Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Sci Rep ; 14(1): 13955, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886398

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by muscle fatigability due to acetylcholine receptor (AChR) autoantibodies. To better characterize juvenile MG (JMG), we analyzed 85 pre- and 132 post-pubescent JMG (with a cutoff age of 13) compared to 721 adult MG patients under 40 years old using a French database. Clinical data, anti-AChR antibody titers, thymectomy, and thymic histology were analyzed. The proportion of females was higher in each subgroup. No significant difference in the anti-AChR titers was observed. Interestingly, the proportion of AChR+ MG patients was notably lower among adult MG patients aged between 30 and 40 years, at 69.7%, compared to over 82.4% in the other subgroups. Thymic histological data were examined in patients who underwent thymectomy during the year of MG onset. Notably, in pre-JMG, the percentage of thymectomized patients was significantly lower (32.9% compared to more than 42.5% in other subgroups), and the delay to thymectomy was twice as long. We found a positive correlation between anti-AChR antibodies and germinal center grade across patient categories. Additionally, only females, particularly post-JMG patients, exhibited the highest rates of lymphofollicular hyperplasia (95% of cases) and germinal center grade. These findings reveal distinct patterns in JMG patients, particularly regarding thymic follicular hyperplasia, which appears to be exacerbated in females after puberty.


Subject(s)
Autoantibodies , Myasthenia Gravis , Receptors, Cholinergic , Thymectomy , Thymus Gland , Humans , Myasthenia Gravis/pathology , Myasthenia Gravis/epidemiology , Female , Male , Adult , France/epidemiology , Thymus Gland/pathology , Thymus Gland/surgery , Adolescent , Autoantibodies/immunology , Autoantibodies/blood , Receptors, Cholinergic/immunology , Young Adult , Child , Cohort Studies , Germinal Center/pathology , Germinal Center/immunology
2.
Commun Biol ; 6(1): 1266, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38092861

ABSTRACT

Culture-adapted human mesenchymal stromal cells (hMSCs) are appealing candidates for regenerative medicine applications. However, these cells implanted in lesions as single cells or tissue constructs encounter an ischemic microenvironment responsible for their massive death post-transplantation, a major roadblock to successful clinical therapies. We hereby propose a paradigm shift for enhancing hMSC survival by designing, developing, and testing an enzyme-controlled, nutritive hydrogel with an inbuilt glucose delivery system for the first time. This hydrogel, composed of fibrin, starch (a polymer of glucose), and amyloglucosidase (AMG, an enzyme that hydrolyze glucose from starch), provides physiological glucose levels to fuel hMSCs via glycolysis. hMSCs loaded in these hydrogels and exposed to near anoxia (0.1% pO2) in vitro exhibited improved cell viability and angioinductive functions for up to 14 days. Most importantly, these nutritive hydrogels promoted hMSC viability and paracrine functions when implanted ectopically. Our findings suggest that local glucose delivery via the proposed nutritive hydrogel can be an efficient approach to improve hMSC-based therapeutic efficacy.


Subject(s)
Hydrogels , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Cell Survival , Glucose/metabolism , Starch/metabolism
6.
Sci Rep ; 10(1): 10681, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32606364

ABSTRACT

One of the main challenges in cell therapy for muscle diseases is to efficiently target the muscle. To address this issue and achieve better understanding of in vivo cell fate, we evaluated the relevance of a non-invasive cell tracking method in the Golden Retriever Muscular Dystrophy (GRMD) model, a well-recognised model of Duchenne Muscular Dystrophy (DMD). Mesoangioblasts were directly labelled with 111In-oxine, and injected through one of the femoral arteries. The scintigraphy images obtained provided the first quantitative mapping of the immediate biodistribution of mesoangioblasts in a large animal model of DMD. The results revealed that cells were trapped by the first capillary filters: the injected limb and the lung. During the days following injection, radioactivity was redistributed to the liver. In vitro studies, performed with the same cells prepared for injecting the animal, revealed prominent cell death and 111In release. In vivo, cell death resulted in 111In release into the vasculature that was taken up by the liver, resulting in a non-specific and non-cell-bound radioactive signal. Indirect labelling methods would be an attractive alternative to track cells on the mid- and long-term.


