Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 74: 64-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24960579

ABSTRACT

Over 4 million individuals in the United States, and over 140 million individuals worldwide, are exposed daily to arsenic-contaminated drinking water. Human exposures can range from below the current limit of 10 µg/L to over 1mg/L, with 100 µg/L promoting disease in a large portion of those exposed. Although increased attention has recently been paid to myopathy following arsenic exposure, the pathogenic mechanisms underlying clinical symptoms remain poorly understood. This study tested the hypothesis that arsenic induces lasting muscle mitochondrial dysfunction and impairs metabolism. Compared to nonexposed controls, mice exposed to drinking water containing 100 µg/L arsenite for 5 weeks demonstrated impaired muscle function, mitochondrial myopathy, and altered oxygen consumption that were concomitant with increased mitochondrial fusion gene transcription. There were no differences in the levels of inorganic arsenic or its monomethyl and dimethyl metabolites between controls and exposed muscles, confirming that arsenic does not accumulate in muscle. Nevertheless, muscle progenitor cells isolated from exposed mice recapitulated the aberrant myofiber phenotype and were more resistant to oxidative stress, generated more reactive oxygen species, and displayed autophagic mitochondrial morphology, compared to cells isolated from nonexposed mice. These pathological changes from a possible maladaptive oxidative stress response provide insight into declines in muscle functioning caused by exposure to this common environmental contaminant.


Subject(s)
Arsenic/toxicity , Energy Metabolism/drug effects , Muscle, Skeletal/drug effects , Muscular Diseases/chemically induced , Myofibrils/pathology , Stem Cells/drug effects , Animals , Autophagy , Cells, Cultured , Environmental Exposure/adverse effects , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Muscular Diseases/metabolism , Oxidative Stress , Phenotype , Reactive Oxygen Species/metabolism , Stem Cells/metabolism , Stem Cells/ultrastructure
2.
Sci Rep ; 4: 4826, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24769917

ABSTRACT

Understanding stem cell (SC) population dynamics is essential for developing models that can be used in basic science and medicine, to aid in predicting cells fate. These models can be used as tools e.g. in studying patho-physiological events at the cellular and tissue level, predicting (mal)functions along the developmental course, and personalized regenerative medicine. Using time-lapsed imaging and statistical tools, we show that the dynamics of SC populations involve a heterogeneous structure consisting of multiple sub-population behaviors. Using non-Gaussian statistical approaches, we identify the co-existence of fast and slow dividing subpopulations, and quiescent cells, in stem cells from three species. The mathematical analysis also shows that, instead of developing independently, SCs exhibit a time-dependent fractal behavior as they interact with each other through molecular and tactile signals. These findings suggest that more sophisticated models of SC dynamics should view SC populations as a collective and avoid the simplifying homogeneity assumption by accounting for the presence of more than one dividing sub-population, and their multi-fractal characteristics.


Subject(s)
Models, Biological , Models, Statistical , Stem Cells/cytology , Stem Cells/physiology , Animals , Cell Division , Cell Proliferation , Humans , Mice , Rats , Time-Lapse Imaging
3.
J Orthop Res ; 31(7): 1089-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23553740

ABSTRACT

We have previously reported the high regenerative potential of murine muscle-derived stem cells (mMDSCs) that are capable of differentiating into multiple mesodermal cell lineages, including myogenic, endothelial, chondrocytic, and osteoblastic cells. Recently, we described a putative human counterpart of mMDSCs, the myogenic endothelial cells (MECs), in adult human skeletal muscle, which efficiently repair/regenerate the injured and dystrophic skeletal muscle as well as the ischemic heart in animal disease models. Nevertheless it remained unclear whether human MECs, at the clonal level, preserve mMDSC-like chondrogenic and osteogenic potentials and classic stem cell characteristics including high proliferation and resistance to stress. Herein, we demonstrated that MECs, sorted from fresh postnatal human skeletal muscle biopsies, can be grown clonally and exhibit robust resistance to oxidative stress with no tumorigeneity. MEC clones were capable of differentiating into chondrocytes and osteoblasts under inductive conditions in vitro and participated in cartilage and bone formation in vivo. Additionally, adipogenic and angiogenic potentials of clonal MECs (cMECs) were observed. Overall, our study showed that cMECs not only display typical properties of adult stem cells but also exhibit chondrogenic and osteogenic capacities in vitro and in vivo, suggesting their potential applications in articular cartilage and bone repair/regeneration.


Subject(s)
Cell Differentiation/physiology , Chondrogenesis/physiology , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Osteogenesis/physiology , Adipocytes/cytology , Adult , Animals , Biopsy , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Endothelium/cytology , Endothelium/physiology , Humans , In Vitro Techniques , Male , Mice , Mice, SCID , Osteoblasts/cytology , Oxidative Stress/physiology , Transplantation, Heterologous
4.
PLoS One ; 8(3): e54922, 2013.
Article in English | MEDLINE | ID: mdl-23526927

ABSTRACT

Cellular therapy is a potential approach to improve the regenerative capacity of damaged or diseased skeletal muscle. However, its clinical use has often been limited by impaired donor cell survival, proliferation and differentiation following transplantation. Additionally, functional improvements after transplantation are all-too-often negligible. Because the host microenvironment plays an important role in the fate of transplanted cells, methods to modulate the microenvironment and guide donor cell behavior are warranted. The purpose of this study was to investigate whether the use of neuromuscular electrical stimulation (NMES) for 1 or 4 weeks following muscle-derived stem cell (MDSC) transplantation into dystrophic skeletal muscle can modulate the fate of donor cells and enhance their contribution to muscle regeneration and functional improvements. Animals submitted to 4 weeks of NMES after transplantation demonstrated a 2-fold increase in the number of dystrophin+ myofibers as compared to control transplanted muscles. These findings were concomitant with an increased vascularity in the MDSC+NMES group when compared to non-stimulated counterparts. Additionally, animals subjected to NMES (with or without MDSC transplantation) presented an increased maximal specific tetanic force when compared to controls. Although cell transplantation and/or the use of NMES resulted in no changes in fatigue resistance, the combination of both MDSC transplantation and NMES resulted in a faster recovery from fatigue, when compared to non-injected and non-stimulated counterparts. We conclude that NMES is a viable method to improve MDSC engraftment, enhance dystrophic muscle strength, and, in combination with MDSC transplantation, improve recovery from fatigue. These findings suggest that NMES may be a clinically-relevant adjunct approach for cell transplantation into skeletal muscle.


Subject(s)
Electric Stimulation Therapy/methods , Muscular Dystrophy, Animal/therapy , Myoblasts, Skeletal/transplantation , Animals , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Development , Muscle Strength , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Neuromuscular Junction/physiopathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Regeneration , Stem Cell Niche
5.
Tissue Eng Part A ; 18(3-4): 232-41, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21854253

ABSTRACT

Muscle-derived stem cells (MDSCs) isolated from murine skeletal tissue by the preplate method have displayed the capability to commit to the myogenic lineage and regenerate more efficiently than myoblasts in skeletal and cardiac muscle in murine Duchenne Muscular Dystrophy mice (mdx). However, until now, these studies have not been translated to human muscle cells. Here, we describe the isolation, by a preplate technique, of candidate human MDSCs, which exhibit myogenic and regenerative characteristics similar to their murine counterparts. Using the preplate isolation method, we compared cells that adhere faster to the flasks, preplate 2 (PP2), and cells that adhere slower, preplate 6 (PP6). The human PP6 cells express several markers of mesenchymal stem cells and are distinct from human PP2 (a myoblast-like population) based on their expression of CD146 and myogenic markers desmin and CD56. After transplantation to the gastrocnemius muscle of mdx/SCID mice, we observe significantly higher levels of PP6 cells participating in muscle regeneration as compared with the transplantation of PP2 cells. This study supports some previous findings related to mouse preplate cells, and also identifies some differences between mouse and human muscle preplate cells.


Subject(s)
Cell Separation/methods , Muscle Cells/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration/physiology , Animals , Cell Adhesion , Cell Fusion , Cell Proliferation , Humans , Mice , Mice, Inbred mdx , Mice, SCID , Muscle Cells/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Phenotype , Regeneration/genetics
6.
Biotechnol Prog ; 27(4): 913-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21692197

ABSTRACT

Automated time-lapsed microscopy provides unique research opportunities to visualize cells and subcellular components in experiments with time-dependent parameters. As accessibility to these systems is increasing, we review here their use in cell science with a focus on stem cell research. Although the use of time-lapsed imaging to answer biological questions dates back nearly 150 years, only recently have the use of an environmentally controlled chamber and robotic stage controllers allowed for high-throughput continuous imaging over long periods at the cell and subcellular levels. Numerous automated imaging systems are now available from both companies that specialize in live cell imaging and from major microscope manufacturers. We discuss the key components of robots used for time-lapsed live microscopic imaging, and the unique data that can be obtained from image analysis. We show how automated features enhance experimentation by providing examples of uniquely quantified proliferation and migration live cell imaging data. In addition to providing an efficient system that drastically reduces man-hours and consumes fewer laboratory resources, this technology greatly enhances cell science by providing a unique dataset of temporal changes in cell activity.


Subject(s)
Cell Biology , Diagnostic Imaging/methods , Animals , Humans , Time-Lapse Imaging/methods
7.
Stem Cells Dev ; 20(3): 451-63, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20923371

ABSTRACT

Perivascular multipotent mesenchymal progenitors exist in a variety of tissues, including the placenta. Here, we suggest that the abundant vasculature present in the human placenta can serve as a source of myogenic cells to regenerate skeletal muscle. Chorionic villi dissected from the mid-gestation human placenta were first transplanted intact into the gastrocnemius muscles of SCID/mdx mice, where they participated in muscle regeneration by producing myofibers expressing human dystrophin and spectrin. In vitro-cultured placental villi released rapidly adhering and migratory CD146+CD34⁻CD45⁻CD56⁻ cells of putative perivascular origin that expressed mesenchymal stem cell markers. CD146+CD34⁻CD45⁻CD56⁻ perivascular cells isolated and purified from the placental villi by flow cytometry were indeed highly myogenic in culture, and generated dystrophin-positive myofibers, and they promoted angiogenesis after transplantation into SCID/mdx mouse muscles. These observations confirm the existence of mesenchymal progenitor cells within the walls of human blood vessels, and suggest that the richly vascularized human placenta is an abundant source of perivascular myogenic cells able to migrate within dystrophic muscle and regenerate myofibers.


Subject(s)
Muscle, Skeletal/physiology , Placenta/cytology , Regeneration , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Nuclear/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Shape , Cells, Cultured , Chorionic Villi/metabolism , Chorionic Villi/transplantation , Dystrophin/metabolism , Female , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/cytology , Neovascularization, Physiologic , Placenta/blood supply , Pregnancy , Spectrin/metabolism , Tissue Culture Techniques , Transcription, Genetic
8.
J Biol Chem ; 285(43): 33549-33566, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20729206

ABSTRACT

m-Calpain plays a critical role in cell migration enabling rear de-adhesion of adherent cells by cleaving structural components of the adhesion plaques. Growth factors and chemokines regulate keratinocyte, fibroblast, and endothelial cell migration by modulating m-calpain activity. Growth factor receptors activate m-calpain secondary to phosphorylation on serine 50 by ERK. Concurrently, activated m-calpain is localized to its inner membrane milieu by binding to phosphatidylinositol 4,5-bisphosphate (PIP(2)). Opposing this, CXCR3 ligands inhibit cell migration by blocking m-calpain activity secondary to a PKA-mediated phosphorylation in the C2-like domain. The failure of m-calpain activation in the absence of PIP(2) points to a key regulatory role, although whether this PIP(2)-mediated membrane localization is regulatory for m-calpain activity or merely serves as a docking site for ERK phosphorylation is uncertain. Herein, we report the effects of two CXCR3 ligands, CXCL11/IP-9/I-TAC and CXCL10/IP-10, on the EGF- and VEGF-induced redistribution of m-calpain in human fibroblasts and endothelial cells. The two chemokines block the tail retraction and, thus, the migration within minutes, preventing and reverting growth factor-induced relocalization of m-calpain to the plasma membrane of the cells. PKA phosphorylation of m-calpain blocks the binding of the protease to PIP(2). Unexpectedly, we found that this was due to membrane anchorage itself and not merely serine 50 phosphorylation, as the farnesylation-induced anchorage of m-calpain triggers a strong activation of this protease, leading notably to an increased cell death. Moreover, the ERK and PKA phosphorylations have no effect on this membrane-anchored m-calpain. However, the presence of PIP(2) is still required for the activation of the anchored m-calpain. In conclusion, we describe a novel mechanism of m-calpain activation by interaction with the plasma membrane and PIP(2) specifically, this phosphoinositide acting as a cofactor for the enzyme. The phosphorylation of m-calpain by ERK and PKA by growth factors and chemokines, respectively, act in cells to regulate the enzyme only indirectly by controlling its redistribution.


Subject(s)
Calpain/metabolism , Cell Membrane/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Inositol Phosphates/metabolism , Animals , Calpain/genetics , Cell Death/drug effects , Cell Death/physiology , Cell Line , Cell Membrane/genetics , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/cytology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Humans , Inositol Phosphates/genetics , Mice , Phosphorylation/physiology , Protein Structure, Tertiary , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Receptors, Growth Factor/agonists , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Tissue Eng Part A ; 16(3): 839-49, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19788347

ABSTRACT

Muscle-derived stem-cell (MDSC) transplantation presents a promising method for the treatment of muscle injuries. This study investigated the ability of exercise to enhance MDSC transplantation into the injured muscle. Mice were divided into four groups: contusion + phosphate-buffered saline (C + PBS; n = 14 muscles), C + MDSC transplantation (n = 12 muscles), C + PBS + treadmill running (C + PBS + TM; n = 17 muscles), and C + MDSC + TM (n = 13 muscles). One day after injury, the TM groups began running for 1 or 5 weeks. Two days after injury, muscles of C + MDSC and C + MDSC + TM groups were injected with MDSCs. One or 5 weeks later, the number and differentiation of transplanted MDSCs, myofiber regeneration, collagen I formation, and vascularity were assessed histologically. In vitro, MDSCs were subjected to mechanical stimulation, and growth kinetics were quantified. In vitro, mechanical stimulation decreased the MDSC population doubling time (18.6 +/- 1.6 h) and cell division time (10.9 +/- 0.7 h), compared with the controls (population doubling time: 23.0 +/- 3.4 h; cell division time: 13.3 +/- 1.1 h) (p = 0.01 and 0.03, respectively). In vivo, 5 weeks of TM increased the myogenic contribution of transplanted MDSCs, compared with the controls (p = 0.02). C + MDSC, C + PBS + TM, and C + MDSC + TM demonstrated decreased fibrosis at 5 weeks, compared with the C + PBS controls (p = 0.00, p = 0.03, and p = 0.02, respectively). Results suggest that the mechanical stimulation favors MDSC proliferation, both in vitro and in vivo, and that exercise enhances MDSC transplantation after injury.


Subject(s)
Muscle, Skeletal/pathology , Physical Conditioning, Animal , Stem Cell Transplantation , Wound Healing , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Collagen/metabolism , Female , Fluorescent Antibody Technique , Kinetics , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/blood supply , Regeneration , Transduction, Genetic , beta-Galactosidase/metabolism
10.
J Biomed Biotechnol ; 2009: 789526, 2009.
Article in English | MEDLINE | ID: mdl-20037738

ABSTRACT

Human umbilical cord blood is an excellent primitive source of noncontroversial stem cells for treatment of hematologic disorders; meanwhile, new stem cell candidates in the umbilical cord (UC) tissue could provide therapeutic cells for nonhematologic disorders. We show novel in situ characterization to identify and localize a panel of some markers expressed by mesenchymal stromal cells (MSCs; CD44, CD105, CD73, CD90) and CD146 in the UC. We describe enzymatic isolation and purification methods of different UC cell populations that do not require manual separation of the vessels and stroma of the coiled, helical-like UC tissue. Unique quantitation of in situ cell frequency and stromal cell counts upon harvest illustrate the potential to obtain high numerical yields with these methods. UC stromal cells can differentiate to the osteogenic and chondrogenic lineages and, under specific culturing conditions, they exhibit high expandability with unique long-term stability of their phenotype. The remarkable stability of the phenotype represents a novel finding for human MSCs, from any source, and supports the use of these cells as highly accessible stromal cells for both basic studies and potentially therapeutic applications such as allogeneic clinical use for musculoskeletal disorders.


Subject(s)
Cell Culture Techniques/methods , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Tissue Engineering/methods , CD146 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Phenotype
11.
Spine (Phila Pa 1976) ; 34(23): 2486-93, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19841610

ABSTRACT

STUDY DESIGN: We demonstrated the differentiation of notochordal cells by direct observation using a live automated cell imaging system. We also hypothesized that notochordal cells have characteristics of chondrocyte-like cells. OBJECTIVE: To determine characteristics of notochordal cells by matrix protein expression and their differentiation using a live automated cell imager. SUMMARY OF BACKGROUND DATA: Although notochordal cells are critical to homeostasis of intervertebral disc, their fate has not been extensively studied and there is little evidence of notochordal cells as progenitors. METHODS: Notochordal cells purified from rabbit nucleus pulposus were isolated after serial filtration. Notochordal cells in 3-dimensional culture were compared to chondrocyte-like cells by S sulfate incorporation into proteoglycan and reverse transcription polymerase chain reaction for gene expression(collagen II and aggrecan). Notochordal cells in 2-D culture were used for immunocytochemical staining (collagen II, aggrecan, and SOX9) and time-lapsed cell tracking study. RESULTS: Notochordal cells were capable of proteoglycan production at a rate comparable to chondrocyte-like cells (108% +/- 22.6% to chondrocyte-like cells) and expressed collagen II, aggrecan, and SOX9. In time-lapsed cell tracking analysis, notochordal cells were slower in population doubling time than chondrocyte-like cells and differentiated into 3 morphologically distinct cell types: vacuolated cells (area: 2392 +/- 507.1 microm, velocity: 0.09 +/- 0.01 microm/min); giant cells (area: 12678 +/- 1637.0 microm, velocity: 0.08 +/- 0.01 microm/min) which grew rapidly without cell division; polygonal cells (area: 3053 +/- 751.2 microm, 0.14 +/- 0.01 microm/min) morphologically similar to typical differentiation type of chondrocyte-like cells (area: 2671 +/- 235.6 microm, 0.19 +/- 0.01 microm/min). Rarely, notochordal cells formed clusters analogous to that observed in vivo. CONCLUSION: These studies demonstrate a chondrocyte phenotype of notochordal cells and are the first direct evidence of notochordal cell differentiation, suggesting that they may act as progenitor cells, which has the potential to lead to their use in novel approaches to regeneration of degenerative intervertebral disc.


Subject(s)
Cell Differentiation/physiology , Diagnostic Imaging/methods , Intervertebral Disc/physiology , Notochord/physiology , Aggrecans/genetics , Aggrecans/metabolism , Animals , Cell Culture Techniques , Cell Differentiation/genetics , Cell Shape/genetics , Cell Shape/physiology , Cells, Cultured , Chondrocytes/metabolism , Collagen/genetics , Collagen/metabolism , Immunohistochemistry , Intervertebral Disc/cytology , Intervertebral Disc/metabolism , Notochord/cytology , Notochord/metabolism , Phenotype , Proteoglycans/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/metabolism
12.
Exp Cell Res ; 315(17): 2963-73, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19607826

ABSTRACT

The objective of the present study was to evaluate how different ligand interactions of profilin-1 (Pfn1), an actin-binding protein that is upregulated during capillary morphogenesis of vascular endothelial cells (VEC), contribute to migration and capillary forming ability of VEC. We adopted a knockdown-knockin experimental system to stably express either fully functional form or mutants of Pfn1 that are impaired in binding to two of its major ligands, actin (H119E mutant) and proteins containing polyproline domains (H133S mutant), in a human dermal microvascular cell line (HmVEC) against near-null endogenous Pfn1 background. We found that silencing endogenous Pfn1 expression in HmVEC leads to slower random migration, reduced velocity of membrane protrusion and a significant impairment in matrigel-induced cord formation. Only re-expression of fully functional but not any of the two ligand-binding deficient mutants of Pfn1 rescues the above defects. We further show that loss of Pfn1 expression in VEC inhibits three-dimensional capillary morphogenesis, MMP2 secretion and ECM invasion. VEC invasion through ECM is also inhibited when actin and polyproline interactions of Pfn1 are disrupted. Together, these experimental data demonstrate that Pfn1 regulates VEC migration, invasion and capillary morphogenesis through its interaction with both actin and proline-rich ligands.


Subject(s)
Capillaries/cytology , Capillaries/physiology , Cell Movement/physiology , Endothelium, Vascular/physiology , Profilins/metabolism , Actins/metabolism , Endothelium, Vascular/cytology , Gelatin/analysis , Gene Silencing , Humans , Morphogenesis/physiology , Peptides/metabolism , Phalloidine/analysis , Profilins/deficiency , Profilins/genetics , Umbilical Veins/cytology , Umbilical Veins/growth & development , Umbilical Veins/physiology
13.
Mol Ther ; 17(10): 1788-98, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19603004

ABSTRACT

We have isolated a population of muscle-derived stem cells (MDSCs) that, when compared with myoblasts, display an improved regeneration capacity, exhibit better cell survival, and improve myogenesis and angiogenesis. In addition, we and others have observed that the origin of the MDSCs may reside within the blood vessel walls (endothelial cells and pericytes). Here, we investigated the role of vascular endothelial growth factor (VEGF)-mediated angiogenesis in MDSC transplantation-based skeletal muscle regeneration in mdx mice (an animal model of muscular dystrophy). We studied MDSC and MDSC transduced to overexpress VEGF; no differences were observed in vitro in terms of phenotype or myogenic differentiation. However, after in vivo transplantation, we observe an increase in angiogenesis and endogenous muscle regeneration as well as a reduction in muscle fibrosis in muscles transplanted with VEGF-expressing cells when compared to control cells. In contrast, we observe a significant decrease in vascularization and an increase in fibrosis in the muscles transplanted with MDSCs expressing soluble forms-like tyrosine kinase 1 (sFlt1) (VEGF-specific antagonist) when compared to control MDSCs. Our results indicate that VEGF-expressing cells do not increase the number of dystrophin-positive fibers in the injected mdx muscle, when compared to the control MDSCs. Together the results suggest that the transplantation of VEGF-expressing MDSCs improved skeletal muscle repair through modulation of angiogenesis, regeneration and fibrosis in the injected mdx skeletal muscle.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/therapy , Stem Cells/cytology , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Stem Cell Transplantation/methods , Stem Cells/physiology , Transduction, Genetic , Vascular Endothelial Growth Factor A/genetics
14.
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
15.
Cell Biochem Biophys ; 52(2): 85-102, 2008.
Article in English | MEDLINE | ID: mdl-18841496

ABSTRACT

The field of regenerative medicine offers hope for the development of a cell-based therapy for the repair of articular cartilage (AC). Yet, the greatest challenge in the use of stem cells for tissue repair, is understanding how the cells respond to stimuli and using that knowledge to direct cell fate. Novel methods that utilize stem cells in cartilage regeneration will require specific spatio-temporal controls of the biochemical and biophysical signaling environments. Current chondrogenic differentiation research focuses on the roles of biochemical stimuli like growth factors, hormones, and small molecules, and the role of the physical environment and mechanical stimuli, such as compression and shear stress, which likely act through mechanical receptors. Numerous signals are associated with chondrogenic-like activity of cells in different systems, however many variables for a controlled method still need to be optimized; e.g., spatial and temporal application of the stimuli, and time of transplantation of an engineered construct. Understanding the necessary microenvironmental signals for cell differentiation will advance cell therapy for cartilage repair.


Subject(s)
Cartilage, Articular/cytology , Cell Differentiation/physiology , Chondrogenesis/physiology , Stem Cells/cytology , Tissue Engineering , Animals , Cartilage, Articular/physiology , Colony-Forming Units Assay/methods , Hormones/physiology , Humans , Mechanoreceptors/physiology , Regeneration/physiology , Stem Cells/physiology
16.
Cell Stem Cell ; 3(3): 301-13, 2008 Sep 11.
Article in English | MEDLINE | ID: mdl-18786417

ABSTRACT

Mesenchymal stem cells (MSCs), the archetypal multipotent progenitor cells derived in cultures of developed organs, are of unknown identity and native distribution. We have prospectively identified perivascular cells, principally pericytes, in multiple human organs including skeletal muscle, pancreas, adipose tissue, and placenta, on CD146, NG2, and PDGF-Rbeta expression and absence of hematopoietic, endothelial, and myogenic cell markers. Perivascular cells purified from skeletal muscle or nonmuscle tissues were myogenic in culture and in vivo. Irrespective of their tissue origin, long-term cultured perivascular cells retained myogenicity; exhibited at the clonal level osteogenic, chondrogenic, and adipogenic potentials; expressed MSC markers; and migrated in a culture model of chemotaxis. Expression of MSC markers was also detected at the surface of native, noncultured perivascular cells. Thus, blood vessel walls harbor a reserve of progenitor cells that may be integral to the origin of the elusive MSCs and other related adult stem cells.


Subject(s)
Adult Stem Cells/cytology , Fetal Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Pericytes/cytology , Adolescent , Adult , Aged , Cell Movement , Fetus , Flow Cytometry , Humans , Middle Aged , Muscle Development
17.
Methods Cell Biol ; 86: 295-309, 2008.
Article in English | MEDLINE | ID: mdl-18442653

ABSTRACT

We have identified with molecular markers and purified by flow cytometry two populations of cells that are developmentally and anatomically related to blood vessel walls in human tissues: myoendothelial cells, found in skeletal muscle and coexpressing markers of endothelial and myogenic cells, and pericytes--aka mural cells--which surround endothelial cells in capillaries and microvessels. Purified myoendothelial cells and pericytes exhibit multilineage developmental potential and differentiate, in culture and in vivo, into skeletal myofibers, bone, cartilage, and adipocytes. Myoendothelial cells and pericytes can be cultured on the long term with sustained marker expression and differentiation potential and clonal populations thereof have been derived. Yet, these blood vessel wall-derived progenitors exhibit no tendency to malignant transformation upon extended culture. Our results suggest that multipotent progenitor cells, such as mesenchymal stem cells, previously isolated retrospectively from diverse cultured adult tissues are derived from a subset of perivascular cells. We present in this chapter the main strategies and tactics used to purify, culture on the long term, and phenotypically characterize these novel multipotent cells.


Subject(s)
Blood Vessels/cytology , Cell Culture Techniques/methods , Endothelial Cells/cytology , Multipotent Stem Cells/cytology , Muscle, Skeletal/cytology , Pericytes/cytology , Stem Cells/cytology , Adult , Cell Culture Techniques/instrumentation , Cell Separation/methods , Cells, Cultured , Endothelium, Vascular/cytology , Fetus/anatomy & histology , Flow Cytometry , Genotype , Humans , Phenotype
18.
Crit Rev Eukaryot Gene Expr ; 18(2): 173-88, 2008.
Article in English | MEDLINE | ID: mdl-18304031

ABSTRACT

Sex is well known to influence life expectancy and disposition to disease. Stem and progenitor cells are believed to persist throughout life, and they contribute to the repair and healthy maintenance of tissue; consequently, sex-related differences demonstrated by stem cells may provide insight to sex-related differences in aging, disease, and healing. However, cell sex is an often overlooked variable in stem cell biology.


Subject(s)
Adult Stem Cells/physiology , Muscle, Skeletal/physiology , Sex Characteristics , Aging/physiology , Animals , Cell Culture Techniques , Disease/etiology , Estrogens/physiology , Female , Gene Expression/physiology , Humans , Male , Models, Biological , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/metabolism , Organ Size , Oxidative Stress/physiology , Pericytes/physiology , Satellite Cells, Skeletal Muscle/physiology
19.
Biomaterials ; 29(7): 825-33, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18035412

ABSTRACT

There is a clinical need for a tissue-engineered vascular graft (TEVG), and combining stem cells with biodegradable tubular scaffolds appears to be a promising approach. The goal of this study was to characterize the incorporation of muscle-derived stem cells (MDSCs) within tubular poly(ester urethane) urea (PEUU) scaffolds in vitro to understand their interaction, and to evaluate the mechanical properties of the constructs for vascular applications. Porous PEUU scaffolds were seeded with MDSCs using our recently described rotational vacuum seeding device, and cultured inside a spinner flask for 3 or 7 days. Cell viability, number, distribution and phenotype were assessed along with the suture retention strength and uniaxial mechanical behavior of the TEVGs. The seeding device allowed rapid even distribution of cells within the scaffolds. After 3 days, the constructs appeared completely populated with cells that were spread within the polymer. Cells underwent a population doubling of 2.1-fold, with a population doubling time of 35 h. Stem cell antigen-1 (Sca-1) expression by the cells remained high after 7 days in culture (77+/-20% vs. 66+/-6% at day 0) while CD34 expression was reduced (19+/-12% vs. 61+/-10% at day 0) and myosin heavy chain expression was scarce (not quantified). The estimated burst strength of the TEVG constructs was 2127+/-900 mm Hg and suture retention strength was 1.3+/-0.3N. We conclude from this study that MDSCs can be rapidly seeded within porous biodegradable tubular scaffolds while maintaining cell viability and high proliferation rates and without losing stem cell phenotype for up to 7 days of in-vitro culture. The successful integration of these steps is thought necessary to provide rapid availability of TEVGs, which is essential for clinical translation.


Subject(s)
Biocompatible Materials , Blood Vessel Prosthesis , Muscles/cytology , Stem Cells/cytology , Tissue Engineering/methods , Animals , Cell Separation , Cells, Cultured , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...