Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell Rep ; 43(3): 113854, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38412099

ABSTRACT

The definition of cell metabolic profile is essential to ensure skeletal muscle fiber heterogeneity and to achieve a proper equilibrium between the self-renewal and commitment of satellite stem cells. Heme sustains several biological functions, including processes profoundly implicated with cell metabolism. The skeletal muscle is a significant heme-producing body compartment, but the consequences of impaired heme homeostasis on this tissue have been poorly investigated. Here, we generate a skeletal-muscle-specific feline leukemia virus subgroup C receptor 1a (FLVCR1a) knockout mouse model and show that, by sustaining heme synthesis, FLVCR1a contributes to determine the energy phenotype in skeletal muscle cells and to modulate satellite cell differentiation and muscle regeneration.


Subject(s)
Membrane Transport Proteins , Satellite Cells, Skeletal Muscle , Mice , Animals , Membrane Transport Proteins/metabolism , Heme/metabolism , Mice, Knockout , Muscle, Skeletal/metabolism , Energy Metabolism , Satellite Cells, Skeletal Muscle/metabolism , Cell Differentiation/physiology
2.
Stem Cell Res ; 64: 102889, 2022 10.
Article in English | MEDLINE | ID: mdl-35961103

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked syndrome that affects skeletal and cardiac muscle and is caused by mutation of the dystrophin gene. Induced pluripotent stem cells (iPSCs) were generated from dermal fibroblasts by electroporation with episomal vectors containing the reprogramming factors (OCT4, SOX2, LIN28, KLF4, and l-MYC). The donor carried an out-of-frame deletion of exons 45-50 of the dystrophin gene. The established iPSC line exhibited normal morphology, expressed pluripotency markers, had normal karyotype and possessed trilineage differentiation potential.


Subject(s)
Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Humans , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Induced Pluripotent Stem Cells/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Exons/genetics , Cell Differentiation , Fibroblasts/metabolism , Cellular Reprogramming
3.
Cell Mol Life Sci ; 79(5): 259, 2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35474498

ABSTRACT

Delayed wound healing and chronic skin lesions represent a major health problem. Over the past years, growth factors mediated by platelet-rich plasma (PRP) and cell-based therapies were developed as effective and affordable treatment able to improve wound healing capacity. We have advanced existing concepts to develop a highly efficient high-throughput protocol with proven application for the isolation of PRP and pro-angiogenic cells (AngioPRP). This protocol outlines the effectiveness of AngioPRP in promoting the critical healing process including wound closure, re-epithelialization, granulation tissue growth, and blood vessel regeneration. We coupled this effect with normalization of mechanical properties of rescued mouse wounds, which is sustained by a correct arrangement of elastin and collagen fibers. Proteomic analysis of treated wounds demonstrated a fingerprint of AngioPRP based on the up-regulation of detoxification pathway of glutathione metabolism, correlated to a decrease in inflammatory response. Overall, these results have enabled us to provide a framework for how AngioPRP supports wound healing, opening avenues for further clinical advances.


Subject(s)
Blood Platelets , Platelet-Rich Plasma , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Platelet-Rich Plasma/metabolism , Proteomics , Wound Healing/physiology
4.
Sci Transl Med ; 13(607)2021 08 18.
Article in English | MEDLINE | ID: mdl-34408077

ABSTRACT

Friedreich ataxia (FRDA) is caused by the reduced expression of the mitochondrial protein frataxin (FXN) due to an intronic GAA trinucleotide repeat expansion in the FXN gene. Although FRDA has no cure and few treatment options, there is research dedicated to finding an agent that can curb disease progression and address symptoms as neurobehavioral deficits, muscle endurance, and heart contractile dysfunctions. Because oxidative stress and mitochondrial dysfunctions are implicated in FRDA, we demonstrated the systemic delivery of catalysts activity of gold cluster superstructures (Au8-pXs) to improve cell response to mitochondrial reactive oxygen species and thereby alleviate FRDA-related pathology in mesenchymal stem cells from patients with FRDA. We also found that systemic injection of Au8-pXs ameliorated motor function and cardiac contractility of YG8sR mouse model that recapitulates the FRDA phenotype. These effects were associated to long-term improvement of mitochondrial functions and antioxidant cell responses. We related these events to an increased expression of frataxin, which was sustained by reduced autophagy. Overall, these results encourage further optimization of Au8-pXs in experimental clinical strategies for the treatment of FRDA.


