102 results on '"Vilquin JT"'
Search Results
2. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial: first randomized placebo-controlled study of myoblast transplantation.
- Author
-
Menasché P, Alfieri O, Janssens S, McKenna W, Reichenspurner H, Trinquart L, Vilquin JT, Marolleau JP, Seymour B, Larghero J, Lake S, Chatellier G, Solomon S, Desnos M, and Hagège AA
- Published
- 2008
3. Courrier : glucocorticoïde et myogenèse
- Author
-
Poindron, P, primary, Warter, JM, additional, Braun, S, additional, Vilquin, JT, additional, Tranchant, C, additional, and Labouret, P, additional
- Published
- 1989
- Full Text
- View/download PDF
4. Skeletal myoblast transplantation in ischemic heart failure: long-term follow-up of the first phase I cohort of patients.
- Author
-
Hagège AA, Marolleau JP, Vilquin JT, Alhéritière A, Peyrard S, Duboc D, Abergel E, Messas E, Mousseaux E, Schwartz K, Desnos M, Menasché P, Hagège, Albert A, Marolleau, Jean-Pierre, Vilquin, Jean-Thomas, Alhéritière, Armelle, Peyrard, Séverine, Duboc, Denis, Abergel, Eric, and Messas, Emmanuel
- Published
- 2006
5. Comparison of juvenile and adult myasthenia gravis in a French cohort with focus on thymic histology.
- Author
-
Truffault F, Auger L, Dragin N, Vilquin JT, Fadel E, Thomas de Montpreville V, Mansuet-Lupo A, Regnard JF, Alifano M, Sharshar T, Behin A, Eymard B, Bolgert F, Demeret S, Berrih-Aknin S, and Le Panse R
- Subjects
- Humans, Female, Male, Adult, France epidemiology, Adolescent, Young Adult, Child, Cohort Studies, Germinal Center pathology, Germinal Center immunology, Myasthenia Gravis pathology, Myasthenia Gravis epidemiology, Thymus Gland pathology, Thymus Gland surgery, Thymectomy, Autoantibodies immunology, Autoantibodies blood, Receptors, Cholinergic immunology
- 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., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. Enzyme-controlled, nutritive hydrogel for mesenchymal stromal cell survival and paracrine functions.
- Author
-
Denoeud C, Luo G, Paquet J, Boisselier J, Wosinski P, Moya A, Diallo A, Larochette N, Marinesco S, Meiller A, Becquart P, Moussi H, Vilquin JT, Logeart-Avramoglou D, Gand A, Larreta-Garde V, Pauthe E, Potier E, and Petite H
- Subjects
- Humans, Cell Survival, Glucose metabolism, Starch metabolism, Hydrogels, Mesenchymal Stem Cells metabolism
- 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% pO
2 ) 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., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
7. [CAR-T cells: therapeutic weapons to fight muscle fibrosis?]
- Author
-
Bayer Wildberger A and Vilquin JT
- Subjects
- Humans, Fibrosis, Muscles, Immunotherapy, Adoptive, Weapons, T-Lymphocytes
- Published
- 2022
- Full Text
- View/download PDF
8. [Exosomes: Multi-faceted natural intercellular messengers as new candidates for the treatment of myopathies?]
- Author
-
Bayer-Wildberger A, Lorant J, and Vilquin JT
- Subjects
- Humans, Proteins, Exosomes, Muscular Diseases therapy
- Published
- 2021
- Full Text
- View/download PDF
9. The authors reply: Comment on: "Aldehyde dehydrogenases contribute to skeletal muscle homeostasis in healthy, aging, and Duchenne muscular dystrophy patients" by Etienne et al.
- Author
-
Etienne J, Joanne P, Catelain C, Riveron S, Bayer Wildberger A, Lafable J, Punzon I, Blot S, Agbulut O, and Vilquin JT
- Subjects
- Aging, Aldehydes, Homeostasis, Humans, Muscle, Skeletal, Oxidoreductases, Muscular Dystrophy, Duchenne
- Published
- 2020
- Full Text
- View/download PDF
10. Aldehyde dehydrogenases contribute to skeletal muscle homeostasis in healthy, aging, and Duchenne muscular dystrophy patients.
- Author
-
Etienne J, Joanne P, Catelain C, Riveron S, Bayer AC, Lafable J, Punzon I, Blot S, Agbulut O, and Vilquin JT
- Subjects
- Homeostasis, Humans, Aldehyde Dehydrogenase metabolism, Healthy Aging physiology, Muscle, Skeletal metabolism, Muscular Dystrophy, Duchenne physiopathology
- 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., (© 2020 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)- Published
- 2020
- Full Text
- View/download PDF
11. In vivo stem cell tracking using scintigraphy in a canine model of DMD.
- Author
-
Barthélémy I, Thibaud JL, de Fornel P, Cassano M, Punzón I, Mauduit D, Vilquin JT, Devauchelle P, Sampaolesi M, and Blot S
- Subjects
- 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, Cell Movement physiology, Muscular Dystrophy, Animal pathology, Muscular Dystrophy, Duchenne pathology, Stem Cells pathology
- 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
111 In-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 and111 In release. In vivo, cell death resulted in111 In 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.- Published
- 2020
- Full Text
- View/download PDF
12. In Vivo Myoblasts Tracking Using the Sodium Iodide Symporter Gene Expression in Dogs.
- Author
-
Punzón I, Mauduit D, Holvoet B, Thibaud JL, de Fornel P, Deroose CM, Blanchard-Gutton N, Vilquin JT, Sampaolesi M, Barthélémy I, and Blot S
- 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 m Tc-pertechnetate (99 m TcO4 - ) 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., (© 2020 The Authors.)- Published
- 2020
- Full Text
- View/download PDF
13. The Muscle Is Not a Passive Target in Myasthenia Gravis.
- Author
-
Vilquin JT, Bayer AC, Le Panse R, and Berrih-Aknin S
- 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., (Copyright © 2019 Vilquin, Bayer, Le Panse and Berrih-Aknin.)
- Published
- 2019
- Full Text
- View/download PDF
14. [Cell therapy in muscular disorders: the future lies in comparisons of progenitors].
- Author
-
Vilquin JT and Braun S
- Subjects
- Animals, Cell Differentiation, Cell- and Tissue-Based Therapy methods, Cells, Cultured, Humans, Muscle, Skeletal cytology, Muscle, Skeletal physiology, Myoblasts physiology, Reference Standards, Regenerative Medicine standards, Regenerative Medicine trends, Stem Cells cytology, Cell- and Tissue-Based Therapy trends, Muscular Diseases therapy, Physiology, Comparative methods, Physiology, Comparative standards, Physiology, Comparative trends, Stem Cells classification, Stem Cells physiology
- 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., (© 2019 médecine/sciences – Inserm.)
- Published
- 2019
- Full Text
- View/download PDF
15. Long-Term Engraftment (16 Years) of Myoblasts in a Human Infarcted Heart.
- Author
-
Crahès M, Bories MC, Vilquin JT, Marolleau JP, Desnos M, Larghero J, Soulat G, Bruneval P, Hagège AA, and Menasché P
- Subjects
- 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, Myoblasts, Skeletal transplantation, Myocardial Infarction therapy
- 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., (© 2018 The Authors. Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.)
- Published
- 2018
- Full Text
- View/download PDF
16. Modelling human myoblasts survival upon xenotransplantation into immunodeficient mouse muscle.
- Author
-
Praud C, Vauchez K, Zongo P, and Vilquin JT
- Subjects
- Animals, Cell Survival, Humans, Mice, Mice, Inbred Strains, Mice, SCID, Muscle Fibers, Skeletal cytology, Muscle Fibers, Skeletal metabolism, Cell Transplantation, Models, Biological, Muscle, Skeletal cytology, Muscle, Skeletal surgery, Myoblasts cytology, Myoblasts transplantation, Transplantation, Heterologous
- 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., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. [Cell therapies for cardiopathies: the shift of paradigms].
- Author
-
Vilquin JT and Etienne J
- Subjects
- Cardiomyopathy, Dilated therapy, Embryonic Stem Cells, Heart Failure therapy, Humans, Myoblasts, Myocytes, Cardiac, Stem Cell Transplantation, Cardiomyopathies therapy, Cell- and Tissue-Based Therapy, Heart Diseases therapy
- 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., (© 2016 médecine/sciences – Inserm.)
- Published
- 2016
- Full Text
- View/download PDF
18. [Not Available].
- Author
-
Vilquin JT
- Published
- 2015
- Full Text
- View/download PDF
19. Myoblasts and embryonic stem cells differentially engraft in a mouse model of genetic dilated cardiomyopathy.
- Author
-
Catelain C, Riveron S, Papadopoulos A, Mougenot N, Jacquet A, Vauchez K, Yada E, Pucéat M, Fiszman M, Butler-Browne G, Bonne G, and Vilquin JT
- Subjects
- 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, Cardiomyopathy, Dilated therapy, Embryonic Stem Cells transplantation, Myoblasts transplantation
- 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.
- Published
- 2013
- Full Text
- View/download PDF
20. A novel genetic variant in the transcription factor Islet-1 exerts gain of function on myocyte enhancer factor 2C promoter activity.
- Author
-
Friedrich FW, Dilanian G, Khattar P, Juhr D, Gueneau L, Charron P, Fressart V, Vilquin JT, Isnard R, Gouya L, Richard P, Hammoudi N, Komajda M, Bonne G, Eschenhagen T, Dubourg O, Villard E, and Carrier L
- Subjects
- 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, Cardiomyopathies genetics, LIM-Homeodomain Proteins genetics, MADS Domain Proteins metabolism, Muscular Dystrophy, Emery-Dreifuss genetics, Myogenic Regulatory Factors metabolism, Transcription Factors genetics
- 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.
- Published
- 2013
- Full Text
- View/download PDF
21. Converting pathological cells to therapeutic ones: an odyssey through pluripotency.
- Author
-
Vilquin JT
- Subjects
- Animals, Humans, Embryonic Stem Cells physiology, Induced Pluripotent Stem Cells physiology, Muscle, Skeletal physiopathology, Muscular Dystrophy, Duchenne therapy, Myoblasts, Skeletal transplantation
- Published
- 2012
- Full Text
- View/download PDF
22. Endoplasmic reticulum stress does not mediate palmitate-induced insulin resistance in mouse and human muscle cells.
- Author
-
Hage Hassan R, Hainault I, Vilquin JT, Samama C, Lasnier F, Ferré P, Foufelle F, and Hajduch E
- Subjects
- Animals, Biomarkers metabolism, Cell Line, Cells, Cultured, Endoribonucleases metabolism, Glycogen metabolism, Glycosylation drug effects, Humans, Mice, Muscle Cells drug effects, Muscle Fibers, Skeletal drug effects, Muscle Fibers, Skeletal metabolism, Myoblasts drug effects, Myoblasts metabolism, Protein Processing, Post-Translational drug effects, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Tunicamycin pharmacology, Endoplasmic Reticulum Stress drug effects, Insulin Resistance, Muscle Cells metabolism, Palmitic Acid adverse effects
- Abstract
Aims/hypothesis: Recent experiments in liver and adipocyte cell lines indicate that palmitate can induce endoplasmic reticulum (ER) stress. Since it has been shown that ER stress can interfere with insulin signalling, our hypothesis was that the deleterious action of palmitate on the insulin signalling pathway in muscle cells could also involve ER stress., Methods: We used C2C12 and human myotubes that were treated either with palmitate or tunicamycin. Total lysates and RNA were prepared for western blotting or quantitative RT-PCR respectively. Glycogen synthesis was assessed by [¹⁴C]glucose incorporation., Results: Incubation of myotubes with palmitate or tunicamycin inhibited insulin-stimulated protein kinase B (PKB)/ v-akt murine thymoma viral oncogene homologue 1 (Akt). In parallel, an increase in ER stress markers was observed. Pre-incubation with chemical chaperones that reduce ER stress only prevented tunicamycin but not palmitate-induced insulin resistance. We hypothesised that ER stress activation levels induced by palmitate may not be high enough to induce insulin resistance, in contrast with tunicamycin-induced ER stress. Indeed, tunicamycin induced a robust activation of the inositol-requiring enzyme 1 (IRE-1)/c-JUN NH₂-terminal kinase (JNK) pathway, leading to serine phosphorylation of insulin receptor substrate 1 (IRS-1) and a decrease in IRS-1 tyrosine phosphorylation. In contrast, palmitate only induced a very weak activation of the IRE1/JNK pathway, with no IRS1 serine phosphorylation., Conclusions/interpretation: These data show that insulin resistance induced by palmitate is not related to ER stress in muscle cells.
- Published
- 2012
- Full Text
- View/download PDF
23. Cell therapy for muscular dystrophies: advances and challenges.
- Author
-
Vilquin JT, Catelain C, and Vauchez K
- Subjects
- 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, Treatment Outcome, Muscle, Skeletal surgery, Muscular Dystrophies surgery, Myoblasts, Skeletal transplantation, Stem Cell Transplantation adverse effects
- 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.
- Published
- 2011
- Full Text
- View/download PDF
24. Distinction between two populations of islet-1-positive cells in hearts of different murine strains.
- Author
-
Khattar P, Friedrich FW, Bonne G, Carrier L, Eschenhagen T, Evans SM, Schwartz K, Fiszman MY, and Vilquin JT
- Subjects
- 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, Homeodomain Proteins metabolism, Myocardium cytology
- 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.
- Published
- 2011
- Full Text
- View/download PDF
25. Current advances in cell therapy strategies for muscular dystrophies.
- Author
-
Negroni E, Vallese D, Vilquin JT, Butler-Browne G, Mouly V, and Trollet C
- Subjects
- Humans, Cell- and Tissue-Based Therapy, Embryonic Stem Cells transplantation, Muscular Dystrophies therapy
- 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.
- Published
- 2011
- Full Text
- View/download PDF
26. Characterization of distinct mesenchymal-like cell populations from human skeletal muscle in situ and in vitro.
- Author
-
Lecourt S, Marolleau JP, Fromigué O, Vauchez K, Andriamanalijaona R, Ternaux B, Lacassagne MN, Robert I, Boumédiene K, Chéreau F, Marie P, Larghéro J, Fiszman M, and Vilquin JT
- Subjects
- Biomarkers analysis, Biomarkers metabolism, Cell Culture Techniques, Cell Differentiation physiology, Cell Lineage physiology, Cell Separation methods, Cells, Cultured, Clone Cells, Gene Expression, Humans, Immunophenotyping, In Situ Hybridization, Fluorescence, Magnetics, Mesenchymal Stem Cells immunology, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells physiology, Microspheres, Muscle, Skeletal immunology, Muscle, Skeletal metabolism, Muscle, Skeletal physiology, Mesenchymal Stem Cells cytology, Muscle, Skeletal cytology
- Abstract
Human skeletal muscle is an essential source of various cellular progenitors with potential therapeutic perspectives. We first used extracellular markers to identify in situ the main cell types located in a satellite position or in the endomysium of the skeletal muscle. Immunohistology revealed labeling of cells by markers of mesenchymal (CD13, CD29, CD44, CD47, CD49, CD62, CD73, CD90, CD105, CD146, and CD15 in this study), myogenic (CD56), angiogenic (CD31, CD34, CD106, CD146), hematopoietic (CD10, CD15, CD34) lineages. We then analysed cell phenotypes and fates in short- and long-term cultures of dissociated muscle biopsies in a proliferation medium favouring the expansion of myogenic cells. While CD56(+) cells grew rapidly, a population of CD15(+) cells emerged, partly from CD56(+) cells, and became individualized. Both populations expressed mesenchymal markers similar to that harboured by human bone marrow-derived mesenchymal stem cells. In differentiation media, both CD56(+) and CD15(+) cells shared osteogenic and chondrogenic abilities, while CD56(+) cells presented a myogenic capacity and CD15(+) cells presented an adipogenic capacity. An important proportion of cells expressed the CD34 antigen in situ and immediately after muscle dissociation. However, CD34 antigen did not persist in culture and this initial population gave rise to adipogenic cells. These results underline the diversity of human muscle cells, and the shared or restricted commitment abilities of the main lineages under defined conditions., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
27. Aldehyde dehydrogenase activity identifies a population of human skeletal muscle cells with high myogenic capacities.
- Author
-
Vauchez K, Marolleau JP, Schmid M, Khattar P, Chapel A, Catelain C, Lecourt S, Larghéro J, Fiszman M, and Vilquin JT
- Subjects
- Adipogenesis physiology, Cell Differentiation physiology, Cells, Cultured, Flow Cytometry, Humans, Immunohistochemistry, In Vitro Techniques, Phenotype, Aldehyde Dehydrogenase metabolism, Muscle Cells cytology, Muscle Development physiology, Muscle, Skeletal cytology
- Abstract
Aldehyde dehydrogenase 1A1 (ALDH) activity is one hallmark of human bone marrow (BM), umbilical cord blood (UCB), and peripheral blood (PB) primitive progenitors presenting high reconstitution capacities in vivo. In this study, we have identified ALDH(+) cells within human skeletal muscles, and have analyzed their phenotypical and functional characteristics. Immunohistofluorescence analysis of human muscle tissue sections revealed rare endomysial cells. Flow cytometry analysis using the fluorescent substrate of ALDH, Aldefluor, identified brightly stained (ALDH(br)) cells with low side scatter (SSC(lo)), in enzymatically dissociated muscle biopsies, thereafter abbreviated as SMALD(+) (for skeletal muscle ALDH(+)) cells. Phenotypical analysis discriminated two sub-populations according to CD34 expression: SMALD(+)/CD34(-) and SMALD(+)/CD34(+) cells. These sub-populations did not initially express endothelial (CD31), hematopoietic (CD45), and myogenic (CD56) markers. Upon sorting, however, whereas SMALD(+)/CD34(+) cells developed in vitro as a heterogeneous population of CD56(-) cells able to differentiate in adipoblasts, the SMALD(+)/CD34(-) fraction developed in vitro as a highly enriched population of CD56(+) myoblasts able to form myotubes. Moreover, only the SMALD(+)/CD34(-) population maintained a strong myogenic potential in vivo upon intramuscular transplantation. Our results suggest that ALDH activity is a novel marker for a population of new human skeletal muscle progenitors presenting a potential for cell biology and cell therapy.
- Published
- 2009
- Full Text
- View/download PDF
28. Discrepancies between the fate of myoblast xenograft in mouse leg muscle and NMR label persistency after loading with Gd-DTPA or SPIOs.
- Author
-
Baligand C, Vauchez K, Fiszman M, Vilquin JT, and Carlier PG
- Subjects
- Animals, Cell Death, Cell Survival, Cell Transplantation methods, Cells, Cultured, Contrast Media toxicity, Disease Models, Animal, Ferrosoferric Oxide toxicity, Flow Cytometry, Gadolinium DTPA toxicity, Half-Life, Hindlimb, Humans, Macrophages, Mice, Mice, Inbred C57BL, Myoblasts cytology, Myoblasts drug effects, Nanoparticles, Phagocytosis, Prussian Blue Reaction, Time Factors, Transplantation, Heterologous methods, Contrast Media pharmacokinetics, Ferrosoferric Oxide pharmacokinetics, Gadolinium DTPA pharmacokinetics, Magnetic Resonance Spectroscopy, Myoblasts transplantation, Transplantation Immunology
- Abstract
1H-NMR (nuclear magnetic resonance) imaging is regularly proposed to non-invasively monitor cell therapy protocols. Prior to transplantation, cells must be loaded with an NMR contrast agent (CA). Most studies performed so far make use of superparamagnetic iron oxide particles (SPIOs), mainly for favorable detection sensitivity. However, in the case of labeled cell death, SPIO recapture by inflammatory cells might introduce severe bias. We investigated whether NMR signal changes induced by preloading with SPIOs or the low molecular weight gadolinium (Gd)-DTPA accurately monitored the outcome of transplanted cells in a murine model of acute immunologic rejection. CA-loaded human myoblasts were grafted in the tibialis anterior of C57BL/6 mice. NMR imaging was repeated regularly until 3 months post-transplantation. Label outcome was evaluated by the size of the labeled area and its relative contrast to surrounding tissue. In parallel, immunohistochemistry assessed the presence of human cells. Data analysis revealed that CA-induced signal changes did not strictly reflect the graft status. Gd-DTPA label disappeared rapidly yet with a 2-week delay compared with immunohistochemical evaluation. More problematically, SPIO label was still visible after 3 months, grossly overestimating cell survival (<1 week). SPIOs should be used with extreme caution to evaluate the presence of grafted cells in vivo and could hardly be recommended for the long-term monitoring of cell transplantation protocols.
- Published
- 2009
- Full Text
- View/download PDF
29. Differentiation potential of human muscle-derived cells towards chondrogenic phenotype in alginate beads culture.
- Author
-
Andriamanalijaona R, Duval E, Raoudi M, Lecourt S, Vilquin JT, Marolleau JP, Pujol JP, Galera P, and Boumediene K
- Subjects
- Alginates, CD56 Antigen metabolism, Cells, Cultured, Humans, Immunohistochemistry, Muscle, Skeletal metabolism, Phenotype, Cell Differentiation physiology, Chondrocytes cytology, Chondrogenesis physiology, Muscle, Skeletal cytology
- Abstract
Objective: The aim of this study was to evaluate the differentiation potential of two populations of muscle-derived cells (CD56- and CD56+) towards chondrogenic phenotype in alginate beads culture and to compare the effect of transforming growth factor beta 1 (TGFbeta1) on the differentiation process in these populations., Methods: Muscle CD56- and CD56+ cells were cultured in alginate beads, in a chondrogenic medium, containing or not TGFbeta1 (10 ng/ml). Cultures were maintained for 3, 7, 14 or 21 days in a humidified culture incubator. At harvest, one culture of each set was fixed for alcian blue staining and aggrecan detection. The steady-state level of matrix macromolecules mRNA was assessed by real-time polymerase chain reaction (PCR). Protein detection was performed by western-blot analysis. The binding activity of nuclear extracts to Cbfa1 DNA sequence was also evaluated by electrophoretic mobility shift assays (EMSA)., Results: Chondrogenic differentiation of both CD56+ and CD56- muscle-derived cells was improved in alginate scaffold, even without growth factor, as suggested by increased chondrogenesis markers expression during the culture. Furthermore, TGFbeta1 enhanced the differentiation process and allowed to maintain a high expression of markers of mature chondrocytes. Of importance, the combination of alginate and TGFbeta1 treatment resulted in a further down-regulation of collagen type I and type X, as well as Cbfa1 both expression and binding activity., Conclusions: Thus, alginate scaffold and chondrogenic medium are sufficient to lead both populations CD56+ and CD56- towards chondrogenic differentiation. Moreover, TGFbeta1 enhances this process and allows to maintain the chondrogenic phenotype by inhibiting terminal differentiation, particularly for CD56- cells.
- Published
- 2008
- Full Text
- View/download PDF
30. Myoblast xenotransplantation as a tool to evaluate the appropriateness of nanoparticular versus cellular trackers.
- Author
-
Praud C, Vauchez K, Lombes A, Fiszman MY, and Vilquin JT
- Subjects
- Animals, Cell Differentiation, Cell Proliferation, Cells, Cultured, Dendrimers chemistry, Humans, Mice, Mice, Inbred C57BL, Muscular Diseases therapy, Myoblasts cytology, Tibia, Ferric Compounds chemistry, Muscle, Skeletal pathology, Myoblasts transplantation, Nanoparticles chemistry, Transplantation, Heterologous
- Abstract
Myoblast transplantation is being considered as a potential strategy to improve muscle function in myopathies; hence, it is important to identify the transplanted cells and to have available efficient reagents to track these cells. We first validated a human to mouse xenotransplantation model warranting the complete and rapid rejection of the cells. We then used this model to assess the appropriateness of a nanoparticle reagent to track the transplanted cells. Human myoblasts were loaded with ferrite nanoparticles and injected into the tibialis muscle of immunocompetent mice. Upon collection and histological analysis of muscle sections at different time points, we observed the total disappearance of the human cells within 6 days while ferrite particles remained detectable and colocalized with mouse infiltrating and neighboring cells at the injection site. These results suggest that the use of exogenous markers such as ferrite nanoparticles may lead to false-positive results and misinterpretation of cell fate.
- Published
- 2008
- Full Text
- View/download PDF
31. Ex vivo generation of mature and functional human smooth muscle cells differentiated from skeletal myoblasts.
- Author
-
Le Ricousse-Roussanne S, Larghero J, Zini JM, Barateau V, Foubert P, Uzan G, Liu X, Lacassagne MN, Ternaux B, Robert I, Benbunan M, Vilquin JT, Vauchez K, Tobelem G, and Marolleau JP
- Subjects
- Animals, Biomarkers analysis, Cells, Cultured, Drug Combinations, Epithelial Cells physiology, Female, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Muscle Fibers, Skeletal physiology, Myoblasts, Skeletal drug effects, Myoblasts, Skeletal metabolism, Myoblasts, Skeletal transplantation, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle transplantation, Neovascularization, Physiologic, Transplantation, Heterologous, Vascular Endothelial Growth Factor A pharmacology, Cell Differentiation, Collagen pharmacology, Laminin pharmacology, Myoblasts, Skeletal physiology, Myocytes, Smooth Muscle physiology, Proteoglycans pharmacology, Tissue Culture Techniques
- Abstract
We described the ex vivo production of mature and functional human smooth muscle cells (SMCs) derived from skeletal myoblasts. Initially, myoblasts expressed all myogenic cell-related markers such as Myf5, MyoD and Myogenin and differentiate into myotubes. After culture in a medium containing vascular endothelial growth factor (VEGF), these cells were shown to have adopted a differentiated SMC identity as demonstrated by alphaSMA, SM22alpha, calponin and smooth muscle-myosin heavy chain expression. Moreover, the cells cultured in the presence of VEGF did not express MyoD anymore and were unable to fuse in multinucleated myotubes. We demonstrated that myoblasts-derived SMCs (MDSMCs) interacted with endothelial cells to form, in vitro, a capillary-like network in three-dimensional collagen culture and, in vivo, a functional vascular structure in a Matrigel implant in nonobese diabetic-severe combined immunodeficient mice. Based on the easily available tissue source and their differentiation into functional SMCs, these data argue that skeletal myoblasts might represent an important tool for SMCs-based cell therapy.
- Published
- 2007
- Full Text
- View/download PDF
32. Mesenchymal stem cells in bone and cartilage repair: current status.
- Author
-
Vilquin JT and Rosset P
- Subjects
- Bone Substitutes, Cell Differentiation, Cell Division, Humans, Bone and Bones physiology, Cartilage physiology, Mesenchymal Stem Cells cytology, Regeneration
- Abstract
The progression of rheumatoid pathologies, degenerative diseases, traumatologies, and their cortege of increasing medical, social and economical needs, has mandated the development of tissue repair and engineering technologies in orthopedic medicine. Mesenchymal stem cells (MSCs) are multipotent cells that can be extracted from large and relatively easily accessible compartments of the body, especially the bone marrow, and such cells are able to differentiate into adipogenic, chondrogenic and osteogenic precursors. The concept of using MSCs to repair tissues has progressively evolved, and the goal of cell-mediated therapy is to prolong the natural physiological abilities of healing, or substitute them, when these are lacking, failing or progressing too slowly. In recent years, the first clinical trials on the utility of MSCs, with or without scaffolds and/or growth factors, have been initiated. In this review, the authors focus on findings from preclinical research, clinical trials and case reports involving bone and cartilage repairs. New perspectives are considered regarding uses of cell types, cell delivery approaches and growth factors. They also consider the stringent conditions, constraints and considerations necessary to take cell-mediated therapy from bench to bedside.
- Published
- 2006
- Full Text
- View/download PDF
33. Autologous myoblast transplantation for chronic ischemic mitral regurgitation.
- Author
-
Messas E, Bel A, Morichetti MC, Carrion C, Handschumacher MD, Peyrard S, Vilquin JT, Desnos M, Bruneval P, Carpentier A, Menasché P, Levine RA, and Hagège AA
- Subjects
- Animals, Chronic Disease, Cicatrix, Disease Models, Animal, Echocardiography, Injections, Intralesional, Mitral Valve Insufficiency etiology, Myocardial Contraction, Sheep, Stroke Volume, Transplantation, Autologous, Ventricular Remodeling physiology, Cell Transplantation methods, Mitral Valve Insufficiency physiopathology, Mitral Valve Insufficiency therapy, Myoblasts, Skeletal transplantation, Myocardial Infarction complications
- Abstract
Objectives: This study was designed to assess whether post-myocardial infarction (MI) in-scar transplantation of skeletal myoblasts (SM) could reduce chronic ischemic mitral regurgitation (MR) by decreasing left ventricular (LV) remodeling., Background: Extensive work has confirmed the relationship between ischemic MR and post-myocardial infarction (MI) remodeling of the LV., Methods: An infero-posterior MI was created in 13 sheep, thereby resulting in increasing MR. Two months post-MI, the animals were randomized and in-scar injected with expanded autologous SM (n = 6, mean: 251 x 10(6) cells) or culture medium only (n = 7). Three-dimensional echocardiography was performed at baseline, before transplantation, and for two months thereafter (sacrifice), with measurements of LV end-diastolic and end-systolic volumes (ESV), ejection fraction (EF), MR stroke volume, and leaflet tethering distance; wall motion score index (WMSi) was assessed by two-dimensional echo., Results: Measurements were similar between groups at baseline and before transplantation. At sacrifice, transplantation was found to have reduced MR progression (regurgitant volume change: -1.83 +/- 0.32 ml vs. 5.9 +/- 0.7 ml in control group, p < 0.0001) and tethering distance (-0.41 +/- 0.09 cm vs. 0.44 +/- 0.12 cm in control group, p < 0.001), with significant improvement of EF (2.01 +/- 0.94% vs. -4.86 +/- 2.23%, p = 0.02), WMSi (-0.25 +/- 0.11 vs. 0.13 +/- 0.03 in controls, p < 0.01) and a trend to a lesser increase in ESV (23.3 +/- 3.5 ml vs. 35.4 +/- 4.2 ml in control group, p = 0.055)., Conclusions: Autologous skeletal myoblast transplantation attenuates mild-to-moderate chronic ischemic MR, which otherwise is progressive, by decreasing tethering distance and improving EF and wall motion score, thereby enhancing valve coaptation. These data shed additional light on the mechanism by which skeletal myoblast transplantation may be cardioprotective.
- Published
- 2006
- Full Text
- View/download PDF
34. Normal growth and regenerating ability of myoblasts from unaffected muscles of facioscapulohumeral muscular dystrophy patients.
- Author
-
Vilquin JT, Marolleau JP, Sacconi S, Garcin I, Lacassagne MN, Robert I, Ternaux B, Bouazza B, Larghero J, and Desnuelle C
- Subjects
- Animals, Biomarkers analysis, Biopsy, CD56 Antigen analysis, Case-Control Studies, Cell Differentiation, Cell Proliferation, Cells, Cultured, Desmin analysis, Electromyography, Feasibility Studies, Humans, Mice, Mice, SCID, Microscopy, Phase-Contrast, Muscular Dystrophy, Facioscapulohumeral therapy, Myoblasts immunology, Myoblasts transplantation, Patient Selection, Regeneration, Telomere ultrastructure, Transplantation, Autologous, Muscular Dystrophy, Facioscapulohumeral pathology, Myoblasts pathology
- Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disease characterized by a typical regional distribution, featuring composed patterns of clinically affected and unaffected muscles. No treatment is available for this condition, in which the pathophysiological mechanism is still unknown. Autologous transfer of myoblasts from unaffected to affected territories could be considered as a potential strategy to delay or stop muscle degeneration. To evaluate the feasibility of this concept, we explored and compared the growth and differentiation characteristics of myoblasts prepared from phenotypically unaffected muscles of five FSHD patients and 10 control donors. According to a clinically approved procedure, 10(9) cells of a high degree of purity were obtained within 16-23 days. More than 80% of these cells were myoblasts, as demonstrated by labeling of the muscle markers CD56 and desmin. FSHD myoblasts presented a doubling time equivalent to that of control cells; they kept high proliferation ability and did not show early telomere shortening. In vitro, these cells were able to differentiate and to express muscle-specific antigens. In vivo, they participated to muscle structures when injected into immunodeficient mice. These data suggest that myoblasts expanded from unaffected FSHD muscles may be suitable tools in view of autologous cell transplantation clinical trials.
- Published
- 2005
- Full Text
- View/download PDF
35. Myoblast transplantation: clinical trials and perspectives. Mini-review.
- Author
-
Vilquin JT
- Subjects
- Animals, Cell Movement physiology, Cell Survival physiology, Heart Failure surgery, Humans, Muscular Dystrophy, Facioscapulohumeral surgery, Regeneration, Tissue Engineering, Transplantation, Autologous, Urinary Incontinence, Stress surgery, Muscular Dystrophy, Duchenne surgery, Myoblasts transplantation
- Abstract
Myoblast transplantation (MT) is proposed to challenge the repair of striated muscles in various pathological conditions. Indeed, myoblasts are of easy procurement and expansion from small muscle biopsies. They are naturally able to participate to skeletal muscle regeneration through cell-cell fusion, leading to the formation of new or hybrid muscle fibers. In an autologous context, MT relies on the localized increased muscle generative capacity brought by large number of myoblasts : cells may be transferred from a clinically unaffected area to the disabled territory. In an heterologous context, MT further allows to transfer a non-pathologic, donor genome into the disabled recipient muscle fibers. Autologous approaches are then developed as localized repairs are mandated, while heterologous approaches are used in the context of generalized, advanced degenerative diseases. Whatever the immunological situation, however, MT is hampered by several issues, mainly the poor survival and the limited migratory ability of the myoblasts. Some solutions are emerging, which recently allowed the set-up of new clinical trials in dedicated indications.
- Published
- 2005
36. Can cold or heat shock improve skeletal myoblast engraftment in infarcted myocardium?
- Author
-
Maurel A, Azarnoush K, Sabbah L, Vignier N, Le Lorc'h M, Mandet C, Bissery A, Garcin I, Carrion C, Fiszman M, Bruneval P, Hagege A, Carpentier A, Vilquin JT, and Menasché P
- Subjects
- Adenoviridae genetics, Animals, Cell Differentiation genetics, Cell Division genetics, Cold Temperature, Female, Genetic Markers, HSP70 Heat-Shock Proteins metabolism, Hot Temperature, Male, Muscle, Skeletal cytology, Myoblasts, Skeletal cytology, Phenotype, Rats, Rats, Inbred Lew, Y Chromosome, beta-Galactosidase genetics, Cryopreservation, Graft Survival, Heat-Shock Response, Myoblasts, Skeletal transplantation, Myocardial Infarction therapy
- Abstract
Objective: Cell death remains a major limitation of skeletal myoblast (SM) transplantation but the patterns of cell survival and proliferation in heart and their potential modulation by thermic stresses like heat shock (HS) and cryopreservation (Cryo) are still incompletely characterized., Methods: To track SMs in situ, we developed a dual-marker system based on the semiconservative expression of the foreign soluble protein, beta-Galactosidase (beta-Gal) and the constitutive expression of the Y chromosome in a myocardial infarction model. Control medium or Lewis male rat SMs (fresh or subjected to Cryo or HS) were injected in Lewis female rats., Results: There was a massive cell loss early after transplantation in the fresh group, which was only partially compensated for by a subsequent proliferation. Conversely, both Cryo and HS significantly improved early cell survival but blunted subsequent proliferation so that, at 15 days posttransplantation, the total number of engrafted donor-derived Y-positive cells did not differ significantly between the three groups. Most of them expressed a skeletal muscle phenotype., Conclusions: These data confirm the high death rate of in-scar transplanted myoblasts, demonstrate the ability of those that survive to proliferate and differentiate along the myogenic pathway but do not support the efficacy of either Cryo or HS for increasing the ultimate magnitude of myoblast engraftment.
- Published
- 2005
- Full Text
- View/download PDF
37. Skeletal myoblast transplantation through a catheter-based coronary sinus approach: an effective means of improving function of infarcted myocardium.
- Author
-
Brasselet C, Morichetti MC, Messas E, Carrion C, Bissery A, Bruneval P, Vilquin JT, Lafont A, Hagège AA, Menasché P, and Desnos M
- Subjects
- Animals, Cardiac Catheterization methods, Myocardial Infarction physiopathology, Recovery of Function, Sheep, Ventricular Function, Left physiology, Myoblasts, Skeletal transplantation, Myocardial Infarction therapy
- Abstract
Aims: This study was designed to assess the functional effects of a transvenous coronary sinus technique of skeletal myoblast delivery in infarcted myocardium., Methods and Results: An anterior myocardial infarction was created percutaneously in 14 sheep. Simultaneously, a muscle biopsy was harvested and expanded. Two weeks later, sheep were instrumented percutaneously with a dedicated catheter incorporating an extendable needle for puncture of the venous wall and, under endovascular ultrasound guidance, a microcatheter was advanced through the needle into the target scar for cell delivery. Following the baseline echocardiographic assessment of left ventricular (LV) function, sheep were randomly allocated to receive four-staged in-scar injections of either autologous cells (n=7) or culture medium (n=7). Two months later, LV function was reassessed blindly and hearts were explanted for subsequent histological and immunohistochemical analysis. There were no acute procedural complications. Baseline LV ejection fraction (EF) was significantly lower in transplanted sheep than in controls [38% (35-48) vs. 51% (38-55), respectively, P=0.03; median (range)]. Two months later, LVEF was significantly higher in the transplanted group than in controls [50% (47-56) vs. 39% (36-47), respectively, P=0.002]. Clusters of myoblasts were identified by histology and immunohistochemistry in three of the seven transplanted sheep., Conclusion: These data suggest the functional efficacy of the transvenous coronary sinus technique as a less invasive means of cell delivery to infarcted myocardium.
- Published
- 2005
- Full Text
- View/download PDF
38. [The possible place of autologus cell therapy in facioscapulohumeral muscular dystrophy].
- Author
-
Desnuelle C, Sacconi S, Marolleau JP, Larghero J, and Vilquin JT
- Subjects
- Case-Control Studies, Cell Differentiation physiology, Cell Proliferation, Cells, Cultured, Humans, Male, Middle Aged, Myoblasts, Skeletal physiology, Transplantation, Autologous, Muscular Dystrophy, Facioscapulohumeral pathology, Muscular Dystrophy, Facioscapulohumeral therapy, Myoblasts, Skeletal pathology, Myoblasts, Skeletal transplantation
- Abstract
Facioscapulohumeral dystrophy (FSHD), one of the most common forms of muscular dystrophy, derives its name from the patients' selective, often asymmetric clinical distribution of muscle weakness. Interestingly, affected and non affected areas can coexist in the same patient for many years. The molecular hallmark is total deletion of the subtelomeric D4Z4 repeat on chromosome 4q. There is no specific treatment. Gene therapy is unlikely to be feasible, as no alterations have been found in the genes located in this subtelomeric region. Muscular dystrophies are characterized by the coexistence of genetically induced muscle degeneration and compensatory muscle regeneration by myoblast proliferation from satellite cells; muscle weakness and atrophy appears when this mechanism is overwhelmed. Cell therapy with autologous myoblasts would, in theory, be a simple way of boosting the regenerative process and of preventing or delaying muscle degeneration. This approach might also avoid the use of toxic immunotherapies. By using a recent very-high-yield cell culture method, we analyzed the proliferation and differentiation of myoblasts obtained from FSHD patients, both ex vivo and in vivo (by intramuscular injection to immunodeficient mice). Myoblasts were obtained by muscle biopsy from five FSHD patients harboring the D4Z4 deletion. We selected the vastus lateralis muscle, which exhibited no clinical, radiological or pathological signs of dystrophy. The growth characteristics of these cells were compared with those of cells from normal control muscles, based on the culture yield, phenotypic characterization with anti-CD56 and anti-desmin antibodies, and the capacity for differentiation (myotube production in vitro and human dystrophin expression one month after injection to Rag2 immunodeficient mice). Patients' cells recovered from 1 g of muscle biopsy specimen resembled control cells in terms of their growth kinetics, culture yield, and capacity to differentiate and produce mature muscle cells. These results indicate that myoblasts taken from unaffected muscle of patients with FSHD warrant testing in a human cell therapy trial.
- Published
- 2005
39. Does the functional efficacy of skeletal myoblast transplantation extend to nonischemic cardiomyopathy?
- Author
-
Pouly J, Hagège AA, Vilquin JT, Bissery A, Rouche A, Bruneval P, Duboc D, Desnos M, Fiszman M, Fromes Y, and Menasché P
- Subjects
- Animals, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated pathology, Cardiomyopathy, Dilated physiopathology, Cells, Cultured transplantation, Cricetinae, Female, Fibrosis, Heart Ventricles physiopathology, Injections, Male, Mesocricetus, Myocardium pathology, Random Allocation, Sarcoglycans deficiency, Single-Blind Method, Transplantation, Heterotopic, Ventricular Function, Left, Cardiomyopathy, Dilated therapy, Muscle, Skeletal cytology, Myoblasts transplantation
- Abstract
Background: The benefits of skeletal myoblast (SM) transplantation on infarcted myocardium have been investigated extensively; however, little is known about its effects in nonischemic cardiomyopathy models. To address this issue, we tested SM transplantation in CHF147 Syrian hamsters, a strain characterized by a delta-sarcoglycan deficiency that phenotypically features the human setting of primary dilated cardiomyopathy., Methods and Results: Cell culture techniques were used to prepare approximately 5x10(6) muscle cells from autologous tibialis anterior muscle, of which 50% were SMs (desmin staining). The cells were injected in 6 sites across the left ventricular wall (n=14). Control animals (n=12) received equivalent volumes of culture medium. Left ventricular systolic function was assessed in a blinded fashion from 2D echocardiographic left ventricular fractional area change, before transplantation, and 4 weeks later. Explanted hearts were processed for the detection of myotubes and quantification of fibrosis. Baseline functional data did not differ between the 2 groups. Four weeks after transplantation, 6 of the 10 surviving grafted hamsters were improved compared with 0 of the 8 survivors of the control group. This translated into a 6% decrease in fractional area change in controls compared with a 24% increase in cell-transplanted hamsters (P=0.001). Engrafted myotubes were consistently detected in all SM transplanted hearts by immunohistochemistry, whereas fibrosis was not worsened by cell injections., Conclusions: These data suggest that the functional benefits of SM transplantation might extend to nonischemic cardiomyopathy.
- Published
- 2004
- Full Text
- View/download PDF
40. Comparison of human skeletal myoblasts and bone marrow-derived CD133+ progenitors for the repair of infarcted myocardium.
- Author
-
Agbulut O, Vandervelde S, Al Attar N, Larghero J, Ghostine S, Léobon B, Robidel E, Borsani P, Le Lorc'h M, Bissery A, Chomienne C, Bruneval P, Marolleau JP, Vilquin JT, Hagège A, Samuel JL, and Menasché P
- Subjects
- AC133 Antigen, Animals, Antigens, CD, Antigens, Surface analysis, Colony-Forming Units Assay, Fluorescent Antibody Technique, Graft Survival, Heart physiology, Humans, In Situ Hybridization, Fluorescence, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardium cytology, Polymerase Chain Reaction, Rats, Rats, Nude, Stroke Volume, Ventricular Function, Left, Glycoproteins analysis, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells immunology, Myoblasts, Skeletal transplantation, Myocardial Infarction therapy, Peptides analysis, Regeneration
- Abstract
Objectives: The present study was designed as a face-to-face functional comparison of human skeletal myoblasts (SMs) and CD133(+) bone marrow-derived hematopoietic progenitors in an animal model of semichronic myocardial infarction., Background: Compared with SMs, bone marrow-derived cells have the advantage of plasticity and might more effectively regenerate ischemic cardiac tissue. However, few data exist on the comparative efficacy of these two cell types in semichronic infarcts., Methods: A myocardial infarction was created by coronary ligation in 32 nude rats. Ten days later, rats received in-scar injections of human SMs, CD133(+) progenitors, or culture medium. Left ventricular function was assessed before and one month after transplantation by echocardiography and pressure-volume loops. Immunofluorescence, polymerase chain reaction, and in situ hybridization were used to detect cells grafted in the hearts., Results: One month after transplantation, left ventricular ejection fraction decreased by 8 +/- 4% in controls, whereas it increased by 7 +/- 3% in CD133(+)-grafted hearts (p = 0.0015 vs. controls) and further by 15 +/- 5% in SM-treated hearts (p = 0.008 vs. controls). Systolic indices yielded by pressure-volume loops paralleled these data. Engrafted myotubes were identified in all SM-treated hearts by immunofluorescence, whereas in CD133(+)-grafted hearts, few human cells were only detected by polymerase chain reaction., Conclusions: In the setting of postinfarction scars, the transplantation of bone marrow-derived CD133(+) progenitors improves cardiac function, but this benefit is not superior to that afforded by myogenic cells.
- Published
- 2004
- Full Text
- View/download PDF
41. [Cell transplantation in heart failure management].
- Author
-
Vilquin JT and Marolleau JP
- Subjects
- Animals, Clinical Trials as Topic, Humans, Myocytes, Cardiac transplantation, Cell Transplantation methods, Heart Failure surgery
- Abstract
Heart failure is becoming a major issue for public health in western countries and the effect of currently available therapies is limited. Therefore cell transplantation was developed as an alternative strategy to improve cardiac structure and function. This review describes the multiple cell types and clinical trials considered for use in this indication. Most studies have been developed in models of post-ischemic heart failure. The transplantation of fetal or neonatal cardiomyocytes has proven to be functionally successful, but ethical as well as immunological and technical reasons make their clinical use limited. Recent reports, however, suggested that adult autologous cardiomyocytes could be prepared from stem cells present in various tissues (bone marrow, vessels, adult heart itself, adipose tissue). Alternatively, endothelial progenitors originating from bone marrow or peripheral blood could promote the neoangiogenesis within the scar tissue. Hematopietic stem cells prepared from bone marrow or peripheral blood have been proposed but their differentiation ability seems limited. Finally, the transplantation of skeletal muscle cells (myoblasts) in the infarcted area improved myocardial function, in correlation with the development of skeletal muscle tissue in various animal models. The latter results paved the way for the development of a first phase I clinical trial of myoblast transplantation in patients with severe post-ischemic heart failure. It required the scale-up of human cell production according to good manufacturing procedures, started in june 2000 in Paris and was terminated in november 2001, and was followed by several others. The results were encouraging and prompted the onset of a blinded, multicentric phase II clinical trial for skeletal muscle cells transplantation. Meanwhile, phase I clinical trials also evaluate the safeness and efficacy of various cell types originating from the bone marrow or the peripheral blood. However, potential side effects related to the biological properties of the cells or the delivery procedures are being reported. High quality clinical trials supported by strong pre-clinical data will help to evaluate the role of cell therapy as a potential treatment for heart failure.
- Published
- 2004
- Full Text
- View/download PDF
42. Electrotransfer at MR imaging: tool for optimization of gene transfer protocols--feasibility study in mice.
- Author
-
Paturneau-Jouas M, Parzy E, Vidal G, Carlier PG, Wary C, Vilquin JT, de Kerviler E, Schwartz K, and Leroy-Willig A
- Subjects
- Animals, Contrast Media, Electroporation, Feasibility Studies, Gadolinium DTPA, Mice, Mice, Inbred C57BL, Muscle, Skeletal cytology, Plasmids, Transfection methods, Gene Transfer Techniques, Magnetic Resonance Imaging
- Abstract
Purpose: To test, by using an electrotransfer protocol for the transfection of skeletal muscle with naked plasmid complementary DNA, whether in vivo magnetic resonance (MR) imaging can help delineate either the spatial extent of the electric field when contrast agent is injected intraperitoneally or the transfection area when contrast agent is injected locally., Materials and Methods: Three groups of five mice each were examined at 4 T. Gadopentetate dimeglumine was injected intraperitoneally before electroporation in group 1 and after electroporation in group 2. In group 3, gadopentetate dimeglumine was coinjected in situ with plasmid pCMV-beta Gal in the gastrocnemius muscle before electroporation. MR imaging and muscle preparation for histologic examination were performed 3 days later. On T1-weighted images, increase of muscle signal intensity was determined in regions of interest (ROIs) of treated legs and compared with contralateral ROIs. Comparison of signal intensity increase between groups 1 and 2 was performed with Kruskal-Wallis test., Results: In groups 1 and 3, T1-weighted images of treated muscle showed zones of strongly increased signal intensity. In corresponding ROIs of groups 1, 2, and 3, the mean T1-weighted signal intensity increase at day 3 was 1.64 +/- 0.20 (SD), 1.16 +/- 0.06, and 1.58 +/- 0.17, respectively. The difference between groups 1 and 2 (ie, gadopentetate dimeglumine injected before and after electrotransfer) was significant (P <.001) both without and with correction for T2 variation (1.47 +/- 0.19 and 1.04 +/- 0.09, respectively). In group 3, after in situ coinjection of gadopentetate dimeglumine and plasmid, the area of increased signal intensity revealed at ex vivo MR imaging of the muscle showed a reasonable concordance with the transfected area revealed with beta-galactosidase on histologic sections., Conclusion: In vivo and ex vivo results indicate that atraumatic visualization of the permeabilized and transfected area is possible.
- Published
- 2003
- Full Text
- View/download PDF
43. Myoblasts transplanted into rat infarcted myocardium are functionally isolated from their host.
- Author
-
Leobon B, Garcin I, Menasche P, Vilquin JT, Audinat E, and Charpak S
- Subjects
- Animals, Animals, Newborn, Cell Differentiation, Cell Transplantation, Cells, Cultured, Electrophysiology, Green Fluorescent Proteins, Luminescent Proteins metabolism, Microscopy, Fluorescence, Microscopy, Video, Muscle, Skeletal cytology, Muscles metabolism, Muscles physiology, Rats, Rats, Wistar, Transplantation, Homologous, Myoblasts cytology, Myoblasts physiology, Myoblasts, Skeletal cytology, Myocardial Infarction metabolism, Myocardium pathology
- Abstract
Survival and differentiation of myogenic cells grafted into infarcted myocardium have raised the hope that cell transplantation becomes a new therapy for cardiovascular diseases. The approach was further supported by transplantation of skeletal myoblasts, which was shown to improve cardiac performance in several animal species. Despite the success of myoblast transplantation and its recent trial in human, the mechanism responsible for the functional improvement remains unclear. Here, we used intracellular recordings coupled to video and fluorescence microscopy to establish whether myoblasts, genetically labeled with enhanced GFP and transplanted into rat infarcted myocardium, retain excitable and contractile properties, and participate actively to cardiac function. Our results indicate that grafted myoblasts differentiate into peculiar hyperexcitable myotubes with a contractile activity fully independent of neighboring cardiomyocytes. We conclude that mechanisms other than electromechanical coupling between grafted and host cells are involved in the improvement of cardiac function.
- Published
- 2003
- Full Text
- View/download PDF
44. Building the translational highway: toward new partnerships between academia and the private sector.
- Author
-
Schwartz K and Vilquin JT
- Subjects
- Academies and Institutes, Private Sector, Technology Transfer
- Published
- 2003
- Full Text
- View/download PDF
45. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction.
- Author
-
Menasché P, Hagège AA, Vilquin JT, Desnos M, Abergel E, Pouzet B, Bel A, Sarateanu S, Scorsin M, Schwartz K, Bruneval P, Benbunan M, Marolleau JP, and Duboc D
- Subjects
- Adult, Aged, Cardiac Surgical Procedures adverse effects, Cell Count, Cells, Cultured, Coronary Artery Bypass methods, Defibrillators, Implantable, Echocardiography, Doppler, Endpoint Determination, Feasibility Studies, Heart Failure etiology, Humans, Male, Middle Aged, Myocardial Ischemia complications, Myocardial Ischemia surgery, Safety, Stroke Volume physiology, Tachycardia, Ventricular etiology, Tachycardia, Ventricular therapy, Transplantation, Autologous, Treatment Outcome, Ventricular Dysfunction, Left etiology, Cardiac Surgical Procedures methods, Heart Failure surgery, Myoblasts, Skeletal transplantation, Postoperative Complications, Ventricular Dysfunction, Left surgery
- Abstract
Objectives: This phase I trial was designed to assess the feasibility and safety of autologous skeletal myoblast transplantation in patients with severe ischemic cardiomyopathy., Background: Experimentally, myoblast grafting into postinfarction myocardial scars improves left ventricular function., Methods: Ten patients were included on the basis of the following criteria: 1) severe left ventricular dysfunction (ejection fraction < or = 35%); 2) the presence of a postinfarction akinetic and nonviable scar, as assessed by dobutamine echocardiography and 18-fluorodeoxyglucose positron emission tomography; and 3) an indication of coronary bypass in remote areas. Skeletal myoblasts were grown from a biopsy taken at the thigh., Results: An average of 871 x 10(6) cells (86% of myoblasts) were obtained after a mean period of 16 days and implanted uneventfully across the scar at the time of bypass. Except for one patient whose early death was unrelated to the cell transplantation, all patients had an uncomplicated postoperative course. Four patients showed delayed episodes of sustained ventricular tachycardia and were implanted with an internal defibrillator. At an average follow-up of 10.9 months, the mean New York Heart Association functional class improved from 2.7 +/- 0.2 preoperatively to 1.6 +/- 0.1 postoperatively (p < 0.0001), and the ejection fraction increased from 24 +/- 1% to 32 +/- 1% (p < 0.02). A blinded echocardiographic analysis showed that 63% of the cell-implanted scars (14 of 22) demonstrated improved systolic thickening. One noncardiac death occurred 17.5 months after transplantation., Conclusions: These preliminary data suggest the feasibility and safety of autologous skeletal myoblast transplantation in severe ischemic cardiomyopathy, with the caveat of an arrhythmogenic potential. New-onset contraction of akinetic and nonviable segments suggests a functional efficacy that requires confirmation by randomized studies.
- Published
- 2003
- Full Text
- View/download PDF
46. Long-term (1 year) functional and histological results of autologous skeletal muscle cells transplantation in rat.
- Author
-
Al Attar N, Carrion C, Ghostine S, Garcin I, Vilquin JT, Hagège AA, and Menasché P
- Subjects
- Animals, Echocardiography, Male, Models, Animal, Myocardial Infarction physiopathology, Myocardium metabolism, Myosins metabolism, Protein Isoforms metabolism, Rats, Rats, Wistar, Stroke Volume, Time Factors, Muscle Fibers, Skeletal transplantation, Myocardial Infarction pathology, Myocardial Infarction surgery, Myocardium pathology, Ventricular Function, Left
- Abstract
Background: Several studies have demonstrated the short-term benefits of autologous skeletal muscle cell transplantation on postinfarction left ventricular function. The present experiments were designed to assess the long-term effects of the procedure., Methods and Results: Thirteen Wistar rats that had undergone skeletal muscle cell transplantation (n=6) or injection of control culture medium (n=7) in isoforms areas after myocardial infarction created by coronary artery ligation and survived for 1 year were functionally assessed by combining echocardiography and pressure-volume loops. At 1 year after transplantation, both contractile and relaxation indices were significantly improved in the skeletal muscle cell-grafted group compared with controls. One-year echocardiographic measurements of ejection fraction were similar to those recorded 2 months after the procedure. The stability of the functional outcome contrasted with a decrease in the number of histologically detectable skeletal myotubes over time. However, the proportion of the slow and composite (fast and slow) myosin isoforms expressed by skeletal muscle fibers still present after 1 year was greater than that found in animals sacrificed after 2 months., Conclusion: The functional benefits of autologous skeletal muscle cell transplantation are sustained over time and are associated with either selection, preservation or an increased expression of slow myosin heavy chain isoforms. The discrepancy between maintenance of this improvement and the decay in the engrafted myotubes suggests protective mechanisms operative from the early post-transplantation stage and possibly involving modulation of extracellular matrix remodelling or paracrinally induced maturation of putative cardiac resident stem cells.
- Published
- 2003
- Full Text
- View/download PDF
47. Spontaneous muscular dystrophy caused by a retrotransposal insertion in the mouse laminin alpha2 chain gene.
- Author
-
Besse S, Allamand V, Vilquin JT, Li Z, Poirier C, Vignier N, Hori H, Guénet JL, and Guicheney P
- Subjects
- Animals, Base Sequence, Blotting, Western, DNA Transposable Elements genetics, DNA, Complementary analysis, Disease Models, Animal, Fluorescent Antibody Technique methods, Homozygote, Humans, Laminin deficiency, Laminin metabolism, Mice, Mice, Mutant Strains, Molecular Sequence Data, Muscle, Skeletal pathology, Muscular Dystrophies congenital, Muscular Dystrophies metabolism, Muscular Dystrophy, Animal metabolism, Mutation, Phenotype, Retroelements physiology, Reverse Transcriptase Polymerase Chain Reaction methods, Sequence Analysis, DNA methods, Laminin genetics, Muscle, Skeletal metabolism, Muscular Dystrophies genetics, Muscular Dystrophy, Animal genetics, Retroelements genetics
- Abstract
We identified a novel spontaneous mouse model of human congenital muscular dystrophy with laminin alpha2 chain deficiency, named dy(Pas)/dy(Pas). Homozygous animals rapidly developed a progressive muscular dystrophy leading to premature death. Immunohistological and biochemical analyses demonstrated the absence of laminin alpha2 chain expression in skeletal muscle. Analysis of the laminin alpha2 chain cDNA showed the insertion of the long terminal repeat of an intracisternal A-particle gene. In addition, a 6.1 kb insertion composed of retrotransposon elements was identified in the Lama2 sequence. The dy(Pas)/dy(Pas) mouse is thus the first spontaneous mutant with a complete laminin alpha2 chain deficiency in which the mutation has been identified.
- Published
- 2003
- Full Text
- View/download PDF
48. Laminin alpha2 deficiency and muscular dystrophy; genotype-phenotype correlation in mutant mice.
- Author
-
Guo LT, Zhang XU, Kuang W, Xu H, Liu LA, Vilquin JT, Miyagoe-Suzuki Y, Takeda S, Ruegg MA, Wewer UM, and Engvall E
- Subjects
- Animals, DNA Mutational Analysis, Desmin genetics, Disease Models, Animal, Fluorescent Antibody Technique methods, Gene Expression, Humans, Immunoblotting methods, Laminin chemistry, Laminin deficiency, Laminin genetics, Mice, Mice, Mutant Strains genetics, Mice, Mutant Strains metabolism, Mice, Transgenic, Muscle, Skeletal metabolism, Muscular Dystrophies genetics, Muscular Dystrophies pathology, Peripheral Nerves metabolism, Promoter Regions, Genetic, Protein Structure, Tertiary physiology, Protein Subunits immunology, Protein Subunits metabolism, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins immunology, Genotype, Laminin metabolism, Muscular Dystrophies metabolism, Phenotype
- Abstract
Deficiency of laminin alpha2 is the cause of one of the most severe muscular dystrophies in humans and other species. It is not yet clear how particular mutations in the laminin alpha2 chain gene affect protein expression, and how abnormal levels or structure of the protein affect disease. Animal models may be valuable for such genotype-phenotype analysis and for determining mechanism of disease as well as function of laminin. Here, we have analyzed protein expression in three lines of mice with mutations in the laminin alpha2 chain gene and in two lines of transgenic mice overexpressing the human laminin alpha2 chain gene in skeletal muscle. The dy(3K)/dy(3K) experimental mutant mice are completely deficient in laminin alpha2; the dy/dy spontaneous mutant mice have small amounts of apparently normal laminin; and the dy(W)/dy(W) mice express even smaller amounts of a truncated laminin alpha2, lacking domain VI. Interestingly, all mutants lack laminin alpha2 in peripheral nerve. We have demonstrated previously, that overexpression of the human laminin alpha2 in skeletal muscle in dy(2J)/dy(2J) and dy(W)/dy(W) mice under the control of a striated muscle-specific creatine kinase promoter substantially prevented the muscular dystrophy in these mice. However, dy(W)/dy(W) mice, expressing the human laminin alpha2 under the control of the striated muscle-specific portion of the desmin promoter, still developed muscular dystrophy. This failure to rescue is apparently because of insufficient production of laminin alpha2. This study provides additional evidence that the amount of laminin alpha2 is most critical for the prevention of muscular dystrophy. These data may thus be of significance for attempts to treat congenital muscular dystrophy in human patients.
- Published
- 2003
- Full Text
- View/download PDF
49. Viability and differentiation of autologous skeletal myoblast grafts in ischaemic cardiomyopathy.
- Author
-
Hagège AA, Carrion C, Menasché P, Vilquin JT, Duboc D, Marolleau JP, Desnos M, and Bruneval P
- Subjects
- Aged, Coronary Artery Bypass, Humans, Male, Microscopy, Confocal, Myoblasts, Skeletal pathology, Myocardial Infarction pathology, Myocardium pathology, Ventricular Dysfunction, Left pathology, Cell Differentiation physiology, Cell Survival physiology, Myoblasts, Skeletal transplantation, Myocardial Infarction surgery, Ventricular Dysfunction, Left surgery
- Abstract
Autologous skeletal myoblast transplantation might improve postinfarction ventricular function, but graft viability and differentiation (ie, proof of concept) has not been shown. A 72-year-old man had autologous cultured myoblasts from his vastus lateralis injected to an area of transmural inferior myocardial infarction in non-reperfused scar tissue. He showed improvement in symptoms and left-ventricular ejection fraction. When he died 17.5 months after the procedure, the grafted post-infarction scar showed well developed skeletal myotubes with a preserved contractile apparatus. 65% of myotubes expressed the slow myosin isoform and 33% coexpressed the slow and fast isoforms (vs 44% and 0.6%, respectively, in skeletal muscle). Myoblast grafts can survive and show a switch to slow-twitch fibres, which might allow sustained improvement in cardiac function.
- Published
- 2003
- Full Text
- View/download PDF
50. Cell transplantation for post-ischemic heart failure.
- Author
-
Vilquin JT, Marolleau JP, Hagege A, Menasche P, Fiszman M, and Schwartz K
- Subjects
- Bone Marrow Transplantation, Clinical Trials as Topic, Humans, Muscle, Skeletal cytology, Myocardium cytology, Peripheral Blood Stem Cell Transplantation, Cell Transplantation, Heart Failure therapy, Myocardial Ischemia complications, Neovascularization, Physiologic
- Abstract
Post-ischemic heart failure is becoming a major issue for public health in occidental countries and therapeutical options are limited. Therefore cell transplantation was developed as an alternative strategy to improve cardiac structure and function. This review describes the multiple cell types and clinical trials considered for use in this indication. The transplantation of fetal or neonatal cardiomyocytes has proven to be functionally successful, but ethical as well as technical reasons make their clinical use limited. Recent reports, however, suggested that adult autologous cardiomyocytes could be prepared from stem cells present in various mesenchymal tissues. Alternatively, endothelial progenitors originating from bone marrow or peripheral blood could promote the neoangiogenesis within the scar tissue. Finally, the transplantation of skeletal muscle cells (SMC) in the infarcted area improved myocardial function, in correlation with the development of skeletal muscle tissue in various animal models. The latter results paved the way for the development of a first phase I clinical trial of SMC transplantation in patients with severe ischemic heart failure. It required the scale-up of human cell production according to Good Manufacturing Procedures, it started in June 2000 in Paris and was terminated in November 2001, and it was followed by several others. The results were encouraging and prompted the onset of a blinded, multicentric phase II clinical trial for SMC transplantation. Meanwhile, clinical trials also evaluate the safety and efficacy of various cells types originating from the bone marrow.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.