, skeletal muscles show reduced mass and functional capacity largely due to loss of the regenerative ability of satellite cells (SCs), the quiescent stem cells located beneath the basal lamina surrounding each myofiber. While both the external environment and intrinsic properties of SCs appear to contribute to the age-related SC deficiency, the latter ones have been poorly investigated especially in humans. In the present work, we analyzed several parameters of SCs derived from biopsies of vastus lateralis muscle from healthy non-trained young (28.7 ± 5.9 years; n = 10) and aged (77.3 ± 6.4 years; n = 11) people. Compared with young SCs, aged SCs showed impaired differentiation when cultured in differentiation medium, and exhibited the following: (1) reduced proliferation; (2) higher expression levels of S100B, a negative regulator of myoblast differentiation; (3) undetectable levels in growth medium of full-length RAGE (receptor for advanced glycation end products), a multiligand receptor of the immunoglobulin superfamily, the engagement of which enhances myoblast differentiation; and (4) lower expression levels of the transcription factors, MyoD and Pax7. Also, either overexpression of full-length RAGE or knockdown of S100B in aged SCs resulted in enhanced differentiation, while overexpression of either a non-transducing mutant of RAGE (RAGEΔcyto) or S100B in young SCs resulted in reduced differentiation compared with controls. Moreover, while aged SCs maintained the ability to respond to mitogenic factors (e.g., bFGF and S100B), they were no longer able to secrete these factors, unlike young SCs. These data support a role for intrinsic factors, besides the extracellular environment in the defective SC function in aged skeletal muscles.


Subject(s)
Aging/metabolism , Nerve Growth Factors/metabolism , Receptors, Immunologic/metabolism , S100 Proteins/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Adult , Aged , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Humans , MAP Kinase Signaling System , Male , MyoD Protein/metabolism , Oncogene Protein v-akt , PAX7 Transcription Factor/metabolism , Receptor for Advanced Glycation End Products , S100 Calcium Binding Protein beta Subunit , Satellite Cells, Skeletal Muscle/physiology , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Exp Gerontol ; 44(8): 523-31, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19457451

ABSTRACT

Sarcopenia is the age-related loss of muscle mass, strength and function. Human muscle proteins are synthesized at a slower rate in the elderly than in young adults, leading to atrophy and muscle mass loss with a decline in the functional capability. Additionally, aging is accompanied by a decrease in the ability of muscle tissue to regenerate following injury or overuse due to the impairment of intervening satellite cells, in which we previously reported oxidative damage evidences. The aim of the present study was to determine the effects of aging on myoblasts and myotubes obtained from human skeletal muscle, and characterize the transcriptional profile as molecular expression patterns in relation to age-dependent modifications in their regenerative capacity. Our data show that the failure to differentiate does not depend on reduced myogenic cell number, but difficulty to complete the differentiation program. Data reported here suggested the following findings: (i) oxidative damage accumulation in molecular substrates, probably due to impaired antioxidant activity and insufficient repair capability, (ii) limited capability of elderly myoblasts to execute a complete differentiation program; restricted fusion, possibly due to altered cytoskeleton turnover and extracellular matrix degradation and (iii) activation of atrophy mechanism by activation of a specific FOXO-dependent program.


Subject(s)
Aging/physiology , Cell Differentiation , Muscle Fibers, Skeletal/physiology , Myoblasts/physiology , Regeneration/physiology , Sarcopenia/physiopathology , Satellite Cells, Skeletal Muscle/physiology , Adult , Aged , Aged, 80 and over , Aging/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Sarcopenia/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
8.
Free Radic Res ; 43(2): 138-48, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19115119

ABSTRACT

Oxidative stress is linked to several human diseases, including diabetes. However, the intracellular signal transduction pathways regulated by reactive oxygen species (ROS) remain to be established. Deleterious effects of ROS stem from interactions with various ion transport proteins such as ion channels and pumps, primarily altering Ca(2 +) homeostasis and inducing cell dysfunction. This study characterized the Ca(2 +) transport system in lymphocytes of patients with type-2 diabetes, evaluating the possible correlation between cell modifications and the existence of specific oxidative stress damage. Lymphocytes from type-2 diabetes patients displayed oxidative stress features (accumulation of some ROS species, membrane peroxidation, increase in protein carbonyls, increase in SOD and Catalase activity) and Ca(2 +) dyshomeostasis (modified voltage-dependent and inositol 1,4,5-triphosphate-mediated Ca(2 +) channel activities, decrease in Ca(2 +) pumps activity). The data support a correlation between oxidative damage and alterations in intracellular Ca(2 +) homeostasis, possibly due to modification of the ionic control in lymphocytes of type-2 diabetes patients.


Subject(s)
Calcium/blood , Diabetes Mellitus, Type 2/blood , Oxidative Stress/physiology , Aged , Calcium Signaling , Case-Control Studies , Cell Membrane/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Homeostasis , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Middle Aged , Reactive Oxygen Species/metabolism , Signal Transduction
9.
Cell Metab ; 8(5): 425-36, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19046573

ABSTRACT

The antioxidant enzyme superoxide dismutase 1 (SOD1) is a critical player of the antioxidative defense whose activity is altered in several chronic diseases, including amyotrophic lateral sclerosis. However, how oxidative insult affects muscle homeostasis remains unclear. This study addresses the role of oxidative stress on muscle homeostasis and function by the generation of a transgenic mouse model expressing a mutant SOD1 gene (SOD1(G93A)) selectively in skeletal muscle. Transgenic mice developed progressive muscle atrophy, associated with a significant reduction in muscle strength, alterations in the contractile apparatus, and mitochondrial dysfunction. The analysis of molecular pathways associated with muscle atrophy revealed that accumulation of oxidative stress served as signaling molecules to initiate autophagy, one of the major intracellular degradation mechanisms. These data demonstrate that skeletal muscle is a primary target of SOD1(G93A) -mediated toxicity and disclose the molecular mechanism whereby oxidative stress triggers muscle atrophy.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Superoxide Dismutase/physiology , Animals , Autophagy/physiology , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle Contraction , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Mutation , Nerve Degeneration/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Sarcolemma/pathology , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
10.
Mol Cell ; 17(4): 603-9, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15721263

ABSTRACT

Myosin VI moves processively along actin with a larger step size than expected from the size of the motor. Here, we show that the proximal tail (the approximately 80-residue segment following the IQ domain) is not a rigid structure but, rather, a flexible domain that permits the heads to separate. With a GCN4 coiled coil inserted in the proximal tail, the heads are closer together in electron microscopy (EM) images, and the motor takes shorter processive steps. Single-headed myosin VI S1 constructs take nonprocessive 12 nm steps, suggesting that most of the processive step is covered by a diffusive search for an actin binding site. Based on these results, we present a mechanical model that describes stepping under an applied load.


Subject(s)
Actins/metabolism , Myosin Type V/metabolism , Actins/chemistry , Actins/ultrastructure , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Electron , Myosin Type V/chemistry , Myosin Type V/genetics , Plasmids , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL