Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
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
2.
Am J Pathol ; 179(5): 2501-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21924229

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic progressive muscle disease resulting from the lack of dystrophin and without effective treatment. Adult stem cell populations have given new impetus to cell-based therapy of neuromuscular diseases. One of them, muscle-derived stem cells, isolated based on delayed adhesion properties, contributes to injured muscle repair. However, these data were collected in dystrophic mice that exhibit a relatively mild tissue phenotype and clinical features of DMD patients. Here, we characterized canine delayed adherent stem cells and investigated the efficacy of their systemic delivery in the clinically relevant DMD animal model to assess potential therapeutic application in humans. Delayed adherent stem cells, named MuStem cells (muscle stem cells), were isolated from healthy dog muscle using a preplating technique. In vitro, MuStem cells displayed a large expansion capacity, an ability to proliferate in suspension, and a multilineage differentiation potential. Phenotypically, they corresponded to early myogenic progenitors and uncommitted cells. When injected in immunosuppressed dystrophic dogs, they contributed to myofiber regeneration, satellite cell replenishment, and dystrophin expression. Importantly, their systemic delivery resulted in long-term dystrophin expression, muscle damage course limitation with an increased regeneration activity and an interstitial expansion restriction, and persisting stabilization of the dog's clinical status. These results demonstrate that MuStem cells could provide an attractive therapeutic avenue for DMD patients.


Subject(s)
Muscle Cells/transplantation , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dogs , Dystrophin/metabolism , Immunosuppressive Agents/pharmacology , Injections, Intramuscular , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Stem Cells/cytology , Transplantation, Homologous
3.
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
4.
Muscle Nerve ; 42(4): 584-95, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20734311

ABSTRACT

Myoblast migration requires matrix metalloproteinase (MMP) activity but the contribution of individual MMPs or tissue inhibitors of matrix metalloproteinase (TIMPs), particularly MMP-9 and TIMP-1, is lacking. Using two clones derived for differential regulation of MMP-2, MMP-9, and TIMP-1, we correlated protein expression with cell migration. MMP/TIMP regulation was determined by zymography, western blots, and quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Cell migration was compared in vitro and after grafting into nude-mdx mouse muscles. C2M9 clones produced high MMP-9 and low MMP-2, and migrated better than C2F clones, which secreted low MMP-9, but overexpressed MMP-2 and TIMP-1. Improvement of C2F invasion by MMP-9 and inhibition of C2M9 migration by MMP-9 inhibitor I confirmed the role of MMP-9 and pointed to potential inhibition by TIMP-1. Higher complementation achieved by C2M9 grafts corroborated the beneficial effect of MMP-9 overexpression. Modulation of MMP-9 expression opens perspectives for improved efficacy of cell therapy for muscular dystrophies.


Subject(s)
Cell Movement/physiology , Cell Transplantation , Matrix Metalloproteinase 9/metabolism , Muscle Development/physiology , Myoblasts/physiology , Myoblasts/transplantation , Animals , Cell Fusion , Cell Line , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Myoblasts/enzymology , Tissue Inhibitor of Metalloproteinases/metabolism , Up-Regulation
5.
Am J Physiol Cell Physiol ; 299(3): C706-13, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20592245

ABSTRACT

Muscular dystrophies are often associated with significant cardiac disease that can be the prominent feature associated with gene mutations in sarcoglycan. Cardiac cell death is a main feature of cardiomyopathy in sarcoglycan deficiency and may arise as a cardiomyocyte intrinsic process that remains unclear. Deficiency of delta-sarcoglycan (delta-SG) induces disruption of the dystrophin-associated glycoprotein complex, a known cause of membrane instability that may explain cardiomyocytes cytosolic Ca2+ increase. In this study we assessed the hypothesis that cytosolic Ca2+ increase triggers cardiomyocyte death through mitochondrial Ca2+ overload and dysfunction in the delta-SG-deficient CHF147 hamster. We showed that virtually all isolated CHF147 ventricular myocytes exhibited elevated cytosolic and mitochondrial Ca2+ levels by the use of the Fura-2 and Rhod-2 fluorescent probes. Observation of living cells with Mito-Tracker red lead to the conclusion that approximately 15% of isolated CHF147 cardiomyocytes had disorganized mitochondria. Transmission electron microscope imaging showed mitochondrial swelling associated with crest and membrane disruption. Analysis of the mitochondrial permeability transition pore (MPTP) activity using calcein revealed that mitochondria of CHF147 ventricular cells were twofold leakier than wild types, whereas reactive oxygen species production was unchanged. Bax, Bcl-2, and LC3 expression analysis by Western blot indicated that the intrinsic apoptosis and the cell death associated to autophagy pathways were not significantly activated in CHF147 hearts. Our results lead to conclusion that cardiomyocytes death in delta-SG-deficient animals is an intrinsic phenomenon, likely related to Ca2+-induced necrosis. In this process Ca2+ overload-induced MPTP activation and mitochondrial disorganization may have an important role.


Subject(s)
Calcium/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Myocytes, Cardiac/metabolism , Sarcoglycans/metabolism , Animals , Cell Death , Cricetinae , Cytosol/metabolism , Heart Ventricles/cytology , In Vitro Techniques , Male , Mesocricetus , Microtubule-Associated Proteins/biosynthesis , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Reactive Oxygen Species/metabolism , Sarcoglycans/genetics
6.
Exp Cell Res ; 316(15): 2513-26, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20430024

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.


Subject(s)
Mesenchymal Stem Cells/cytology , Muscle, Skeletal/cytology , 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
7.
Mol Ther ; 17(11): 1948-58, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19738599

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.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Muscle Cells/cytology , Muscle Development/physiology , Muscle, Skeletal/cytology , Adipogenesis/physiology , Cell Differentiation/physiology , Cells, Cultured , Flow Cytometry , Humans , Immunohistochemistry , In Vitro Techniques , Phenotype
8.
J Gene Med ; 8(8): 1048-55, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16779861

ABSTRACT

BACKGROUND: The hamster strain CHF147 presents a progressive dilated cardiomyopathy (DCM) due to a large deletion of the delta-sarcoglycan gene that leads to heart failure. This cardiomyopathy induces premature death. We have previously shown that a short-term treatment using IGF-1 preserves cardiac structure and improves function of the CHF147 hamster. METHODS: In the current study, we measured long-term effects of short-term treatment with recombinant human IGF-1 (rhIGF-1) in CHF147 hamsters. CHF147 hamsters (7-8 months old) were implanted under the skin with an osmotic pump filled either with saline or with recombinant human IGF-1 at a total dose of 25 microg. The osmotic pump allowed a continuous delivery of the protein for a mean duration of 19 days. RESULTS: We observed a significant increase in overall survival, as well as preservation of cardiac function, in the rhIGF-1-treated group. At the time of death, hearts of treated animals did not present any macroscopical or histological differences compared to those of sham hamsters. These results show that rhIGF-1 treatment slows down the evolution of the DCM in the CHF147 hamster. Moreover, the low dose treatment did not increase IGF-1 serum levels. CONCLUSIONS: This study is the first one reporting beneficial effects of IGF-1 treatment on survival of an animal model presenting DCM. Our results raise hopes for a new therapeutic approach of this pathology.


Subject(s)
Insulin-Like Growth Factor I/therapeutic use , Longevity/drug effects , Sarcoglycans/deficiency , Animals , Body Weight/drug effects , Cardiomyopathy, Dilated/drug therapy , Cricetinae , Echocardiography , Electrocardiography , Gene Deletion , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Life Expectancy , Male , Mesocricetus/genetics , Organ Size/drug effects , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Sarcoglycans/genetics , Time Factors
9.
Transplantation ; 80(5): 660-5, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16177642

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.


Subject(s)
Cryopreservation , Graft Survival , Heat-Shock Response , Myoblasts, Skeletal/transplantation , Myocardial Infarction/therapy , 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
10.
Basic Res Cardiol ; 100(2): 161-70, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15611844

ABSTRACT

Dilated cardiomyopathies (DCM) are due to progressive dilatation of the cardiac cavities and thinning of the ventricular walls and lead unavoidably to heart failure. They represent a major cause for heart transplantation and, therefore, defining an efficient symptomatic treatment for DCM remains a challenge. We have taken advantage of the hamster strain CHF147 that displays progressive cardiomyopathy leading to heart failure to test whether stimulation of a hypertrophic pathway could delay the process of dilatation.Six month old CHF147 hamsters were treated with IGF-1 so that we could compare the efficacy of systemic administration of human recombinant IGF-1 protein (rh IGF-1) at low dose to that of direct myocardial injections of a plasmid DNA containing IGF-1 cDNA (pCMV-IGF1).IGF-1 treatment did not induce a significant variation of ventricle mass, but preserved left ventricular (LV) wall thickness and delayed dilatation of cardiac cavities when compared to non-treated hamsters. Together with this reduction of dilatation, we also noted a reduction in the amount of interstitial collagen. Furthermore, IGF-1 treatment induced beneficial effects on cardiac function since treated hamsters presented improved cardiac output and stroke volume, decreased end diastolic pressure when compared to nontreated hamsters and also showed a trend towards increased contractility (dP/dt(max)).This study provides evidence that IGF-1 treatment induces beneficial structural and functional effects on DCM of CHF147 hamsters, hence making this molecule a promising candidate for future gene therapy of heart failure due to DCM.


Subject(s)
Cardiomyopathy, Dilated/therapy , Cardiotonic Agents , Genetic Therapy , Heart Failure/prevention & control , Insulin-Like Growth Factor I , Myocardium/metabolism , Sarcoglycans/deficiency , Animals , Cardiac Output , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Cardiotonic Agents/administration & dosage , Collagen/metabolism , Cricetinae , Disease Models, Animal , Fibrosis , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Injections , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Mesocricetus , Myocardial Contraction , Myocardium/pathology , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Sarcoglycans/genetics , Stroke Volume , Ventricular Function, Left , Ventricular Pressure
11.
Circulation ; 110(12): 1626-31, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15364802

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.


Subject(s)
Cardiomyopathy, Dilated/therapy , Muscle, Skeletal/cytology , Myoblasts/transplantation , 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
12.
Cardiovasc Res ; 63(2): 293-304, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15249187

ABSTRACT

OBJECTIVE: Cardiac myosin-binding protein C (cMyBP-C) gene mutations are involved in familial hypertrophic cardiomyopathy (FHC). Many of these mutations produce truncated proteins, which are unstable in the cardiac tissue of patients, suggesting that haploinsufficiency could account for the development of the phenotype. However, existing mouse models of cMyBP-C gene mutations have represented hypomorphic alleles without evidence of asymmetric septal hypertrophy, a key FHC phenotypic feature. In the present study, we generated a new model of cMyBP-C null mice and characterized the phenotype in both homozygotes and heterozygotes at different ages. METHODS: The mouse model was based upon the targeted deletion of exons 1 and 2, which contain the transcription initiation site, and the phenotype was determined by molecular, functional and morphological analyses. RESULTS: Herein, we demonstrate that inactivation of one or two mouse cMyBP-C alleles leads to different cardiac disorders at different post-natal time windows. The homozygous cMyBP-C null mice do not express the cMyBP-C gene, develop eccentric left ventricular hypertrophy with decreased fractional shortening at 3-4 months of age and a markedly impaired relaxation after 9 months. This is associated with myocardial disarray and an increase of interstitial fibrosis. The heterozygous cMyBP-C null mice present a slight but significant decrease of cMyBP-C amount and develop asymmetric septal hypertrophy associated with fibrosis at 10-11 months of age. CONCLUSION: These data provide evidence that heterozygous cMyBP-C null mice represent the first model with a key feature of human FHC that is asymmetric septal hypertrophy.


Subject(s)
Cardiomegaly/pathology , Carrier Proteins/genetics , Myocardium/pathology , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/metabolism , Carrier Proteins/metabolism , Echocardiography , Heart Septum , Heterozygote , Homozygote , Immunohistochemistry/methods , Mice , Mice, Knockout , Models, Animal , Myocardium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...