Subject(s)
Cell Movement/physiology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/pathology , Stem Cells/pathology , Animals , Cell Differentiation/physiology , Cell Tracking/methods , Disease Models, Animal , Dogs , Dystrophin/metabolism , Female , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Radionuclide Imaging/methods , Stem Cells/metabolism , Tissue Distribution/physiology
7.
Mol Ther Methods Clin Dev ; 17: 317-327, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32577429

ABSTRACT

Stem cell-based therapies are a promising approach for the treatment of degenerative muscular diseases; however, clinical trials have shown inconclusive and even disappointing results so far. Noninvasive cell monitoring by medicine imaging could improve the understanding of the survival and biodistribution of cells following injection. In this study, we assessed the canine sodium iodide symporter (cNIS) reporter gene as an imaging tool to track by single-photon emission computed tomography (SPECT/CT) transduced canine myoblasts after intramuscular (IM) administrations in dogs. cNIS-expressing cells kept their myogenic capacities and showed strong 99 mTc-pertechnetate (99 mTcO4 -) uptake efficiency both in vitro and in vivo. cNIS expression allowed visualization of cells by SPECT/CT along time: 4 h, 48 h, 7 days, and 30 days after IM injection; biopsies collected 30 days post administration showed myofiber's membranes expressing cNIS. This study demonstrates that NIS can be used as a reporter to track cells in vivo in the skeletal muscle of large animals. Our results set a proof of concept of the benefits NIS-tracking tool may bring to the already challenging cell-based therapies arena in myopathies and pave the way to a more efficient translation to the clinical setting from more accurate pre-clinical results.

8.
J Cachexia Sarcopenia Muscle ; 11(4): 1047-1069, 2020 08.
Article in English | MEDLINE | ID: mdl-32157826

ABSTRACT

BACKGROUND: Aldehyde dehydrogenases (ALDHs) are key players in cell survival, protection, and differentiation via the metabolism and detoxification of aldehydes. ALDH activity is also a marker of stem cells. The skeletal muscle contains populations of ALDH-positive cells amenable to use in cell therapy, whose distribution, persistence in aging, and modifications in myopathic context have not been investigated yet. METHODS: The Aldefluor® (ALDEF) reagent was used to assess the ALDH activity of muscle cell populations, whose phenotypic characterizations were deepened by flow cytometry. The nature of ALDH isoenzymes expressed by the muscle cell populations was identified in complementary ways by flow cytometry, immunohistology, and real-time PCR ex vivo and in vitro. These populations were compared in healthy, aging, or Duchenne muscular dystrophy (DMD) patients, healthy non-human primates, and Golden Retriever dogs (healthy vs. muscular dystrophic model, Golden retriever muscular dystrophy [GRMD]). RESULTS: ALDEF+ cells persisted through muscle aging in humans and were equally represented in several anatomical localizations in healthy non-human primates. ALDEF+ cells were increased in dystrophic individuals in humans (nine patients with DMD vs. five controls: 14.9 ± 1.63% vs. 3.6 ± 0.39%, P = 0.0002) and dogs (three GRMD dogs vs. three controls: 10.9 ± 2.54% vs. 3.7 ± 0.45%, P = 0.049). In DMD patients, such increase was due to the adipogenic ALDEF+ /CD34+ populations (11.74 ± 1.5 vs. 2.8 ± 0.4, P = 0.0003), while in GRMD dogs, it was due to the myogenic ALDEF+ /CD34- cells (3.6 ± 0.6% vs. 1.03 ± 0.23%, P = 0.0165). Phenotypic characterization associated the ALDEF+ /CD34- cells with CD9, CD36, CD49a, CD49c, CD49f, CD106, CD146, and CD184, some being associated with myogenic capacities. Cytological and histological analyses distinguished several ALDH isoenzymes (ALDH1A1, 1A2, 1A3, 1B1, 1L1, 2, 3A1, 3A2, 3B1, 3B2, 4A1, 7A1, 8A1, and 9A1) expressed by different cell populations in the skeletal muscle tissue belonging to multinucleated fibres, or myogenic, endothelial, interstitial, and neural lineages, designing them as potential new markers of cell type or of metabolic activity. Important modifications were noted in isoenzyme expression between healthy and DMD muscle tissues. The level of gene expression of some isoenzymes (ALDH1A1, 1A3, 1B1, 2, 3A2, 7A1, 8A1, and 9A1) suggested their specific involvement in muscle stability or regeneration in situ or in vitro. CONCLUSIONS: This study unveils the importance of the ALDH family of isoenzymes in the skeletal muscle physiology and homeostasis, suggesting their roles in tissue remodelling in the context of muscular dystrophies.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Healthy Aging/physiology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Homeostasis , Humans
9.
Med Sci (Paris) ; 35 Hors série n° 2: 7-10, 2019 Nov.
Article in French | MEDLINE | ID: mdl-31859623

ABSTRACT

Cell therapy approaches dedicated to the treatment of dystrophinopathies and involving essentially myoblasts and mesoangioblasts have produced mitigated clinical results. If several types of alternative progenitors have been developed, no standardized comparison has been carried out yet to investigate their regenerative efficacy in vivo, at least at a local level. A comparative study has therefore been designed recently aiming at giving a new impetus to this therapeutic field.


TITLE: Thérapie cellulaire des maladies musculaires - Un avenir à l'aune d'une comparaison des progéniteurs. ABSTRACT: Les approches de thérapie cellulaire des dystrophinopathies basées sur l'utilisation de myoblastes ou de mésoangioblastes se sont traduites par des résultats cliniques mitigés. De nombreux candidats cellulaires alternatifs ont été décrits, mais aucune comparaison standardisée n'a pu encore établir leurs efficacités, ne serait-ce qu'en vue d'une régénération musculaire localisée. Une étude comparative a donc été décidée récemment et pourrait permettre de donner un nouvel élan à cette approche.


Subject(s)
Cell- and Tissue-Based Therapy/trends , Muscular Diseases/therapy , Physiology, Comparative , Stem Cells/classification , Stem Cells/physiology , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myoblasts/physiology , Physiology, Comparative/methods , Physiology, Comparative/standards , Physiology, Comparative/trends , Reference Standards , Regenerative Medicine/standards , Regenerative Medicine/trends , Stem Cells/cytology
10.
Front Neurol ; 10: 1343, 2019.
Article in English | MEDLINE | ID: mdl-31920954

ABSTRACT

Myasthenia gravis (MG) is a rare autoimmune disease mediated by pathogenic antibodies (Ab) directed against components of the neuromuscular junction (NMJ), mainly the acetylcholine receptor (AChR). The etiological mechanisms are not totally elucidated, but they include a combination of genetic predisposition, triggering event(s), and hormonal components. MG disease is associated with defective immune regulation, chronic cell activation, inflammation, and the thymus is frequently abnormal. MG is characterized by muscle fatigability that is very invalidating and can be life-threatening when respiratory muscles are affected. MG is not cured, and symptomatic treatments with acetylcholinesterase inhibitors and immunosuppressors are life-long medications associated with severe side effects (especially glucocorticoids). While the muscle is the ultimate target of the autoimmune attack, its place and role are not thoroughly described, and this mini-review will focus on the cascade of pathophysiologic mechanisms taking place at the NMJ and its consequences on the muscle biology, function, and regeneration in myasthenic patients, at the histological, cellular, and molecular levels. The fine structure of the synaptic cleft is damaged by the Ab binding that is coupled to focal complement-dependent lysis in the case of MG with anti-AChR antibodies. Cellular and molecular reactions taking place in the muscle involve several cell types as well as soluble factors. Finally, the regenerative capacities of the MG muscle tissue may be altered. Altogether, the studies reported in this review demonstrate that the muscle is not a passive target in MG, but interacts dynamically with its environment in several ways, activating mechanisms of compensation that limit the pathogenic mechanisms of the autoantibodies.

11.
Stem Cells Transl Med ; 7(10): 705-708, 2018 10.
Article in English | MEDLINE | ID: mdl-30211981

ABSTRACT

We report the case of a patient who had undergone injections of myoblasts in an infarct area 16 years before being referred for heart transplantation. The pathological examination of the explanted heart found persisting myotubes embedded in fibrosis. This finding supports the ability of myoblasts to survive in harsh environments, which can make them appealing candidates for transplantation in diseases requiring supply of new myogenic cells. Stem Cells Translational Medicine 2018;7:705-708.


Subject(s)
Myoblasts, Skeletal/transplantation , Myocardial Infarction/therapy , Adult , Fibrosis , Humans , Male , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myosin Heavy Chains , Troponin T/metabolism , Ventricular Function, Left/physiology
12.
Exp Cell Res ; 364(2): 217-223, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29458172

ABSTRACT

Cell transplantation has been challenged in several clinical indications of genetic or acquired muscular diseases, but therapeutic success were mitigated. To understand and improve the yields of tissue regeneration, we aimed at modelling the fate of CD56-positive human myoblasts after transplantation. Using immunodeficient severe combined immunodeficiency (SCID) mice as recipients, we assessed the survival, integration and satellite cell niche occupancy of human myoblasts by a triple immunohistochemical labelling of laminin, dystrophin and human lamin A/C. The counts were integrated into a classical mathematical decline equation. After injection, human cells were essentially located in the endomysium, then they disappeared progressively from D0 to D28. The final number of integrated human nuclei was grossly determined at D2 after injection, suggesting that no more efficient fusion between donor myoblasts and host fibers occurs after the resolution of the local damages created by needle insertion. Almost 1% of implanted human cells occupied a satellite-like cell niche. Our mathematical model validated by histological counting provided a reliable quantitative estimate of human myoblast survival and/or incorporation into SCID muscle fibers. Informations brought by histological labelling and this mathematical model are complementary.


Subject(s)
Cell Transplantation , Models, Biological , Muscle, Skeletal/cytology , Muscle, Skeletal/surgery , Myoblasts/cytology , Myoblasts/transplantation , Transplantation, Heterologous , Animals , Cell Survival , Humans , Mice , Mice, Inbred Strains , Mice, SCID , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism
13.
Med Sci (Paris) ; 32 Hors série n°2: 30-39, 2016 Nov.
Article in French | MEDLINE | ID: mdl-27869075

ABSTRACT

Heart failure is a major concern for public health systems, and several approaches of cellular therapy are being investigated with the goal of improving the function of these failing hearts. Many cell types have been used (skeletal myoblasts, hematopoietic, endothelial or mesenchymal progenitors, cardiac cells…), most often in the indication of post-ischemic heart failure rather than in the indication of genetic dilated cardiomyopathy. It is easier, indeed, to target a restricted area than the whole myocardium. Several clinical trials have reported slight but encouraging functional benefits, but their interpretations were frequently limited by the small sizes of cohorts, and by the biological variabilities inherent to the patients status and to the biology of the cells. These trials also shed light on unexpected mechanisms of action of the cells, which are changing the concepts and methodologies of the studies. The functional benefits observed would be due, indeed, to the secretion of trophic factors by the cells, instead of their true structural and mechanical integration within the myocardial tissue. Accordingly, the new generations of clinical trials aim at improving the size and homogeneity of the patient cohorts to increase the statistical power. On the other hand, several studies are associating or conditionning cells with biomaterials or cocktails of cytokines to improve their survival and their biological efficacy. In parallel, bio-engineering investigates several ways to support cells in vitro and in vivo, to sustain the architectural structure of the failing myocardium, to produce ex vivo some true substitutive cardiac tissue, or to purely replace the cells by their active secreted products. Several therapeutic devices should emerge from these researches, and the choice of their respective use will be ultimately guided by the medical indication.


Subject(s)
Cardiomyopathies/therapy , Cell- and Tissue-Based Therapy , Heart Diseases/therapy , Cardiomyopathy, Dilated/therapy , Embryonic Stem Cells , Heart Failure/therapy , Humans , Myoblasts , Myocytes, Cardiac , Stem Cell Transplantation
14.
Med Sci (Paris) ; 31 Spec No 3: 30-1, 2015 Nov.
Article in French | MEDLINE | ID: mdl-26546929
15.
Mol Ther ; 21(5): 1064-75, 2013 May.
Article in English | MEDLINE | ID: mdl-23439500

ABSTRACT

The functional and architectural benefits of embryonic stem cells (ESC) and myoblasts (Mb) transplantations into infarcted myocardium have been investigated extensively. Whereas ESC repopulated fibrotic areas and contributed to myocardial regeneration, Mb exerted their effects through paracrine secretions and scar remodeling. This therapeutic perspective, however, has been less explored in the setting of nonischemic dilated cardiomyopathies (DCMs). Our aim was to compare the integration and functional efficacy of ESC committed to cardiac fate by bone morphogenic protein 2 (BMP-2) pretreatment and Mb used as gold standard following their transplantation into the myocardium of a mouse model of laminopathy exhibiting a progressive and lethal DCM. After 4 and 8 weeks of transplantation, stabilization was observed in Mb-transplanted mice (P = 0.008) but not in groups of ESC-transplanted or medium-injected animals, where the left ventricular fractional shortening (LVFS) decreased by 32 ± 8% and 41 ± 8% respectively. Engrafted differentiated cells were consistently detected in myocardia of mice receiving Mb, whereas few or no cells were detected in the hearts of mice receiving ESC, except in two cases where teratomas were formed. These data suggest that committed ESC fail to integrate in DCM where scar tissue is absent to provide the appropriate niche, whereas the functional benefits of Mb transplantation might extend to nonischemic cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/therapy , Embryonic Stem Cells/transplantation , Myoblasts/transplantation , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/physiopathology , Cell Differentiation , Cell Line , Cell- and Tissue-Based Therapy , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Graft Survival , Male , Mice , Muscle Development , Myoblasts/cytology , Myoblasts/metabolism , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology , Phenotype
16.
Eur J Heart Fail ; 15(3): 267-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23152444

ABSTRACT

AIMS: The transcription factor Islet-1 (ISL1) is a marker of cardiovascular progenitors and is essential for mammalian cardiogenesis. An ISL1 haplotype has recently been associated with congenital heart disease. In this study we evaluated whether ISL1 variants are associated with hypertrophic (HCM), dilated (DCM), arrhythmogenic right ventricular cardiomyopathy (ARVC), or with Emery-Dreifuss muscular dystrophy (EDMD). METHODS AND RESULTS: The six exon and intron boundaries of ISL1 were screened for genetic variants in a cohort of 454 index cases. Eleven exonic variants were identified in HCM, DCM, ARVC, and/or EDMD. Out of the five novel variants, two are located in the 5'-untranslated region, two are silent (p.Arg171Arg and p.Asn189Asn), and one is a missense (p.Asn252Ser). The latter was identified in the homozygous state in one DCM patient, and in the heterozygous state in 11 relatives, who did not present with DCM but often with cardiovascular features. This variant was found in one HCM patient also carrying a MYH7 mutation and in 3/96 North-African Caucasian control individuals, but was absent in 138 European Caucasian control individuals. We investigated the effect of the ISL1 wild type and p.Asn252Ser mutant on myocyte enhancer factor 2C (Mef2c) promoter activity, an established ISL1 target. Mef2c promoter activity was ∼4-fold higher in the presence of wild-type and ∼6-fold higher in the presence of mutant ISL1 in both HEK and CHO cells. CONCLUSION: This study describes a new gain-of-function p.Asn252Ser variant in the human ISL1 gene, which could potentially lead to greater activation of downstream targets involved in cardiac development, dilation, and hypertrophy.


Subject(s)
Cardiomyopathies/genetics , LIM-Homeodomain Proteins/genetics , MADS Domain Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/genetics , Myogenic Regulatory Factors/metabolism , Transcription Factors/genetics , 5' Untranslated Regions/genetics , Adult , Animals , Arrhythmogenic Right Ventricular Dysplasia/genetics , CHO Cells , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Case-Control Studies , Cohort Studies , Cricetinae , Cricetulus , Exons , Female , Gene Transfer Techniques , Genetic Predisposition to Disease , HEK293 Cells , Heterozygote , Homozygote , Humans , Introns , MEF2 Transcription Factors , Male , Mutation, Missense , Pedigree , Polymorphism, Single Nucleotide , Promoter Regions, Genetic
18.
Curr Opin Organ Transplant ; 16(6): 640-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001714

ABSTRACT

PURPOSE OF REVIEW: Cell therapy is considered a potential therapeutic avenue for the treatment of skeletal muscle diseases. Heterologous and autologous approaches have been attempted in the context, respectively, of generalized degenerative disease and of localized repairs. Cell transplantation trials, however, have been hampered by poor survival and limited migratory ability of the cells. This article reviews recent problems including the identification of new putative cellular candidates, the combination of complementary genetic or pharmacological therapeutic approaches, and the set up of clinical trials. RECENT FINDINGS: Deeper investigations identified anoikis, oxidative stress, fusion inability and some administration methodologies as causes of early massive cell death. It was proposed to adapt the injection strategies or to combine them with genetic modifications of the cells or pharmacological interventions on the environment to improve the success of implantation. New myogenic cell types have been identified, mainly in the family of perivascular cells, which can be administered systemically. New concepts have emerged regarding the correction of gene expression (use of lentiviral vectors, set-up of exon skipping, direct DNA repair, etc.). SUMMARY: Initial cell transplantation trials dedicated to the repair of striated muscles in muscular dystrophies produced mitigated results and underlined some limitations of cellular candidates under study. The research and identification of new stem cell candidates, the invention of new molecular strategies for correction of gene expression, the development of complementary approaches to improve transplantation success, have been justified by the unmet medical needs. These efforts led to new preclinical and clinical trials based on these concepts.


Subject(s)
Muscle, Skeletal/surgery , Muscular Dystrophies/surgery , Myoblasts, Skeletal/transplantation , Stem Cell Transplantation , Animals , Combined Modality Therapy , Genetic Therapy , Humans , Muscle Development , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Neuromuscular Agents/therapeutic use , Recovery of Function , Stem Cell Transplantation/adverse effects , Treatment Outcome
19.
Expert Opin Biol Ther ; 11(2): 157-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21219234

ABSTRACT

INTRODUCTION: Muscular dystrophies are a heterogeneous group of genetic diseases characterized by muscle weakness, wasting and degeneration. Cell therapy consists of delivering myogenic precursor cells to damaged tissue for the complementation of missing proteins and/or the regeneration of new muscle fibres. AREAS COVERED: We focus on human candidate cells described so far (myoblasts, mesoangioblasts, pericytes, myoendothelial cells, CD133(+) cells, aldehyde-dehydrogenase-positive cells, mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells), gene-based strategies developed to modify cells prior to injection, animal models (dystrophic and/or immunodeficient) used for pre-clinical studies, and clinical trials that have been performed using cell therapy strategies. The approaches are reviewed in terms of feasibility, hurdles, potential solutions and/or research areas from where the solution may come and potential application in terms of types of dystrophies and targets. EXPERT OPINION: Cell therapy for muscular dystrophies should be put in the context of which dystrophy or muscle group is targeted, what tools are available at hand, but even more importantly what can cell therapy bring as compared with and/or in combination with other therapeutic strategies. The solution will probably be the right dosage of these combinations adapted to each dystrophy, or even to each type of mutation within a dystrophy.


Subject(s)
Cell- and Tissue-Based Therapy , Embryonic Stem Cells/transplantation , Muscular Dystrophies/therapy , Humans
20.
Stem Cells Dev ; 20(6): 1043-52, 2011 06.
Article in English | MEDLINE | ID: mdl-20942609

ABSTRACT

Islet-1 expression identifies populations of progenitor cells in embryonic, fetal, and newborn murine hearts that are able to give rise to all cardiac cell lineages ex vivo and in vivo. Using systematic immunohistochemistry, we investigated whether islet-1-positive cells are present in adult mouse heart from the perspective of their potential therapeutic utility. The presence, localization, and nature of islet-1-positive cells were assessed in mice of different strains, ages, and conditions. Islet-1-positive cells were present in mouse heart from postnatal day 1 to young adulthood. Depending on the strain, these cells were organized in either 1 or 2 types of clusters localized to restricted areas, at a distance of 6%-35% of the heart length from the base. The first type of cluster was present in all strains and consisted of neural crest-derived cells that formed cardiac ganglia. The number of cells remained stable (a few hundred) from neonatal up to adult ages, and variations were noted between strains regarding their long-term persistency. The second type of cluster was essentially present in 129SvJ or Balb/C strains and absent from the other strains tested (C57BL/6J, C3H, SJL). It consisted of cells expressing highly ordered sarcomeric actin, consistent with their having cardiomyocyte identity. These cells disappeared in animals older than 4 months. Neither the number nor the type of islet-1-positive cells varied with time in a mouse model of dilated cardiomyopathy. Our studies demonstrate that islet-1-positive cells are relatively few in number in adult murine heart, being localized in restricted and rather inaccessible areas, and can represent both neural crest and cardiomyocyte lineages.


Subject(s)
Homeodomain Proteins/metabolism , Myocardium/cytology , Animals , Biomarkers/metabolism , Cell Aggregation , Cell Count , Cell Proliferation , Gene Knock-In Techniques , LIM-Homeodomain Proteins , Mice , Mice, Inbred Strains , Models, Animal , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurofilament Proteins/metabolism , Organ Specificity , Transcription Factors , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...