Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cells ; 13(9)2024 May 04.
Article in English | MEDLINE | ID: mdl-38727321

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deficiency of the survival motor neuron (SMN) protein. Although SMA is a genetic disease, environmental factors contribute to disease progression. Common pathogen components such as lipopolysaccharides (LPS) are considered significant contributors to inflammation and have been associated with muscle atrophy, which is considered a hallmark of SMA. In this study, we used the SMNΔ7 experimental mouse model of SMA to scrutinize the effect of systemic LPS administration, a strong pro-inflammatory stimulus, on disease outcome. Systemic LPS administration promoted a reduction in SMN expression levels in CNS, peripheral lymphoid organs, and skeletal muscles. Moreover, peripheral tissues were more vulnerable to LPS-induced damage compared to CNS tissues. Furthermore, systemic LPS administration resulted in a profound increase in microglia and astrocytes with reactive phenotypes in the CNS of SMNΔ7 mice. In conclusion, we hereby show for the first time that systemic LPS administration, although it may not precipitate alterations in terms of deficits of motor functions in a mouse model of SMA, it may, however, lead to a reduction in the SMN protein expression levels in the skeletal muscles and the CNS, thus promoting synapse damage and glial cells' reactive phenotype.


Subject(s)
Disease Models, Animal , Lipopolysaccharides , Muscular Atrophy, Spinal , Animals , Lipopolysaccharides/pharmacology , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/metabolism , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 1 Protein/genetics , Mice, Inbred C57BL , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Inflammation/pathology
2.
Neurol Res ; 42(3): 209-221, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32048570

ABSTRACT

Objectives: Natalizumab (NTZ), a treatment indicated for patients with highly active Relapsing - Remitting Multiple Sclerosis (RRMS), is known to induce increased relative frequency of lymphocytes. Progressive Multifocal Leukoencephalitis (PML) is a rare but serious adverse event related to NTZ. Moreover, reduced L-selectin (CD62L) expression in T-cells in cryopreserved samples of patients with RRMS under NTZ has been proposed as a biomarker of pre-PML state. We explore the association between L-selectin expression in T-cells and hematological parameters in freshly processed samples of patients with RRMS under NTZ.Methods: We studied L-selectin expression in patients with: RRMS under NTZ (n=34), fingolimod (FTY, n=14), interferon-beta (IFNß, n=22), glatiramer acetate (GA, N=17); in 9 patients with secondary progressive (SP) MS and in 6 healthy controls. Twenty-two patients under NTZ and 6 patients under FTY were followed for 18 months. One NTZ-treated patient developed PML during the study.Results: Patients under NTZ exhibited increased relative frequency of lymphocytes (40.02±1.45) compared to patients under first-line treatment (30.57±1.68, p<0.001) and to patients with SPMS (29±1.56, p=0.02), and a lower mean L-selectin expression in (69.39±1.73) compared to patients under first-line treatment (79.1±1.17, p=0.003). A negative correlation between the relative frequency of CD4+CD62L+ T-cells and the absolute lymphocyte counts (Pearson's r=0.367, p=0.033) was observed.Discussion: We hereby provide mechanistic insight in a possible pathway implicated in NTZ-related PML risk. These results further underline the need for thorough validation of L-selectin expression in T-cells as a potential pre-PML biomarker.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Immunologic Factors/adverse effects , L-Selectin/metabolism , Leukoencephalopathy, Progressive Multifocal/chemically induced , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Natalizumab/adverse effects , Adult , Brain/drug effects , Brain/pathology , Female , Humans , Leukoencephalopathy, Progressive Multifocal/immunology , Leukoencephalopathy, Progressive Multifocal/pathology , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Risk Factors
3.
Prion ; 7(6): 488-95, 2013.
Article in English | MEDLINE | ID: mdl-24247293

ABSTRACT

Prions are notorious for their extraordinary resistance to traditional methods of decontamination, rendering their transmission a public health risk. Iatrogenic Creutzfeldt-Jakob disease (iCJD) via contaminated surgical instruments and medical devices has been verified both experimentally and clinically. Standard methods for prion inactivation by sodium hydroxide or sodium hypochlorite have failed, in some cases, to fully remove prion infectivity, while they are often impractical for routine applications. Prion accumulation in peripheral tissues and indications of human-to-human bloodborne prion transmission, highlight the need for novel, efficient, yet user-friendly methods of prion inactivation. Here we show both in vitro and in vivo that homogenous photocatalytic oxidation, mediated by the photo-Fenton reagent, has the potential to inactivate the pathological prion isoform adsorbed on metal substrates. Photocatalytic oxidation with 224 µg mL(-1) Fe (3+), 500 µg mL(-1) h(-1) H 2O 2, UV-A for 480 min lead to 100% survival in golden Syrian hamsters after intracranial implantation of stainless steel wires infected with the 263K prion strain. Interestingly, photocatalytic treatment of 263K infected titanium wires, under the same experimental conditions, prolonged the survival interval significantly, but failed to eliminate infectivity, a result that we correlate with the increased adsorption of PrP(Sc) on titanium, in comparison to stainless steel. Our findings strongly indicate that our, user--and environmentally--friendly protocol can be safely applied to the decontamination of prion infected stainless steel surfaces.


Subject(s)
Decontamination/methods , Prions/isolation & purification , Prostheses and Implants/virology , Stainless Steel/chemistry , Titanium/chemistry , Animals , Catalysis , Cricetinae , Humans , Hydrogen Peroxide/chemistry , Iron/chemistry , Mesocricetus , Oxidants, Photochemical/chemistry , Photolysis , Prions/physiology
4.
Neurobiol Dis ; 44(3): 327-39, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21827858

ABSTRACT

AIMS: Central nervous system complications including cognitive impairment are an early manifestation of diabetes mellitus, also evident in animal models. NAP (generic name, davunetide), a neuroprotective peptide was tested here for its ability to prevent diabetes-related brain pathologies in the streptozotocin injected diabetes rat model. METHODS: Diabetes was induced by an intraperitoneal streptozotocin injection (55 mg/kg). Intranasal NAP or vehicle was administered daily starting on the day following streptozotocin injection. Cognitive assessment was performed 12 weeks after diabetes induction, using the Morris water maze paradigm. Brain structural integrity was assessed on the 15th week of diabetes by magnetic resonance T2 scan. Characterization of cellular populations, apoptosis and synaptic density was performed 16 weeks after diabetes induction, using immunohistochemical markers and quantified in the prefrontal cortex, the cerebral cortex and the hippocampus of both hemispheres. RESULTS: Impaired spatial memory of the diabetic rats was observed in the water maze by attenuated learning curve and worsened performance in the probe memory test. NAP treatment significantly improved both measurements. T2 magnetic resonance imaging revealed atrophy in the prefrontal cortex of the diabetes rat group, which was prevented by NAP treatment. Immunohistochemical analysis showed that NAP treatment protected against major loss of the synaptic marker synaptophysin and astrocytic apoptosis, resulting from streptozotocin treatment. CONCLUSIONS: Our results show for the first time protective effects for NAP (davuentide) in a diabetes rat model at the behavioral and structural levels against one of the most severe complications of diabetes.


Subject(s)
Central Nervous System Diseases/etiology , Central Nervous System Diseases/prevention & control , Diabetes Mellitus, Experimental/complications , Neuroprotective Agents/therapeutic use , Oligopeptides/therapeutic use , Animals , Caspase 3/metabolism , Central Nervous System Diseases/pathology , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Lectins/metabolism , Magnetic Resonance Imaging/methods , Male , Maze Learning/drug effects , Mental Disorders/drug therapy , Mental Disorders/etiology , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Social Behavior , Space Perception/drug effects , Synaptophysin/metabolism , Time Factors
5.
Brain Res ; 1390: 126-41, 2011 May 16.
Article in English | MEDLINE | ID: mdl-21406188

ABSTRACT

BACKGROUND AND PURPOSE: Experimental autoimmune encephalomyelitis (EAE) is a widely used model of multiple sclerosis (MS) and both conditions have been reported to exhibit reduced endocannabinoid activity. The purpose of this study was to address the effect of exogenously administered 2-arachidonoylglycerol (2AG), an endocannabinoid receptor ligand, on acute phase and chronic disability in EAE. EXPERIMENTAL APPROACH: Acute and chronic EAE models were induced in susceptible mice and 2AG-treatment was applied for 14 days from day of disease induction. KEY RESULTS: 2AG-treatment ameliorated acute phase of disease with delay of disease onset in both EAE models and reduced disease mortality and long-term (70 days post-induction) clinical disability in chronic EAE. Reduced axonal pathology in the chronic EAE- (p<0.0001) and increased activation and ramification of microglia in the 2AG-treated acute EAE- (p<0.05) model were noticed. The latter was accompanied by a 2- to 4-fold increase of the M2-macrophages in the perivascular infiltrations (p<0.001) of the 2AG-treated animals in the acute (day 22), although not the chronic (day 70), EAE model. Expression of cannabinoid receptors 1 (CB1R) and 2 (CB2R) was increased in 2AG-treated animals of acute EAE vs. controls (p<0.05). In addition, ex vivo viability assays exhibited reduced proliferation of activated lymph node cells when extracted from 2AG-treated EAE animals, whereas a dose-dependent response of activated lymphocytes to 2AG-treatment in vitro was noticed. CONCLUSION AND IMPLICATIONS: Our data indicate for the first time that 2AG treatment may provide direct (via CBRs) and immune (via M2 macrophages) mediated neuroprotection in EAE.


Subject(s)
Arachidonic Acids/therapeutic use , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glycerides/therapeutic use , Acute Disease , Animals , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/pathology , Endocannabinoids , Female , Mice , Mice, Inbred C57BL , Random Allocation
6.
Exp Neurol ; 230(1): 78-89, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440544

ABSTRACT

Autologous bone marrow stromal cells (BMSCs) offer significant practical advantages for potential clinical applications in multiple sclerosis (MS). Based on recent experimental data, a number of clinical trials have been designed for the intravenous (IV) and/or intrathecal (ITH) administration of BMSCs in MS patients. Delivery of BMSCs in the cerebrospinal fluid via intracerebroventricular (ICV) transplantation is a useful tool to identify mechanisms underlying the migration and function of these cells. In the current study, BMSCs were ICV administered in severe and mild EAE, as well as naive animals; neural precursor cells (NPCs) served as cellular controls. Our data indicated that ICV-transplanted BMSCs significantly ameliorated mild though not severe EAE. Moreover, BMSCs exerted significant anti-inflammatory effect on spinal cord with concomitant reduced axonopathy only in the mild EAE model. BMSCs migrated into the brain parenchyma and, depending on their cellular density, within brain parenchyma formed cellular masses characterized by focal inflammation, demyelination, axonal loss and increased collagen-fibronectin deposition. These masses were present in 64% of ICV BMASC-transplanted severe EAE animals whereas neither BMSCs transplanted in mild EAE cases nor the NPCs exhibited similar behavior. BMSCs possibly exerted their fibrogenic effect via both paracrine and autocrine manner, at least partly due to up-regulation of connective tissue growth factor (CTGF) under the trigger of TGFb1. Our findings are of substantial relevance for clinical trials in MS, particularly regarding the possibility that ICV transplanted BMSCs entering the inflamed central nervous system may exhibit - under conditions - a local pathology of yet unknown consequences.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/surgery , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/physiology , Animals , Area Under Curve , Brain/pathology , Cell Survival/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/mortality , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Injections, Intraventricular/adverse effects , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Severity of Illness Index , Spinal Cord/pathology , Transforming Growth Factor beta/metabolism , Transplantation, Autologous/adverse effects , Tumor Necrosis Factor-alpha/metabolism
7.
Exp Neurol ; 230(1): 16-26, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20420833

ABSTRACT

Transplanted Neural Precursor Cells (NPCs) are capable of long-distance migration inside the inflamed CNS, but exhibit limited myelinating capacities in animal models of Multiple Sclerosis (MS). Inflammation seems to be both beneficial for the recruitment and migration of NPCs and restrictive for their terminal differentiation. In the present study, a set of transplantation experiments was applied in order to investigate the migratory potential, the differentiation pattern and long-term survival of NPCs in Experimental Autoimmune Encephalomyelitis (EAE) mice, the animal model of MS. The in vitro differentiation potential of NPCs in the presence of either pro- (TNFa, INFγ) or anti- (TGFb) inflammatory cytokines was also analyzed. According to the in vivo results obtained, at the acute phase of EAE only a small fraction of transplanted NPCs succeed to differentiate, whereas at chronic phase most of them followed a differentiation process to glial cell lineage along white matter tracts. However, this differentiation was not fully completed, since 8 months after their transplantation a number of NPCs remained as pre-oligodendrocytes. Glial differentiation of NPCs was also found to be inhibited or promoted following their treatment with TNFa or TGFb respectively, in vitro. Our findings suggest that inflammation triggers migration whereas the anti-inflammatory component is a prerequisite for NPCs to follow glial differentiation thereby providing myelinating oligodendrocytes. It is speculated that the fine balance between the pro- and anti-inflammatory determinants in the CNS may be a key factor for transplanted NPCs to exhibit a better therapeutic effect in EAE and MS. This article is part of a Special Issue entitled "Interaction between repair, disease, & inflammation."


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/surgery , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Neurons/physiology , Animals , Animals, Newborn , Antigens/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/physiology , Cytokines/pharmacology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein , Glycoproteins/adverse effects , Green Fluorescent Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/drug effects , Neurons/drug effects , Peptide Fragments/adverse effects , Proteoglycans/metabolism , Severity of Illness Index , Statistics, Nonparametric , Time Factors
8.
Neuroreport ; 22(2): 68-72, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21150806

ABSTRACT

Recent studies on neural precursor cell (NPC) transplantation in multiple sclerosis animal models reveal that these cells exert their therapeutic effect mainly because of immunomodulation rather than cell replacement. In this study intraventricularly transplanted NPCs in mice, induced experimental autoimmune encephalomyelitis, the animal model of multiple sclerosis, improved the clinical symptoms and suppressed inflammation in the brain by enhancing the apoptosis of inflammatory cells. However, the same treatment failed to reduce significantly the inflammatory cells in the spinal cord, the pathology of which predominantly determines the clinical manifestation of experimental autoimmune encephalomyelitis. Our findings suggest that immunosuppression is rather a local phenomenon and thus, bystander neuroprotective mechanisms triggered by NPC intraventricular transplantation should be accountable for their therapeutic effect.


Subject(s)
Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Inflammation/etiology , Neural Stem Cells/transplantation , Spinal Cord/pathology , Stem Cell Transplantation/adverse effects , Animals , Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/immunology , Inflammation/pathology , Mice , Neural Stem Cells/pathology , Neurons/immunology , Neurons/pathology , Spinal Cord/immunology
9.
FASEB J ; 23(12): 4266-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19723705

ABSTRACT

Despite efforts aimed at developing novel therapeutics for traumatic brain injury (TBI), no specific pharmacological agent is currently clinically available. Here, we show that the pan-histone deacetylase (HDAC) inhibitor ITF2357, a compound shown to be safe and effective in humans, improves functional recovery and attenuates tissue damage when administered as late as 24 h postinjury. Using a well-characterized, clinically relevant mouse model of closed head injury (CHI), we demonstrate that a single dose of ITF2357 administered 24 h postinjury improves neurobehavioral recovery from d 6 up to 14 d postinjury (improved neurological score vs. vehicle; P< or =0.05), and that this functional benefit is accompanied by decreased neuronal degeneration, reduced lesion volume (22% reduction vs. vehicle; P< or =0.01), and is preceded by increased acetylated histone H3 levels and attenuation of injury-induced decreases in cytoprotective heat-shock protein 70 kDa and phosphorylated Akt. Moreover, reduced glial accumulation and activation were observed 3 d postinjury, and total p53 levels at the area of injury and caspase-3 immunoreactivity within microglia/macrophages at the trauma area were elevated, suggesting enhanced clearance of these cells via apoptosis following treatment. Hence, our findings underscore the relevance of HDAC inhibitors for ameliorating trauma-induced functional deficits and warrant consideration of applying ITF2357 for this indication.


Subject(s)
Apoptosis/drug effects , Brain Injuries/drug therapy , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Neuroglia/drug effects , Animals , Brain/pathology , Caspase 3/metabolism , Hydroxamic Acids/administration & dosage , Male , Mice , Neuroprotective Agents/pharmacology
10.
Neurobiol Dis ; 34(2): 381-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19264130

ABSTRACT

NAP (NAPVSIPQ) provides broad neuroprotection through microtubule interaction. Here, NAP was investigated for neuroprotection in an in vivo tauopathy model. Transgenic mice (2-month-old) that express the human double mutant tau protein [P301S;K257T] fused to the tau promoter, were subjected to daily intranasal drug treatment for approximately 5 months. Results showed increased performance in the NAP-treated mice compared to controls, as demonstrated in the Morris water maze, (p<0.05). Treatment continued for 5 additional months and mouse cortices were biochemically analyzed. Protein extraction identified increased tau protein content in the heat-stable soluble fraction, which contains microtubule-associated tau, in the 1-year-old NAP-treated mice as compared to vehicle-controls. Tau phosphorylation (Ser 202) increased in the tau-transgenic mice compared to control mice, and was significantly reduced in NAP-treated mice. The current studies show for the first time activity for NAP in a "pure" tauopathy model, positioning it as a promising drug candidate in multiple neurodegenerative tauopathies.


Subject(s)
Brain/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Tauopathies/drug therapy , tau Proteins/drug effects , Animals , Brain/metabolism , Brain/pathology , Cytoprotection/drug effects , Cytoprotection/physiology , Disease Models, Animal , Humans , In Vitro Techniques , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Transgenic , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neuroprotective Agents/therapeutic use , Oligopeptides/therapeutic use , Phosphorylation/drug effects , Solubility/drug effects , Tauopathies/genetics , Tauopathies/metabolism , Treatment Outcome , tau Proteins/metabolism
11.
Exp Neurol ; 198(2): 275-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16472805

ABSTRACT

Stem cell transplantation was introduced as a mean of cell replacement therapy, but the mechanism by which it confers clinical improvement in experimental models of neurological diseases is not clear. Here, we transplanted neural precursor cells (NPCs) into the ventricles of mice at day 6 after induction of chronic experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). Transplanted cells migrated into white matter tracts and attenuated the clinical course of disease. NPC transplantation down-regulated the inflammatory brain process at the acute phase of disease, as indicated by a reduction in the number of perivascular infiltrates and of brain CD3+ T cells, an increase in the number and proportion of regulatory T cells and a reduction in the expression of ICAM-1 and LFA-1 in the brain. Demyelination and acute axonal injury in this model are considered to result mainly from the acute inflammatory process and correlate well with the chronic neurological residua. In consequence to inhibition of brain inflammation, precursor cell transplantation attenuated the primary demyelinating process and reduced the acute axonal injury. As a result, the size of demyelinated areas and extent of chronic axonal pathology were reduced in the transplanted brains. We suggest that the beneficial effect of transplanted NPCs in chronic EAE is mediated, in part, by decreasing brain inflammation and reducing tissue injury.


Subject(s)
Encephalitis/surgery , Encephalomyelitis, Autoimmune, Experimental/surgery , Gene Expression Regulation/physiology , Neurons/physiology , Stem Cell Transplantation , Amyloid beta-Protein Precursor , Animals , Animals, Newborn , Antigens/metabolism , Axons/pathology , Blotting, Northern/methods , Bromodeoxyuridine/pharmacokinetics , Disease Models, Animal , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/complications , Epidermal Growth Factor/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Intercellular Adhesion Molecule-1/metabolism , Intermediate Filament Proteins/metabolism , Ki-1 Antigen/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/pathology , Nerve Tissue Proteins/metabolism , Nestin , Neural Cell Adhesion Molecule L1/metabolism , Neurons/drug effects , O Antigens/metabolism , Phosphopyruvate Hydratase/metabolism , Proteoglycans/metabolism , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Sialic Acids/metabolism , Stem Cell Transplantation/methods
12.
Clin Neurol Neurosurg ; 108(3): 241-4, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16388896

ABSTRACT

Both inflammatory and neurodegenerative components may contribute to the clinical profile of multiple sclerosis (MS) leading to irreversible deficits when they exceed the threshold of compensation. The mechanisms leading to tissue injury in MS are complex. Inflammation appears to be caused by overactive pro-inflammatory T-helper 1 cells, initiating an inflammatory cascade with several cellular and molecular immune components participating in the pathogenetic mechanism. Current treatments are most effective in the inflammatory phase of the disease since they may interfere with various stages of the immune cascade. Recent evidence has emerged that inflammation may not only be destructive, but may also play a part in tissue repair. This has opened up a new aspect of our knowledge of the role of the inflammatory process in MS. Data regarding the role of regulatory cells in particular, imply that specific immunomodulatory strategies that support the function of these particular cellular subpopulations may participate in the downregulation of autoimmune responses in MS.


Subject(s)
Autoimmunity/physiology , Inflammation/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Forkhead Transcription Factors/physiology , Humans , Multiple Sclerosis/physiopathology , T-Lymphocytes, Regulatory/physiology
13.
Clin Neurol Neurosurg ; 108(3): 250-4, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16413962

ABSTRACT

In chronic inflammatory diseases like multiple sclerosis (MS), neuroprotection refers to strategies aimed at prevention of the irreversible damage of various neuronal and glial cell populations, and promoting regeneration. It is increasingly recognized that MS progression, in addition to demyelination, leads to substantial irreversible damage to, and loss of neurons, resulting in brain atrophy and cumulative disability. One of the most promising neuroprotective strategies involves the use of bone marrow derived stem cells. Both hematopoietic and non-hematopoietic (stromal) cells can, under certain circumstances, differentiate into cells of various neuronal and glial lineages. Neuronal stem cells have also been reported to suppress EAE by exerting direct in situ immunomodulating effects, in addition to their ability to provide a potential source for remyelination and neuroregeneration. Preliminary results from our laboratory indicate that intravenous or intracerebral/intraventricular injection of bone marrow derived stromal cells could differentiate in neuronal/glial cells and suppress the clinical signs of chronic EAE. Both bone marrow and neuronal stem cells may therefore have a therapeutic potential in MS. It seems that future treatment strategies for MS should combine immunomodulation with neuroprotective modalities to achieve maximal clinical benefit.


Subject(s)
Immunologic Factors/therapeutic use , Multiple Sclerosis/therapy , Neuroprotective Agents/therapeutic use , Stem Cell Transplantation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...