Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 371
Filter
1.
Food Chem Toxicol ; 77: 120-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25592784

ABSTRACT

A toxicity/toxicokinetic swine-adapted infant formula feeding study was conducted in Domestic Yorkshire Crossbred Swine from lactation day 3 for 28 consecutive days during the preweaning period at carrageenan concentrations of 0, 300, 1000 and 2250 ppm under GLP guidelines. This study extends the observations in newborn baboons (McGill et al., 1977) to piglets and evaluates additional parameters: organ weights, clinical chemistry, special gastrointestinal tract stains (toluidine blue, Periodic Acid-Schiff), plasma levels of carrageenan; and evaluation of potential immune system effects. Using validated methods, immunophenotyping of blood cell types (lymphocytes, monocytes, B cells, helper T cells, cytotoxic T cells, mature T cells), sandwich immunoassays for blood cytokine evaluations (IL-6, IL-8, IL1ß, TNF-α), and immunohistochemical staining of the gut for IL-8 and TNF-α were conducted. No treatment-related adverse effects at any carrageenan concentration were found on any parameter. Glucosuria in a few animals was not considered treatment-related. The high dose in this study, equivalent to ~430 mg/kg/day, provides an adequate margin of exposure for human infants, as affirmed by JECFA and supports the safe use of carrageenan for infants ages 0-12 weeks and older and infants with special medical needs.


Subject(s)
Carrageenan/pharmacokinetics , Gastrointestinal Tract/drug effects , Immune System/drug effects , Infant Formula/chemistry , Animals , Animals, Newborn , Body Weight/drug effects , Carrageenan/adverse effects , Carrageenan/blood , Dose-Response Relationship, Drug , Female , Interleukin-1beta/blood , Interleukin-6/blood , Interleukin-8/blood , Male , Organ Size/drug effects , Swine , Toxicity Tests , Tumor Necrosis Factor-alpha/blood
2.
J Vet Intern Med ; 25(5): 1089-96, 2011.
Article in English | MEDLINE | ID: mdl-21781161

ABSTRACT

OBJECTIVES: To describe the clinical phenotype of a new motor disorder in Labrador Retrievers. ANIMALS AND METHODS: Case series study. Seven young male Labrador Retrievers presented for evaluation of stiff gait. RESULTS: All affected dogs had generalized muscular stiffness, persistent at rest and resulting in restricted joint movements. They showed a forward flexed posture, festinating gait, and bradykinesia. Signs developed between 2 and 16 months of age and tended to stabilize in adulthood. Needle electromyogram in the conscious state showed continuous motor unit activity in resting epaxial and proximal limb muscles. This activity was abolished by general anesthesia. Muscle and nerve histopathology was normal. In 2 dogs necropsied, astrocytosis was evident throughout the spinal cord gray matter, reticular formation and caudate nuclei. Decreased neuronal counts were selectively found in the spinal cord Rexed's lamina VII, but not in VIII and IX. Pedigree analysis showed that the affected dogs were from 5 related litters. CONCLUSIONS AND CLINICAL IMPORTANCE: This new hypertonicity syndrome in Labrador Retrievers is unique because of the selective distribution of the histological lesions, the lack of progression in adulthood, and its exclusive occurrence in male dogs. Pedigree analysis suggests an X-linked hereditary disease, although other modes of inheritance cannot be ruled out with certainty. We hypothesize that altered output from basal nuclei and reticular formation together with motor neuron disinhibition caused by a decreased number of spinal cord interneurons leads to the muscular stiffness.


Subject(s)
Dog Diseases/genetics , Movement Disorders/veterinary , Muscle Rigidity/veterinary , Animals , Dog Diseases/diagnosis , Dog Diseases/physiopathology , Dogs , Electromyography/veterinary , Gait/physiology , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/physiopathology , Genetic Diseases, X-Linked/veterinary , Male , Movement Disorders/diagnosis , Movement Disorders/genetics , Movement Disorders/physiopathology , Muscle Rigidity/diagnosis , Muscle Rigidity/genetics , Muscle Rigidity/physiopathology , Muscle, Skeletal/pathology , Pedigree
3.
Cell Transplant ; 17(7): 865-73, 2008.
Article in English | MEDLINE | ID: mdl-19044212

