Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci Res ; 88(8): 1632-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20091773

ABSTRACT

Activated microglia can influence the survival of neural cells through the release of cytotoxic factors. Here, we investigated the interaction between Toll-like receptor 4 (TLR4)-activated microglia and oligodendrocytes or their precursor cells (OPC). Primary rat or N9 microglial cells were activated by exposure to TLR4-specifc lipopolysaccharide (LPS), resulting in mitogen-activated protein kinase activation, increased CD68 and inducible nitric oxide synthase expression, and release of the proinflammatory cytokines tumor necrosis factor (TNF) and interleukin-6 (IL-6). Microglial conditioned medium (MGCM) from LPS-activated microglia attenuated primary OPC proliferation without inducing cell death. The microglial-induced inhibition of OPC proliferation was reversed by stimulating group III metabotropic glutamate receptors in microglia with the agonist L-AP4. In contrast to OPC, LPS-activated MGCM enhanced the survival of mature oligodendrocytes. Further investigation suggested that TNF and IL-6 released from TLR4-activated microglia might contribute to the effect of MGCM on OPC proliferation, insofar as TNF depletion of LPS-activated MGCM reduced the inhibition of OPC proliferation, and direct addition of TNF or IL-6 attenuated or increased proliferation, respectively. OPC themselves were also found to express proteins involved in TLR4 signalling, including TLR4, MyD88, and MAL. Although LPS stimulation of OPC did not induce proinflammatory cytokine release or affect their survival, it did trigger JNK phosphorylation, suggesting that TLR4 signalling in these cells is active. These findings suggest that OPC survival may be influenced not only by factors released from endotoxin-activated microglia but also through a direct response to endotoxins. This may have consequences for myelination under conditions in which microglial activation and cerebral infection are both implicated. , Inc.


Subject(s)
Cell Proliferation , Microglia/physiology , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Fibroblast Growth Factor 2/pharmacology , Gangliosides/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Ki-67 Antigen/metabolism , Lipopolysaccharides/pharmacology , Microglia/chemistry , Microglia/drug effects , Myelin Basic Protein/metabolism , Neuroblastoma , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stem Cells/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
2.
Cell Cycle ; 8(7): 1069-79, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19270512

ABSTRACT

The potential of mesenchymal stem cells (MSC) to differentiate into neural lineages has raised the possibility of autologous cell transplantation as a therapy for neurodegenerative diseases. We have identified a population of circulating human fetal mesenchymal stem cells (hfMSC) that are highly proliferative and can readily differentiate into mesodermal lineages such as bone, cartilage, fat and muscle. Here, we demonstrate for the first time that primary hfMSC can differentiate into cells with an oligodendrocyte phenotype both in vitro and in vivo. By exposing hfMSC to neuronal conditioned medium or by introducing the pro-oligodendrocyte gene, Olig-2, hfMSC adopted an oligodendrocyte-like morphology, expressed oligodendrocyte markers and appeared to mature appropriately in culture. Importantly we also demonstrate the differentiation of a clonal population of hfMSC into both mesodermal (bone) and ectodermal (oligodendrocyte) lineages. In the developing murine brain transplanted hfMSC integrated into the parenchyma but oligodendrocyte differentiation of these naïve hfMSC was very low. However, the proportion of cells expressing oligodendrocyte markers increased significantly (from 0.2% to 4%) by preexposing the cells to differentiation medium in vitro prior to transplantation. Importantly, the process of in vivo differentiation occurred without cell fusion. These findings suggest that hfMSC may provide a potential source of oligodendrocytes for study and potential therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Mesenchymal Stem Cells/physiology , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Oligodendroglia/physiology , Animals , Brain/cytology , Brain/growth & development , Culture Media, Conditioned/pharmacology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Transcription Factors/metabolism , Zebrafish Proteins
3.
Cell Tissue Res ; 327(1): 67-82, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16868787

ABSTRACT

The alpha- and beta-dystrobrevins (DBs) belong to a family of dystrophin-related and dystrophin-associated proteins that are members of the dystrophin-associated protein complex (DAPC). This complex provides a link between the cytoskeleton and the extracellular matrix or other cells. However, specific functions of the two dystrobrevins remain largely unknown, with alpha-DB being believed to have a role mainly in skeletal muscle. Here, we describe previously unknown expression patterns and the localisation and molecular characteristics of alpha-DB isoforms in non-muscle mouse tissues. We demonstrate a highly specific sub-cellular distribution of alpha-DB in organs forming blood-tissue barriers. We show alpha-DB expression and localisation in testicular Sertoli cells, stomach and respiratory epithelia and provide electron-microscopic evidence for its immunolocalisation in these cells and in the central nervous system. Moreover, we present the molecular characterisation of alpha-DB transcript in these tissues and provide evidence for a distinct heterogeneity of associations between alpha-DB and dystrophins and utrophin in normal and dystrophic non-muscle tissues. Together, our results indicate that alpha-DB, in addition to its role in skeletal muscle, may also be required for the proper function of specific non-muscle tissues and that disruption of DAPC might lead to tissue-blood barrier abnormalities.


Subject(s)
Dystrophin-Associated Proteins/metabolism , Epithelium/metabolism , Gastric Mucosa/metabolism , Muscular Dystrophy, Duchenne/metabolism , Sertoli Cells/metabolism , Animals , Blood-Air Barrier/metabolism , Blood-Air Barrier/ultrastructure , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/ultrastructure , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/ultrastructure , Blood-Testis Barrier/metabolism , Blood-Testis Barrier/ultrastructure , Disease Models, Animal , Dystrophin-Associated Proteins/genetics , Epithelium/ultrastructure , Fluorescent Antibody Technique, Indirect , Gastric Mucosa/ultrastructure , Gene Expression , Gene Silencing , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Duchenne/pathology , RNA, Messenger/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/ultrastructure , Sertoli Cells/ultrastructure
4.
FASEB J ; 20(6): 610-20, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581969

ABSTRACT

Pathological cellular hallmarks of Duchenne muscular dystrophy (DMD) include, among others, abnormal calcium homeostasis. Changes in the expression of specific receptors for extracellular ATP in dystrophic muscle have been recently documented: here, we demonstrate that at the earliest, myoblast stage of developing dystrophic muscle a purinergic dystrophic phenotype arises. In myoblasts of a dystrophin-negative muscle cell line established from the mdx mouse model of DMD but not in normal myoblasts, exposure to extracellular ATP triggered a strong increase in cytoplasmic Ca2+ concentrations. Influx of extracellular Ca2+ was stimulated by ATP and BzATP and inhibited by zinc, Coomassie Brilliant Blue-G, and KN-62, demonstrating activation of P2X7 receptors. Significant expression of P2X4 and P2X7 proteins was immunodetected in dystrophic myoblasts. Therefore, full-length dystrophin appears, surprisingly, to play an important role in myoblasts in controlling responses to ATP. Our results suggest that altered function of P2X receptors may be an important contributor to pathogenic Ca2+ entry in dystrophic mouse muscle and may have implications for the pathogenesis of muscular dystrophies. Treatments aiming at inhibition of specific ATP receptors could be of a potential therapeutic benefit.


Subject(s)
Adenosine Triphosphate/pharmacology , Myoblasts, Skeletal/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Line , Dystrophin/metabolism , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal , RNA, Messenger/metabolism , Receptors, Purinergic P2/genetics , Utrophin/metabolism
5.
Neuromuscul Disord ; 15(3): 225-36, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15725584

ABSTRACT

Using a combination of molecular and immunohistochemical methods, we have obtained evidence for a distinctive change in the expression patterns of ATP-gated (P2X) receptor subunits in dystrophic muscle from both Duchenne muscular dystrophy (DMD) patients and the mdx mouse model. In control myofibres there was no staining for any P2X subtype studied here, although P2X1 stained the smooth muscle of the blood vessels and P2X6 nerves and the tunica intima in small arteries. In contrast, P2X1 and P2X6 were co-expressed strongly in small regenerating muscle fibres in the dystrophic muscles, whereas this expression decreased in fully regenerated fibres. Moreover, immunoreactivity for the P2X2 receptor re-appeared in dystrophic muscle, where it co-localised with the Type 1 fibres. There is, thus, a burst of production of several P2X receptor subtypes in regenerating dystrophic muscle, which may have implications for drug targets for this muscle pathology.


Subject(s)
Dystrophin/deficiency , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Receptors, Purinergic P2/metabolism , Animals , Blotting, Northern/methods , Blotting, Western/methods , Embryo, Mammalian , Gene Expression Regulation , Humans , Immunohistochemistry/methods , Male , Methyl Green/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , RNA, Messenger/biosynthesis , Rats , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X2 , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment/methods , Sequence Analysis, Protein/methods , Succinate Dehydrogenase/metabolism , Vasoactive Intestinal Peptide/metabolism
6.
Neurobiol Dis ; 15(2): 212-20, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006691

ABSTRACT

Following molecular and immunohistochemical analysis of the purinergic P2X4 receptor subunit in dystrophin-deficient muscle we have identified a distinct subpopulation of P2X4-positive cells infiltrating the dystrophic fibres. These cells were absent from normal muscle and rarely present in the dystrophic muscle taken before and after the onset of degeneration. We have identified these P2X4-positive cells as macrophages, demonstrating for the first time that human and mouse tissue macrophages express P2X4 in addition to P2X7 receptor subunits both in vitro and in situ. Moreover, we have demonstrated that the increase in the P2X4 expression is yet another feature of an inflammatory response identified in DNA arrays of dystrophic muscle. Immunohistochemical analysis failed to localise discernible expression of P2X4 protein in adult skeletal or cardiac muscle fibres, whilst myoblasts in culture expressed low levels of this subunit, as detected by RT-PCR and Western blotting. In light of the involvement of macrophages in the dystrophic process, the function of P2X receptors and their role in the Duchenne pathology as well as their potential role in therapeutic applications are discussed.


Subject(s)
Inflammation/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Receptors, Purinergic P2/metabolism , Alternative Splicing/genetics , Animals , Cells, Cultured , Disease Models, Animal , Humans , Immunohistochemistry , Infant , Infant, Newborn , Inflammation/pathology , Inflammation/physiopathology , Macrophages/cytology , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Mutation/genetics , Myoblasts/cytology , Myoblasts/metabolism , Myocardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X4 , Receptors, Purinergic P2X7 , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...