Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Curr Microbiol ; 77(8): 1448-1456, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32198535

ABSTRACT

Coix lachrymal-jobi L. var. ma-yuen Stapf of Gramineae are annual or perennial herbs and an important food-medicine homologous plants of high value in nutrition, health protection, and comprehensive utilization. In recent years, the revival of researches on its roles in food and medicinal applications of this underutilized grass for food security and economic empowerment of rural communities has been seen . In this research, Coix kernel endophytic fungi were isolated and identified by fungal colony morphology observation combined with the PCR-amplified fungal internal transcribed spacer (ITS) sequence analyses. All together six isolates to five species of Coix endophytic fungi and two isolates to the genus level were identified from the kernels of six Coix cultivars: Penicillium expansum, Penicillium polonicum, Cladosporium cladosporioides, Alternaria alternata, Aspergillus flavus, and two genera of Aspergillus and Fusarium. Potential benefits and harms analyses showed that Penicillium expansum, Aspergillus oryzae, and Cladosporium cladosporioides can produce a variety of beneficial composite enzymes and have an extensive application in microbial chemistry, food science, and fermentation, whereas Penicillium, Aspergillus flavus, Alternaria alternate, and Fusarium can produce corresponding toxins harmful to plants, animals, and humans. These results not only provided a basis for the targeted prevention of contamination in the tissue culture of Coix kernels by the addition of specific antibiotics, but also enriched the endophytic fungi resource pool of Gramineae crops and suggested new ideas for the improvement, cultivation, post-harvest seeds/kernels storage, and the development of new natural drugs.


Subject(s)
Coix/microbiology , Fungi/classification , Seeds/microbiology , Aspergillus/classification , Aspergillus/isolation & purification , China , Cladosporium/classification , Cladosporium/isolation & purification , DNA, Fungal/genetics , DNA, Intergenic/genetics , Endophytes/classification , Endophytes/isolation & purification , Fungi/isolation & purification , Genome, Fungal , Penicillium/classification , Penicillium/isolation & purification
2.
Clin Orthop Relat Res ; 473(9): 2908-19, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26070773

ABSTRACT

BACKGROUND: Extremity trauma is the most common injury seen in combat hospitals as well as in civilian trauma centers. Major skeletal muscle injuries that are complicated by ischemia often result in substantial muscle loss, residual disability, or even amputation, yet few treatment options are available. A therapy that would increase skeletal muscle tolerance to hypoxic damage could reduce acute myocyte loss and enhance preservation of muscle mass in these situations. QUESTIONS/PURPOSES: In these experiments, we investigated (1) whether cobalt protoporphyrin (CoPP), a pharmacologic inducer of cytoprotective heme oxygenase-1 (HO-1), would upregulate HO-1 expression and activity in skeletal muscle, tested in muscle-derived stem cells (MDSCs); and (2) whether CoPP exposure would protect MDSCs from cell death during in vitro hypoxia/reoxygenation. Then, using an in vivo mouse model of hindlimb ischemia/reperfusion injury, we examined (3) whether CoPP pharmacotherapy would reduce skeletal muscle damage when delivered after injury; and (4) whether it would alter the host inflammatory response to injury. METHODS: MDSCs were exposed in vitro to a single dose of 25 µΜ CoPP and harvested over 24 to 96 hours, assessing HO-1 protein expression by Western blot densitometry and HO-1 enzyme activity by cGMP levels. To generate hypoxia/reoxygenation stress, MDSCs were treated in vitro with phosphate-buffered saline (vehicle), CoPP, or CoPP plus an HO-1 inhibitor, tin protoporphyrin (SnPP), and then subjected to 5 hours of hypoxia (< 0.5% O2) followed by 24 hours of reoxygenation and evaluated for apoptosis. In vivo, hindlimb ischemia/reperfusion injury was produced in mice by unilateral 2-hour tourniquet application followed by 24 hours of reperfusion. In three postinjury treatment groups (n = 7 mice/group), CoPP was administered intraperitoneally during ischemia, at the onset of reperfusion, or 1 hour later. Two control groups of mice with the same injury received phosphate-buffered saline (vehicle) or the HO-1 inhibitor, SnPP. Myocyte damage in the gastrocnemius and tibialis anterior muscles was determined by uptake of intraperitoneally delivered Evans blue dye (EBD), quantified by image analysis. On serial sections, inflammation was gauged by the mean myeloperoxidase staining intensity per unit area over the entirety of each muscle. RESULTS: In MDSCs, a single exposure to CoPP increased HO-1 protein expression and enzyme activity, both of which were sustained for 96 hours. CoPP treatment of MDSCs reduced apoptotic cell populations by 55% after in vitro hypoxia/reoxygenation injury (from a mean of 57.3% apoptotic cells in vehicle-treated controls to 25.7% in CoPP-treated cells, mean difference 31.6%; confidence interval [CI], 28.1-35.0; p < 0.001). In the hindlimb ischemia/reperfusion model, CoPP delivered during ischemia produced a 38% reduction in myocyte damage in the gastrocnemius muscle (from 86.4% ± 7% EBD(+) myofibers in vehicle-treated, injured controls to 53.2% EBD(+) in CoPP-treated muscle, mean difference 33.2%; 95% CI, 18.3, 48.4; p < 0.001). A 30% reduction in injury to the gastrocnemius was seen with drug delivery at the onset of reperfusion (to 60.6% ± 13% EBD(+) with CoPP treatment, mean difference 25.8%; CI, 12.2-39.4; p < 0.001). In the tibialis anterior, however, myocyte damage was decreased only when CoPP was given at the onset of reperfusion, resulting in a 27% reduction in injury (from 78.8% ± 8% EBD(+) myofibers in injured controls to 58.3% ± 14% with CoPP treatment, mean difference 20.5%; CI, 6.1-35.0; p = 0.004). Delaying CoPP delivery until 1 hour after tourniquet release obviated the protective effect in both muscles. Mean MPO staining intensity per unit area, indicating the host inflammatory response, decreased by 27-34% across both the gastrocnemius and tibialis anterior muscles when CoPP was given either during ischemia or at the time of reperfusion. Delaying drug delivery until 1 hour after the start of reperfusion abrogated this antiinflammatory effect. CONCLUSIONS: CoPP can decrease skeletal muscle damage when given early in the course of ischemia/reperfusion injury and also provide protection for regenerative stem cell populations. CLINICAL RELEVANCE: Pharmacotherapy with HO-1 inducers, delivered in the field, on hospital arrival, or during trauma surgery, may improve preservation of muscle mass and muscle-inherent stem cells after severe ischemic limb injury.


Subject(s)
Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Protoporphyrins/pharmacology , Reperfusion Injury/prevention & control , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Cytoprotection , Disease Models, Animal , Enzyme Induction , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/biosynthesis , Hindlimb , Inflammation Mediators/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Regeneration/drug effects , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Stem Cells/enzymology , Stem Cells/pathology , Time Factors
3.
Stem Cells Transl Med ; 3(6): 734-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24736402

ABSTRACT

Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) can regenerate infarcted myocardium. However, when implanted into acutely infarcted hearts, few cells survive the first week postimplant. To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). When hESC-CMs were challenged with an in vitro hypoxia/reoxygenation injury, mimicking cell transplantation into an ischemic site, survival was significantly greater among cells pretreated with CoPP versus phosphate-buffered saline (PBS)-pretreated controls. Compared with PBS-pretreated cells, CoPP-pretreated hESC-CM preparations exhibited higher levels of HO-1 expression, Akt phosphorylation, and vascular endothelial growth factor production, with reduced apoptosis, and a 30% decrease in intracellular reactive oxygen species. For in vivo translation, 1 × 10(7) hESC-CMs were pretreated ex vivo with CoPP or PBS and then injected intramyocardially into rat hearts immediately following acute infarction (permanent coronary ligation). At 1 week, hESC-CM content, assessed by quantitative polymerase chain reaction for human Alu sequences, was 17-fold higher in hearts receiving CoPP- than PBS-pretreated cells. On histomorphometry, cardiomyocyte graft size was 2.6-fold larger in hearts receiving CoPP- than PBS-pretreated cells, occupying up to 12% of the ventricular area. Vascular density of host-perfused human-derived capillaries was significantly greater in grafts composed of CoPP- than PBS-pretreated cells. Taken together, these experiments demonstrate that ex vivo pretreatment of hESC-CMs with a single dose of CoPP before intramyocardial implantation more than doubled resulting graft size and improved early graft vascularization in acutely infarcted hearts. These findings open the door for delivery of these, or other, stem cells during acute interventional therapy following myocardial infarction or ischemia.


Subject(s)
Embryonic Stem Cells/drug effects , Embryonic Stem Cells/transplantation , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/transplantation , Protoporphyrins/pharmacology , Regeneration , Animals , Apoptosis/drug effects , Cell Differentiation , Cell Line , Cell Proliferation , Disease Models, Animal , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/pathology , Enzyme Induction , Female , Graft Survival , Heme Oxygenase-1/biosynthesis , Humans , Male , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Nude , Reactive Oxygen Species/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism
4.
Mol Biol Cell ; 20(1): 509-20, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19005220

ABSTRACT

Stem cells are classically defined by their multipotent, long-term proliferation, and self-renewal capabilities. Here, we show that increased antioxidant capacity represents an additional functional characteristic of muscle-derived stem cells (MDSCs). Seeking to understand the superior regenerative capacity of MDSCs compared with myoblasts in cardiac and skeletal muscle transplantation, our group hypothesized that survival of the oxidative and inflammatory stress inherent to transplantation may play an important role. Evidence of increased enzymatic and nonenzymatic antioxidant capacity of MDSCs were observed in terms of higher levels of superoxide dismutase and glutathione, which appears to confer a differentiation and survival advantage. Further when glutathione levels of the MDSCs are lowered to that of myoblasts, the transplantation advantage of MDSCs over myoblasts is lost when transplanted into both skeletal and cardiac muscles. These findings elucidate an important cause for the superior regenerative capacity of MDSCs, and provide functional evidence for the emerging role of antioxidant capacity as a critical property for MDSC survival post-transplantation.


Subject(s)
Antioxidants/metabolism , Muscle, Skeletal/cytology , Myoblasts/physiology , Myocytes, Cardiac/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Cell Death/physiology , Cell Differentiation/physiology , Hydrogen Peroxide/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Myoblasts/cytology , Myocytes, Cardiac/cytology , Oxidants/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Stem Cells/cytology , Tumor Necrosis Factor-alpha/metabolism
5.
Nat Biotechnol ; 25(9): 1025-34, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17767154

ABSTRACT

We document anatomic, molecular and developmental relationships between endothelial and myogenic cells within human skeletal muscle. Cells coexpressing myogenic and endothelial cell markers (CD56, CD34, CD144) were identified by immunohistochemistry and flow cytometry. These myoendothelial cells regenerate myofibers in the injured skeletal muscle of severe combined immunodeficiency mice more effectively than CD56+ myogenic progenitors. They proliferate long term, retain a normal karyotype, are not tumorigenic and survive better under oxidative stress than CD56+ myogenic cells. Clonally derived myoendothelial cells differentiate into myogenic, osteogenic and chondrogenic cells in culture. Myoendothelial cells are amenable to biotechnological handling, including purification by flow cytometry and long-term expansion in vitro, and may have potential for the treatment of human muscle disease.


Subject(s)
Endothelial Cells/cytology , Muscle, Skeletal/cytology , Adolescent , Adult , Aged , Animals , Biomarkers/metabolism , CD56 Antigen , Cell Proliferation , Cell Survival , Cells, Cultured , Child , Clone Cells , Flow Cytometry , Humans , Mice , Mice, SCID , Middle Aged , Muscle, Skeletal/physiology , Neoplasms/pathology , Regeneration , Time Factors
6.
J Cell Biol ; 177(1): 73-86, 2007 Apr 09.
Article in English | MEDLINE | ID: mdl-17420291

ABSTRACT

We have shown that muscle-derived stem cells (MDSCs) transplanted into dystrophic (mdx) mice efficiently regenerate skeletal muscle. However, MDSC populations exhibit heterogeneity in marker profiles and variability in regeneration abilities. We show here that cell sex is a variable that considerably influences MDSCs' regeneration abilities. We found that the female MDSCs (F-MDSCs) regenerated skeletal muscle more efficiently. Despite using additional isolation techniques and cell cloning, we could not obtain a male subfraction with a regeneration capacity similar to that of their female counterparts. Rather than being directly hormonal or caused by host immune response, this difference in MDSCs' regeneration potential may arise from innate sex-related differences in the cells' stress responses. In comparison with F-MDSCs, male MDSCs have increased differentiation after exposure to oxidative stress induced by hydrogen peroxide, which may lead to in vivo donor cell depletion, and a proliferative advantage for F-MDSCs that eventually increases muscle regeneration. These findings should persuade researchers to report cell sex, which is a largely unexplored variable, and consider the implications of relying on cells of one sex.


Subject(s)
Muscle, Skeletal/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Cell Differentiation , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/cytology , Oligonucleotide Array Sequence Analysis , Regeneration/genetics , Sex Factors , Stem Cell Transplantation , Stem Cells/classification
7.
Tissue Eng ; 12(7): 1891-901, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16889519

ABSTRACT

Human, rat, and mouse studies have demonstrated the existence of a population of adipose-derived adult stem (ADAS) cells that can undergo multilineage differentiation in vitro. However, it remains unclear whether these cells maintain their multilineage potential in vivo. The aim of this study was to examine the in vitro and in vivo characteristics and behavior of a potential population of murine ADAS (muADAS) cells isolated from the visceral fat of the abdominal cavity of C57BL/10J mice. We used flow cytometry to examine the cells' expression of CD29, CD31, CD45, CD34, CD44, CD144, CD146, Flk1, and Sca-1. The isolated cell population was CD45 negative, which precludes contamination by hematopoietic cells, but was partially positive for Sca-1 and CD34: 2 stem-cell markers. After induction in conditioned medium, the muADAS cells gained the ability to undergo adipogenic, osteogenic, chondrogenic, myogenic, and hematopoietic differentiation in vitro. The muADAS cells readily differentiated to form bone and cartilage in vivo for up to 24 weeks, but their ability to regenerate muscle or reconstitute bone marrow was found to be limited.


Subject(s)
Adipose Tissue/physiology , Cell Differentiation/physiology , Chondrogenesis/physiology , Osteogenesis/physiology , Stem Cells/physiology , Adipose Tissue/cytology , Animals , Antigens, CD/metabolism , Cells, Cultured , Mice , Mice, SCID , Stem Cells/cytology , Time Factors
9.
J Gene Med ; 6(2): 155-65, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14978769

ABSTRACT

BACKGROUND: Adenoviral gene transfer to adult skeletal muscle is hindered by several major limitations, including host immune responses and maturation-dependent loss of myofiber infectivity. Ex vivo gene delivery is more efficient than direct viral injection in surmounting maturation-dependent adenoviral transduction. Here we investigated the use of helper cells to improve the efficiency of ex vivo gene transfer to adult mouse skeletal muscle. METHODS: New producer cells carrying the E1 gene of adenovirus type 5 (E32 cells) were developed using primary myoblasts from mdx mice. The E32 cells and 293 cells were infected with an E1-deleted first-generation adenovirus carrying the LacZ gene. These transduced helper cells were injected into the skeletal muscle of adult mdx and SCID mice. RESULTS: LacZ-positive mature myofibers were detected in the skeletal muscle of adult mice sacrificed 5 days post-injection. The gene transfer efficiency using 293 cells and E32 cells was 6.2 and 3.6 times higher than myoblast-mediated gene transfer, respectively. Ex vivo gene transfer of these cell types led to a better outcome than did direct adenoviral injection. CONCLUSIONS: We achieved more efficient adenoviral gene transduction by using 293 and E32 helper cells than by myoblast-mediated gene transfer and direct viral injection. These helper cells also enabled adenoviral gene transfer to mature myofibers. The mechanisms by which this method facilitated adenoviral gene transfer to mature myofibers remains unclear; however, we hypothesize that the in vivo occurrence of cytopathic effects (CPE) in the transduced 293 and E32 helper cell populations facilitated the improved adenoviral transduction of myofibers.


Subject(s)
Adenoviridae , Genetic Therapy , Genetic Vectors , Helper Viruses , Muscle, Skeletal/metabolism , Animals , Cell Fusion , Genes, Reporter , In Vitro Techniques , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/therapy
10.
Methods Mol Biol ; 246: 301-8, 2004.
Article in English | MEDLINE | ID: mdl-14970600

ABSTRACT

Type 1 herpes simplex virus (HSV-1)-based vectors, which are naturally capable of carrying large DNA fragments like the 14 kb dystrophin cDNA, have been studied for their ability to transduce muscle cells. These vectors can persist in the host cell in a nonintegrated state and can be prepared at adequately high titers (10(7)-10(9) PFU/mL). They also infect myoblasts, myotubes, and immature myofibers efficiently. The major disadvantage of the first-generation HSV vectors is their relatively high cytotoxicity, which hampers long-term transgene expression. Second-generation mutants defective for multiple immediate early (IE) genes (e.g., ICP4, ICP22, and ICP27) display substantially reduced cytotoxicity in vitro, which improves the duration of transgene expression (6-11). In this chapter, we describe a new method of gene delivery using second-generation HSV-1 vectors. This procedure should enable an investigator to transduce normal mouse muscle cells, both in vitro and in vivo. We explain the conditions for muscle cell isolation, transduction in vitro and in vivo, and the technique for evaluating transduction efficiency (beta-galactosidase; beta-gal) using histology or the beta-gal assay (ONPG) method.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Muscle, Skeletal/metabolism , Simplexvirus/genetics , Animals , Mice , Mice, Inbred C57BL , Transduction, Genetic
11.
Cell Cycle ; 3(2): 104-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14712064

ABSTRACT

Researchers have identified 2 types of stem cells in skeletal muscle: satellite cells and multipotent stem cells (MPSCs). The latter category includes different cell populations isolated by various researchers using several techniques. The methods used to isolate these cells appear to influence the stem cell characteristics of the MPSCs. Although MPSCs and satellite cells could represent different stages of maturation of the same progenitor cells, they also could represent distinct populations of stem cells that exist in skeletal muscle. This article summarizes the recent developments in muscle-derived stem cell research.


Subject(s)
Cell Differentiation/physiology , Multipotent Stem Cells/cytology , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Animals , Blood Vessels/cytology , Bone Marrow Cells/cytology , Cell Separation , Mice , Muscle, Skeletal/physiology , Regeneration/physiology , Stem Cell Transplantation
12.
Birth Defects Res C Embryo Today ; 69(3): 230-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14671776

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating X-linked muscle disease characterized by progressive muscle weakness caused by the lack of dystrophin expression at the sarcolemma of muscle fibers. Although various approaches to delivering dystrophin in dystrophic muscle have been investigated extensively (e.g., cell and gene therapy), there is still no treatment that alleviates the muscle weakness in this common inherited muscle disease. The transplantation of myoblasts can enable transient delivery of dystrophin and improve the strength of injected dystrophic muscle, but this approach has various limitations, including immune rejection, poor cellular survival rates, and the limited spread of the injected cells. The isolation of muscle cells that can overcome these limitations would enhance the success of myoblast transplantation significantly. The efficiency of cell transplantation might be improved through the use of stem cells, which display unique features, including (1) self-renewal with production of progeny, (2) appearance early in development and persistence throughout life, and (3) long-term proliferation and multipotency. For these reasons, the development of muscle stem cells for use in transplantation or gene transfer (ex vivo approach) as treatment for patients with muscle disorders has become more attractive in the past few years. In this paper, we review the current knowledge regarding the isolation and characterization of stem cells isolated from skeletal muscle by highlighting their biological features and their relationship to satellite cells as well as other populations of stem cells derived from other tissues. We also describe the remarkable ability of stem cells to regenerate skeletal muscle and their potential use to alleviate the muscle weakness associated with DMD.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Muscles/cytology , Muscular Dystrophy, Duchenne/therapy , Regeneration , Stem Cells/physiology , Animals , Cell Differentiation , Cell Transplantation , Cells, Cultured , Dystrophin/physiology , Genetic Therapy/methods , Humans , Models, Biological , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/pathology
13.
Hum Gene Ther ; 14(16): 1535-46, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14577915

ABSTRACT

Duchenne's muscular dystrophy (DMD) is a lethal muscle disease caused by a lack of dystrophin expression at the sarcolemma of muscle fibers. We investigated retroviral vector delivery of dystrophin in dystrophin-deficient DMD(mdx) (hereafter referred to as mdx) mice via an ex vivo approach using mdx muscle-derived stem cells (MDSCs). We generated a retrovirus carrying a functional human mini-dystrophin (RetroDys3999) and used it to stably transduce mdx MDSCs obtained by the preplate technique (MD3999). These MD3999 cells expressed dystrophin and continued to express stem cell markers, including CD34 and Sca-1. MD3999 cells injected into mdx mouse skeletal muscle were able to deliver dystrophin. Though a relatively low number of dystrophin-positive myofibers was generated within the gastrocnemius muscle, these fibers persisted for up to 24 weeks postinjection. The injection of cells from additional MDSC/Dys3999 clones into mdx skeletal muscle resulted in varying numbers of dystrophin-positive myofibers, suggesting a differential regenerating capacity among the clones. At 2 and 4 weeks postinjection, the infiltration of CD4- and CD8-positive lymphocytes and a variety of cytokines was detected within the injected site. These data suggest that the transplantation of retrovirally transduced mdx MDSCs can enable persistent dystrophin restoration in mdx skeletal muscle; however, the differential regenerating capacity observed among the MDSC/Dys3999 clones and the postinjection immune response are potential challenges facing this technology.


Subject(s)
Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Stem Cell Transplantation , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Transplantation , Cells, Cultured , Dystrophin/deficiency , Gene Expression , Genetic Therapy , Injections, Intramuscular , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/immunology , Retroviridae/genetics , Transplantation, Isogeneic
14.
Nat Cell Biol ; 5(7): 640-6, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12792651

ABSTRACT

Muscle-derived stem cells (MDSCs) can differentiate into multiple lineages, including haematopoietic lineages. However, it is unknown whether MDSCs preserve their myogenic potential after differentiation into other lineages. To address this issue, we isolated from dystrophic muscle a population of MDSCs that express stem-cell markers and can differentiate into various lineages. After systemic delivery of three MDSC clones into lethally irradiated mice, we found that differentiation of the donor cells into various lineages of the haematopoietic system resulted in repopulation of the recipients' bone marrow. Donor-derived bone-marrow cells, isolated from these recipients by fluorescence-activated cell sorting (FACS), also repopulated the bone marrow of secondary, lethally irradiated, recipients and differentiated into myogenic cells both in vitro and in vivo in normal mdx mice. These findings demonstrate that MDSC clones retain their myogenic potential after haematopoietic differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Clone Cells/cytology , Hematopoietic Stem Cells/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Stem Cells/cytology , Animals , Biomarkers , Bone Marrow Cells/radiation effects , Cells, Cultured , Clone Cells/transplantation , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Myoblasts/cytology , Myoblasts/physiology , Stem Cells/physiology
15.
J Cell Sci ; 115(Pt 22): 4361-74, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12376567

ABSTRACT

Characterization of myogenic subpopulations has traditionally been performed independently of their functional performance following transplantation. Using the preplate technique, which separates cells based on their variable adhesion characteristics, we investigated the use of cell surface proteins to potentially identify progenitors with enhanced regeneration capabilities. Based on previous studies, we used cell sorting to investigate stem cell antigen-1 (Sca-1) and CD34 expression on myogenic populations with late adhesion characteristics. We compared the regeneration efficiency of these sorted progenitors, as well as those displaying early adhesion characteristics, by quantifying their ability to regenerate skeletal muscle and restore dystrophin following transplantation into allogenic dystrophic host muscle. Identification and utilization of late adhering populations based on CD34 expression led to differential regeneration, with CD34-positive populations exhibiting significant improvements in dystrophin restoration compared with both their CD34-negative counterparts and early adhering cell populations. Regenerative capacity was found to correspond to the level of myogenic commitment, defined by myogenic regulatory factor expression, and the rate and degree of induced cell differentiation and fusion. These results demonstrate the ability to separate definable subpopulations of myogenic progenitors based on CD34 expression and reveal the potential implications of defining myogenic cell behavioral and phenotypic characteristics in relation to their regenerative capacity in vivo.


Subject(s)
Antigens, Surface/metabolism , Membrane Fusion/physiology , Muscle, Skeletal/growth & development , Myoblasts/metabolism , Regeneration/physiology , Sarcolemma/metabolism , Animals , Antigens, CD34/immunology , Antigens, CD34/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , Antigens, Surface/immunology , Cell Adhesion/immunology , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Separation/methods , Cells, Cultured , Dystrophin/biosynthesis , Dystrophin/deficiency , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscular Dystrophies/therapy , Myoblasts/cytology , Myoblasts/transplantation , Myogenic Regulatory Factors/metabolism , Phenotype , Sarcolemma/immunology , Tissue Transplantation/methods
16.
Microsc Res Tech ; 58(1): 45-51, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12112422

ABSTRACT

Transfer of therapeutic genes into muscle tissue has promise for the treatment of a variety of muscular dystrophies. Various vectors have been used to deliver genes to skeletal muscle but their application has faced several major limitations including: (1) the lack of transgene persistence caused by the immune rejection of transduced myofibers and/or vector toxicity, and (2) the maturation dependence of viral transduction. While the immunorejection and/or cytotoxic problems are being overcome with the development of new vectors, maturation-dependent viral transduction is still a major hurdle in gene transfer to skeletal muscle. Poor adenoviral transduction in mature myofibers has been attributed to: (1) the extracellular matrix of mature myofibers may form a physical barrier and prevent the passage of large viral particles; (2) viral receptors are down-regulated with muscle maturation; and (3) loss of myoblasts with muscle maturation, which serve as intermediaries in the viral transduction. In this review, we will focus on recent developments in overcoming those hurdles of gene therapy in skeletal muscle, especially to adenovirus (Ad), including: (1) new mutant vectors lacking all viral genes to decrease immunogenicity, and hence, improve persistence of transgene expression in muscle in vivo; (2) using tissue specific promoters to evade immunorejection; (3) permeabilization of the extracellular matrix; (4) modifying the viral receptors in mature myofibers; and (5) myoblast or muscle stem cell mediated ex vivo gene transfer.


Subject(s)
Adenoviridae/genetics , Muscle, Skeletal/metabolism , Transduction, Genetic/methods , Transgenes/genetics , Animals , Extracellular Matrix/metabolism , Genetic Vectors/genetics , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/immunology , Organ Specificity , Receptors, Virus/metabolism , Stem Cells/metabolism
17.
J Cell Biol ; 157(5): 851-64, 2002 May 27.
Article in English | MEDLINE | ID: mdl-12021255

ABSTRACT

Three populations of myogenic cells were isolated from normal mouse skeletal muscle based on their adhesion characteristics and proliferation behaviors. Although two of these populations displayed satellite cell characteristics, a third population of long-time proliferating cells expressing hematopoietic stem cell markers was also identified. This third population comprises cells that retain their phenotype for more than 30 passages with normal karyotype and can differentiate into muscle, neural, and endothelial lineages both in vitro and in vivo. In contrast to the other two populations of myogenic cells, the transplantation of the long-time proliferating cells improved the efficiency of muscle regeneration and dystrophin delivery to dystrophic muscle. The long-time proliferating cells' ability to proliferate in vivo for an extended period of time, combined with their strong capacity for self-renewal, their multipotent differentiation, and their immune-privileged behavior, reveals, at least in part, the basis for the improvement of cell transplantation. Our results suggest that this novel population of muscle-derived stem cells will significantly improve muscle cell-mediated therapies.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration/physiology , Stem Cell Transplantation , Stem Cells/cytology , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Separation , Dystrophin/physiology , Endothelial Growth Factors/pharmacology , Hematopoietic Stem Cell Transplantation , In Vitro Techniques , Lymphokines/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/immunology , Muscular Dystrophy, Animal/pathology , Nerve Growth Factor/pharmacology , Stem Cells/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...