ABSTRACT

The adult bone marrow contains a population of multipotent mesenchymal stromal cells (MSCs), defined by plastic adherence, expression of stromal cell surface markers, and differentiation into mesenchymal lineages. There has been much interest in the possible therapeutic use of MSCs in the treatment of demyelinating diseases of the central nervous system. One therapeutic possibility is that these cells may be able to remyelinate when directly injected into the demyelinated spinal cord. Here we examine the effects of direct transplantation of green fluorescent protein (GFP)-labeled MSCs into a model of focal spinal cord demyelination induced by ethidium bromide. We demonstrate that direct intralesional injection of undifferentiated MSCs does not lead to remyelination. Furthermore, we report that transplanted MSCs migrate into areas of normal tissue, deposit collagen, and are associated with axonal damage. These findings support the need for further experimental evaluation of the safety and efficacy of direct parenchymal injection of MSCs into demyelinated lesions and highlight an important issue regarding potential clinical consequences of culture heterogeneity of MSCs between centers.


Subject(s)
Demyelinating Diseases , Mesenchymal Stem Cell Transplantation , Spinal Cord , Animals , Biomarkers/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/therapy , Disease Models, Animal , Green Fluorescent Proteins , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Cord/pathology
4.
Brain ; 131(Pt 6): 1464-77, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18490361

ABSTRACT

In multiple sclerosis, demyelination of the CNS axons is associated with axonal injury and degeneration, which is now accepted as the major cause of neurological disability in the disease. Although the kinetics and the extent of axonal damage have been described in detail, the mechanisms by which it occurs are as yet unclear; one suggestion is failure of remyelination. The goal of this study was to test the hypothesis that failure of prompt remyelination contributes to axonal degeneration following demyelination. Remyelination was inhibited by exposing the brain to 40 Gy of X-irradiation prior to cuprizone intoxication and this resulted in a significant increase in the extent of axonal degeneration and loss compared to non-irradiated cuprizone-fed mice. To exclude the possibility that this increase was a consequence of the X-irradiation and to highlight the significance of remyelination, we restored remyelinating capacity to the X-irradiated mouse brain by transplanting of GFP-expressing embryo-derived neural progenitors. Restoring the remyelinating capacity in these mice resulted in a significant increase in axon survival compared to non-transplanted, X-irradiated cuprizone-intoxicated mice. Our results support the concept that prompt remyelination protects axons from demyelination-associated axonal loss and that remyelination failure contributes to the axon loss that occurs in multiple sclerosis.


Subject(s)
Axons/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/physiology , Nerve Regeneration , Animals , Corpus Callosum/pathology , Cuprizone , Demyelinating Diseases , Diffusion Magnetic Resonance Imaging , Female , Green Fluorescent Proteins/analysis , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Multiple Sclerosis/pathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Stem Cell Transplantation , X-Rays
5.
J Comp Pathol ; 138(2-3): 160-4, 2008.
Article in English | MEDLINE | ID: mdl-18295785

ABSTRACT

l-2-Hydroxyglutaric aciduria (l-2-HGA) is a hereditary neurometabolic disorder reported in human beings and dogs. An 11-month-old Staffordshire bull terrier was suspected to have the disease, on the basis of clinical signs and magnetic resonance imaging findings. l-2-HGA was confirmed by urinary organic analysis and DNA testing and the dog was humanely destroyed. Post-mortem findings consisted only of microscopical lesions in the brain, characterized by marked spongiform changes and predominantly affecting the grey matter of the cerebral cortex, thalamus, cerebellum and brainstem. The spongiform changes were characterized by well-demarcated, clear vacuoles located at perineuronal and perivascular sites. Immunohistochemical and ultrastructural examination confirmed that the affected cells were astrocytes.


Subject(s)
Brain Diseases, Metabolic, Inborn/veterinary , Brain/pathology , Dog Diseases/pathology , Glutaryl-CoA Dehydrogenase/deficiency , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Biomarkers/metabolism , Brain/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic, Inborn/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , DNA Mutational Analysis , Dog Diseases/genetics , Dog Diseases/urine , Dogs , Euthanasia, Animal , Fatal Outcome , Glial Fibrillary Acidic Protein/metabolism , Glutarates/urine , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , Magnetic Resonance Imaging/veterinary , Male , Mutation , Vacuoles/ultrastructure
6.
Curr Top Microbiol Immunol ; 318: 193-212, 2008.
Article in English | MEDLINE | ID: mdl-18219819

ABSTRACT

Remyelination is the regenerative process by which demyelinated axons are reinvested with new myelin sheaths. It is associated with functional recovery and maintenance of axonal health. It occurs as a spontaneous regenerative response following demyelination in a range of pathologies including traumatic injury as well as primary demyelinating disease such as multiple sclerosis (MS). Experimental models of demyelination based on the use of toxins, while not attempting to accurately mimic a disease with complex etiology and pathogenesis such as MS, have nevertheless proven extremely useful for studying the biology of remyelination. In this chapter, we review the main toxin models of demyelination, drawing attention to their differences and how they can be used to study different aspects of remyelination. We also describe the optimal use of these models, highlighting potential pitfalls in interpretation, and how remyelination can be unequivocally recognized. Finally, we discuss the role of toxin models alongside viral and immune-mediated models of demyelination.


Subject(s)
Antibodies/pharmacology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Ethidium/pharmacology , Galactosylceramides/immunology , Lysophosphatidylcholines/pharmacology , Myelin Sheath/physiology , Animals , Antibodies/immunology , Cats , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Rabbits , Rats , Spinal Cord/pathology
7.
J Neurol Sci ; 265(1-2): 43-6, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17826797

ABSTRACT

The relative merits of endogenous and exogenous oligodendrocyte progenitor cells (OPCs) for remyelination are compared in terms of their ability to repopulate OPC-depleted tissue and generate remyelinating oligodendrocytes. Exogenous neonatal OPCs can repopulate OPC-depleted tissue 5-10 times faster than endogenous cells and as a result are capable of more extensive remyelination. Both endogenous and exogenous cells will only repopulate normal tissue if there is extensive depletion of the local OPC population and both show reduced ability to generate remyelinating cells in the absence of acute inflammation. When endogenous OPCs are depleted by X-irradiation during cuprizone intoxication, where there is a combination of astrocytosis and acute demyelination, endogenous but not exogenous embryo-derived OPCs fail to repopulate the OPC-depleted cortex.


Subject(s)
Demyelinating Diseases/therapy , Nerve Regeneration/physiology , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Humans , Stem Cell Transplantation
8.
J Neurol Sci ; 265(1-2): 1-4, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17459413

ABSTRACT

In order to devise a strategy to enhance remyelination in multiple sclerosis (MS) it is necessary to understand the cause of remyelination failure in MS. A case is made that areas of chronic demyelination arise because of concurrent loss of oligodendrocyte progenitor cells (OPCs) and oligodendrocytes and that because of the slow rate of repopulation that occurs in old individuals the recruited OPCs are not exposed to the acute inflammatory environment required to generate remyelinating oligodendrocytes. Based on this analysis the case is made that only areas of acute demyelination will be amenable to transplant-mediated remyelination. An analysis of the many cells that could be used to provide a source of remyelinating cells would indicate that structural repair of the CNS in MS would likely only be possible if neural precursors were used and the most promising route for their introduction would appear to be by intraventricular injection. Both neural precursors and mesenchymal stromal cells can be immunomodulatory and neuroprotective following intravenous injection; however, only neural precursors are likely to be able to contribute to structural repair of the damaged nervous system.


Subject(s)
Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Oligodendroglia/physiology , Regeneration/physiology , Stem Cells/physiology , Animals , Disease Models, Animal , Humans , Multiple Sclerosis/therapy , Stem Cell Transplantation
9.
Brain ; 130(Pt 8): 2175-85, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17550908

ABSTRACT

Cell transplant therapies are currently under active consideration for a number of degenerative diseases. In the immune-mediated demyelinating-neurodegenerative disease multiple sclerosis (MS), only the myelin sheaths of the CNS are lost, while Schwann cell myelin of the PNS remains normal. This, and the finding that Schwann cells can myelinate CNS axons, has focussed interest on Schwann cell transplants to repair myelin in MS. However, the experimental use of these cells for myelin repair in animal models has revealed a number of problems relating to the incompatibility between peripheral glial cells and the CNS glial environment. Here, we have tested whether these difficulties can be avoided by using an earlier stage of the Schwann cell lineage, the Schwann cell precursor (SCP). For direct comparison of these two cell types, we implanted Schwann cells from post-natal rat nerves and SCPs from embryo day 14 (E14) rat nerves into the CNS under various experimental conditions. Examination 1 and 2 months later showed that in the presence of naked CNS axons, both types of cell form myelin that antigenically and ultrastructurally resembles that formed by Schwann cells in peripheral nerves. In terms of every other parameter we studied, however, the cells in these two implants behaved remarkably differently. As expected from previous work, Schwann cell implants survive poorly unless the cells find axons to myelinate, the cells do not migrate significantly from the implantation site, fail to integrate with host oligodendrocytes and astrocytes, and form little myelin when challenged with astrocyte-rich environment in the retina. Following SCP implantation, on the other hand, the cells survive well, migrate through normal CNS tissue, interface smoothly and intimately with host glial cells and myelinate extensively among the astrocytes of the retina. Furthermore, when implanted at a distance from a demyelinated lesion, SCPs but not Schwann cells migrate through normal CNS tissue to reach the lesion and generate new myelin. These features of SCP implants are all likely to be helpful attributes for a myelin repair cell. Since these cells also form Schwann cell myelin that is arguably likely to be resistant to MS pathology, they share some of the main advantages of Schwann cells without suffering from the disadvantages that render Schwann cells less than ideal candidates for transplantation into MS lesions.


Subject(s)
Multiple Sclerosis/therapy , Myelin Sheath/physiology , Nerve Regeneration , Schwann Cells/transplantation , Stem Cell Transplantation/methods , Animals , Astrocytes/physiology , Cell Movement , Cell Survival , Female , Multiple Sclerosis/physiopathology , Rats , Rats, Sprague-Dawley , Retina/cytology , Schwann Cells/cytology , Schwann Cells/physiology , Spinal Cord/cytology
10.
Glia ; 54(8): 880-90, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17006890

ABSTRACT

Transplantation of oligodendrocyte precursor cells (OPCs) results in efficient remyelination in animal models of demyelination. However, the experiments so far undertaken have not addressed the need for tissue-type matching to achieve graft-mediated remyelination. Examination of MHC expression (main determinant of allograft rejection) by OPCs showed nondetectable levels under standard culture conditions and upregulation of MHC Class I expression only upon exposure to interferon gamma. We therefore hypothesized that MHC matching of OPC grafts may not be crucial to achieve transplant-mediated remyelination. Transplant experiments performed using a nonself repairing toxin-induced demyelination model showed that, similarly to allogeneic neurons, survival of allogeneic oligodendrocyte lineage cells is influenced by donor-host haplotype combination and graft composition. Transplantation of allogeneic mixed glial cell cultures resulted in remyelination failure by 1 month postengraftment due to a rejection response targeting both myelinating oligodendrocytes and OPCs, suggesting that inflammation-induced upregulation of OPC MHC I expression results in susceptibility to cytotoxic T cell attack. In contrast, remyelination persisted for at least 2 months following transplantation of OPC-enriched cultures whose overall MHC expression level was significantly decreased. While OPC-enriched preparations elicited delayed type hypersensitivity responses in hosts sensitized to alloantigens, allografting of such preparations into a central nervous system demyelinating lesion did not result in recipient priming. We conclude that while allografted oligodendrocyte lineage cells become targets of a graft rejection response once this response has been initiated, transplantation of OPC-enriched preparations can evade priming against alloantigens and graft rejection. This finding indicates that close tissue matching may not be an essential requirement for successful transplant-mediated remyelination.


Subject(s)
Demyelinating Diseases/therapy , Graft Rejection/immunology , Graft Survival/immunology , Nerve Regeneration/immunology , Oligodendroglia/physiology , Oligodendroglia/transplantation , Stem Cell Transplantation/methods , Stem Cells/immunology , Animals , Cells, Cultured , Coculture Techniques , Demyelinating Diseases/physiopathology , Disease Models, Animal , Down-Regulation/immunology , Female , Graft Rejection/prevention & control , Haplotypes/immunology , Histocompatibility Antigens/immunology , Histocompatibility Testing/methods , Histocompatibility Testing/standards , Immunity, Cellular/immunology , Immunosuppression Therapy/methods , Myelin Sheath/immunology , Oligodendroglia/cytology , Rats , Rats, Inbred Lew , Stem Cells/cytology , T-Lymphocytes/immunology
11.
J Neuroimmunol ; 175(1-2): 69-76, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16626812

ABSTRACT

Axon loss is recognised as a significant contributor to the progression of the disability associated with multiple sclerosis. Although evidence of axon damage is found in areas of chronic demyelination it is more frequently seen in association with acute demyelination. This study compares the incidence of axon degeneration associated with the areas undergoing demyelination in young adult (8-10 weeks) and aged (6-7 months) C57BL/6 mice in cuprizone intoxication; a widely used model of demyelination. The incidence of axon transection, as indicated by the presence of SMI 32 positive axonal spheroids, and evidence of axon loss in the medial corpus callosum, were significantly greater in aged mice, as was the magnitude of the macrophage and astrocyte response to demyelination. Aged C57BL/6 mice are thus more prone to axon degeneration in association with demyelination than young adult mice. A retrospective study indicated that the incidence of axon degeneration was much higher in C57BL/6 mice than in the Swiss albino mice used in the early cuprizone intoxication studies which were fed much higher doses of cuprizone. These results indicate both a genetic and age susceptibility to demyelination-associated axon transection.


Subject(s)
Aging/immunology , Axons/immunology , Axons/pathology , Cuprizone/toxicity , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Aging/drug effects , Animals , Axons/drug effects , Axons/ultrastructure , Corpus Callosum/drug effects , Corpus Callosum/immunology , Corpus Callosum/pathology , Corpus Callosum/ultrastructure , Demyelinating Diseases/chemically induced , Demyelinating Diseases/physiopathology , Female , Mice , Mice, Inbred C57BL
12.
Philos Trans R Soc Lond B Biol Sci ; 360(1461): 1775-95, 2005 Sep 29.
Article in English | MEDLINE | ID: mdl-16147541

ABSTRACT

Remyelination of demyelinated central nervous system (CNS) axons is considered as a potential treatment for multiple sclerosis, and it has been achieved in experimental models of demyelination by transplantation of pro-myelinating cells. However, the experiments undertaken have not addressed the need for tissue-type matching in order to achieve graft-mediated remyelination since they were performed in conditions in which the chance for graft rejection was minimized. This article focuses on the factors determining survival of allogeneic oligodendrocyte lineage cells and their contribution to the remyelination of demyelinating CNS lesions. The immune status of the CNS as well as the suitability of different models of demyelination for graft rejection studies are discussed, and ways of enhancing allogeneic oligodendrocyte-mediated remyelination are presented. Finally, the effects of glial graft rejection on host remyelination are described, highlighting the potential benefits of the acute CNS inflammatory response for myelin repair.


Subject(s)
Central Nervous System/immunology , Demyelinating Diseases/therapy , Graft Rejection/immunology , Immunity, Cellular , Models, Immunological , Oligodendroglia/cytology , Oligodendroglia/transplantation , Central Nervous System/metabolism , Demyelinating Diseases/immunology , Humans , Lymphocytes/immunology , Major Histocompatibility Complex/immunology
13.
Neuropathol Appl Neurobiol ; 31(4): 374-83, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16008821

ABSTRACT

Some, but not all, chronically demyelinated multiple sclerosis (MS) lesions are depleted of oligodendrocyte progenitor cells (OPCs) suggesting that OPCs are destroyed during the process of demyelination and some factor impedes OPC repopulation of the depleted tissue. The chronically demyelinated axons in MS lie in an astrocytic environment and it has been proposed that this might impede entry of OPCs into such regions. By depleting a short length of spinal cord of its OPCs using 40 Gy of X-irradiation in both normal rats and rats with progressive myelin loss accompanied by an astrocytosis (taiep rats), we investigated whether such changes affect the ability of OPCs to repopulate OPC-depleted tissue. In both taiep and normal rats, the rate of repopulation decreases with age, but no difference was detected in the rate at which OPCs repopulated normally myelinated and chronically demyelinated and astrocytosed tissue. This indicates that, if the astrocytic environment of the taiep central nervous system (CNS) is comparable to that found in MS lesions, then the presence of chronically demyelinated axons and astrocytosis in chronic MS lesions does not represent a barrier to repopulation of the tissue by OPCs. However, similar to the situation in the normal adult rodent CNS, the rate of repopulation by endogenous OPCs in aged taiep rats is very slow, approximately 0.2 mm per week.


Subject(s)
Gliosis/pathology , Multiple Sclerosis/pathology , Oligodendroglia/cytology , Spinal Cord/cytology , Spinal Cord/radiation effects , Stem Cells/cytology , Age Factors , Animals , Brain/cytology , Brain/pathology , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Oligodendroglia/radiation effects , Rats , Spinal Cord/pathology , Stem Cells/radiation effects
14.
Neuropathol Appl Neurobiol ; 31(2): 105-14, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15771704

ABSTRACT

Some, but not all chronically demyelinated MS lesions are depleted of oligodendrocyte progenitor cells (OPCs) suggesting that OPCs are destroyed during the process of demyelination and some factor impedes OPC repopulation of the depleted tissue. The chronically demyelinated axons in MS lie in an astrocytic environment and it has been proposed that this might impede entry of OPCs into such regions. By depleting a short length of spinal cord of its OPCs using 40 Gy of X-irradiation in both normal rats and rats with progressive myelin loss accompanied by an astrocytosis (taiep rats), we investigated whether such changes affect the ability of OPCs to repopulate OPC-depleted tissue. In both taiep and normal rats, the rate of repopulation decreases with age, but no difference was detected in the rate at which OPCs repopulated normally myelinated and chronically demyelinated and astrocytosed tissue. This indicates that, if the astrocytic environment of the taiep CNS is comparable to that found in MS lesions, then the presence of chronically demyelinated axons and astrocytosis in chronic MS lesions does not represent a barrier to repopulation of the tissue by OPCs. However, similar to the situation in the normal adult rodent CNS, the rate of repopulation by endogenous OPCs in aged taiep rats is very slow, approximately 0.2 mm per week.


Subject(s)
Demyelinating Diseases/pathology , Oligodendroglia/cytology , Oligodendroglia/physiology , Spinal Cord/pathology , Stem Cells/physiology , Age Factors , Animals , Disease Models, Animal , Female , Gliosis/pathology , Male , Multiple Sclerosis/pathology , Oligodendroglia/radiation effects , Rats , Spinal Cord/radiation effects
15.
Brain ; 128(Pt 3): 528-39, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15699059

ABSTRACT

A major challenge in multiple sclerosis research is to understand the cause or causes of remyelination failure and to devise ways of ameliorating its consequences. This requires appropriate experimental models. Although there are many models of acute demyelination, at present there are few suitable models of chronic demyelination. The taiep rat is a myelin mutant that shows progressive myelin loss and, by 1 year of age, its CNS tissue has many features of chronic areas of demyelination in multiple sclerosis: chronically demyelinated axons present in an astrocytic environment in the absence of acute inflammation. Using the taiep rat and a combination of X-irradiation and cell transplantation, it has been possible to address a number of questions concerning remyelination failure in chronic multiple sclerosis lesions, such as whether chronically demyelinated axons have undergone changes that render them refractory to remyelination and why remyelination is absent when oligodendrocyte progenitor cells (OPCs) are present. Our experiments show that (i) transplanted OPCs will not populate OPC-containing areas of chronic demyelination; (ii) myelination competent OPCs can repopulate OPC-depleted chronically demyelinated astrocytosed tissue, but this repopulation does not result in remyelination--closely resembling the situation found in some multiple sclerosis plaques; and (iii) the induction of acute inflammation in this non-remyelinating situation results in remyelination. Thus, we can conclude that axonal changes induced by chronic demyelination are unlikely to contribute to remyelination failure in multiple sclerosis. Rather, remyelination fails either because OPCs fail to repopulate areas of demyelination or because if OPCs are present they are unable to generate remyelinating oligodendrocytes owing to the presence of inhibitory factors and/or a lack of the stimuli required to activate these cells to generate remyelinating oligodendrocytes. This non-remyelinating situation can be transformed to a remyelinating one by the induction of acute inflammation.


Subject(s)
Inflammation/physiopathology , Multiple Sclerosis/physiopathology , Myelin Sheath/physiology , Nerve Regeneration , Acute Disease , Animals , Cell Division , Chronic Disease , Disease Models, Animal , Disease Progression , Multiple Sclerosis/pathology , Myelin Sheath/genetics , Myelin Sheath/ultrastructure , Oligodendroglia/physiology , Oligodendroglia/transplantation , Oligodendroglia/ultrastructure , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Spinal Cord/ultrastructure , Stem Cell Transplantation , Stem Cells/physiology
16.
Neuropathol Appl Neurobiol ; 31(1): 1-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15634226

ABSTRACT

In certain experimental and naturally occurring pathological situations in the central nervous system (CNS), demyelinated axons are remyelinated by Schwann cells. It has always been assumed that these Schwann cells are derived from Schwann cells associated with peripheral nerves. However, it has become apparent that CNS precursors can give rise to Schwann cells in vitro and following transplantation into astrocyte-free areas of demyelination in vivo. This paper compares the behaviour of remyelinating Schwann cells following transplantation of peripheral nerve derived Schwann cells over, and into, astrocyte-depleted areas of demyelination to that which follows transplantation of CNS cells and that seen in normally remyelinating ethidium bromide induced demyelinating lesions. It concludes that while the examination of normally remyelinating lesions can not resolve the origin of the remyelinating Schwann cells, the results from transplantation studies provide strong evidence that the Schwann cells that remyelinate CNS axons are most likely generated from CNS precursors. In addition these studies also indicate that the precursors that give rise to these Schwann cells are the same cells that give rise to remyelinating oligodendrocytes.


Subject(s)
Astrocytes/cytology , Axons/ultrastructure , Central Nervous System/cytology , Myelin Sheath/physiology , Oligodendroglia/cytology , Schwann Cells/cytology , Animals , Cell Lineage , Central Nervous System/physiology , Humans , Myelin Sheath/ultrastructure , Schwann Cells/ultrastructure , Stem Cells/cytology , Stem Cells/physiology
17.
Eur J Neurosci ; 20(6): 1445-60, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15355312

ABSTRACT

There is a long-standing controversy as to whether oligodendrocytes may be capable of cell division and thus contribute to remyelination. We recently published evidence that a subpopulation of myelin oligodendrocyte glycoprotein (MOG)-expressing cells in the adult rat spinal cord co-expressed molecules previously considered to be restricted to oligodendrocyte progenitors [G. Li et al. (2002) Brain Pathol., 12, 463-471]. To further investigate the properties of MOG-expressing cells, anti-MOG-immunosorted cells were grown in culture and transplanted into acute demyelinating lesions. The immunosorting protocol yielded a cell preparation in which over 98% of the viable cells showed anti-MOG- and O1-immunoreactivity; 12-15% of the anti-MOG-immunosorted cells co-expressed platelet-derived growth factor alpha receptor (PDGFRalpha) or the A2B5-epitope. When cultured in serum-free medium containing EGF and FGF-2, 15-18% of the anti-MOG-immunosorted cells lost anti-MOG- and O1-immunoreactivity and underwent cell division. On removal of these growth factors, cells differentiated into oligodendrocytes, or astrocytes and Schwann cells when the differentiation medium contained BMPs. Transplantation of anti-MOG-immunosorted cells into areas of acute demyelination immediately after isolation resulted in the generation of remyelinating oligodendrocytes and Schwann cells. Our studies indicate that the adult rat CNS contains a significant number of oligodendrocyte precursors that express MOG and galactocerebroside, molecules previously considered restricted to mature oligodendrocytes. This may explain why myelin-bearing oligodendrocytes were considered capable of generating remyelinating cells. Our study also provides evidence that the adult oligodendrocyte progenitor can be considered as a source of the Schwann cells that remyelinate demyelinated CNS axons following concurrent destruction of oligodendrocytes and astrocytes.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/cytology , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Regeneration/physiology , Oligodendroglia/physiology , Animals , Axons/drug effects , Axons/physiology , Axons/ultrastructure , Benzimidazoles/metabolism , Blotting, Western/methods , Bone Morphogenetic Proteins/pharmacology , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , Cyclophilins/genetics , Cyclophilins/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/therapy , Epidermal Growth Factor/pharmacology , Ethidium , Female , Flow Cytometry/methods , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Vitro Techniques , Intermediate Filament Proteins/metabolism , Microscopy, Electron, Scanning Transmission/methods , Myelin Proteins , Myelin Sheath/radiation effects , Myelin Sheath/ultrastructure , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Nerve Tissue Proteins/metabolism , Nestin , Octamer Transcription Factor-6 , Oligodendroglia/ultrastructure , Rats , Receptors, Platelet-Derived Growth Factor/metabolism , Stem Cells/physiology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transplants
18.
J Neurotrauma ; 21(8): 1044-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15319003

ABSTRACT

Following spinal cord trauma there is controversy as to whether myelin-supporting oligodendrocytes at a distance from areas of spinal cord damage undergo apoptosis. To examine the response of oligodendrocytes to axon degeneration, we counted the number of oligodendrocytes and oligodendrocyte precursors in the dorsal funiculi during the course of Wallerian degeneration. Axons were disrupted at T13 and the number of labelled cells in the dorsal funiculi counted at T12, 4 days and 2, 4, and 8 weeks after injury using riboprobes to exon-3b of the PLP gene whose expression is considered to restricted to myelin-supporting oligodendrocytes, PDGFRalpha which is regarded as a marker of oligodendrocyte precursors, and MOG a marker previously used to identify myelin-supporting oligodendrocytes. We found that the number of PLP-exon-3b labelled cells remained constant during the course of Wallerian degeneration while the number of cells labelled with the riboprobes to PDGFRalpha and MOG increased. Significantly the number of MOG-positive cells was increased at times when the number of PDGFRalpha labelled cells was highest. The number of PDGFRalpha labelled cells decreased with time while the number PLP-exon-3b labelled cells remained constant. It is therefore possible that the apoptotic oligodendrocytes identified in previous studies could represent degenerating oligodendrocyte precursors or their progeny rather than degenerating myelin-supporting oligodendrocytes.


Subject(s)
Myelin Sheath/metabolism , Oligodendroglia/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Stem Cells/metabolism , Wallerian Degeneration/metabolism , Animals , Biomarkers , Cell Count , DNA Probes , Exons/genetics , Female , Gene Expression Regulation/genetics , Genetic Markers/genetics , Myelin Proteins , Myelin Proteolipid Protein/genetics , Myelin-Associated Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein , Rats , Receptor, Platelet-Derived Growth Factor beta/genetics , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Up-Regulation/genetics , Wallerian Degeneration/genetics
19.
J Neuropathol Exp Neurol ; 62(9): 908-16, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14533780

ABSTRACT

Rates of remyelination decline with age and this has been attributed to slower recruitment of oligodendrocyte progenitor cells (OPCs) into areas of demyelination and slower differentiation of OPCs into remyelinating oligodendrocytes. When considering causes for reduced recruitment rates, intrinsic causes (alterations in biological properties of OPCs) need to be separated from extrinsic causes (age-related differences in the lesion environment). Using 40 Gy of X-irradiation to deplete tissue of its endogenous OPC-population, we examined the effects of age on the rate at which adult rat OPCs colonize OPC-depleted tissue. We found a significant reduction in the rate of colonization between 2 and 10 months of age (0.6 mm/week versus 0.38 mm/week). To determine if this represented an intrinsic property of OPCs or was due to changes in the environment that the cells were recolonizing, OPCs from 10-month-old animals were transplanted into 2-month-old hosts and OPCs from 2-month-old animals were transplanted into 10-month-old hosts. These experiments showed that the transplanted OPCs retained their age-related rate of colonization, indicating that the decline in colonizing rates of OPCs with age reflects an intrinsic property of OPCs. This age-related decline in the ability of OPCs to repopulate OPC-depleted tissue has implications for understanding remyelination failure in multiple sclerosis (MS) and developing therapies for remyelination failure.


Subject(s)
Aging/pathology , Oligodendroglia/cytology , Stem Cells/cytology , Animals , Male , Rats , Stem Cell Transplantation/methods
20.
J Neurosci Res ; 73(6): 787-93, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12949904

ABSTRACT

We have attempted to extend a previously described rat model of focal oligodendrocyte progenitor cell (OPC) depletion, using 40 Gy X-irradiation (Chari and Blakemore [2002] Glia 37:307-313), to the adult mouse spinal cord, to examine the ability of OPCs present in adjacent normal areas to colonise areas of progenitor depletion. In contrast to rat, OPCs in the mouse spinal cord appeared to be a comparatively radiation-resistant population, as 30-35% of OPCs survived in X-irradiated tissue (whereas <1% of OPCs survive in X-irradiated rat spinal cord). The numbers of surviving OPCs remained constant with time indicating that this population was incapable of regenerating itself in response to OPC loss. Additionally, these OPCs did not contribute to remyelination of axons when demyelinating lesions were placed in X-irradiated tissue, suggesting that the surviving cells are functionally impaired. Importantly, the length of the OPC-depleted area did not diminish with time, as would be expected if progressive repopulation of OPC-depleted areas by OPCs from normal areas was occurring. Our findings therefore raise the possibility that the presence of a residual dysfunctional OPC population may inhibit colonisation of such areas by normal OPCs.


Subject(s)
Oligodendroglia/physiology , Regeneration/physiology , Spinal Cord/metabolism , Stem Cells/physiology , Animals , Antigens/metabolism , Cell Count , Cell Survival/radiation effects , Cells, Cultured , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Enzyme Inhibitors/toxicity , Ethidium/toxicity , Female , Immunohistochemistry/methods , In Situ Hybridization/methods , Mice , Mice, Inbred C57BL , Microscopy, Electron/instrumentation , Microscopy, Electron/methods , Oligodendroglia/metabolism , Oligodendroglia/radiation effects , Oligodendroglia/ultrastructure , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Schwann Cells/metabolism , Schwann Cells/radiation effects , Schwann Cells/ultrastructure , Spinal Cord/cytology , Spinal Cord/radiation effects , Spinal Cord/ultrastructure , Stem Cells/metabolism , Stem Cells/radiation effects , Time Factors , X-Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL