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










Publication year range
1.
Biochimie ; 196: 121-122, 2022 05.
Article in English | MEDLINE | ID: mdl-35358661

Subject(s)
Regeneration
2.
Biochimie ; 196: 131-142, 2022 May.
Article in English | MEDLINE | ID: mdl-35063588

ABSTRACT

A wound is considered hard-to-heal when, despite the appropriate clinical analysis and intervention, the wound area reduces by less than a third at four weeks and complete healing fails to occur within 12 weeks. The most prevalent hard-to-heal wounds are associated with underlying metabolic diseases or vascular insufficiency and include arterial, venous, pressure and diabetic foot ulcers. Their common features include an abnormal immune response and extended inflammatory phase, a subdued proliferation phase due to cellular insufficiencies and finally an almost non-existent remodeling phase. Advances in wound care technology, tested in both pre-clinical models and clinical trials, have paved the way for improved treatment options, focused on regeneration. These interventions have been shown to limit the extent of ongoing inflammatory damage, decrease bacterial load, promote angiogenesis and deposition of granulation tissue, and stimulate keratinocyte migration thereby promoting re-epithelialization in these wounds. The current review discusses these hard-to-heal wounds in the context of their underlying pathology and potential of advanced treatment options, which if applied promptly as a standard of care, could reduce morbidity, promote quality of life, and alleviate the burden on a strained health system.


Subject(s)
Diabetic Foot , Leg Ulcer , Diabetic Foot/drug therapy , Humans , Leg Ulcer/therapy , Quality of Life , Wound Healing
3.
Physiol Rep ; 9(2): e14704, 2021 01.
Article in English | MEDLINE | ID: mdl-33463904

ABSTRACT

Skeletal muscle regeneration is a complex process influenced by non-myogenic macrophages and fibroblasts, which acquire different phenotypes in response to changes in the injury milieu or changes in experimental conditions. In vitro, serum stimulates the differentiation of fibroblasts into myofibroblasts, while lipopolysaccharide (LPS) stimulates the polarization of unstimulated (M0) macrophages to acquire an M1 pro-inflammatory phenotype. We characterized these phenotypes using morphology (with circularity as shape descriptor; perfect circularity = 1.0) and phenotype-specific markers. Myofibroblasts (high α-smooth muscle actin [SMA] expression) had high circularity (mean 0.60 ± 0.03). Their de-differentiation to fibroblasts (low α-SMA expression) significantly lessened circularity (0.47 ± 0.01 and 0.35 ± 0.02 in 2% or 0% serum culture media respectively (p < 0.05). Unstimulated (M0) macrophages (no CD86 expression) had high circularity (0.72 ± 0.02) which decreased when stimulated to M1 macrophages (CD86 expression) (LPS; 0.61 ± 0.02; p < 0.05). Utilizing these established conditions, we then co-cultured M1 macrophages with myofibroblasts or myoblasts. M1 macrophages significantly decreased relative myofibroblast numbers (from 223 ± 22% to 64 ± 7%), but not myoblast numbers. This pro-inflammatory co-culture model was used to rapidly screen the following four compounds for ability to prevent M1 macrophage-mediated decrease in myofibroblast numbers: L-NAME (inducible nitric oxide synthase inhibitor), SB203580 (p38 mitogen-activated protein kinase inhibitor), SP600125 (c-Jun N-terminal kinase inhibitor) and LY294002 (phosphoinositide 3-kinase [PI3K] inhibitor). We found that LY294002 rescued myofibroblasts and decreased macrophage numbers. Myofibroblast rescue did not occur with L-NAME, SB203580 or SP600125 incubation. In conclusion, these data suggest a PI3K-associated mechanism whereby myofibroblasts can be rescued, despite simulated pro-inflammatory conditions.


Subject(s)
Inositol/analogs & derivatives , Macrophages/drug effects , Muscle, Skeletal/drug effects , Myofibroblasts/drug effects , Phosphatidylinositol 3-Kinases/chemistry , Signal Transduction/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Inositol/pharmacology , Macrophages/metabolism , Macrophages/pathology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphatidylinositol 3-Kinases/metabolism
4.
Wound Repair Regen ; 28(4): 506-516, 2020 07.
Article in English | MEDLINE | ID: mdl-32281194

ABSTRACT

The advancement of autologous mesenchymal stem cell (MSC) therapy for the treatment of non-healing diabetic wounds is hampered by endogenous MSC dysfunction and limited viability of cells post-transplantation into the pathological wound environment. The development of effective strategies to restore the functional capabilities of these impaired MSCs prior to transplantation may be a key to their ultimate success as wound repair mediators. The current study therefore investigated whether antioxidant preconditioning [7.5 mM N-acetylcysteine (NAC) + 0.6 mM ascorbic 2-phosphate (AAP)] could restore the growth rate, migration ability and viability of impaired MSCs and whether this restored state is maintained in the presence of diabetic wound fluid (DWF). Healthy control (source: wild type, C57BL/6J mice) (n = 12) and impaired/diabetic MSCs (source: obese prediabetic, B6.Cg-Lepob/J mice) (n = 12) were isolated from the bone marrow of mice. Treatment groups post-isolation were as follow: (a) No treatment (baseline phenotype): MSCs expanded in standard growth media (SGM) (±8 days) and only exposed to growth media. (b) DWF (baseline response): MSCs expanded in SGM (±8 days) followed by exposure to DWF (24 hours, 48 hours, 96 hours). (c) Antioxidant preconditioning (preconditioned phenotype): MSCs expanded in the presence of NAC/AAP (±8 days). (d) Antioxidant preconditioning + DWF (preconditioned response): MSCs expanded in the presence of NAC/AAP (±8 days) followed by exposure to DWF (24 hours, 48 hours, 96 hours). The results demonstrated that expansion of MSCs (both healthy control and impaired diabetic) in the presence of combined NAC/AAP treatment improved ex vivo MSC viability and protected MSCs in the presence of DWF. Despite improved viability, AAP/NAC could however not rescue the reduced proliferation and migration capacity of impaired diabetic MSCs. The protective effect of NAC/AAP preconditioning against the toxicity of DWF could however be a potential strategy to improve cell number post-transplantation.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/analogs & derivatives , Cell Movement/drug effects , Cell Proliferation/drug effects , Mesenchymal Stem Cells/drug effects , Prediabetic State/metabolism , Wounds and Injuries/metabolism , Animals , Ascorbic Acid/pharmacology , Case-Control Studies , Cell Survival/drug effects , Diabetes Mellitus/metabolism , Exudates and Transudates , Mesenchymal Stem Cell Transplantation , Mice , Mice, Obese , Osteogenesis , Transplantation, Autologous
5.
Stem Cells Dev ; 27(23): 1646-1657, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30187827

