Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Elife ; 132024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842166

ABSTRACT

Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.


Subject(s)
Cell Survival , Endothelial Cells , Proto-Oncogene Proteins c-akt , Signal Transduction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Animals , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Mice , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Endothelial Cells/metabolism , Endothelial Cells/physiology , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/physiology , Mice, Inbred C57BL , Male
2.
Methods Mol Biol ; 2640: 453-462, 2023.
Article in English | MEDLINE | ID: mdl-36995613

ABSTRACT

Skeletal muscle is a highly ordered tissue composed of a complex network of a diverse variety of cells. The dynamic spatial and temporal interaction between these cells during homeostasis and during times of injury gives the skeletal muscle its regenerative capacity. To properly understand the process of regeneration, a three-dimensional (3-D) imaging process must be conducted. With the advancement of imaging and computing technology, it has become powerful to analyze spatial data from confocal microscope images. In order to prepare whole tissue skeletal muscle samples for confocal imaging, the muscle must be subjected to tissue clearing. With the use of an ideal optical clearing protocol - one that minimizes light scattering via refractive index mismatching - a more accurate 3-D image of the muscle can be produced as it does not involve the physical sectioning of the muscle. While there have been several protocols relating to the study of 3-D biology in whole tissue, these protocols have primarily been focused on the nervous system. In this chapter, we present a new method for skeletal muscle tissue clearing. In addition, this protocol aims to outline the specific parameters required for taking 3-D images of immunofluorescence-stained skeletal muscle samples using a confocal microscope.


Subject(s)
Imaging, Three-Dimensional , Muscle, Skeletal , Microscopy, Confocal , Fluorescent Antibody Technique , Imaging, Three-Dimensional/methods
3.
Methods Mol Biol ; 2640: 463-477, 2023.
Article in English | MEDLINE | ID: mdl-36995614

ABSTRACT

Skeletal muscle is a highly ordered tissue composed of a complex network of a diverse variety of cells. The dynamic spatial and temporal interaction between these cells during homeostasis and during times of injury gives the skeletal muscle its regenerative capacity. In order to properly understand the process of regeneration, a three-dimensional (3-D) imaging process must be conducted. While there have been several protocols studying 3-D imaging, it has primarily been focused on the nervous system. This protocol aims to outline the workflow for rendering a 3-D image of the skeletal muscle using spatial data from confocal microscope images. This protocol uses the ImageJ, Ilastik, and Imaris software for 3-D rendering and computational image analysis as both are relatively easy to use and have powerful segmentation capabilities.


Subject(s)
Imaging, Three-Dimensional , Satellite Cells, Skeletal Muscle , Satellite Cells, Skeletal Muscle/physiology , Muscle, Skeletal/physiology , Image Processing, Computer-Assisted , Muscle Development/physiology , Cell Differentiation
4.
Nat Metab ; 4(2): 180-189, 2022 02.
Article in English | MEDLINE | ID: mdl-35228746

ABSTRACT

Adult skeletal muscle is a highly plastic tissue that readily reduces or gains its mass in response to mechanical and metabolic stimulation; however, the upstream mechanisms that control muscle mass remain unclear. Notch signalling is highly conserved, and regulates many cellular events, including proliferation and differentiation of various types of tissue stem cell via cell-cell contact. Here we reveal that multinucleated myofibres express Notch2, which plays a crucial role in disuse- or diabetes-induced muscle atrophy. Mechanistically, in both atrophic conditions, the microvascular endothelium upregulates and releases the Notch ligand, Dll4, which then activates muscular Notch2 without direct cell-cell contact. Inhibition of the Dll4-Notch2 axis substantively prevents these muscle atrophy and promotes mechanical overloading-induced muscle hypertrophy in mice. Our results illuminate a tissue-specific function of the endothelium in controlling tissue plasticity and highlight the endothelial Dll4-muscular Notch2 axis as a central upstream mechanism that regulates catabolic signals from mechanical and metabolic stimulation, providing a therapeutic target for muscle-wasting diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium-Binding Proteins , Muscular Atrophy , Animals , Endothelium , Mice , Muscle, Skeletal , Receptor, Notch2
6.
J Cell Biol ; 220(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34009269

ABSTRACT

Circadian rhythms regulate cell proliferation and differentiation, but circadian control of tissue regeneration remains elusive at the molecular level. Here, we show that proper myoblast differentiation and muscle regeneration are regulated by the circadian master regulators Per1 and Per2. Depletion of Per1 or Per2 suppressed myoblast differentiation in vitro and muscle regeneration in vivo, demonstrating their nonredundant functions. Both Per1 and Per2 were required for the activation of Igf2, an autocrine promoter of myoblast differentiation, accompanied by Per-dependent recruitment of RNA polymerase II, dynamic histone modifications at the Igf2 promoter and enhancer, and the promoter-enhancer interaction. This circadian epigenetic priming created a preferred time window for initiating myoblast differentiation. Consistently, muscle regeneration was faster if initiated at night, when Per1, Per2, and Igf2 were highly expressed compared with morning. This study reveals the circadian timing as a significant factor for effective muscle cell differentiation and regeneration.


Subject(s)
Circadian Rhythm/genetics , Insulin-Like Growth Factor II/genetics , Period Circadian Proteins/genetics , Regeneration/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mice, Knockout , Muscle, Skeletal/growth & development , Myoblasts/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics
7.
PLoS Genet ; 15(12): e1008468, 2019 12.
Article in English | MEDLINE | ID: mdl-31877123

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease in which the dystrophin coding for a membrane stabilizing protein is mutated. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice. Recent DMD transcriptomics revealed the defects were correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with constitutive (CAGCreERTM:Flt1LoxP/LoxP) and endothelial cell-specific conditional gene knockout mice (Cdh5CreERT2:Flt1LoxP/LoxP) for Flt1 (VEGFR1) which is a decoy receptor for VEGF. Here, we showed that while constitutive deletion of Flt1 is detrimental to the skeletal muscle function, endothelial cell-specific Flt1 deletion resulted in increased vascular density, increased satellite cell number and improvement in the DMD-associated phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Muscular Dystrophy, Duchenne/drug therapy , Peptides/administration & dosage , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Endothelial Cells/metabolism , Gene Knockout Techniques , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Organ Specificity , Peptides/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-1/genetics
8.
Muscle Nerve ; 59(5): 594-602, 2019 05.
Article in English | MEDLINE | ID: mdl-30698289

ABSTRACT

INTRODUCTION: The vasculature and blood flow in muscle are perturbed in Duchenne muscular dystrophy (DMD) and its mdx mouse model. MicroRNA-92a (miR-92a) is enriched in endothelial cells, especially during ischemic injury. METHODS: Because antagonizing miR-92a was shown to result in increased proliferation and migration of endothelial cells and recovery from ischemia, we assessed the effects of Antagomir-92a in vitro in muscle stem cell culture and in vivo in mdx mice. RESULTS: miR-92a was found to be highly expressed in muscle endothelial cells and satellite cells. Treatment with Antagomir-92a increased capillary density and tissue perfusion, which was accompanied by an increase in satellite cells. However, Antagomir-92a-treated mdx mice showed no histological improvement and had worse muscle function. Antagomir-92a suppressed myogenic differentiation in satellite cell culture. DISCUSSION: AntagomiR-92a improves the vasculature but not the muscle in mdx mice, possibly due to its side effects on satellite cell differentiation. Muscle Nerve 59:594-594, 2019.


Subject(s)
Antagomirs/pharmacology , MicroRNAs/antagonists & inhibitors , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Hand Strength , Laser-Doppler Flowmetry , Mice , Mice, Inbred mdx , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/cytology
9.
Cell Rep ; 25(9): 2537-2551.e8, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30485818

ABSTRACT

Fkbp5 is a widely expressed peptidyl prolyl isomerase that serves as a molecular chaperone through conformational changes of binding partners. Although it regulates diverse protein functions, little is known about its roles in myogenesis. We found here that Fkbp5 plays critical roles in myoblast differentiation through two mechanisms. First, it sequesters Cdk4 within the Hsp90 storage complex and prevents the formation of the cyclin D1-Cdk4 complex, which is a major inhibitor of differentiation. Second, Fkbp5 promotes cis-trans isomerization of the Thr172-Pro173 peptide bond in Cdk4 and inhibits phosphorylation of Thr172, an essential step for Cdk4 activation. Consistent with these in vitro findings, muscle regeneration is delayed in Fkbp5-/- mice. The related protein Fkbp4 also sequesters Cdk4 within the Hsp90 complex but does not isomerize Cdk4 or induce Thr173 phosphorylation despite its highly similar sequence. This study demonstrates protein isomerization as a critical regulatory mechanism of myogenesis by targeting Cdk4.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 4/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Cell Line , Cell Proliferation , HSP90 Heat-Shock Proteins/metabolism , Isomerism , Male , Mice, Knockout , Muscles/physiology , Peptides/metabolism , Proline/metabolism , Protein Binding , Regeneration , Tacrolimus Binding Proteins/deficiency
10.
Elife ; 72018 10 04.
Article in English | MEDLINE | ID: mdl-30284969

ABSTRACT

Adult skeletal muscle maintenance and regeneration depend on efficient muscle stem cell (MuSC) functions. The mechanisms coordinating cell cycle with activation, renewal, and differentiation of MuSCs remain poorly understood. Here, we investigated how adult MuSCs are regulated by CDKN1c (p57kip2), a cyclin-dependent kinase inhibitor, using mouse molecular genetics. In the absence of CDKN1c, skeletal muscle repair is severely impaired after injury. We show that CDKN1c is not expressed in quiescent MuSCs, while being induced in activated and proliferating myoblasts and maintained in differentiating myogenic cells. In agreement, isolated Cdkn1c-deficient primary myoblasts display differentiation defects and increased proliferation. We further show that the subcellular localization of CDKN1c is dynamic; while CDKN1c is initially localized to the cytoplasm of activated/proliferating myoblasts, progressive nuclear translocation leads to growth arrest during differentiation. We propose that CDKN1c activity is restricted to differentiating myoblasts by regulated cyto-nuclear relocalization, coordinating the balance between proliferation and growth arrest.


Subject(s)
Adult Stem Cells/cytology , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Muscle Development/genetics , Animals , Cell Cycle Checkpoints/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Myoblasts/cytology , Myoblasts/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/cytology
11.
Cell Stem Cell ; 23(4): 530-543.e9, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290177

ABSTRACT

Skeletal muscle is a complex tissue containing tissue resident muscle stem cells (satellite cells) (MuSCs) important for postnatal muscle growth and regeneration. Quantitative analysis of the biological function of MuSCs and the molecular pathways responsible for a potential juxtavascular niche for MuSCs is currently lacking. We utilized fluorescent reporter mice and muscle tissue clearing to investigate the proximity of MuSCs to capillaries in 3 dimensions. We show that MuSCs express abundant VEGFA, which recruits endothelial cells (ECs) in vitro, whereas blocking VEGFA using both a vascular endothelial growth factor (VEGF) inhibitor and MuSC-specific VEGFA gene deletion reduces the proximity of MuSCs to capillaries. Importantly, this proximity to the blood vessels was associated with MuSC self-renewal in which the EC-derived Notch ligand Dll4 induces quiescence in MuSCs. We hypothesize that MuSCs recruit capillary ECs via VEGFA, and in return, ECs maintain MuSC quiescence though Dll4.


Subject(s)
Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Vascular Endothelial Growth Factors/metabolism , Animals , Cells, Cultured , Coculture Techniques , Mice , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Niche
12.
Cell Rep ; 22(8): 2118-2132, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466738

ABSTRACT

Circadian rhythms regulate cell proliferation and differentiation; however, little is known about their roles in myogenic differentiation. Our synchronized differentiation studies demonstrate that myoblast proliferation and subsequent myotube formation by cell fusion occur in circadian manners. We found that one of the core regulators of circadian rhythms, Cry2, but not Cry1, is critical for the circadian patterns of these two critical steps in myogenic differentiation. This is achieved through the specific interaction between Cry2 and Bclaf1, which stabilizes mRNAs encoding cyclin D1, a G1/S phase transition regulator, and Tmem176b, a transmembrane regulator for myogenic cell fusion. Myoblasts lacking Cry2 display premature cell cycle exit and form short myotubes because of inefficient cell fusion. Consistently, muscle regeneration is impaired in Cry2-/- mice. Bclaf1 knockdown recapitulated the phenotypes of Cry2 knockdown: early cell cycle exit and inefficient cell fusion. This study uncovers a post-transcriptional regulation of myogenic differentiation by circadian rhythms.


Subject(s)
Cell Differentiation , Circadian Rhythm , Cryptochromes/metabolism , Cyclin D1/genetics , Membrane Proteins/metabolism , Muscle Development , RNA Stability/genetics , Repressor Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Fusion , Cell Line , Cyclin D1/metabolism , Gene Expression Regulation , Mice, Knockout , Muscles/metabolism , Myoblasts/cytology , Myoblasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regeneration
13.
Biotechniques ; 63(2): 72-76, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28803542

ABSTRACT

Viral vector-mediated foreign gene expression in cultured cells has been extensively used in stem cell studies to explore gene function. However, it is difficult to obtain high-quality stem cells and primary cells after viral vector infection. Here, we describe a new protocol for high-efficiency retroviral infection of primary muscle stem cell (satellite cell) cultures. We compared multiple commercially available transfection reagents to determine which was optimal for retroviral infections of primary myoblasts. Centrifugation force was also tested, and a spin infection protocol with centrifugation at 2800 × g for 90 min had the highest infection efficiency for primary myoblasts. We confirmed that infected muscle stem cells maintain cell proliferation and the capacity for in vitro and in vivo myogenic differentiation. Our new, efficient retroviral infection protocol for muscle stem cells can be applied to molecular biology experiments as well as translational studies.


Subject(s)
Genetic Vectors/genetics , Muscle Development , Myoblasts/cytology , Retroviridae/genetics , Stem Cells/cytology , Transfection/methods , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Myoblasts/metabolism , Stem Cells/metabolism
14.
Appl Microbiol Biotechnol ; 101(4): 1581-1592, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27915375

ABSTRACT

Alginate is a marine non-food-competing polysaccharide that has potential applications in biorefinery. Owing to its large size (molecular weight >300,000 Da), alginate cannot pass through the bacterial cell membrane. Therefore, bacteria that utilize alginate are presumed to have an enzyme that degrades extracellular alginate. Recently, Vibrio algivorus sp. SA2T was identified as a novel alginate-decomposing and alginate-utilizing species. However, little is known about the mechanism of alginate degradation and metabolism in this species. To address this issue, we screened the V. algivorus genomic DNA library for genes encoding polysaccharide-decomposing enzymes using a novel double-layer plate screening method and identified alyB as a candidate. Most identified alginate-decomposing enzymes (i.e., alginate lyases) must be concentrated and purified before extracellular alginate depolymerization. AlyB of V. algivorus heterologously expressed in Escherichia coli depolymerized extracellular alginate without requiring concentration or purification. We found seven homologues in the V. algivorus genome (alyB, alyD, oalA, oalB, oalC, dehR, and toaA) that are thought to encode enzymes responsible for alginate transport and metabolism. Introducing these genes into E. coli enabled the cells to assimilate soluble alginate depolymerized by V. algivorus AlyB as the sole carbon source. The alginate was bioconverted into L-lysine (43.3 mg/l) in E. coli strain AJIK01. These findings demonstrate a simple and novel screening method for identifying polysaccharide-degrading enzymes in bacteria and provide a simple alginate biocatalyst and fermentation system with potential applications in industrial biorefinery.


Subject(s)
Alginates/metabolism , Polysaccharide-Lyases/metabolism , Vibrio/enzymology , Vibrio/metabolism , Glucuronic Acid/metabolism , Hexuronic Acids/metabolism
15.
Methods Mol Biol ; 1460: 129-40, 2016.
Article in English | MEDLINE | ID: mdl-27492170

ABSTRACT

Skeletal muscle is a highly ordered yet complex tissue containing several cell types that interact with each other in order to maintain structure and homeostasis. It is also a highly regenerative tissue that responds to damage in a highly intricate but stereotypic manner, with distinct spatial and temporal kinetics. Proper examination of this process requires one to look at the three-dimensional orientation of the cellular and subcellular components, which can be accomplished through tissue clearing. While there has been a recent surge of protocols to study biology in whole tissue, it has primarily focused on the nervous system. This chapter describes the workflow for whole mount analysis of murine skeletal muscle for LacZ reporters, fluorescent reporters and immunofluorescence staining. Using this technique, we are able to visualize LacZ reporters more effectively in deep tissue samples, and to perform fluorescent imaging with a depth greater than 1700 µm.


Subject(s)
Fluorescent Antibody Technique , Genes, Reporter , Green Fluorescent Proteins/metabolism , Muscle, Skeletal/metabolism , beta-Galactosidase/metabolism , Animals , Biomarkers , Green Fluorescent Proteins/genetics , Mice , Microscopy, Fluorescence/methods , Staining and Labeling , beta-Galactosidase/genetics
16.
PLoS One ; 10(3): e0120325, 2015.
Article in English | MEDLINE | ID: mdl-25775477

ABSTRACT

Duchenne muscular dystrophy (DMD), the most common and severe type of dystrophinopathy, is an X-linked recessive genetic disease caused by the absence of dystrophin, which leads to fragility and vulnerability of the sarcolemma to mechanical stretching with increased membrane permeability. Currently, glucocorticoids such as prednisolone are the only medication available for DMD. However, molecular pathways responsible for this effect are still unclear. In addition, it remains unclear whether sex-related factors, including pregnancy and the postpartum period, affect the phenotype of dystrophinopathy. Here, we report the amelioration of muscle membrane permeability in the diaphragm muscle of pregnant and postpartum, but not in nulliparous, mdx mice, an animal model for DMD, during the physiological surge of corticosterone, the most abundant glucocorticoid in rodents. Cultures of single muscle fibers and myotubes isolated from mdx mouse diaphragm demonstrate resistance to hypo-osmotic shock when treated with corticosterone but not with estradiol or progesterone. This corticosterone-mediated resistance was diminished by an antagonist of corticosterone, indicating that the glucocorticoid-glucocorticoid receptor axis plays a role in this membrane stabilization effect on muscle. Moreover, subcutaneous injection of corticosterone into mdx mice showed decreased membrane permeability. This is the first report to demonstrate that pregnancy-related resistance to muscle fiber damage in mdx mice due to the membrane stabilization effect of corticosterone. We also propose that this membrane stabilization effect is exerted through annexin A1 up-regulation as the molecular mechanisms of glucocorticoid effects on DMD muscle. Furthermore, single muscle fiber culture studies provide a sensitive chemical screening platform for muscular dystrophies.


Subject(s)
Cell Membrane/metabolism , Corticosterone/blood , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Phenotype , Animals , Annexin A1/genetics , Annexin A1/metabolism , Cell Membrane/drug effects , Cell Membrane Permeability , Corticosterone/pharmacology , Female , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Osmotic Pressure , Pregnancy , Up-Regulation
17.
J Vis Exp ; (86)2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24747722

ABSTRACT

Muscle satellite cells are a stem cell population required for postnatal skeletal muscle development and regeneration, accounting for 2-5% of sublaminal nuclei in muscle fibers. In adult muscle, satellite cells are normally mitotically quiescent. Following injury, however, satellite cells initiate cellular proliferation to produce myoblasts, their progenies, to mediate the regeneration of muscle. Transplantation of satellite cell-derived myoblasts has been widely studied as a possible therapy for several regenerative diseases including muscular dystrophy, heart failure, and urological dysfunction. Myoblast transplantation into dystrophic skeletal muscle, infarcted heart, and dysfunctioning urinary ducts has shown that engrafted myoblasts can differentiate into muscle fibers in the host tissues and display partial functional improvement in these diseases. Therefore, the development of efficient purification methods of quiescent satellite cells from skeletal muscle, as well as the establishment of satellite cell-derived myoblast cultures and transplantation methods for myoblasts, are essential for understanding the molecular mechanisms behind satellite cell self-renewal, activation, and differentiation. Additionally, the development of cell-based therapies for muscular dystrophy and other regenerative diseases are also dependent upon these factors. However, current prospective purification methods of quiescent satellite cells require the use of expensive fluorescence-activated cell sorting (FACS) machines. Here, we present a new method for the rapid, economical, and reliable purification of quiescent satellite cells from adult mouse skeletal muscle by enzymatic dissociation followed by magnetic-activated cell sorting (MACS). Following isolation of pure quiescent satellite cells, these cells can be cultured to obtain large numbers of myoblasts after several passages. These freshly isolated quiescent satellite cells or ex vivo expanded myoblasts can be transplanted into cardiotoxin (CTX)-induced regenerating mouse skeletal muscle to examine the contribution of donor-derived cells to regenerating muscle fibers, as well as to satellite cell compartments for the examination of self-renewal activities.


Subject(s)
Flow Cytometry/methods , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Mice , Mice, Transgenic , Muscular Dystrophy, Animal/therapy , Regeneration/physiology
18.
PLoS One ; 7(7): e41736, 2012.
Article in English | MEDLINE | ID: mdl-22848585

ABSTRACT

Skeletal myoblast transplantation has therapeutic potential for repairing damaged heart. However, the optimal conditions for this transplantation are still unclear. Recently, we demonstrated that satellite cell-derived myoblasts lacking the MyoD gene (MyoD(-/-)), a master transcription factor for skeletal muscle myogenesis, display increased survival and engraftment compared to wild-type controls following transplantation into murine skeletal muscle. In this study, we compare cell survival between wild-type and MyoD(-/-) myoblasts after transplantation into infarcted heart. We demonstrate that MyoD(-/-) myoblasts display greater resistance to hypoxia, engraft with higher efficacy, and show a larger improvement in ejection fraction than wild-type controls. Following transplantation, the majority of MyoD(-/-) and wild-type myoblasts form skeletal muscle fibers while cardiomyocytes do not. Importantly, the transplantation of MyoD(-/-) myoblasts induces a high degree of angiogenesis in the area of injury. DNA microarray data demonstrate that paracrine angiogenic factors, such as stromal cell-derived factor-1 (SDF-1) and placental growth factor (PlGF), are up-regulated in MyoD(-/-) myoblasts. In addition, over-expression and gene knockdown experiments demonstrate that MyoD negatively regulates gene expression of these angiogenic factors. These results indicate that MyoD(-/-) myoblasts impart beneficial effects after transplantation into an infarcted heart, potentially due to the secretion of paracrine angiogenic factors and enhanced angiogenesis in the area of injury. Therefore, our data provide evidence that a genetically engineered myoblast cell type with suppressed MyoD function is useful for therapeutic stem cell transplantation.


Subject(s)
MyoD Protein/genetics , Myoblasts/transplantation , Myocardial Infarction/physiopathology , Myocardium/pathology , Neovascularization, Physiologic , Stem Cell Transplantation , Ventricular Dysfunction, Left/surgery , Animals , Cell Differentiation , Cell Hypoxia , Cell Proliferation , Cell Survival , Chemokine CXCL12/genetics , Coculture Techniques , Endothelial Cells/pathology , Female , Gene Expression Regulation , Mice , Mice, Inbred BALB C , Muscle, Skeletal/pathology , Myoblasts/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocardium/metabolism , Placenta Growth Factor , Pregnancy Proteins/genetics
19.
Nucleic Acids Res ; 39(20): 9034-46, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21785135

ABSTRACT

Modification of complex microbial cellular processes is often necessary to obtain organisms with particularly favorable characteristics, but such experiments can take many generations to achieve. In the present article, we accelerated the experimental evolution of Escherichia coli populations under selection for improved growth using one of the restriction-modification systems, which have shaped bacterial genomes. This resulted in faster evolutionary changes in both the genome and bacterial growth. Transcriptome/genome analysis at various stages enabled prompt identification of sequential genome rearrangements and dynamic gene-expression changes associated with growth improvement. The changes were related to cell-to-cell communication, the cell death program, as well as mass production and energy consumption. These observed changes imply that improvements in microorganism population growth can be achieved by inactivating the cellular mechanisms regulating fraction of active cells in a population. Some of the mutations were shown to have additive effects on growth. These results open the way for the application of evolutionary genome engineering to generate organisms with desirable properties.


Subject(s)
DNA Restriction-Modification Enzymes/metabolism , Escherichia coli/genetics , Evolution, Molecular , Genetic Engineering/methods , Genome, Bacterial , Adaptation, Physiological/genetics , DNA Restriction-Modification Enzymes/genetics , Escherichia coli/growth & development , Mutation , Phenotype , Transcriptome
20.
Stem Cells ; 29(3): 505-16, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21425413

ABSTRACT

Expression of the four transcription factors, that is, Oct4, Sox2, cMyc, and Klf4 has been shown to generate induced pluripotent stem cells (iPSCs) from many types of specialized differentiated somatic cells. It remains unclear, however, whether fully committed skeletal muscle progenitor cells (myoblasts) have the potency to undergo reprogramming to develop iPSCs in line with previously reported cases. To test this, we have isolated genetically marked myoblasts derived from satellite cell of adult mouse muscles using the Cre-loxP system (Pax7-CreER:R26R and Myf5-Cre:R26R). On infection with retroviral vectors expressing the four factors, these myoblasts gave rise to myogenic lineage tracer lacZ-positive embryonic stem cell (ESC)-like colonies. These cells expressed ESC-specific genes and were competent to differentiate into all three germ layers and germ cells, indicating the successful generation of myoblast-derived iPSCs. Continuous expression of the MyoD gene, a master transcription factor for skeletal muscle specification, inhibited this reprogramming process in myoblasts. In contrast, reprogramming myoblasts isolated from mice lacking the MyoD gene led to an increase in reprogramming efficiency. Our data also indicated that Oct4 acts as a transcriptional suppressor of MyoD gene expression through its interaction with the upstream enhancer region. Taken together, these results indicate that suppression of MyoD gene expression by Oct4 is required for the initial reprogramming step in the development of iPSCs from myoblasts. This data suggests that the skeletal muscle system provides a well-defined differentiation model to further elaborate on the effects of iPSC reprogramming in somatic cells.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/physiology , MyoD Protein/genetics , Myoblasts/physiology , Octamer Transcription Factor-3/physiology , Animals , Cells, Cultured , Down-Regulation/genetics , Female , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Mice, Inbred ICR , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , MyoD Protein/metabolism , MyoD Protein/physiology , Myoblasts/cytology , Myoblasts/metabolism , Octamer Transcription Factor-3/metabolism , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...