Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Publication year range
1.
FASEB J ; 38(13): e23797, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38963344

ABSTRACT

The role of N-glycosylation in the myogenic process remains poorly understood. Here, we evaluated the impact of N-glycosylation inhibition by Tunicamycin (TUN) or by phosphomannomutase 2 (PMM2) gene knockdown, which encodes an enzyme essential for catalyzing an early step of the N-glycosylation pathway, on C2C12 myoblast differentiation. The effect of chronic treatment with TUN on tibialis anterior (TA) and extensor digitorum longus (EDL) muscles of WT and MLC/mIgf-1 transgenic mice, which overexpress muscle Igf-1Ea mRNA isoform, was also investigated. TUN-treated and PMM2 knockdown C2C12 cells showed reduced ConA, PHA-L, and AAL lectin binding and increased ER-stress-related gene expression (Chop and Hspa5 mRNAs and s/uXbp1 ratio) compared to controls. Myogenic markers (MyoD, myogenin, and Mrf4 mRNAs and MF20 protein) and myotube formation were reduced in both TUN-treated and PMM2 knockdown C2C12 cells. Body and TA weight of WT and MLC/mIgf-1 mice were not modified by TUN treatment, while lectin binding slightly decreased in the TA muscle of WT (ConA and AAL) and MLC/mIgf-1 (ConA) mice. The ER-stress-related gene expression did not change in the TA muscle of WT and MLC/mIgf-1 mice after TUN treatment. TUN treatment decreased myogenin mRNA and increased atrogen-1 mRNA, particularly in the TA muscle of WT mice. Finally, the IGF-1 production and IGF1R signaling pathways activation were reduced due to N-glycosylation inhibition in TA and EDL muscles. Decreased IGF1R expression was found in TUN-treated C2C12 myoblasts which was associated with lower IGF-1-induced IGF1R, AKT, and ERK1/2 phosphorylation compared to CTR cells. Chronic TUN-challenge models can help to elucidate the molecular mechanisms through which diseases associated with aberrant N-glycosylation, such as Congenital Disorders of Glycosylation (CDG), affect muscle and other tissue functions.


Subject(s)
Cell Differentiation , Endoplasmic Reticulum Chaperone BiP , Muscle, Skeletal , Myoblasts , Receptor, IGF Type 1 , Signal Transduction , Tunicamycin , Animals , Mice , Glycosylation , Myoblasts/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Tunicamycin/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/genetics , Muscle, Skeletal/metabolism , Muscle Development/physiology , Cell Line , Mice, Transgenic , Endoplasmic Reticulum Stress , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics
2.
Brain ; 146(11): 4425-4436, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37327376

ABSTRACT

Amyotrophic lateral sclerosis (ALS), the major adult-onset motor neuron disease, has been viewed almost exclusively as a disease of upper and lower motor neurons, with muscle changes interpreted as a consequence of the progressive loss of motor neurons and neuromuscular junctions. This has led to the prevailing view that the involvement of muscle in ALS is only secondary to motor neuron loss. Skeletal muscle and motor neurons reciprocally influence their respective development and constitute a single functional unit. In ALS, multiple studies indicate that skeletal muscle dysfunction might contribute to progressive muscle weakness, as well as to the final demise of neuromuscular junctions and motor neurons. Furthermore, skeletal muscle has been shown to participate in disease pathogenesis of several monogenic diseases closely related to ALS. Here, we move the narrative towards a better appreciation of muscle as a contributor of disease in ALS. We review the various potential roles of skeletal muscle cells in ALS, from passive bystanders to active players in ALS pathophysiology. We also compare ALS to other motor neuron diseases and draw perspectives for future research and treatment.


Subject(s)
Amyotrophic Lateral Sclerosis , Adult , Humans , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Muscle, Skeletal/pathology , Neuromuscular Junction/pathology , Muscle Weakness
3.
EMBO J ; 37(18)2018 09 14.
Article in English | MEDLINE | ID: mdl-30177572

ABSTRACT

Myogenesis is a highly regulated process that involves the conversion of progenitor cells into multinucleated myofibers. Besides proteins and miRNAs, long noncoding RNAs (lncRNAs) have been shown to participate in myogenic regulatory circuitries. Here, we characterize a murine chromatin-associated muscle-specific lncRNA, Charme, which contributes to the robustness of the myogenic program in vitro and in vivo In myocytes, Charme depletion triggers the disassembly of a specific chromosomal domain and the downregulation of myogenic genes contained therein. Notably, several Charme-sensitive genes are associated with human cardiomyopathies and Charme depletion in mice results in a peculiar cardiac remodeling phenotype with changes in size, structure, and shape of the heart. Moreover, the existence of an orthologous transcript in human, regulating the same subset of target genes, suggests an important and evolutionarily conserved function for Charme Altogether, these data describe a new example of a chromatin-associated lncRNA regulating the robustness of skeletal and cardiac myogenesis.


Subject(s)
Muscle Development , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/metabolism , Ventricular Remodeling , Animals , Humans , Mice , Muscle Fibers, Skeletal/pathology , Myocytes, Cardiac/pathology , RNA, Long Noncoding/genetics
4.
Sensors (Basel) ; 22(18)2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36146227

ABSTRACT

Tissue engineering is a multidisciplinary approach focused on the development of innovative bioartificial substitutes for damaged organs and tissues. For skeletal muscle, the measurement of contractile capability represents a crucial aspect for tissue replacement, drug screening and personalized medicine. To date, the measurement of engineered muscle tissues is rather invasive and not continuous. In this context, we proposed an innovative sensor for the continuous monitoring of engineered-muscle-tissue contractility through an embedded technique. The sensor is based on the calibrated deflection of one of the engineered tissue's supporting pins, whose movements are measured using a noninvasive optical method. The sensor was calibrated to return force values through the use of a step linear motor and a micro-force transducer. Experimental results showed that the embedded sensor did not alter the correct maturation of the engineered muscle tissue. Finally, as proof of concept, we demonstrated the ability of the sensor to capture alterations in the force contractility of the engineered muscle tissues subjected to serum deprivation.


Subject(s)
Muscle Contraction , Tissue Engineering , Muscle, Skeletal/physiology
5.
Hum Mol Genet ; 26(14): 2781-2790, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28472288

ABSTRACT

Oxidative stress is involved in the pathogenesis of Duchenne muscular dystrophy (DMD), an X-linked genetic disorder caused by mutations in the dystrophin gene and characterized by progressive, lethal muscle degeneration and chronic inflammation. In this study, we explored the expression and signaling pathway of a master player of the anti-oxidant and anti-inflammatory response, namely NF-E2-related Factor 2, in muscle biopsies of DMD patients. We classified DMD patients in two age groups (Class I, 0-2 years and Class II, 2-9 years), in order to evaluate the antioxidant pathway expression during the disease progression. We observed that altered enzymatic antioxidant responses, increased levels of oxidized glutathione and oxidative damage are differently modulated in the two age classes of patients and well correlate with the severity of pathology. Interestingly, we also observed a modulation of relevant markers of the inflammatory response, such as heme oxygenase 1 and Inteleukin-6 (IL-6), suggesting a link between oxidative stress and chronic inflammatory response. Of note, using a transgenic mouse model, we demonstrated that IL-6 overexpression parallels the antioxidant expression profile and the severity of dystrophic muscle observed in DMD patients. This study advances our understanding of the pathogenic mechanisms underlying DMD and defines the critical role of oxidative stress on muscle wasting with clear implications for disease pathogenesis and therapy in human.


Subject(s)
Muscular Dystrophy, Duchenne/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Animals , Antioxidants/metabolism , Child , Child, Preschool , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Female , Glutathione/genetics , Glutathione/metabolism , Humans , Infant , Infant, Newborn , Inflammation/genetics , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Signal Transduction
6.
Curr Genomics ; 20(1): 24-37, 2019 Jan.
Article in English | MEDLINE | ID: mdl-31015789

ABSTRACT

The peculiar ability of skeletal muscle tissue to operate adaptive changes during post-natal de-velopment and adulthood has been associated with the existence of adult somatic stem cells. Satellite cells, occupying an exclusive niche within the adult muscle tissue, are considered bona fide stem cells with both stem-like properties and myogenic activities. Indeed, satellite cells retain the capability to both maintain the quiescence in uninjured muscles and to be promptly activated in response to growth or re-generative signals, re-engaging the cell cycle. Activated cells can undergo myogenic differentiation or self-renewal moving back to the quiescent state. Satellite cells behavior and their fate decision are finely controlled by mechanisms involving both cell-autonomous and external stimuli. Alterations in these regu-latory networks profoundly affect muscle homeostasis and the dynamic response to tissue damage, con-tributing to the decline of skeletal muscle that occurs under physio-pathologic conditions. Although the clear myogenic activity of satellite cells has been described and their pivotal role in muscle growth and regeneration has been reported, a comprehensive picture of inter-related mechanisms guiding muscle stem cell activity has still to be defined. Here, we reviewed the main regulatory networks determining satellite cell behavior. In particular, we focused on genetic and epigenetic mechanisms underlining satel-lite cell maintenance and commitment. Besides intrinsic regulations, we reported current evidences about the influence of environmental stimuli, derived from other cell populations within muscle tissue, on satel-lite cell biology.

7.
Sensors (Basel) ; 19(23)2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31766596

ABSTRACT

In this paper, the characterization of the main techniques and transducers employed to measure local and global strains induced by uniaxial loading of murine tibiae is presented. Micro strain gauges and digital image correlation (DIC) were tested to measure local strains, while a moving coil motor-based length transducer was employed to measure relative global shortening. Local strain is the crucial parameter to be measured when dealing with bone cell mechanotransduction, so we characterized these techniques in the experimental conditions known to activate cell mechanosensing in vivo. The experimental tests were performed using tibia samples excised from twenty-two C57BL/6 mice. To evaluate measurement repeatability we computed the standard deviation of ten repetitive compressions to the mean value. This value was lower than 3% for micro strain gauges, and in the range of 7%-10% for DIC and the length transducer. The coefficient of variation, i.e., the standard deviation to the mean value, was about 35% for strain gauges and the length transducer, and about 40% for DIC. These results provided a comprehensive characterization of three methodologies for local and global bone strain measurement, suggesting a possible field of application on the basis of their advantages and limitations.


Subject(s)
Tibia/physiology , Animals , Biomechanical Phenomena/physiology , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Stress, Mechanical , Transducers , Weight-Bearing/physiology
8.
Stem Cells ; 35(3): 725-738, 2017 03.
Article in English | MEDLINE | ID: mdl-27612437

ABSTRACT

The transcription factor MEF2C (Myocyte Enhancer Factor 2C) plays an established role in the early steps of myogenic differentiation. However, the involvement of MEF2C in adult myogenesis and in muscle regeneration has not yet been systematically investigated. Alternative splicing of mammalian MEF2C transcripts gives rise to two mutually exclusive protein variants: MEF2Cα2 which exerts a positive control of myogenic differentiation, and MEF2Cα1, in which the α1 domain acts as trans-repressor of the MEF2C pro-differentiation activity itself. However, MEF2Cα1 variants are persistently expressed in differentiating cultured myocytes, suggesting a role in adult myogenesis. We found that overexpression of both MEF2Cα1/α2 proteins in a mouse model of muscle injury promotes muscle regeneration and hypertrophy, with each isoform promoting different stages of myogenesis. Besides the ability of MEF2Cα2 to increase differentiation, we found that overexpressed MEF2Cα1 enhances both proliferation and differentiation of primary myoblasts, and activates the AKT/mTOR/S6K anabolic signaling pathway in newly formed myofibers. The multiple activities of MEF2Cα1 are modulated by phosphorylation of Ser98 and Ser110, two amino acid residues located in the α1 domain of MEF2Cα1. These specific phosphorylations allow the interaction of MEF2Cα1 with the peptidyl-prolyl isomerase PIN1, a regulator of MEF2C functions. Overall, in this study we established a novel regulatory mechanism in which the expression and the phosphorylation of MEF2Cα1 are critically required to sustain the adult myogenesis. The described molecular mechanism will represent a new potential target for the development of therapeutical strategies to treat muscle-wasting diseases. Stem Cells 2017;35:725-738.


Subject(s)
Alternative Splicing/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Regeneration , Aging/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Hypertrophy , MEF2 Transcription Factors/chemistry , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Myoblasts/metabolism , NIH 3T3 Cells , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Phosphorylation , Protein Binding , Protein Domains , Satellite Cells, Skeletal Muscle/metabolism , Serine/metabolism
9.
Curr Genomics ; 19(5): 356-369, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30065611

ABSTRACT

Muscle homeostasis is guaranteed by a delicate balance between synthesis and degradation of cell proteins and its alteration leads to muscle wasting and diseases. In this review, we describe the major anabolic pathways that are involved in muscle growth and homeostasis and the proteolytic systems that are over-activated in muscle pathologies. Modulation of these pathways comprises an attractive target for drug intervention.

10.
Hum Mol Genet ; 24(21): 6041-53, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26251044

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by progressive lethal muscle degeneration and chronic inflammatory response. The mdx mouse strain has served as the animal model for human DMD. However, while DMD patients undergo extensive necrosis, the affected muscles of adult mdx mice rapidly regenerates and regains structural and functional integrity. The basis for the mild effects observed in mice compared with the lethal consequences in humans remains unknown. In this study, we provide evidence that interleukin-6 (IL-6) is causally linked to the pathogenesis of muscular dystrophy. We report that forced expression of IL-6, in the adult mdx mice, recapitulates the severe phenotypic characteristics of DMD in humans. Increased levels of IL-6 exacerbate the dystrophic muscle phenotype, sustaining inflammatory response and repeated cycles of muscle degeneration and regeneration, leading to exhaustion of satellite cells. The mdx/IL6 mouse closely approximates the human disease and more faithfully recapitulates the disease progression in humans. This study promises to significantly advance our understanding of the pathogenic mechanisms that lead to DMD.


Subject(s)
Interleukin-6/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Animals , Down-Regulation , Interleukin-6/genetics , Mice , Mice, Inbred mdx , Muscle Development , Muscle, Skeletal/pathology , Phenotype , Protein Serine-Threonine Kinases/metabolism , Satellite Cells, Skeletal Muscle/pathology , Stem Cells/pathology , NF-kappaB-Inducing Kinase
11.
Mol Ther ; 23(7): 1189-1200, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896247

ABSTRACT

Insulin-like growth factor 1 (IGF-1) is a potent enhancer of tissue regeneration, and its overexpression in muscle injury leads to hastened resolution of the inflammatory phase. Here, we show that monocytes/macrophages constitute an important initial source of IGF-1 in muscle injury, as conditional deletion of the IGF-1 gene specifically in mouse myeloid cells (ϕIGF-1 CKO) blocked the normal surge of local IGF-1 in damaged muscle and significantly compromised regeneration. In injured muscle, Ly6C+ monocytes/macrophages and CD206+ macrophages expressed equivalent IGF-1 levels, which were transiently upregulated during transition from the inflammation to repair. In injured ϕIGF-1 CKO mouse muscle, accumulation of CD206+ macrophages was impaired, while an increase in Ly6C+ monocytes/macrophages was favored. Transcriptional profiling uncovered inflammatory skewing in ϕIGF-1 CKO macrophages, which failed to fully induce a reparative gene program in vitro or in vivo, revealing a novel autocrine role for IGF-1 in modulating murine macrophage phenotypes. These data establish local macrophage-derived IGF-1 as a key factor in inflammation resolution and macrophage polarization during muscle regeneration.


Subject(s)
Insulin-Like Growth Factor I/biosynthesis , Muscle, Skeletal/growth & development , Regeneration/genetics , Wound Healing , Animals , Autocrine Communication/genetics , Gene Expression Regulation, Developmental , Inflammation/genetics , Inflammation/pathology , Insulin-Like Growth Factor I/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Monocytes/metabolism , Muscle, Skeletal/metabolism
12.
Mol Ther ; 23(5): 885-895, 2015 May.
Article in English | MEDLINE | ID: mdl-25669433

ABSTRACT

Although in the last decades the molecular underpinnings of the cell cycle have been unraveled, the acquired knowledge has been rarely translated into practical applications. Here, we investigate the feasibility and safety of triggering proliferation in vivo by temporary suppression of the cyclin-dependent kinase inhibitor, p21. Adeno-associated virus (AAV)-mediated, acute knockdown of p21 in intact skeletal muscles elicited proliferation of multiple, otherwise quiescent cell types, notably including satellite cells. Compared with controls, p21-suppressed muscles exhibited a striking two- to threefold expansion in cellularity and increased fiber numbers by 10 days post-transduction, with no detectable inflammation. These changes partially persisted for at least 60 days, indicating that the muscles had undergone lasting modifications. Furthermore, morphological hyperplasia was accompanied by 20% increases in maximum strength and resistance to fatigue. To assess the safety of transiently suppressing p21, cells subjected to p21 knockdown in vitro were analyzed for γ-H2AX accumulation, DNA fragmentation, cytogenetic abnormalities, ploidy, and mutations. Moreover, the differentiation competence of p21-suppressed myoblasts was investigated. These assays confirmed that transient suppression of p21 causes no genetic damage and does not impair differentiation. Our results establish the basis for further exploring the manipulation of the cell cycle as a strategy in regenerative medicine.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation , Chromosome Aberrations , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dependovirus/classification , Dependovirus/genetics , Fibroblasts , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Genetic Vectors/genetics , Humans , Immunohistochemistry , Mice , Muscle Contraction/genetics , Mutation , RNA Interference , RNA, Small Interfering/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Serogroup , Transduction, Genetic
13.
BMC Bioinformatics ; 16: 289, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26370240

ABSTRACT

BACKGROUND: This study addresses a recurrent biological problem, that is to define a formal clustering structure for a set of tissues on the basis of the relative abundance of multiple alternatively spliced isoforms mRNAs generated by the same gene. To this aim, we have used a model-based clustering approach, based on a finite mixture of multivariate Gaussian densities. However, given we had more technical replicates from the same tissue for each quantitative measurement, we also employed a finite mixture of linear mixed models, with tissue-specific random effects. RESULTS: A panel of human tissues was analysed through quantitative real-time PCR methods, to quantify the relative amount of mRNA encoding different IGF-1 alternative splicing variants. After an appropriate, preliminary, equalization of the quantitative data, we provided an estimate of the distribution of the observed concentrations for the different IGF-1 mRNA splice variants in the cohort of tissues by employing suitable kernel density estimators. We observed that the analysed IGF-1 mRNA splice variants were characterized by multimodal distributions, which could be interpreted as describing the presence of several sub-population, i.e. potential tissue clusters. In this context, a formal clustering approach based on a finite mixture model (FMM) with Gaussian components is proposed. Due to the presence of potential dependence between the technical replicates (originated by repeated quantitative measurements of the same mRNA splice isoform in the same tissue) we have also employed the finite mixture of linear mixed models (FMLMM), which allowed to take into account this kind of within-tissue dependence. CONCLUSIONS: The FMM and the FMLMM provided a convenient yet formal setting for a model-based clustering of the human tissues in sub-populations, characterized by homogeneous values of concentrations of the mRNAs for one or multiple IGF-1 alternative splicing isoforms. The proposed approaches can be applied to any cohort of tissues expressing several alternatively spliced mRNAs generated by the same gene, and can overcome the limitations of clustering methods based on simple comparisons between splice isoform expression levels.


Subject(s)
Algorithms , Alternative Splicing/genetics , Insulin-Like Growth Factor I/genetics , RNA, Messenger/genetics , Cluster Analysis , Female , Gene Expression Profiling , Humans , Male , Normal Distribution , Protein Isoforms , RNA Isoforms , Real-Time Polymerase Chain Reaction
14.
J Biol Chem ; 289(32): 21909-25, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24891504

ABSTRACT

Loss of muscle proteins and the consequent weakness has important clinical consequences in diseases such as cancer, diabetes, chronic heart failure, and in aging. In fact, excessive proteolysis causes cachexia, accelerates disease progression, and worsens life expectancy. Muscle atrophy involves a common pattern of transcriptional changes in a small subset of genes named atrophy-related genes or atrogenes. Whether microRNAs play a role in the atrophy program and muscle loss is debated. To understand the involvement of miRNAs in atrophy we performed miRNA expression profiling of mouse muscles under wasting conditions such as fasting, denervation, diabetes, and cancer cachexia. We found that the miRNA signature is peculiar of each catabolic condition. We then focused on denervation and we revealed that changes in transcripts and microRNAs expression did not occur simultaneously but were shifted. Indeed, whereas transcriptional control of the atrophy-related genes peaks at 3 days, changes of miRNA expression maximized at 7 days after denervation. Among the different miRNAs, microRNA-206 and -21 were the most induced in denervated muscles. We characterized their pattern of expression and defined their role in muscle homeostasis. Indeed, in vivo gain and loss of function experiments revealed that miRNA-206 and miRNA-21 were sufficient and required for atrophy program. In silico and in vivo approaches identified transcription factor YY1 and the translational initiator factor eIF4E3 as downstream targets of these miRNAs. Thus miRNAs are important for fine-tuning the atrophy program and their modulation can be a novel potential therapeutic approach to counteract muscle loss and weakness in catabolic conditions.


Subject(s)
MicroRNAs/genetics , Muscular Atrophy/etiology , Muscular Atrophy/genetics , 3' Untranslated Regions , Animals , Base Sequence , Cachexia/genetics , Cachexia/metabolism , Disease Models, Animal , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Profiling , Male , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Molecular Sequence Data , Muscle Denervation , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Starvation/genetics , Starvation/metabolism , Time Factors , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
15.
Mediators Inflamm ; 2015: 537853, 2015.
Article in English | MEDLINE | ID: mdl-26491230

ABSTRACT

To determine the role of mutant SOD1 gene (SOD1(G93A)) on muscle cell differentiation, we derived C2C12 muscle cell lines carrying a stably transfected SOD1(G93A) gene under the control of a myosin light chain (MLC) promoter-enhancer cassette. Expression of MLC/SOD1(G93A) in C2C12 cells resulted in dramatic inhibition of myoblast differentiation. Transfected SOD1(G93A) gene expression in postmitotic skeletal myocytes downregulated the expression of relevant markers of committed and differentiated myoblasts such as MyoD, Myogenin, MRF4, and the muscle specific miRNA expression. The inhibitory effects of SOD1(G93A) gene on myogenic program perturbed Akt/p70 and MAPK signaling pathways which promote differentiation cascade. Of note, the inhibition of the myogenic program, by transfected SOD1(G93A) gene expression, impinged also the identity of myogenic cells. Expression of MLC/SOD1(G93A) in C2C12 myogenic cells promoted a fibro-adipogenic progenitors (FAPs) phenotype, upregulating HDAC4 protein and preventing the myogenic commitment complex BAF60C-SWI/SNF. We thus identified potential molecular mediators of the inhibitory effects of SOD1(G93A) on myogenic program and disclosed potential signaling, activated by SOD1(G93A), that affect the identity of the myogenic cell population.


Subject(s)
Mitosis , Myoblasts/metabolism , Superoxide Dismutase/genetics , Animals , Cell Differentiation , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Flow Cytometry , Histone Deacetylases/metabolism , Hydroxamic Acids/chemistry , Lipids/chemistry , Mice , MicroRNAs/metabolism , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Myogenic Regulatory Factors/metabolism , Myogenin/metabolism , Phenotype , Signal Transduction , Superoxide Dismutase-1 , Transfection , Up-Regulation
16.
Neuromolecular Med ; 26(1): 5, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38491246

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a rare neuromuscular disease with a wide disease progression. Despite several efforts to develop efficient biomarkers, many concerns about the available ones still need to be addressed. MicroRNA (miR) are non-coding RNAs that can modulate molecular circuits and are involved in ALS pathogenic mechanisms. 22 fast and 23 slow-progressing-defined ALS patients were recruited. ALSFRS-R, strength, respiratory function, nerve conduction studies, and creatine kinase were evaluated at the baseline and after 6 months of follow-up. The mean monthly reduction of the previous variables (progression index - PI) was calculated. MiR206, 133a-3p, 151a-5p, 199a-5p, and 423-3p were dosed. The univariate analysis showed an independent reduction of miR206 and an increase of miR423-3p in patients with a slow slope of ALSFRS-R and weakness, respectively. MiR206 and 423-3p are differently modulated in fast and slow-progressing ALS patients, suggesting a role for microRNAs in prognosis and therapeutic target.


Subject(s)
Amyotrophic Lateral Sclerosis , MicroRNAs , Humans , Amyotrophic Lateral Sclerosis/genetics , Disease Progression , MicroRNAs/genetics , Research Design , Biomarkers
17.
Hum Mol Genet ; 20(9): 1844-53, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21325354

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited motor neuron disease caused by the mutation of the survival motor neuron 1 (SMN1) gene and deficiency of the SMN protein. Severe SMA mice have abnormal motor function and small, immature myofibers early in development suggesting that SMN protein deficiency results in retarded muscle growth. Insulin-like growth factor 1 (IGF-1) stimulates myoblast proliferation, induces myogenic differentiation and generates myocyte hypertrophy in vitro and in vivo. We hypothesized that increased expression of IGF-1 specifically in skeletal muscle would attenuate disease features of SMAΔ7 mice. SMAΔ7 mice overexpressing a local isoform of IGF-1 (mIGF-1) in muscle showed enlarged myofibers and a 40% increase in median survival compared with mIGF-1-negative SMA littermates (median survival = 14 versus 10 days, respectively, log-rank P = 0.025). Surprisingly, this was not associated with a significant improvement in motor behavior. Treatment of both mIGF-1(NEG) and mIGF-1(POS) SMA mice with the histone deacetylase inhibitor, trichostatin A (TSA), resulted in a further extension of survival and improved motor behavior, but the combination of mIGF-1 and TSA treatment was not synergistic. These results show that increased mIGF-1 expression restricted to muscle can modulate the phenotype of SMA mice indicating that therapeutics targeted to muscle alone should not be discounted as potential disease-modifying therapies in SMA. IGF-1 may warrant further investigation in mild SMA animal models and perhaps SMA patients.