Subject(s)
Friedreich Ataxia , Animals , Disease Models, Animal , Gold , Humans , Mice , Reactive Oxygen Species , Trinucleotide Repeat Expansion
5.
Nat Commun ; 12(1): 2099, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833239

ABSTRACT

In Duchenne muscular dystrophy (DMD), sarcolemma fragility and myofiber necrosis produce cellular debris that attract inflammatory cells. Macrophages and T-lymphocytes infiltrate muscles in response to damage-associated molecular pattern signalling and the release of TNF-α, TGF-ß and interleukins prevent skeletal muscle improvement from the inflammation. This immunological scenario was extended by the discovery of a specific response to muscle antigens and a role for regulatory T cells (Tregs) in muscle regeneration. Normally, autoimmunity is avoided by autoreactive T-lymphocyte deletion within thymus, while in the periphery Tregs monitor effector T-cells escaping from central regulatory control. Here, we report impairment of thymus architecture of mdx mice together with decreased expression of ghrelin, autophagy dysfunction and AIRE down-regulation. Transplantation of dystrophic thymus in recipient nude mice determine the up-regulation of inflammatory/fibrotic markers, marked metabolic breakdown that leads to muscle atrophy and loss of force. These results indicate that involution of dystrophic thymus exacerbates muscular dystrophy by altering central immune tolerance.


Subject(s)
Immune Tolerance/immunology , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Dystrophy, Animal/pathology , Thymus Gland/pathology , Animals , Autophagy/physiology , Ghrelin/biosynthesis , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Nude , Muscular Dystrophy, Duchenne/pathology , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/immunology , Thymus Gland/transplantation , Transcription Factors/biosynthesis , AIRE Protein
6.
Cell Rep ; 31(5): 107597, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32375047

ABSTRACT

Fibrosis and fat replacement in skeletal muscle are major complications that lead to a loss of mobility in chronic muscle disorders, such as muscular dystrophy. However, the in vivo properties of adipogenic stem and precursor cells remain unclear, mainly due to the high cell heterogeneity in skeletal muscles. Here, we use single-cell RNA sequencing to decomplexify interstitial cell populations in healthy and dystrophic skeletal muscles. We identify an interstitial CD142-positive cell population in mice and humans that is responsible for the inhibition of adipogenesis through GDF10 secretion. Furthermore, we show that the interstitial cell composition is completely altered in muscular dystrophy, with a near absence of CD142-positive cells. The identification of these adipo-regulatory cells in the skeletal muscle aids our understanding of the aberrant fat deposition in muscular dystrophy, paving the way for treatments that could counteract degeneration in patients with muscular dystrophy.


Subject(s)
Adipogenesis/physiology , Cell Differentiation/physiology , Leydig Cells/cytology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Animals , Fibrosis/metabolism , Fibrosis/pathology , Humans , Male , Mice , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism
7.
Stem Cell Res ; 45: 101819, 2020 05.
Article in English | MEDLINE | ID: mdl-32348941

ABSTRACT

Becker Muscular dystrophy (BMD) is an X-linked syndrome characterized by progressive muscle weakness. BMD is generally less severe than Duchenne Muscular Dystrophy. BMD is caused by mutations in the dystrophin gene that normally give rise to the production of a truncated but partially functional dystrophin protein. We generated an induced pluripotent cell line from dermal fibroblasts of a BMD patient carrying a splice mutation in the dystrophin gene (c.1705-8 T>C). The iPSC cell-line displayed the characteristic pluripotent-like morphology, expressed pluripotency markers, differentiated into cells of the three germ layers and had a normal karyotype.


Subject(s)
Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Dystrophin/genetics , Exons , Humans , Muscular Dystrophy, Duchenne/genetics , Mutation
8.
EMBO Mol Med ; 12(1): e11019, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31793167

ABSTRACT

Duchenne muscular dystrophy (DMD) is a debilitating fatal X-linked muscle disorder. Recent findings indicate that IGFs play a central role in skeletal muscle regeneration and development. Among IGFs, insulinlike growth factor 2 (IGF2) is a key regulator of cell growth, survival, migration and differentiation. The type 2 IGF receptor (IGF2R) modulates circulating and tissue levels of IGF2 by targeting it to lysosomes for degradation. We found that IGF2R and the store-operated Ca2+ channel CD20 share a common hydrophobic binding motif that stabilizes their association. Silencing CD20 decreased myoblast differentiation, whereas blockade of IGF2R increased proliferation and differentiation in myoblasts via the calmodulin/calcineurin/NFAT pathway. Remarkably, anti-IGF2R induced CD20 phosphorylation, leading to the activation of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase (SERCA) and removal of intracellular Ca2+ . Interestingly, we found that IGF2R expression was increased in dystrophic skeletal muscle of human DMD patients and mdx mice. Blockade of IGF2R by neutralizing antibodies stimulated muscle regeneration, induced force recovery and normalized capillary architecture in dystrophic mdx mice representing an encouraging starting point for the development of new biological therapies for DMD.


Subject(s)
Muscle, Skeletal/growth & development , Muscular Dystrophy, Duchenne/drug therapy , Receptor, IGF Type 2/antagonists & inhibitors , Regeneration , Animals , Binding Sites , Child , Humans , Mice , Mice, Inbred mdx , Myoblasts , Young Adult
9.
Front Neurol ; 10: 755, 2019.
Article in English | MEDLINE | ID: mdl-31396142

ABSTRACT

Background: Nutritional compounds can exert both anti-inflammatory and anti-oxidant effects. Since these events exacerbate the pathophysiology of muscular dystrophies, we investigated nutraceutical supplementation as an adjuvant therapy in dystrophic patients, to low costs and easy route of administration. Moreover, this treatment could represent an alternative therapeutic strategy for dystrophic patients who do not respond to corticosteroid treatment. Objective: A 24 weeks randomized double-blind placebo-controlled clinical study was aimed at evaluating the safety and efficacy of daily oral administration of flavonoids- and omega3-based natural supplement (FLAVOMEGA) in patients affected by muscular dystrophy with recognized muscle inflammation. Design: We screened 60 patients diagnosed for Duchenne (DMD), Facioscapulohumeral (FSHD), and Limb Girdle Muscular Dystrophy (LGMD). Using a computer-generated random allocation sequence, we stratified patients in a 2:1:1 ratio (DMD:FSHD:LGMD) to one of two treatment groups: continuous FLAVOMEGA, continuous placebo. Of 29 patients included, only 24 completed the study: 15 were given FLAVOMEGA, 14 placebo. Results: FLAVOMEGA was well tolerated with no reported adverse events. Significant treatment differences in the change from baseline in 6 min walk distance (6MWD; secondary efficacy endpoint) (P = 0.033) and in isokinetic knee extension (P = 0.039) (primary efficacy endpoint) were observed in LGMD and FSHD subjects. Serum CK levels (secondary efficacy endpoint) decreased in all FLAVOMEGA treated groups with significant difference in DMD subjects (P = 0.039). Conclusions: Although the small number of patients and the wide range of disease severity among patients reduced statistical significance, we obtained an optimal profile of safety and tolerability for the compound, showing valuable data of efficacy in primary and secondary endpoints. Trial registration number: NCT03317171 Retrospectively registered 25/10/2017.

10.
Sci Rep ; 8(1): 14659, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279586

ABSTRACT

Duchenne muscular dystrophy (DMD) is one of the most common and severe forms of muscular dystrophy. Oxidative myofibre content, muscle vasculature architecture and exercise tolerance are impaired in DMD. Several studies have demonstrated that nutrient supplements ameliorate dystrophic features, thereby enhancing muscle performance. Here, we report that dietary supplementation with a specific branched-chain amino acid-enriched mixture (BCAAem) increased the abundance of oxidative muscle fibres associated with increased muscle endurance in dystrophic mdx mice. Amelioration of the fatigue index in BCAAem-treated mdx mice was caused by a cascade of events in the muscle tissue, which were promoted by endothelial nitric oxide synthase (eNOS) activation and vascular endothelial growth factor (VEGF) expression. VEGF induction led to recruitment of bone marrow (BM)-derived endothelial progenitors (EPs), which increased the capillary density of dystrophic skeletal muscle. Functionally, BCAAem mitigated the dystrophic phenotype of mdx mice without inducing dystrophin protein expression or replacing the dystrophin-associated glycoprotein (DAG) complex in the membrane, which is typically lost in DMD. BCAAem supplementation could be an effective adjuvant strategy in DMD treatment.


Subject(s)
Amino Acids/administration & dosage , Dietary Supplements , Muscular Dystrophy, Duchenne/diet therapy , Animals , Disease Models, Animal , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Strength/drug effects , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Physical Endurance/drug effects , Vascular Endothelial Growth Factor A/metabolism
11.
Hum Mol Genet ; 26(19): 3682-3698, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28666318

ABSTRACT

α-Dystroglycanopathies are a group of muscular dystrophies characterized by α-DG hypoglycosylation and reduced extracellular ligand-binding affinity. Among other genes involved in the α-DG glycosylation process, fukutin related protein (FKRP) gene mutations generate a wide range of pathologies from mild limb girdle muscular dystrophy 2I (LGMD2I), severe congenital muscular dystrophy 1C (MDC1C), to Walker-Warburg Syndrome and Muscle-Eye-Brain disease. FKRP gene encodes for a glycosyltransferase that in vivo transfers a ribitol phosphate group from a CDP -ribitol present in muscles to α-DG, while in vitro it can be secreted as monomer of 60kDa. Consistently, new evidences reported glycosyltransferases in the blood, freely circulating or wrapped within vesicles. Although the physiological function of blood stream glycosyltransferases remains unclear, they are likely released from blood borne or distant cells. Thus, we hypothesized that freely or wrapped FKRP might circulate as an extracellular glycosyltransferase, able to exert a "glycan remodelling" process, even at distal compartments. Interestingly, we firstly demonstrated a successful transduction of MDC1C blood-derived CD133+ cells and FKRP L276IKI mouse derived satellite cells by a lentiviral vector expressing the wild-type of human FKRP gene. Moreover, we showed that LV-FKRP cells were driven to release exosomes carrying FKRP. Similarly, we observed the presence of FKRP positive exosomes in the plasma of FKRP L276IKI mice intramuscularly injected with engineered satellite cells. The distribution of FKRP protein boosted by exosomes determined its restoration within muscle tissues, an overall recovery of α-DG glycosylation and improved muscle strength, suggesting a systemic supply of FKRP protein acting as glycosyltransferase.


Subject(s)
Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/therapy , Proteins/metabolism , Animals , Disease Models, Animal , Dystroglycans/metabolism , Exosomes , Glycosylation , Glycosyltransferases/metabolism , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Myoblasts/metabolism , Pentosyltransferases , Proteins/genetics , Satellite Cells, Skeletal Muscle/transplantation , Transferases
12.
J Tissue Eng Regen Med ; 11(1): 187-196, 2017 01.
Article in English | MEDLINE | ID: mdl-24799388

ABSTRACT

Loss of skeletal muscle tissue caused by traumatic injury or damage due to myopathies produces a deficit of muscle function for which there is still no clinical treatment. Transplantation of myogenic cells, themselves or combined with materials, has been proposed to increase the regenerative capacity of skeletal muscle but it is hampered by many limitations, such as low cell survival and engraftment or immunological reaction and low biocompatibility of the exogenous materials. Recently, myoblast sheet engineering, obtained with thermoresponsive culture dishes, has attracted attention as a new technique for muscle damage treatment. For this purpose, a series of thermoresponsive hydrogels, constituted by poly(N-isopropylacrylamide-co-2-hydroxyethylmethacrylate) [p(NIPAAM-co-HEMA)] were synthesized by a simple and inexpensive free-radical polymerization of the two co-monomers with a redox initiator. Different ratios of N-isopropylacrylamide (NIPAAm) and 2-hydroxyethylmethacrylate (HEMA) have been examined to evaluate the effects on physicochemical, mechanical and optical hydrogel properties. The murine muscle cell line C2 C12 has been exploited to test the cytotoxicity of the thermoresponsive hydrogels, depending on different synthesis conditions. In this study, we have identified a thermoresponsive hydrogel that allows cell adhesion and viability, together with the detachment of viable sheet of muscle cells, giving the chance to develop further applications for muscle damage and disease. Copyright © 2014 John Wiley & Sons, Ltd.


Subject(s)
Acrylamides/chemistry , Biocompatible Materials/chemistry , Hydrogels/chemistry , Methacrylates/chemistry , Myoblasts/cytology , Tissue Engineering/methods , Animals , Calorimetry, Differential Scanning , Cell Adhesion , Cell Line , Cell Proliferation , Compressive Strength , Glass/chemistry , Materials Testing , Mice , Polymers/chemistry , Silanes/chemistry , Spectroscopy, Fourier Transform Infrared , Temperature
13.
Mol Ther ; 24(11): 1949-1964, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27506452

ABSTRACT

Duchenne muscular dystrophy is the most common genetic muscular dystrophy. It is caused by mutations in the dystrophin gene, leading to absence of muscular dystrophin and to progressive degeneration of skeletal muscle. We have demonstrated that the exon skipping method safely and efficiently brings to the expression of a functional dystrophin in dystrophic CD133+ cells injected scid/mdx mice. Golden Retriever muscular dystrophic (GRMD) dogs represent the best preclinical model of Duchenne muscular dystrophy, mimicking the human pathology in genotypic and phenotypic aspects. Here, we assess the capacity of intra-arterial delivered autologous engineered canine CD133+ cells of restoring dystrophin expression in Golden Retriever muscular dystrophy. This is the first demonstration of five-year follow up study, showing initial clinical amelioration followed by stabilization in mild and severe affected Golden Retriever muscular dystrophy dogs. The occurrence of T-cell response in three Golden Retriever muscular dystrophy dogs, consistent with a memory response boosted by the exon skipped-dystrophin protein, suggests an adaptive immune response against dystrophin.


Subject(s)
AC133 Antigen/metabolism , Adaptive Immunity , Muscular Dystrophy, Animal/therapy , Stem Cell Transplantation/methods , Animals , Cells, Cultured , Disease Models, Animal , Dogs , Follow-Up Studies , Humans , Muscular Dystrophy, Animal/immunology , Stem Cells/metabolism , Transplantation, Autologous , Treatment Outcome
14.
Stem Cells Transl Med ; 5(4): 451-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26956210

ABSTRACT

UNLABELLED: Human placental mesenchymal stromal cells (pMSCs) have never been investigated in intrauterine growth restriction (IUGR). We characterized cells isolated from placental membranes and the basal disc of six IUGR and five physiological placentas. Cell viability and proliferation were assessed every 7 days during a 6-week culture. Expression of hematopoietic, stem, endothelial, and mesenchymal markers was evaluated by flow cytometry. We characterized the multipotency of pMSCs and the expression of genes involved in mitochondrial content and function. Cell viability was high in all samples, and proliferation rate was lower in IUGR compared with control cells. All samples presented a starting heterogeneous population, shifting during culture toward homogeneity for mesenchymal markers and occurring earlier in IUGR than in controls. In vitro multipotency of IUGR-derived pMSCs was restricted because their capacity for adipocyte differentiation was increased, whereas their ability to differentiate toward endothelial cell lineage was decreased. Mitochondrial content and function were higher in IUGR pMSCs than controls, possibly indicating a shift from anaerobic to aerobic metabolism, with the loss of the metabolic characteristics that are typical of undifferentiated multipotent cells. SIGNIFICANCE: This study demonstrates that the loss of endothelial differentiation potential and the increase of adipogenic ability are likely to play a significant role in the vicious cycle of abnormal placental development in intrauterine growth restriction (IUGR). This is the first observation of a potential role for placental mesenchymal stromal cells in intrauterine growth restriction, thus leading to new perspectives for the treatment of IUGR.


Subject(s)
Fetal Growth Retardation/pathology , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic , Placenta/pathology , Case-Control Studies , Cell Differentiation/genetics , Cell Movement/genetics , Cells, Cultured , Colony-Forming Units Assay , Endothelium, Vascular/physiology , Female , Fetal Growth Retardation/genetics , Humans , Microvessels/physiology , Neovascularization, Physiologic/genetics , Placenta/blood supply , Pregnancy
15.
Curr Gene Ther ; 15(6): 563-71, 2015.
Article in English | MEDLINE | ID: mdl-26415573

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by the loss of a functional dystrophin protein; the muscles of DMD patients progressively degenerate as a result of mechanical stress during contractions, and the condition eventually leads to premature death. By means antisense oligonucleotides (AONs), it is possible to modulate pre-mRNA splicing eliminating mutated exons and restoring dystrophin open reading frame. To overcome the hurdles in using AONs for therapeutic interventions, we exerted engineered human DMD stem cells with a lentivirus, which permanently and efficiently delivered the cloned AONs. Here we describe for the first time the exosome-mediated release of AONs from engineered human DMD CD133+ stem cells allowing the rescue of murine dystrophin expression. Finally, upon release, AONs could be internalized by host cells suggesting a potential role of exosomes acting as vesicular carriers for DMD gene therapy.


Subject(s)
Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Stem Cells/cytology , Animals , Bystander Effect/physiology , Cells, Cultured , Dystrophin/biosynthesis , Exons/genetics , Humans , Mice , Mice, SCID , Muscle, Skeletal/pathology , Oligonucleotides, Antisense/genetics , RNA Splicing/genetics
16.
Biomed Res Int ; 2015: 680615, 2015.
Article in English | MEDLINE | ID: mdl-25861640

ABSTRACT

Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, is characterized by muscular wasting caused by dystrophin deficiency that ultimately ends in force reduction and premature death. In addition to primary genetic defect, several mechanisms contribute to DMD pathogenesis. Recently, antioxidant supplementation was shown to be effective in the treatment of multiple diseases including muscular dystrophy. Different mechanisms were hypothesized such as reduced hydroxyl radicals, nuclear factor-κB deactivation, and NO protection from inactivation. Following these promising evidences, we investigated the effect of the administration of a mix of dietary natural polyphenols (ProAbe) on dystrophic mdx mice in terms of muscular architecture and functionality. We observed a reduction of muscle fibrosis deposition and myofiber necrosis together with an amelioration of vascularization. More importantly, the recovery of the morphological features of dystrophic muscle leads to an improvement of the endurance of treated dystrophic mice. Our data confirmed that ProAbe-based diet may represent a strategy to coadjuvate the treatment of DMD.


Subject(s)
Muscle, Skeletal/drug effects , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Polyphenols/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Fibrosis/drug therapy , Fibrosis/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myofibrils/drug effects , Myofibrils/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism
17.
Cell Transplant ; 24(2): 213-22, 2015.
Article in English | MEDLINE | ID: mdl-24268028

ABSTRACT

We previously developed a collagen tube filled with autologous skin-derived stem cells (SDSCs) for bridging long rat sciatic nerve gaps. Here we present a case report describing a compassionate use of this graft for repairing the polyinjured motor and sensory nerves of the upper arms of a patient. Preclinical assessment was performed with collagen/SDSC implantation in rats after sectioning the sciatic nerve. For the patient, during the 3-year follow-up period, functional recovery of injured median and ulnar nerves was assessed by pinch gauge test and static two-point discrimination and touch test with monofilaments, along with electrophysiological and MRI examinations. Preclinical experiments in rats revealed rescue of sciatic nerve and no side effects of patient-derived SDSC transplantation (30 and 180 days of treatment). In the patient treatment, motor and sensory functions of the median nerve demonstrated ongoing recovery postimplantation during the follow-up period. The results indicate that the collagen/SDSC artificial nerve graft could be used for surgical repair of larger defects in major lesions of peripheral nerves, increasing patient quality of life by saving the upper arms from amputation.


Subject(s)
Multiple Trauma/therapy , Peripheral Nerve Injuries/therapy , Stem Cell Transplantation , Stem Cells/cytology , Animals , Brain/diagnostic imaging , Collagen/chemistry , Female , Humans , Insemination, Artificial, Heterologous , Male , Nerve Regeneration , Peripheral Nerve Injuries/diagnostic imaging , Peripheral Nerve Injuries/pathology , Radiography , Rats , Rats, Nude , Recovery of Function , Sciatic Nerve/pathology , Skin/cytology , Transplantation, Autologous , Young Adult
18.
Tissue Eng Part C Methods ; 20(4): 308-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23879738

ABSTRACT

Spatiotemporal interactions play important roles in tissue development and function, especially in stem cell-seeded bioscaffolds. Cells interact with the surface of bioscaffold polymers and influence material-driven control of cell differentiation. In vitro cultures of different human progenitor cells, that is, endothelial colony-forming cells (ECFCs) from a healthy control and a patient with Kaposi sarcoma (an angioproliferative disease) and human CD133+ muscle-derived stem cells (MSH 133+ cells), were seeded onto polyglycolic acid-polylactic acid scaffolds. Three-dimensional (3D) images were obtained by X-ray phase-contrast microtomography (micro-CT) and processed with the Modified Bronnikov Algorithm. The method enabled high spatial resolution detection of the 3D structural organization of cells on the bioscaffold and evaluation of the way and rate at which cells modified the construct at different time points from seeding. The different cell types displayed significant differences in the proliferation rate. In conclusion, X-ray synchrotron radiation phase-contrast micro-CT analysis proved to be a useful and sensitive tool to investigate the spatiotemporal pattern of progenitor cell organization on a bioscaffold.


Subject(s)
Lactic Acid/pharmacology , Polyglycolic Acid/pharmacology , Stem Cells/cytology , Stem Cells/drug effects , Synchrotrons , Tissue Scaffolds/chemistry , X-Ray Microtomography/methods , Cell Count , Cells, Cultured , Colony-Forming Units Assay , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Lactic Acid/chemistry , Muscle Cells/cytology , Muscle Cells/drug effects , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Refractometry , X-Rays
19.
FEBS J ; 280(23): 6045-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24028392

ABSTRACT

The protein dysferlin is abundantly expressed in skeletal and cardiac muscles, where its main function is membrane repair. Mutations in the dysferlin gene are involved in two autosomal recessive muscular dystrophies: Miyoshi myopathy and limb-girdle muscular dystrophy type 2B. Development of effective therapies remains a great challenge. Strategies to repair the dysferlin gene by skipping mutated exons, using antisense oligonucleotides (AONs), may be suitable only for a subset of mutations, while cell and gene therapy can be extended to all mutations. AON-treated blood-derived CD133+ stem cells isolated from patients with Miyoshi myopathy led to partial dysferlin reconstitution in vitro but failed to express dysferlin after intramuscular transplantation into scid/blAJ dysferlin null mice. We thus extended these experiments producing the full-length dysferlin mediated by a lentiviral vector in blood-derived CD133+ stem cells isolated from the same patients. Transplantation of engineered blood-derived CD133+ stem cells into scid/blAJ mice resulted in sufficient dysferlin expression to correct functional deficits in skeletal muscle membrane repair. Our data suggest for the first time that lentivirus-mediated delivery of full-length dysferlin in stem cells isolated from Miyoshi myopathy patients could represent an alternative therapeutic approach for treatment of dysferlinopathies.


Subject(s)
Antigens, CD/metabolism , Distal Myopathies/therapy , Glycoproteins/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/therapy , Oligonucleotides, Antisense/pharmacology , Peptides/metabolism , Stem Cell Transplantation , Stem Cells/cytology , AC133 Antigen , Adult , Animals , Blotting, Western , Cells, Cultured , Distal Myopathies/genetics , Distal Myopathies/pathology , Dysferlin , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Injections, Intramuscular , Lentivirus/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred A , Mice, SCID , Muscle Proteins/genetics , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
20.
Methods Mol Biol ; 1052: 143-52, 2013.
Article in English | MEDLINE | ID: mdl-23640253

ABSTRACT

Micro-CT offers high spatial resolution of the distribution of stem cells and provides rapid reconstruction of 3D images and quantitative volumetric analysis. Together with real-time PCR analysis, micro-CT offers the possibility to obtain a quantification of the number of cells that are able to migrate from the bloodstream inside the muscular tissues. Here, we studied for the first time the kinetics of the human cells injected into the femoral artery of DMD animal model. It is fundamental to determine whether the cells disseminate and entrap only within the capillary system of downstream muscles and/or they are able to reach the non-injected muscles and other organs through blood flow. The efficient transplantation of stem cells to dystrophic-deficient muscle reinforced the utility of intra-arterial delivery of cells as a viable approach for cell-based clinical therapies of neuromuscular diseases.


Subject(s)
Imaging, Three-Dimensional/methods , Stem Cell Transplantation , Transplantation, Heterologous , X-Ray Microtomography/methods , Animals , Cell Movement , Cell- and Tissue-Based Therapy , Cells, Cultured , Humans , Mice , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...