ABSTRACT

Mesenchymal stem cells (MSCs) are a promising therapeutic tool for the treatment of nonhealing diabetic wounds. The pathological nature of the niche microenvironment limits the use of autologous cell therapy in diabetic patients. Prolonged exposure of endogenous MSCs to a pathological microenvironment in vivo reduces their ability to respond to environmental cues. This study investigated the effectiveness of ex vivo antioxidant treatment [N-acetylcysteine (7.5 mM NAC) and Ascorbic acid 2-phosphate (0.6 mM AAP)] to restore the paracrine function of diabetic MSCs. Healthy control [bone marrow stem cells derived from wild-type mice (SCWT)] (source: wild-type C57BL/6J mice) (n = 12) and impaired/dysfunctional [bone marrow stem cells derived from ob/ob mice (SCob)] (source: obese diabetic, B6.Cg-Lepob/J mice) (n = 12) MSCs were isolated. Ex vivo treatment groups (SCWT vs. SCob) were as follows: (1) no treatment (baseline phenotype), (2) stimulated with diabetic wound fluid (DWF) (baseline response), (3) antioxidant preconditioning (preconditioned phenotype), and (4) antioxidant preconditioned with subsequent stimulation with DWF (preconditioned response). The paracrine responsiveness on both the molecular (mRNA expression of 80 cytokines and receptors, quantitative polymerase chain reaction microarray) and protein (23-plex bead-array Luminex assay) level was assessed. At baseline, 31 genes were overexpressed (> × 2-fold) and 39 genes were underexpressed (> × 2-fold) in SCob versus SCWT. In conditioned media, significant differences (P < 0.05) were detected at baseline for two proinflammatory cytokines [tumor necrosis factor alpha (TNFα) and interferon gamma (IFNγ)], four chemokines [keratinocyte chemoattractant (KC), granulocyte colony-stimulating factor (GCSF), Eotaxin, and macrophage chemoattractant protein (MCP1)], and one anti-inflammatory cytokine [interleukin 10 (IL10)]. Following stimulation with DWF, significant differences (P < 0.05) were detected in the secretion of two chemokines [granulocyte macrophage colony-stimulating factor (GMCSF) and Eotaxin], three proinflammatory cytokines (TNFα, IFNγ, and IL9), and four anti-inflammatory cytokines (IL10, IL4, IL13, and IL3). Antioxidant preconditioning significantly dampened the excessive TNFα response observed in SCob and improved the secretion of IL10. Taken together these data suggest that the combined ex vivo treatment of autologous stem cells with NAC and AAP could potentially be an effective strategy to restore the paracrine function of impaired diabetic MSCs before transplantation.


Subject(s)
Antioxidants/pharmacology , Bone Marrow Cells/drug effects , Diabetic Angiopathies/therapy , Mesenchymal Stem Cells/drug effects , Paracrine Communication , Acetylcysteine/pharmacology , Animals , Ascorbic Acid/analogs & derivatives , Ascorbic Acid/pharmacology , Bone Marrow Cells/metabolism , Cytokines/genetics , Cytokines/metabolism , Diabetic Angiopathies/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Transcriptome
6.
Biotechniques ; 64(2): 52-58, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29571282

ABSTRACT

The communication between nonmyogenic cells, such as macrophages and fibroblasts, and myoblasts is crucial for successful skeletal muscle repair. In vitro co-culture methods can be used to increase our understanding of these cellular interactions; however, current protocols are restricted to two, often physically separate, cell populations. Here, we demonstrate a novel, inexpensive in vitro triple co-culture method that facilitates the co-culture of at least three cell populations with some degree of cell-cell contact. Using this method, we determined the effect of macrophages and fibroblasts on myoblast proliferation and migration. A significant increase in myoblast proliferation and migration was observed following co-culture with either macrophages or fibroblasts. However, triple co-culture of macrophages, fibroblasts, and myoblasts revealed that the presence of macrophages prevented fibroblasts from maintaining this positive effect on myoblast migration. Macrophages, on the other hand, continued to promote myoblast proliferation whether in the presence of fibroblasts or not. Our triple co-culture system highlights the significance of multicellular communication in regulating myoblast proliferation and migration and emphasizes the importance of more complex co-culture systems when investigating myogenesis in vitro.


Subject(s)
Coculture Techniques , Fibroblasts/physiology , Macrophages/physiology , Myoblasts/physiology , Wound Healing , Animals , Cell Line , Cell Movement , Cell Proliferation , Mice
7.
Biotechniques ; 61(6): 323-326, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27938324

ABSTRACT

Myoblast fusion, which is essential for muscle development, regeneration, and repair, can be assessed in vitro via the calculation of a fusion index. Traditionally, this requires use of either immunocytochemistry or fluorescently-labeled cytoskeletal staining, followed by microscopy and laborious analysis. The expense and time-consuming nature of the optimization and application of antibody-based techniques such as immunocytochemistry, as well as the need for specialized analytical equipment such as fluorescence microscopes, presents a barrier to the routine analysis of this crucial step during terminal differentiation. Here, we describe (i) a novel use of the commonly available LADD Multiple Stain for visualization of myoblast fusion in vitro; (ii) the optimization of a simple image analysis method to generate quick, quantifiable data representative of a fusion index; and (iii) the use of a protocol combining these two procedures to investigate in vitro myoblast fusion in a simple and efficient manner as proof-of-concept.


Subject(s)
Cell Fusion , Coloring Agents/chemistry , Microscopy/methods , Myoblasts/cytology , Animals , Cell Differentiation/physiology , Cell Line , Coloring Agents/metabolism , Mice , Myoblasts/chemistry , Myoblasts/metabolism , Rosaniline Dyes/chemistry , Rosaniline Dyes/metabolism , Tolonium Chloride/chemistry , Tolonium Chloride/metabolism
8.
Growth Factors ; 33(3): 229-41, 2015.
Article in English | MEDLINE | ID: mdl-26135603

ABSTRACT

Hepatocyte growth factor (HGF) regulates satellite cell activation, proliferation, and differentiation. We analyzed the dose-dependent effects of HGF on myogenesis. Murine C2C12 and human donor-derived skeletal muscle myoblasts were treated with 0, 2, or 10 ng/ml HGF followed by assessment of proliferation and differentiation. HGF (2 ng/ml) significantly promoted cell division, but reduced myogenic commitment and fusion. Conversely, 10 ng/ml HGF reduced proliferative capability, but increased differentiation. c-Met expression analysis revealed significantly decreased expression in differentiating cells cultured with 2 ng/ml HGF, but increased expression in proliferating cells with 10 ng/ml HGF. Mitogen-activated protein kinase (MAPKs: ERK, JNK, or p38K) and phosphatidylinositol-3-kinase (PI3K) inhibition abrogated the HGF-stimulated increase in cell number. Interestingly, PI3K and p38 kinase facilitated the negative effect of HGF on proliferation, while ERK inhibition abrogated the HGF-mediated decrease in differentiation. Dose-dependent effects of HGF are mediated by changes in c-Met expression and downstream MAPK and PI3K signalling.


Subject(s)
Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System/drug effects , Muscle Development/drug effects , Proto-Oncogene Proteins c-met/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Phosphoinositide-3 Kinase Inhibitors , Satellite Cells, Skeletal Muscle/cytology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Biotechniques ; 58(1): 25-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25605577

ABSTRACT

Skeletal muscle injury elicits the activation of satellite cells and their migration to the wound area for subsequent terminal differentiation and tissue integration. However, interstitial fibroblasts recruited to the site of injury promote deposition of fibrotic tissue, which hampers myoblast-mediated muscle regeneration. Currently, analysis of myoblast migration in vitro can be accomplished using chemotactic, cell-exclusion, or wound healing assays. Yet, to investigate cell motility following skeletal muscle damage more accurately, migration assays need to better simulate the repair process. Here we present a protocol for the simultaneous isolation of myoblasts and fibroblasts from the same muscle tissue, ensuring the consistent generation of enriched, purified, and matched cell populations at a low passage number. We then describe a wound assay that uses a novel approach to the co-culture of myoblasts and fibroblasts to mimic the injured environment more closely than other established methods. Using this assay, we demonstrate that fibroblasts are able to increase myoblast migration significantly, validating our new in vitro method. As the observed effect on migration is most likely mediated by secreted factors, our assay could easily be extended to include antibody-based protein analysis of secreted factors in animal or human systems.


Subject(s)
Cell Movement , Coculture Techniques , Fibroblasts/cytology , Myoblasts/cytology , Animals , Mice, Inbred BALB C
10.
Front Physiol ; 4: 349, 2013.
Article in English | MEDLINE | ID: mdl-24348426

ABSTRACT

Bioengineering skeletal muscle often requires customized equipment and intricate casting techniques. One of the major hurdles when initially trying to establish in vitro tissue engineered muscle constructs is the lack of consistency across published methodology. Although this diversity allows for specialization according to specific research goals, lack of standardization hampers comparative efforts. Differences in cell type, number and density, variability in matrix and scaffold usage as well as inconsistency in the distance between and type of adhesion posts complicates initial establishment of the technique with confidence. We describe an inexpensive, but readily adaptable silicone chamber system for the generation of skeletal muscle constructs that can readily be standardized and used to elucidate myoblast behavior in a three-dimensional space. Muscle generation, regeneration and adaptation can also be investigated in this model, which is more advanced than differentiated myotubes.

11.
J Muscle Res Cell Motil ; 34(5-6): 333-47, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24150600

ABSTRACT

Skeletal muscle repair requires the migration of myoblasts (activated satellite cells) both to the injury site and then within the wound to facilitate cellular alignment in preparation for differentiation, fusion and eventual healing. Along this journey, the cells encounter a range of soluble and extracellular matrix factors which regulate their movement and ultimately determine how successful the repair process will be. Sub-optimal migration can lead to a number of scenarios, including reduced myoblast numbers entering the wound, poor alignment and insufficient differentiation to correctly repair the damage. It is therefore critical that all aspects of myoblast migration are understood, particularly in response to the changing growth and matrix factor profile prevalent following skeletal muscle injury. Since 1962, when Boyden first introduced his chemotactic chamber, numerous in vitro migration assays have been developed to mimic the wound more closely. These have increased in complexity to account for the complex micro-environment found in vivo during muscle repair and include a range of modified cell exclusion, chemotactic and three-dimensional assays. This review describes and discusses these advances and highlights the importance they have in expanding our understanding of myoblast migration dynamics.


Subject(s)
Cell Movement/physiology , Muscle, Skeletal/cytology , Myoblasts/cytology , Animals , Cell Differentiation/physiology , Humans
12.
Muscle Nerve ; 48(1): 109-16, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23606058

ABSTRACT

INTRODUCTION: We investigated changes in satellite cell (SC) pool size after an acute bout of strenuous exercise and evaluated the influence of baseline SC count and fiber type. METHODS: Participants completed a downhill running (DHR) intervention (5 × 8 min, 2-min rest; 80% VO2max ; -10% gradient). Muscle biopsies were taken 7 days before VO2max and 7-9 days after the DHR intervention. Delayed-onset muscle soreness (DOMS) and creatine kinase activity (CK) were measured on days 1, 2, 7, and 9 post-DHR. SCs were identified by Pax7 and laminin staining. Relative distribution of MHC isoforms was determined by electrophoresis. RESULTS: DOMS and CK peaked on day 1 post-DHR (P < 0.01). The SC pool increased (26%) after DHR (P = 0.005). SCs/total myonuclei after recovery correlated with baseline SCs (r = 0.979, P = 0.003) and VO2max (r = 0.956, P = 0.011), whereas change in SC pool (Pax7(+) cells/total myonuclei: recovery minus baseline) tended to correlate with percent MHC II (r = 0.848; P = 0.06). CONCLUSION: Interindividual physiological characteristics affect SC pool expansion after a single bout of DHR and are influenced by VO2max .


Subject(s)
Cell Differentiation/physiology , Exercise/physiology , Muscle, Skeletal/cytology , Running/physiology , Satellite Cells, Skeletal Muscle/physiology , Adult , Cell Count/methods , Humans , Male , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Young Adult
13.
J Muscle Res Cell Motil ; 31(5-6): 359-67, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21298471

ABSTRACT

Following muscle injury, the damaged tissue and influx of inflammatory cells stimulate the secretion of growth factors and cytokines to initiate repair processes. This release of chemotactic signaling factors activates resident precursor cells and stimulates their mobilization and migration to the site of injury where terminal differentiation can occur. The three transforming growth factor-ß (TGF-ß) isoforms, and insulin-like growth factor-1 (IGF-1) are among the known regulatory factors released following muscle damage. We investigated the effect of recombinant active TGF-ß1, -ß2, -ß3 and IGF-1 on C2C12 skeletal muscle satellite cell and P19 embryonal carcinoma cell terminal differentiation and migration. C2C12 myoblast fusion as well as P19 embryoid body formation and myogenic differentiation was assessed following 72 h TGF-ß treatment (5 ng/ml), whereas the effect of the TGF-ß isoforms on migration was determined following 7 h incubation. Our results showed that TGF-ß decreases C2C12 myoblast fusion in an isoform-independent manner, whereas in the P19 cell lineage, results demonstrate that TGF-ß1 specifically and significantly increased P19 embryoid body formation, but not expression of Connexin-43 or Myosin Heavy Chain. IGF-1 significantly increased migration compared to TGF-ß isoforms, which, on their own, had no significant effect on the mobilization of either C2C12 or P19 cells. TGF-ß isoforms decreased IGF-1-induced migration of both cell lineages. By distinguishing the factors involved in, and the molecular signals required for, myoblast recruitment during repair processes, strategies can be developed towards improved cell-mediated therapies for muscle injury.


Subject(s)
Cell Differentiation/drug effects , Cell Movement/drug effects , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Transforming Growth Factor beta/pharmacology , Animals , Cell Fusion , Cell Line, Tumor , Cells, Cultured , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Organ Specificity , Protein Isoforms/pharmacology , Stem Cells/cytology , Stem Cells/drug effects
14.
Cytotechnology ; 62(3): 265-77, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20680682

ABSTRACT

The abalone, Haliotis midae, is the most valuable commodity in South African aquaculture. The increasing demand for marine shellfish has stimulated research on the biology and physiology of target species in order to improve knowledge on growth, nutritional requirements and pathogen identification. The slow growth rate and long generation time of abalone restrict efficient design of in vivo experiments. Therefore, in vitro systems present an attractive alternative for short term experimentation. The use of marine invertebrate cell cultures as a standardised and controlled system to study growth, endocrinology and disease contributes to the understanding of the biology of economically important molluscs. This paper investigates the suitability of two different H. midae tissues, larval and haemocyte, for establishing primary cell cultures. Cell cultures are assessed in terms of culture initiation, cell yield, longevity and susceptibility to contamination. Haliotis midae haemocytes are shown to be a more feasible tissue for primary cell culture as it could be maintained without contamination more readily than larval cell cultures. The usefulness of short term primary haemocyte cultures is demonstrated here with a growth factor trial. Haemocyte cultures can furthermore be used to relate phenotypic changes at the cellular level to changes in gene expression at the molecular level.

15.
Stem Cells Dev ; 18(6): 813-30, 2009.
Article in English | MEDLINE | ID: mdl-19265462

ABSTRACT

The ability of unspecialized stem cells to differentiate into mature, specialized cell types has made them attractive as potential agents for enhanced tissue repair and regenerative medicine. This is especially true of diseases and disorders for which no or only partially effective treatments are currently available. Recently, increased focus has been placed on the regenerative potential of satellite cells (myogenic precursor cells found in the adult skeletal muscle) in various muscular disorders, such as dystrophy and myocardial injury following ischemia. Animal studies and clinical trials are in progress using satellite cells as cellular candidates; however, this early rollout in the clinical setting has deflected attention from the potential of other less specialized, but potentially more maliable, stem cell sources. Published data is still lacking on the best methods for identification, isolation, and further expansion or nuclear manipulation of these cells in vitro. Also, although differentiation capacity has been proven in terms of protein expression patterns characteristic of myogenesis, proof of contractile and energetic compatibility between graft and host is more difficult to establish. In this regard, although future animal model studies will be invaluable, they must be designed with short- and long-term functional outcomes in mind. This review moves beyond initial excitement regarding the acknowledged potential of cell therapy and provides a realistic exposition of the themes and specific issues that should be considered in current experimental research study designs.


Subject(s)
Muscle Development , Myocardium/pathology , Regenerative Medicine , Stem Cells/cytology , Wound Healing , Adult Stem Cells/cytology , Animals , Humans
16.
Exp Cell Res ; 315(3): 373-84, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19038250

ABSTRACT

Satellite cells are a quiescent heterogeneous population of mononuclear stem and progenitor cells which, once activated, differentiate into myotubes and facilitate skeletal muscle repair or growth. The Transforming Growth Factor-beta (TGF-beta) superfamily members are elevated post-injury and their importance in the regulation of myogenesis and wound healing has been demonstrated both in vitro and in vivo. Most studies suggest a negative role for TGF-beta on satellite cell differentiation. However, none have compared the effect of these three isoforms on myogenesis in vitro. This is despite known isoform-specific effects of TGF-beta1, -beta2 and -beta3 on wound repair in other tissues. In the current study we compared the effect of TGF-beta1, -beta2 and -beta3 on proliferation and differentiation of the C2C12 myoblast cell-line. We found that, irrespective of the isoform, TGF-beta increased proliferation of C2C12 cells by changing the cellular localisation of PCNA to promote cell division and prevent cell cycle exit. Concomitantly, TGF-beta1, -beta2 and -beta3 delayed myogenic commitment by increasing MyoD degradation and decreasing myogenin expression. Terminal differentiation, as measured by a decrease in myosin heavy chain (MHC) expression, was also delayed. These results demonstrate that TGF-beta promotes proliferation and delays differentiation of C2C12 myoblasts in an isoform-independent manner.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/cytology , Myoblasts/cytology , Transforming Growth Factor beta/physiology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mice , Muscle Development/drug effects , Muscle Development/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , MyoD Protein/metabolism , Myoblasts/drug effects , Myoblasts/physiology , Proliferating Cell Nuclear Antigen/metabolism , Protein Isoforms/pharmacology , Protein Isoforms/physiology , Protein Transport , Transforming Growth Factor beta/pharmacology
17.
Exp Physiol ; 92(1): 207-17, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16945942

ABSTRACT

AMP-activated protein kinase (AMPK) functions as a alpha/beta/gamma heterotrimer to preserve ATP levels and so cell viability during stressful conditions. However, its role in aiding survival of adult skeletal muscle precursor cells is unclear. Using the differentiating mouse C2C12 postnatal skeletal muscle myoblast cell line, we have determined that proteins for the AMPK subunit isoforms alpha2 and gamma2 are constitutively expressed, while those for alpha1, beta1 and beta2 are undetectable in undifferentiated myoblasts but increasingly expressed with differentiation to myotubes. Although the gamma3 subunit is expressed at a low level in myoblasts, it too is expressed increasingly with differentiation to myotubes. The p50 but not the p72 isoform of the embryonic alpha subunit homologue MELK is expressed only in proliferating myoblasts, while the ARK5 alpha subunit homologue is increasingly expressed with differentiation. Myotubes displayed higher basal and stimulated alpha1/alpha2 AMPK activation than myoblasts. Furthermore, serum starvation resulted in less apoptosis of differentiated myotubes than of undifferentiated myoblasts. This reflects, in part, the increased expression of functional AMPK in the myotubes, since specific inhibition of AMPK activity with 6-[4-(2-piperidin-1-ylethoxy)-phenyl]-3-pyridin-4-ylpyrazolo[1,5-alpha] pyrimidine (Compound C) exacerbated the apoptosis resulting from serum withdrawal. If these in vitro events can also occur in vivo, they could have implications for pathologies such as muscle wasting, in which undifferentiated satellite stem cells may be easier apoptotic targets than their differentiated counterparts. Furthermore, these results suggest that when interpreting results from in vitro or in vivo experiments on AMPK, the subunit expression profile should be taken into account.


Subject(s)
Apoptosis , Cell Differentiation , Gene Expression Regulation, Enzymologic , Multienzyme Complexes/metabolism , Myoblasts, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Acetyl-CoA Carboxylase/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Cell Survival , Enzyme Activation , Enzyme Inhibitors/pharmacology , Mice , Multienzyme Complexes/antagonists & inhibitors , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Oligomycins/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Repressor Proteins/metabolism , Time Factors
18.
Brain Res ; 1120(1): 124-30, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-17005162

ABSTRACT

We determined c-Fos immunoreactivity (Fos-IR) in selected hypothalamic nuclei, the organum vasculosum of the laminae terminals (OVLT) and somatosensory cortex of rats after hyperthermia induced by exogenous heat exposure, Gram-negative or Gram-positive pyrogen administration. The magnitude of Fos-IR was similar in thermoregulatory hypothalamic nuclei of rats after heat exposure or lipopolysaccharide (LPS) injection, despite the different origins of the hyperthermias. Heat-induced hyperthermia was associated with increased Fos-IR in the somatosensory cortex. LPS, but not heat exposure or injection of killed Staphylococcus aureus cells activated OVLT neurons. The OVLT may thus not be a port of entry for humoral mediators of Gram-positive bacterial fevers.


Subject(s)
Brain/drug effects , Brain/radiation effects , Hot Temperature , Lipopolysaccharides/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Pyrogens/administration & dosage , Analysis of Variance , Animals , Brain/cytology , Brain/metabolism , Cell Count/methods , Immunohistochemistry/methods , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Time Factors
19.
Regen Med ; 1(2): 223-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-17465806

ABSTRACT

Genetic labeling of human hematopoietic progenitor cells (HPC) and their consecutive fate-mapping in vivo is an approach to answer intriguing questions in stem cell biology. We recently reported efficient transient genetic labeling of human CD34+ HPC with the truncated low-affinity nerve growth factor receptor (DeltaLNGFR) for in vivo application. Here we investigate whether HPC labeling with DeltaLNGFR affects lineage-specific cell differentiation, whether DeltaLNGFR expression is maintained during lineage-specific cell differentiation and which leukemia cell line might be an appropriate cell culture model for human CD34+ HPC. Human CD34+ peripheral blood stem cells and various leukemia cell lines were characterized by immunophenotyping. Cells were transfected using nucleofection. Hematopoietic differentiation was studied by colony-forming assays. DeltaLNGFR expression was assessed using reverse transcription-PCR, immunofluorescence and flow cytometry. Nucleofection was efficient and did not significantly reduce hematopoietic cell differentiation. Mature myeloid cells (CD66b+) derived from human CD34+ HPC and Mutz2 cells maintained DeltaLNGFR expression at a high percentage (70 +/- 2% and 58 +/- 2%, respectively). Mutz2 cells may serve as an in vitro model for human myeloid HPC. The method described herein has been adopted to Good Manufacturing Practices (GMP) guidelines and is ready for in vivo application.


Subject(s)
Antigens, CD34/metabolism , Hematopoietic Stem Cells/metabolism , Tissue Engineering/methods , Cell Differentiation , Cell Line, Tumor , Genetic Markers/genetics , HL-60 Cells , Hematopoietic Stem Cells/cytology , Humans , Immunophenotyping , K562 Cells , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Organisms, Genetically Modified , Transfection
20.
Cardiovasc J S Afr ; 15(4): 184-9; discussion 189, 2004.
Article in English | MEDLINE | ID: mdl-15322576

ABSTRACT

The vast developmental repertoire of embryonic stem cells is well recognised. These primitive stem cells can differentiate in vivo and in vitro into cells of all three embryonic germ layers (endoderm, mesoderm, ectoderm), making them attractive potential agents to target for enhanced tissue repair and regeneration. Adult stem cells on the other hand are considered more restricted in their lineage differentiation capabilities. Recent research has challenged this dogma with the finding that bone marrow-derived stem cells can differentiate into a wide variety of cell types including muscle (skeletal and cardiac). Furthermore, although the myocardium has for decades been regarded as a post-mitotic organ, a series of studies has indicated that a population of stem cells exists which is capable of at least partial reconstitution of the myocardium following an ischaemic insult. It is therefore now accepted that adult stem cells could be used to enhance myocardial repair. This review discusses the current status of adult stem cell research in the light of its potential for improving myocardial repair.


Subject(s)
Myocardial Ischemia/physiopathology , Myocardial Ischemia/therapy , Stem Cell Transplantation/methods , Stem Cells/physiology , Adult , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Chemotaxis/physiology , Clinical Trials as Topic , Humans , Myocytes, Cardiac/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...