Subject(s)
Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Up-Regulation , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Motor Activity , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , SMN Complex Proteins/genetics , SMN Complex Proteins/metabolism
18.
Biogerontology ; 14(3): 273-92, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23666344

ABSTRACT

Although adult skeletal muscle is composed of fully differentiated fibers, it retains the capacity to regenerate in response to injury and to modify its contractile and metabolic properties in response to changing demands. The major role in the growth, remodeling and regeneration is played by satellite cells, a quiescent population of myogenic precursor cells that reside between the basal lamina and plasmalemma and that are rapidly activated in response to appropriate stimuli. However, in pathologic conditions or during aging, the complete regenerative program can be precluded by fibrotic tissue formation and resulting in functional impairment of the skeletal muscle. Our study, along with other studies, demonstrated that although the regenerative program can also be impaired by the limited proliferative capacity of satellite cells, this limit is not reached during normal aging, and it is more likely that the restricted muscle repair program in aging is presumably due to missing signals that usually render the damaged muscle a permissive environment for regenerative activity.


Subject(s)
Aging/physiology , Cell Proliferation , Muscle, Skeletal/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neoplasm Proteins/metabolism , Oxidative Stress/physiology , Sarcopenia/metabolism , Sarcopenia/pathology , Sarcopenia/physiopathology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Telomere/ultrastructure , Tumor Suppressor Protein p53/metabolism , Young Adult
19.
Biogerontology ; 14(3): 261-72, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23666343

ABSTRACT

It is known that adipose tissue mass increases with age, and that a number of hormones, collectively called adipokines, are produced by adipose tissue. For most of them it is not known whether their plasmatic levels change with age. Moreover, it is known that adipose tissue infiltration in skeletal muscle is related to sarcopenia and loss of muscle strength. In this study we investigated the age-related changes of representative adipokines and insulin-like growth factor (IGF)-1 and their effect on muscle strength. We studied the association between circulating levels of adiponectin, leptin, resistin and IGF-1 and muscle strength. This cross-sectional study included 412 subjects of different age (152 subjects aged 18-30 years and 260 subjects aged 69-81 years) recruited within the framework of the European research network project "Myoage". The levels of adiponectin (both in male and female subjects) and leptin (only in males) were significantly higher in old subjects compared to young, while those of IGF-1 were lower in old subjects. In old subjects adiponectin, resistin and the resistin/IGF-1 ratio (but not IGF-1 alone) were inversely associated with quadriceps torque, while only adiponectin was inversely associated with handgrip strength independently from percentage of fat mass, height, age, gender and geographical origin. The ratio of leptin to adiponectin was directly associated with handgrip strength in both young and old subjects. These results suggest that in humans the age-associated loss of strength is associated with the levels of representative adipokines and IGF-1.


Subject(s)
Adipokines/blood , Aging/physiology , Insulin-Like Growth Factor I/metabolism , Muscle Strength/physiology , Muscle, Skeletal/physiology , Adiponectin/blood , Adolescent , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Hand Strength/physiology , Humans , Leptin/blood , Male , Resistin/blood , Signal Transduction/physiology , Young Adult
20.
Cell Metab ; 35(3): 379-381, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36889277

ABSTRACT

Aging results from the combination of complex processes still largely undefined. In this issue, Benjamin et al. use multiomic analysis to reveal a causative role of altered glutathione (GSH) synthesis and metabolism in age-dependent muscle stem cell (MuSC) dysfunction, casting light on novel mechanisms regulating stem cell function and on therapeutic approaches to improve defective regeneration in the aged muscle.


Subject(s)
Muscle, Skeletal , Stem Cells , Muscle, Skeletal/metabolism , Stem Cells/metabolism , Glutathione/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL