Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 2731, 2023 05 12.
Article in English | MEDLINE | ID: mdl-37169793

ABSTRACT

A potential therapeutic target to curb obesity and diabetes is thermogenic beige adipocytes. However, beige adipocytes quickly transition into white adipocytes upon removing stimuli. Here, we define the critical role of cyclin dependent kinase inhibitor 2A (Cdkn2a) as a molecular pedal for the beige-to-white transition. Beige adipocytes lacking Cdkn2a exhibit prolonged lifespan, and male mice confer long-term metabolic protection from diet-induced obesity, along with enhanced energy expenditure and improved glucose tolerance. Mechanistically, Cdkn2a promotes the expression and activity of beclin 1 (BECN1) by directly binding to its mRNA and its negative regulator BCL2 like 1 (BCL2L1), activating autophagy and accelerating the beige-to-white transition. Reactivating autophagy by pharmacological or genetic methods abolishes beige adipocyte maintenance induced by Cdkn2a ablation. Furthermore, hyperactive BECN1 alone accelerates the beige-to-white transition in mice and human. Notably, both Cdkn2a and Becn1 exhibit striking positive correlations with adiposity. Hence, blocking Cdkn2a-mediated BECN1 activity holds therapeutic potential to sustain beige adipocytes in treating obesity and related metabolic diseases.


Subject(s)
Adipocytes, Beige , Adipose Tissue, Beige , Obesity , Animals , Humans , Male , Mice , Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Adiposity/genetics , Adiposity/physiology , Obesity/genetics , Obesity/metabolism , Thermogenesis
2.
Am J Physiol Cell Physiol ; 324(3): C718-C727, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36717102

ABSTRACT

Individual limb muscles have characteristic representation and spatial distribution of muscle fiber types (one slow and up to three fast isoforms) appropriate to their unique anatomical location and function. This distribution can be altered by physiological stimuli such as training (i.e., for increased endurance or force) or pathological conditions such as aging. Our group previously showed that ephrin-A3 is expressed only on slow myofibers, and that adult mice lacking ephrin-A3 have dramatically reduced numbers of slow myofibers due to postnatal innervation of previously slow myofibers by fast motor neurons. In this study, fiber type composition of hindlimb muscles of aged and denervated/reinnervated C57BL/6 and ephrin-A3-/- mice was analyzed to determine whether the loss of slow myofibers persists across the lifespan. Surprisingly, fiber-type composition of ephrin-A3-/- mouse muscles at two years of age was nearly indistinguishable from age-matched C57BL/6 mice. After challenge with nerve crush, the percentage of IIa and I/IIa hybrid myofibers increased significantly in aged ephrin-A3-/- mice. While EphA8, the receptor for ephrin-A3, is present at all neuromuscular junctions (NMJs) on fast fibers in 3-6 mo old C57BL/6 and ephrin-A3-/- mice, this exclusive localization is lost with aging, with EphA8 expression now found on a subset of NMJs on some slow muscle fibers. This return to appropriate fiber-type distribution given time and under use reinforces the role of activity in determining fiber-type representation and suggests that, rather than being a passive baseline, the developmentally and evolutionarily selected fiber type pattern may instead be actively reinforced by daily living.


Subject(s)
Ephrin-A3 , Muscle Fibers, Skeletal , Mice , Animals , Ephrin-A3/metabolism , Mice, Inbred C57BL , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/metabolism , Neuromuscular Junction
3.
Front Cell Dev Biol ; 10: 949532, 2022.
Article in English | MEDLINE | ID: mdl-36211464

ABSTRACT

Acute skeletal muscle injury is followed by satellite cell activation, proliferation, and differentiation to replace damaged fibers with newly regenerated muscle fibers, processes that involve satellite cell interactions with various niche signals. Here we show that satellite cell specific deletion of the chemokine receptor CXCR4, followed by suppression of recombination escapers, leads to defects in regeneration and satellite cell pool repopulation in both the transplantation and in situ injury contexts. Mechanistically, we show that endothelial cells and FAPs express the gene for the ligand, SDF1α, and that CXCR4 is principally required for proper activation and for transit through the first cell division, and to a lesser extent the later cell divisions. In the absence of CXCR4, gene expression in quiescent satellite cells is not severely disrupted, but in activated satellite cells a subset of genes normally induced by activation fail to upregulate normally. These data demonstrate that CXCR4 signaling is essential to normal early activation, proliferation, and self-renewal of satellite cells.

4.
Skelet Muscle ; 11(1): 22, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34481522

ABSTRACT

BACKGROUND: Although muscle regenerative capacity declines with age, the extent to which this is due to satellite cell-intrinsic changes vs. environmental changes has been controversial. The majority of aging studies have investigated hindlimb locomotory muscles, principally the tibialis anterior, in caged sedentary mice, where those muscles are abnormally under-exercised. METHODS: We analyze satellite cell numbers in 8 muscle groups representing locomotory and non-locomotory muscles in young and 2-year-old mice and perform transplantation assays of low numbers of hind limb satellite cells from young and old mice. RESULTS: We find that satellite cell density does not decline significantly by 2 years of age in most muscles, and one muscle, the masseter, shows a modest but statistically significant increase in satellite cell density with age. The tibialis anterior and extensor digitorum longus were clear exceptions, showing significant declines. We quantify self-renewal using a transplantation assay. Dose dilution revealed significant non-linearity in self-renewal above a very low threshold, suggestive of competition between satellite cells for space within the pool. Assaying within the linear range, i.e., transplanting fewer than 1000 cells, revealed no evidence of decline in cell-autonomous self-renewal or regenerative potential of 2-year-old murine satellite cells. CONCLUSION: These data demonstrate the value of comparative muscle analysis as opposed to overreliance on locomotory muscles, which are not used physiologically in aging sedentary mice, and suggest that self-renewal impairment with age is precipitously acquired at the geriatric stage, rather than being gradual over time, as previously thought.


Subject(s)
Myoblasts , Satellite Cells, Skeletal Muscle , Aging , Animals , Cell Count , Cell Self Renewal , Mice , Muscle, Skeletal , Regeneration
5.
Cell Rep ; 28(2): 368-381.e6, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291574

ABSTRACT

Skeletal muscle mass, strength, and regenerative capacity decline with age, with many measures showing a greater deterioration in females around the time estrogen levels decrease at menopause. Here, we show that estrogen deficiency severely compromises the maintenance of muscle stem cells (i.e., satellite cells) as well as impairs self-renewal and differentiation into muscle fibers. Mechanistically, by hormone replacement, use of a selective estrogen-receptor modulator (bazedoxifene), and conditional estrogen receptor knockout, we implicate 17ß-estradiol and satellite cell expression of estrogen receptor α and show that estrogen signaling through this receptor is necessary to prevent apoptosis of satellite cells. Early data from a biopsy study of women who transitioned from peri- to post-menopause are consistent with the loss of satellite cells coincident with the decline in estradiol in humans. Together, these results demonstrate an important role for estrogen in satellite cell maintenance and muscle regeneration in females.


Subject(s)
Estrogens/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Animals , Female , Humans , Mice
6.
Proc Natl Acad Sci U S A ; 116(10): 4346-4351, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30760602

ABSTRACT

Optimal cell-based therapies for the treatment of muscle degenerative disorders should not only regenerate fibers but provide a quiescent satellite cell pool ensuring long-term maintenance and regeneration. Conditional expression of Pax3/Pax7 in differentiating pluripotent stem cells (PSCs) allows the generation of myogenic progenitors endowed with enhanced regenerative capacity. To identify the molecular determinants underlying their regenerative potential, we performed transcriptome analyses of these cells along with primary myogenic cells from several developmental stages. Here we show that in vitro-generated PSC-derived myogenic progenitors possess a molecular signature similar to embryonic/fetal myoblasts. However, compared with fetal myoblasts, following transplantation they show superior myofiber engraftment and ability to seed the satellite cell niche, respond to multiple reinjuries, and contribute to long-term regeneration. Upon engraftment, the transcriptome of reisolated Pax3/Pax7-induced PSC-derived myogenic progenitors changes toward a postnatal molecular signature, particularly in genes involved in extracellular matrix remodeling. These findings demonstrate that Pax3/Pax7-induced myogenic progenitors remodel their molecular signature and functionally mature upon in vivo exposure to the adult muscle environment.


Subject(s)
Muscle Development/physiology , PAX3 Transcription Factor/metabolism , PAX7 Transcription Factor/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Gene Expression Profiling , Mice , Muscle Development/genetics , Muscle, Skeletal , Myoblasts/metabolism , PAX3 Transcription Factor/genetics , PAX7 Transcription Factor/genetics , Transcriptome
7.
Cell Stem Cell ; 23(1): 74-85.e6, 2018 Jul 05.
Article in English | MEDLINE | ID: mdl-29979993

ABSTRACT

Derivation of functional skeletal muscle stem cells from pluripotent cells without genetic modification has proven elusive. Here we show that teratomas formed in adult skeletal muscle differentiate in vivo to produce large numbers of α7-Integrin+ VCAM-1+ myogenic progenitors. When FACS-purified and transplanted into diseased muscles, mouse teratoma-derived myogenic progenitors demonstrate very high engraftment potential. As few as 40,000 cells can reconstitute ∼80% of the tibialis anterior muscle volume. Newly generated fibers are innervated, express adult myosins, and ameliorate dystrophy-related force deficit and fatigability. Teratoma-derived myogenic progenitors also contribute quiescent PAX7+ muscle stem cells, enabling long-term maintenance of regenerated muscle and allowing muscle regeneration in response to subsequent injuries. Transcriptional profiling reveals that teratoma-derived myogenic progenitors undergo embryonic-to-adult maturation when they contribute to the stem cell compartment of regenerated muscle. Thus, teratomas are a rich and accessible source of potent transplantable skeletal muscle stem cells. VIDEO ABSTRACT.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Regeneration , Satellite Cells, Skeletal Muscle/pathology , Teratoma/pathology , Animals , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Satellite Cells, Skeletal Muscle/metabolism , Teratoma/metabolism
8.
Hum Mol Genet ; 27(3): 451-462, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29194514

ABSTRACT

Missense mutations in the dystrophin protein can cause Duchenne muscular dystrophy (DMD) or Becker muscular dystrophy (BMD) through an undefined pathomechanism. In vitro studies suggest that missense mutations in the N-terminal actin-binding domain (ABD1) cause protein instability, and cultured myoblast studies reveal decreased expression levels that can be restored to wild-type with proteasome inhibitors. To further elucidate the pathophysiology of missense dystrophin in vivo, we generated two transgenic mdx mouse lines expressing L54R or L172H mutant dystrophin, which correspond to missense mutations identified in human patients with DMD or BMD, respectively. Our biochemical, histologic and physiologic analysis of the L54R and L172H mice show decreased levels of dystrophin which are proportional to the phenotypic severity. Proteasome inhibitors were ineffective in both the L54R and L172H mice, yet mice homozygous for the L172H transgene were able to express even higher levels of dystrophin which caused further improvements in muscle histology and physiology. Given that missense dystrophin is likely being degraded by the proteasome but whole body proteasome inhibition was not possible, we screened for ubiquitin-conjugating enzymes involved in targeting dystrophin to the proteasome. A myoblast cell line expressing L54R mutant dystrophin was screened with an siRNA library targeting E1, E2 and E3 ligases which identified Amn1, FBXO33, Zfand5 and Trim75. Our study establishes new mouse models of dystrophinopathy and identifies candidate E3 ligases that may specifically regulate dystrophin protein turnover in vivo.


Subject(s)
Dystrophin/genetics , Dystrophin/metabolism , Muscular Dystrophy, Duchenne/genetics , Mutation, Missense/genetics , Animals , Blotting, Western , Cell Line , DNA, Complementary/genetics , Fluorescent Antibody Technique , Forelimb/metabolism , Forelimb/physiology , Humans , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/metabolism , Protein Binding , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Nat Commun ; 8: 15926, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28649987

ABSTRACT

Adipose progenitor cells (APCs) reside in a vascular niche, located within the perivascular compartment of adipose tissue blood vessels. Yet, the signals and mechanisms that govern adipose vascular niche formation and APC niche interaction are unknown. Here we show that the assembly and maintenance of the adipose vascular niche is controlled by PPARγ acting within APCs. PPARγ triggers a molecular hierarchy that induces vascular sprouting, APC vessel niche affinity and APC vessel occupancy. Mechanistically, PPARγ transcriptionally activates PDGFRß and VEGF. APC expression and activation of PDGFRß promotes the recruitment and retention of APCs to the niche. Pharmacologically, targeting PDGFRß disrupts APC niche contact thus blocking adipose tissue expansion. Moreover, enhanced APC expression of VEGF stimulates endothelial cell proliferation and expands the adipose niche. Consequently, APC niche communication and retention are boosted by VEGF thereby impairing adipogenesis. Our data indicate that APCs direct adipose tissue niche expansion via a PPARγ-initiated PDGFRß and VEGF transcriptional axis.


Subject(s)
Adipocytes/metabolism , PPAR gamma/metabolism , Stem Cell Niche , Stem Cells/metabolism , Adipocytes/cytology , Adipogenesis , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Proliferation , Female , Male , Mice , PPAR gamma/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stem Cells/cytology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
PLoS One ; 12(4): e0176190, 2017.
Article in English | MEDLINE | ID: mdl-28441415

ABSTRACT

Pluripotent stem cells (PSC) hold great promise for the treatment of human skeletal muscle diseases. However, it remains challenging to convert PSC to skeletal muscle cells, and the mechanisms by which the master regulatory transcription factor, Pax7, promotes muscle stem (satellite) cell identity are not yet understood. We have taken advantage of PSC-derived skeletal muscle precursor cells (iPax7), wherein the induced expression of Pax7 robustly initiates the muscle program and enables the in vitro generation of precursors that seed the satellite cell compartment upon transplantation. Remarkably, we found that chromatin accessibility in myogenic precursors pre-figures subsequent activation of myogenic differentiation genes. We also found that Pax7 binding is generally restricted to euchromatic regions and excluded from H3K27 tri-methylated regions in muscle cells, suggesting that recruitment of this factor is circumscribed by chromatin state. Further, we show that Pax7 binding induces dramatic, localized remodeling of chromatin characterized by the acquisition of histone marks associated with enhancer activity and induction of chromatin accessibility in both muscle precursors and lineage-committed myoblasts. Conversely, removal of Pax7 leads to rapid reversal of these features on a subset of enhancers. Interestingly, another cluster of Pax7 binding sites is associated with a durably accessible and remodeled chromatin state after removal of Pax7, and persistent enhancer accessibility is associated with subsequent, proximal binding by the muscle regulatory factors, MyoD1 and myogenin. Our studies provide new insights into the epigenetic landscape of skeletal muscle stem cells and precursors and the role of Pax7 in satellite cell specification.


Subject(s)
Chromatin/metabolism , Muscle Cells/metabolism , Muscle Development/physiology , PAX7 Transcription Factor/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Line , Mice , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myogenin/metabolism , PAX7 Transcription Factor/genetics , Satellite Cells, Skeletal Muscle/metabolism
12.
Cell Metab ; 25(1): 166-181, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27889388

ABSTRACT

Cold temperatures induce progenitor cells within white adipose tissue to form beige adipocytes that burn energy and generate heat; this is a potential anti-diabesity therapy. However, the potential to form cold-induced beige adipocytes declines with age. This creates a clinical roadblock to potential therapeutic use in older individuals, who constitute a large percentage of the obesity epidemic. Here we show that aging murine and human beige progenitor cells display a cellular aging, senescence-like phenotype that accounts for their age-dependent failure. Activating the senescence pathway, either genetically or pharmacologically, in young beige progenitors induces premature cellular senescence and blocks their potential to form cold-induced beige adipocytes. Conversely, genetically or pharmacologically reversing cellular aging by targeting the p38/MAPK-p16Ink4a pathway in aged mouse or human beige progenitor cells rejuvenates cold-induced beiging. This in turn increases glucose sensitivity. Collectively, these data indicate that anti-aging or senescence modalities could be a strategy to induce beiging, thereby improving metabolic health in aging humans.


Subject(s)
Adipocytes, Beige/cytology , Adipocytes, Beige/metabolism , Aging/physiology , Cellular Senescence , Cold Temperature , Actins/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Male , Mice, Inbred C57BL , Phenotype , Stem Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Methods Mol Biol ; 1460: 163-79, 2016.
Article in English | MEDLINE | ID: mdl-27492172

ABSTRACT

In response to muscle damage, satellite cells proliferate and undertake both differentiation and self-renewal, generating new functional muscle tissue and repopulating this new muscle with stem cells for future injury responses. For many questions relating to the physiological regulation of satellite cells, quantitative readouts of self-renewal and differentiation can be very useful. There is a particular need for a quantitative assay for satellite cell self-renewal that does not rely solely upon sectioning, staining and counting cells in sections. In this chapter, we provide detailed methods for quantifying the self-renewal and differentiation potential of a given population of satellite cells using an assay involving transplantation into injured, regenerating muscle together with specific markers for donor cell identity and state of differentiation. In particular, using the Pax7-ZsGreen transgene as a marker of satellite cell state, self-renewal can be quantified by FACS on transplanted muscle to actually count the total number of resident satellite cells at time points following transplantation.


Subject(s)
Cell Differentiation , Cell Self Renewal , Flow Cytometry , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Biomarkers , Cardiotoxins/adverse effects , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Self Renewal/drug effects , Cell Self Renewal/radiation effects , Cell Separation/methods , Flow Cytometry/methods , Immunohistochemistry , Immunophenotyping , Mice , Mice, Inbred mdx , Muscle Development , Regeneration , Satellite Cells, Skeletal Muscle/transplantation
14.
Skelet Muscle ; 5: 36, 2015.
Article in English | MEDLINE | ID: mdl-26504514

ABSTRACT

BACKGROUND: Satellite cells (SCs) are indispensable for muscle regeneration and repair; however, due to low frequency in primary muscle and loss of engraftment potential after ex vivo expansion, their use in cell therapy is currently unfeasible. To date, an alternative to this limitation has been the transplantation of SC-derived myogenic progenitor cells (MPCs), although these do not hold the same attractive properties of stem cells, such as self-renewal and long-term regenerative potential. METHODS: We develop a method to expand wild-type and dystrophic fresh isolated satellite cells using transient expression of Pax3. This approach can be combined with genetic correction of dystrophic satellite cells and utilized to promote muscle regeneration when transplanted into dystrophic mice. RESULTS: Here, we show that SCs from wild-type and dystrophic mice can be expanded in culture through transient expression of Pax3, and these expanded activated SCs can regenerate the muscle. We test this approach in a gene therapy model by correcting dystrophic SCs from a mouse lacking dystrophin using a Sleeping Beauty transposon carrying the human µDYSTROPHIN gene. Transplantation of these expanded corrected cells into immune-deficient, dystrophin-deficient mice generated large numbers of dystrophin-expressing myofibers and improved contractile strength. Importantly, in vitro expanded SCs engrafted the SC compartment and could regenerate muscle after secondary injury. CONCLUSION: These results demonstrate that Pax3 is able to promote the ex vivo expansion of SCs while maintaining their stem cell regenerative properties.

15.
Cell Rep ; 8(5): 1484-96, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25176645

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is an enigmatic disease associated with epigenetic alterations in the subtelomeric heterochromatin of the D4Z4 macrosatellite repeat. Each repeat unit encodes DUX4, a gene that is normally silent in most tissues. Besides muscular loss, most patients suffer retinal vascular telangiectasias. To generate an animal model, we introduced a doxycycline-inducible transgene encoding DUX4 and 3' genomic DNA into a euchromatic region of the mouse X chromosome. Without induction, DUX4 RNA was expressed at low levels in many tissues and animals displayed a variety of unexpected dominant leaky phenotypes, including male-specific lethality. Remarkably, rare live-born males expressed DUX4 RNA in the retina and presented a retinal vascular telangiectasia. By using doxycycline to induce DUX4 expression in satellite cells, we observed impaired myogenesis in vitro and in vivo. This mouse model, which shows pathologies due to FSHD-related D4Z4 sequences, is likely to be useful for testing anti-DUX4 therapies in FSHD.


Subject(s)
Genes, Dominant , Genes, X-Linked , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Animals , Cells, Cultured , Disease Models, Animal , Euchromatin/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Transgenic , Muscular Dystrophy, Facioscapulohumeral/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology
16.
Cell Stem Cell ; 12(5): 587-601, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23642367

ABSTRACT

Mesp1 is regarded as the master regulator of cardiovascular development, initiating the cardiac transcription factor cascade to direct the generation of cardiac mesoderm. To define the early embryonic cell population that responds to Mesp1, we performed pulse inductions of gene expression over tight temporal windows following embryonic stem cell differentiation. Remarkably, instead of promoting cardiac differentiation in the initial wave of mesoderm, Mesp1 binds to the Tal1 (Scl) +40 kb enhancer and generates Flk-1+ precursors expressing Etv2 (ER71) and Tal1 that undergo hematopoietic differentiation. The second wave of mesoderm responds to Mesp1 by differentiating into PDGFRα+ precursors that undergo cardiac differentiation. Furthermore, in the absence of serum-derived factors, Mesp1 promotes skeletal myogenic differentiation. Lineage tracing revealed that the majority of yolk sac and many adult hematopoietic cells derive from Mesp1+ precursors. Thus, Mesp1 is a context-dependent determination factor, integrating the stage of differentiation and the signaling environment to specify different lineage outcomes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Heart/embryology , Hematopoietic System/embryology , Mesoderm/embryology , Muscle, Skeletal/embryology , Stem Cells/cytology , Aging/metabolism , Animals , Base Pairing/genetics , Bone Marrow Cells/cytology , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Enhancer Elements, Genetic/genetics , Hematopoiesis , Hematopoietic System/cytology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Inbred mdx , Muscle Development , Muscle, Skeletal/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Binding , Protein Multimerization , Proto-Oncogene Proteins/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Stem Cells/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Time Factors , Transcription Factors/metabolism , Yolk Sac/metabolism
17.
Stem Cells ; 31(8): 1611-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23606600

ABSTRACT

Transplantation of a myogenic cell population into an immunodeficient recipient is an excellent way of assessing the in vivo muscle-generating capacity of that cell population. To facilitate both allogeneic and xenogeneic transplantations of muscle-forming cells in mice, we have developed a novel immunodeficient muscular dystrophy model, the NSG-mdx(4Cv) mouse. The IL2Rg mutation, which is linked to the Dmd gene on the X chromosome, simultaneously depletes NK cells and suppresses thymic lymphomas, issues that limit the utility of the SCID/mdx model. The NSG-mdx(4Cv) mouse presents a muscular dystrophy of similar severity to the conventional mdx mouse. We show that this animal supports robust engraftment of both pig and dog muscle mononuclear cells. The question of whether satellite cells prospectively isolated by flow cytometry can confer a functional benefit upon transplantation has been controversial. Using allogeneic Pax7-ZsGreen donors and NSG-mdx(4Cv) recipients, we demonstrate definitively that as few as 900 FACS-isolated satellite cells can provide functional regeneration in vivo, in the form of an increased mean maximal force-generation capacity in cell-transplanted muscles, compared to a sham-injected control group. These studies highlight the potency of satellite cells to improve muscle function and the utility of the NSG-mdx(4Cv) model for studies on muscle regeneration and Duchenne muscular dystrophy therapy.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Duchenne/surgery , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Dogs , Dystrophin/genetics , Dystrophin/metabolism , Female , Genotype , Heterografts , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Satellite Cells, Skeletal Muscle/cytology , Swine , Transplantation, Homologous
18.
Skelet Muscle ; 2(1): 13, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22703589

ABSTRACT

BACKGROUND: Muscle growth and repair is accomplished by the satellite cell pool, a self-renewing population of myogenic progenitors. Functional heterogeneity within the satellite cell compartment and changes in potential with experimental intervention can be revealed by in vitro colony-forming cell (CFC) assays, however large numbers of colonies need to be assayed to give meaningful data, and manually quantifying nuclei and scoring markers of differentiation is experimentally limiting. METHODS: We present G-Tool, a multiplatform (Java) open-source algorithm that analyzes an ensemble of fluorescent micrographs of satellite cell-derived colonies to provide quantitative and statistically meaningful metrics of myogenic potential, including proliferation capacity and propensity to differentiate. RESULTS: We demonstrate the utility of G-Tool in two applications: first, we quantify the response of satellite cells to oxygen concentration. Compared to 3% oxygen which approximates tissue levels, we find that 21% oxygen, the ambient level, markedly limits the proliferative potential of transit amplifying progeny but at the same time inhibits the rate of terminal myogenic differentiation. We also test whether satellite cells from different muscles have intrinsic differences that can be read out in vitro. Compared to masseter, dorsi, forelimb and hindlimb muscles, we find that the diaphragm satellite cells have significantly increased proliferative potential and a reduced propensity to spontaneously differentiate. These features may be related to the unique always-active status of the diaphragm. CONCLUSIONS: G-Tool facilitates consistent and reproducible CFC analysis between experiments and individuals. It is released under an open-source license that enables further development by interested members of the community.

19.
Cell Stem Cell ; 10(5): 610-9, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22560081

ABSTRACT

A major obstacle in the application of cell-based therapies for the treatment of neuromuscular disorders is obtaining the appropriate number of stem/progenitor cells to produce effective engraftment. The use of embryonic stem (ES) or induced pluripotent stem (iPS) cells could overcome this hurdle. However, to date, derivation of engraftable skeletal muscle precursors that can restore muscle function from human pluripotent cells has not been achieved. Here we applied conditional expression of PAX7 in human ES/iPS cells to successfully derive large quantities of myogenic precursors, which, upon transplantation into dystrophic muscle, are able to engraft efficiently, producing abundant human-derived DYSTROPHIN-positive myofibers that exhibit superior strength. Importantly, transplanted cells also seed the muscle satellite cell compartment, and engraftment is present over 11 months posttransplant. This study provides the proof of principle for the derivation of functional skeletal myogenic progenitors from human ES/iPS cells and highlights their potential for future therapeutic application in muscular dystrophies.


Subject(s)
Dystrophin/metabolism , Embryonic Stem Cells/physiology , Induced Pluripotent Stem Cells/physiology , Muscular Dystrophies/therapy , Satellite Cells, Skeletal Muscle/physiology , Animals , Cell Proliferation , Feasibility Studies , Humans , Mice , Mice, Inbred mdx , Muscle Contraction , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Stem Cell Transplantation , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...