Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 12(10)2023 05 17.
Article in English | MEDLINE | ID: mdl-37408252

ABSTRACT

The Neurovascular Unit (NVU), composed of glia (astrocytes, oligodendrocytes, microglia), neurons, pericytes and endothelial cells, is a dynamic interface ensuring the physiological functioning of the central nervous system (CNS), which gets affected and contributes to the pathology of several neurodegenerative diseases. Neuroinflammation is a common feature of neurodegenerative diseases and is primarily related to the activation state of perivascular microglia and astrocytes, which constitute two of its major cellular components. Our studies focus on monitoring in real time the morphological changes of perivascular astrocytes and microglia, as well as their dynamic interactions with the brain vasculature, under physiological conditions and following systemic neuroinflammation triggering both microgliosis and astrogliosis. To this end, we performed 2-photon laser scanning microscopy (2P-LSM) for intravital imaging of the cortex of transgenic mice visualizing the dynamics of microglia and astroglia following neuroinflammation induced by systemic administration of the endotoxin lipopolysaccharide (LPS). Our results indicate that following neuroinflammation the endfeet of activated perivascular astrocytes lose their close proximity and physiological cross-talk with vasculature, an event that most possibly contributes to a loss of blood-brain barrier (BBB) integrity. At the same time, microglial cells become activated and exhibit a higher extent of physical contact with the blood vessels. These dynamic responses of perivascular astrocytes and microglia are peaking at 4 days following LPS administration; however, they still persist at a lower level at 8 days after LPS injection, revealing incomplete reversal of inflammation affecting the glial properties and interactions within the NVU.


Subject(s)
Astrocytes , Microglia , Animals , Mice , Astrocytes/pathology , Microglia/pathology , Lipopolysaccharides/adverse effects , Neuroinflammatory Diseases , Endothelial Cells/pathology , Brain/pathology , Inflammation/pathology , Mice, Transgenic
2.
Stem Cell Reports ; 18(4): 915-935, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36963393

ABSTRACT

The microRNA (miRNA) miR-124 has been employed supplementary to neurogenic transcription factors (TFs) and other miRNAs to enhance direct neurogenic conversion. The aim of this study was to investigate whether miR-124 is sufficient to drive direct reprogramming of astrocytes to induced neurons (iNs) on its own and elucidate its independent mechanism of reprogramming action. Our data show that miR-124 is a potent driver of the reprogramming switch of astrocytes toward an immature neuronal fate by directly targeting the RNA-binding protein Zfp36L1 implicated in ARE-mediated mRNA decay and subsequently derepressing Zfp36L1 neurogenic interactome. To this end, miR-124 contribution in iNs' production largely recapitulates endogenous neurogenesis pathways, being further enhanced upon addition of the neurogenic compound ISX9, which greatly improves iNs' differentiation and functional maturation. Importantly, miR-124 is potent in guiding direct conversion of reactive astrocytes to immature iNs in vivo following cortical trauma, while ISX9 supplementation confers a survival advantage to newly produced iNs.


Subject(s)
MicroRNAs , Neural Stem Cells , Astrocytes/metabolism , Neurons/metabolism , Cell Differentiation/genetics , Neural Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
3.
Mech Ageing Dev ; 192: 111366, 2020 12.
Article in English | MEDLINE | ID: mdl-32991921

ABSTRACT

Remyelination is a physiological response to demyelinating events aiming to restore saltatory conduction and preserve axonal integrity. Resident oligodendrocyte precursor cells (OPC) of the CNS tissue under appropriate conditions are mobilized to proliferate, migrate, and differentiate, in order to produce new myelin sheaths in the demyelinated lesion. In multiple sclerosis (MS), the most common immune-mediated demyelinating disease, remyelination efficiency declines with increasing age and disease duration. As myelin regeneration attempts in clinical trials so far are scarce, and have been met with limited success, the need to explore new remyelinating strategies is more compelling. Recently, ageing and cellular senescence have been implicated in the pathophysiology of a number of neurodegenerative diseases, including multiple sclerosis. Evidence on OPC senescence brings forward the possibility of exploiting cellular senescence as a possible target for promoting the endogenous remyelinating capacity of the CNS. Here we discuss the data indicating how cellular senescence affects remyelination, and the putative benefits to be drawn through the use of senolytic or senomorphic therapies targeting senescent cell populations in MS.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Molecular Targeted Therapy/trends , Multiple Sclerosis , Remyelination , Drug Discovery , Humans , Multiple Sclerosis/metabolism , Multiple Sclerosis/therapy , Myelin Sheath/drug effects , Myelin Sheath/physiology , Remyelination/drug effects , Remyelination/physiology , Sphingosine 1 Phosphate Receptor Modulators/pharmacology
4.
Cell Rep ; 29(4): 932-945.e7, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31644914

ABSTRACT

Local cues in the adult neurogenic niches dynamically regulate homeostasis in neural stem cells, whereas their identity and associated molecular mechanisms remain poorly understood. Here, we show that corticotropin-releasing hormone (CRH), the major mediator of mammalian stress response and a key neuromodulator in the adult brain, is necessary for hippocampal neural stem cell (hiNSC) activity under physiological conditions. In particular, we demonstrate functionality of the CRH/CRH receptor (CRHR) system in mouse hiNSCs and conserved expression in humans. Most important, we show that genetic deficiency of CRH impairs hippocampal neurogenesis, affects spatial memory, and compromises hiNSCs' responsiveness to environmental stimuli. These deficits have been partially restored by virus-mediated CRH expression. Additionally, we provide evidence that local disruption of the CRH/CRHR system reduces neurogenesis, while exposure of adult hiNSCs to CRH promotes neurogenic activity via BMP4 suppression. Our findings suggest a critical role of CRH in adult neurogenesis, independently of its stress-related systemic function.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Animals , Cell Line , Cells, Cultured , Corticotropin-Releasing Hormone/genetics , Hippocampus/cytology , Hippocampus/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Spatial Memory
5.
Int J Mol Sci ; 19(10)2018 Sep 27.
Article in English | MEDLINE | ID: mdl-30261683

ABSTRACT

Ageing is a major risk factor for developing many neurodegenerative diseases. Cellular senescence is a homeostatic biological process that has a key role in driving ageing. There is evidence that senescent cells accumulate in the nervous system with ageing and neurodegenerative disease and may predispose a person to the appearance of a neurodegenerative condition or may aggravate its course. Research into senescence has long been hindered by its variable and cell-type specific features and the lack of a universal marker to unequivocally detect senescent cells. Recent advances in senescence markers and genetically modified animal models have boosted our knowledge on the role of cellular senescence in ageing and age-related disease. The aim now is to fully elucidate its role in neurodegeneration in order to efficiently and safely exploit cellular senescence as a therapeutic target. Here, we review evidence of cellular senescence in neurons and glial cells and we discuss its putative role in Alzheimer's disease, Parkinson's disease and multiple sclerosis and we provide, for the first time, evidence of senescence in neurons and glia in multiple sclerosis, using the novel GL13 lipofuscin stain as a marker of cellular senescence.


Subject(s)
Aging/genetics , Cellular Senescence , Neurodegenerative Diseases/genetics , Aging/pathology , Animals , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology
6.
Glia ; 64(5): 763-79, 2016 May.
Article in English | MEDLINE | ID: mdl-26712314

ABSTRACT

The central nervous system has limited capacity for regeneration after traumatic injury. Transplantation of neural stem/progenitor cells (NPCs) has been proposed as a potential therapeutic approach while insulin-like growth factor I (IGF-I) has neuroprotective properties following various experimental insults to the nervous system. We have previously shown that NPCs transduced with a lentiviral vector for IGF-I overexpression have an enhanced ability to give rise to neurons in vitro but also in vivo, upon transplantation in a mouse model of temporal lobe epilepsy. Here we studied the regenerative potential of NPCs, IGF-I-transduced or not, in a mouse model of hippocampal mechanical injury. NPC transplantation, with or without IGF-I transduction, rescued the injury-induced spatial learning deficits as revealed in the Morris Water Maze. Moreover, it had beneficial effects on the host tissue by reducing astroglial activation and microglial/macrophage accumulation while enhancing generation of endogenous oligodendrocyte precursor cells. One or two months after transplantation the grafted NPCs had migrated towards the lesion site and in the neighboring myelin-rich regions. Transplanted cells differentiated toward the oligodendroglial, but not the neuronal or astrocytic lineages, expressing the early and late oligodendrocyte markers NG2, Olig2, and CNPase. The newly generated oligodendrocytes reached maturity and formed myelin internodes. Our current and previous observations illustrate the high plasticity of transplanted NPCs which can acquire injury-dependent phenotypes within the host CNS, supporting the fact that reciprocal interactions between transplanted cells and the host tissue are an important factor to be considered when designing prospective cell-based therapies for CNS degenerative conditions.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/surgery , Cell Differentiation/physiology , Inflammation/etiology , Learning Disabilities/etiology , Oligodendroglia/physiology , Stem Cell Transplantation/methods , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Animals , Animals, Newborn , Antigens/metabolism , Antigens, CD/metabolism , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/surgery , Ki-67 Antigen/metabolism , Learning Disabilities/surgery , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Proteoglycans/metabolism
7.
Neurotox Res ; 24(2): 244-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23392957

ABSTRACT

Cuprizone [bis(cyclohexylidenehydrazide)]-induced toxic demyelination is an experimental animal model commonly used to study de- and remyelination in the central nervous system. In this model, mice are fed with the copper chelator cuprizone which leads to oligodendrocyte death with subsequent demyelination. The underlying mechanisms of cuprizone-induced oligodendrocyte death are still unknown, and appropriate in vitro investigations to study these mechanisms are not available. Thus, we studied cuprizone effects on rat primary glial cell cultures and on the neuroblastoma cell line SH-SY5Y. Treatment of cells with different concentrations of cuprizone failed to show effects on the proliferation and survival of SH-SY5Y cells, microglia, astrocytes, and oligodendrocyte precursor cells (OPC). In contrast, differentiated mature oligodendrocytes (OL) were found to be significantly affected by cuprizone treatment. This was accompanied by a reduced mitochondrial potential in cuprizone-treated OL. These results demonstrate that the main toxic target for cuprizone is mature OL, whilst other glial cells including OPC are not or only marginally affected. This explains the selective demyelination induced by cuprizone in vivo.


Subject(s)
Cell Differentiation/drug effects , Chelating Agents/toxicity , Cuprizone/toxicity , Oligodendroglia/drug effects , Oligodendroglia/pathology , Animals , Animals, Newborn , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Rats , Rats, Sprague-Dawley
8.
Stem Cells Transl Med ; 2(3): 185-98, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23417642

ABSTRACT

Temporal lobe epilepsy (TLE) is a major neurological disease, often associated with cognitive decline. Since approximately 30% of patients are resistant to antiepileptic drugs, TLE is being considered as a possible clinical target for alternative stem cell-based therapies. Given that insulin-like growth factor I (IGF-I) is neuroprotective following a number of experimental insults to the nervous system, we investigated the therapeutic potential of neural stem/precursor cells (NSCs) transduced, or not, with a lentiviral vector for overexpression of IGF-I after transplantation in a mouse model of kainic acid (KA)-induced hippocampal degeneration, which represents an animal model of TLE. Exposure of mice to the Morris water maze task revealed that unilateral intrahippocampal NSC transplantation significantly prevented the KA-induced cognitive decline. Moreover, NSC grafting protected against neurodegeneration at the cellular level, reduced astrogliosis, and maintained endogenous granule cell proliferation at normal levels. In some cases, as in the reduction of hippocampal cell loss and the reversal of the characteristic KA-induced granule cell dispersal, the beneficial effects of transplanted NSCs were manifested earlier and were more pronounced when these were transduced to express IGF-I. However, differences became less pronounced by 2 months postgrafting, since similar amounts of IGF-I were detected in the hippocampi of both groups of mice that received cell transplants. Grafted NSCs survived, migrated, and differentiated into neurons-including glutamatergic cells-and not glia, in the host hippocampus. Our results demonstrate that transplantation of IGF-I producing NSCs is neuroprotective and restores cognitive function following KA-induced hippocampal degeneration.


Subject(s)
Cognition , Epilepsy, Temporal Lobe/therapy , Genetic Therapy/methods , Hippocampus/surgery , Kainic Acid , Nerve Degeneration , Neural Stem Cells/transplantation , Neurogenesis , Neurons/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Behavior, Animal , Cell Movement , Cell Proliferation , Cell Survival , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/physiopathology , Epilepsy, Temporal Lobe/psychology , Genetic Vectors , Glutamic Acid/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/metabolism , Neurons/metabolism , Spheroids, Cellular , Time Factors , Transduction, Genetic
9.
Exp Neurol ; 231(2): 223-35, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21756906

ABSTRACT

Epilepsy is a major neurological disease, and patients often show spatial memory deficits. Thus, there is a need of effective new therapeutic approaches. IGF-I has been shown to be neuroprotective following a number of experimental insults to the nervous system, and in a variety of animal models of neurodegenerative diseases. In the present work, we investigated the possible neuroprotective effects of IGF-I following unilateral intrahippocampal administration of kainic acid (KA), an animal model of temporal lobe epilepsy (TLE). KA induced cell death, as shown by FluoroJade B, and extensive cell loss in both the ipsilateral and contralateral CA3 and CA4 areas, as well as granule cell dispersal in the DG, as revealed by Cresyl violet staining. KA also resulted in intense astrogliosis and microgliosis, as assessed by the number of GFAP and CD11b immunopositive cells, respectively, and increased hippocampal neurogenesis. Exposure to the Morris Water Maze task revealed that mice injected with KA were deficient in spatial learning and both short- and long-term memories, when tested in a larger diameter pool, which requires the use of allocentric strategies. When tested in a smaller pool, only long-term memory was impaired. Administration of IGF-I decreased seizure severity, hippocampal neurogenesis, and protected against neurodegeneration at the cellular level as assessed by FluoroJade B and Cresyl violet staining, as well as the number of GFAP and CD11b immunopositive cells. Furthermore, IGF-I abolished the cognitive deficits. Our results support that IGF-I could have a possible therapeutic potential in TLE.


Subject(s)
Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Insulin-Like Growth Factor I/pharmacology , Memory/drug effects , Nerve Degeneration/prevention & control , Animals , Cell Count , Cell Death/drug effects , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/physiopathology , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/pathology , Hippocampus/physiopathology , Kainic Acid/pharmacology , Male , Maze Learning/drug effects , Mice , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/pathology
10.
Brain Pathol ; 20(2): 301-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19371354

ABSTRACT

In multiple sclerosis, demyelination occurs beside the white-matter structures and in the cerebral and cerebellar cortex. We have previously shown that, in the cuprizone model, demyelination is present not only in the corpus callosum but also in the cerebral cortex. Here, we have performed a detailed analysis of the dynamics of de- and remyelination in the cerebellar cortex and white matter at nine timepoints in two cerebellar regions. To induce demyelination, C57BL/6 mice were fed with 0.2% cuprizone for 12 weeks followed by a recovery of 8 weeks. Both cortex and white-matter structures were significantly demyelinated after 12 weeks of cuprizone feeding. Remyelination occurred after withdrawal of cuprizone but was less prominent in the more caudal cerebellar region. Microglia infiltration was prominent in all analyzed cerebellar areas, preceding demyelination by approximately 2-4 weeks, and was delayed in the more caudal cerebellar region. Astrogliosis was also seen but did not reach the extent observed in the cerebrum. In summary, cuprizone feeding provides an excellent model for the investigation of de- and remyelination processes in the cerebellar cortex and white matter. Furthermore, demyelination, microglia and astrocyte changes were different in the cerebellum as compared with the cerebrum, indicating region-dependent pathomechanisms.


Subject(s)
Cerebellar Cortex/physiopathology , Demyelinating Diseases/physiopathology , Animals , Astrocytes/pathology , Astrocytes/physiology , Cerebellar Cortex/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Gliosis/chemically induced , Gliosis/pathology , Gliosis/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Microglia/physiology , Myelin Sheath/pathology , Myelin Sheath/physiology , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/physiology , Oligodendroglia/pathology , Oligodendroglia/physiology , Time Factors
11.
Brain Res ; 1283: 127-38, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19524552

ABSTRACT

Cuprizone feeding is a commonly used model to study experimental de- and remyelination, with the corpus callosum being the most frequently investigated white matter tract. We have previously shown that demyelination is also extensive in the cerebral cortex in the cuprizone model. In the current study, we have performed a detailed analysis of the dynamics of demyelination in the cortex in comparison to the corpus callosum. Prominent and almost complete demyelination in the corpus callosum was observed after 4.5-5 weeks of 0.2% cuprizone feeding, whereas complete cortical demyelination was only observed after 6 weeks of cuprizone feeding. Interestingly, remyelination in the corpus callosum occurred even before the termination of cuprizone administration. Accumulation of microglia in the corpus callosum started as early as week 3 reaching its maximum at week 4.5 and was still significantly elevated at week 6 of cuprizone treatment. Within the cortex only a few scattered activated microglial cells were found. Furthermore, the intensity of astrogliosis, accumulation of oligodendrocyte progenitor cells and nestin positive cells differed between the two areas investigated. The time course and dynamics of demyelination differ in the corpus callosum and in the cortex, suggesting different underlying pathomechanisms.


Subject(s)
Brain/pathology , Demyelinating Diseases/pathology , Nerve Fibers, Myelinated/pathology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Brain/drug effects , Brain/physiopathology , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Chelating Agents/toxicity , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/physiopathology , Disease Models, Animal , Disease Progression , Gliosis/chemically induced , Gliosis/pathology , Gliosis/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Nerve Fibers, Myelinated/drug effects , Nerve Regeneration/physiology , Neurotoxins/toxicity , Oligodendroglia/drug effects , Oligodendroglia/pathology , Stem Cells/drug effects , Stem Cells/pathology , Time Factors
12.
Neurosci Lett ; 451(1): 83-8, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19084049

ABSTRACT

In multiple sclerosis demyelination not only affects the white matter, but also the grey matter of the brain. We have previously reported that in the murine cuprizone model for demyelination lesions occur in addition to the corpus callosum also in the neocortex and hippocampus. In the current study, we provide a detailed characterization of hippocampal demyelination in the cuprizone model. Male C57BL/6 mice were challenged with 0.2% cuprizone for 6 weeks. Defined structures within the hippocampus were investigated at week 0 (control), 3, 4, 4.5, 5, 5.5, and 6. Demyelination affected all hippocampal structures analyzed and was complete after 6 weeks of cuprizone treatment. Between the distinct hippocampal structures the temporal pattern of demyelination varied considerably. Furthermore, infiltration of activated microglia as well as astrogliosis was detected. In summary, cuprizone feeding provides a useful model for studying demyelination processes in the mouse hippocampus.


Subject(s)
Cuprizone/toxicity , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Nerve Fibers, Myelinated/pathology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Biomarkers/analysis , Biomarkers/metabolism , Chelating Agents/toxicity , Demyelinating Diseases/chemically induced , Disease Models, Animal , Gliosis/chemically induced , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/drug effects , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Nerve Fibers, Myelinated/drug effects , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurotoxins/toxicity , Wallerian Degeneration/chemically induced , Wallerian Degeneration/pathology , Wallerian Degeneration/physiopathology
13.
Glia ; 56(10): 1104-13, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18442092

ABSTRACT

Unravelling the factors that can positively influence remyelination is one of the major challenges in multiple sclerosis research. Expression of the chemokine receptor CXCR2 on oligodendrocytes both in vitro and in MS lesions has suggested a possible role for CXCR2 in the recruitment of oligodendrocyte precursor cells (OPC). To investigate the function of CXCR2 during remyelination in vivo, we studied this receptor in cuprizone-induced demyelination and subsequent remyelination. We found that CXCR2 is constitutively expressed on OPC, whereas on macrophages/microglia CXCR2 is upregulated upon activation during demyelination. Hence, the expression of CXCR2 is differentially regulated in oligodendrocytes and macrophages/microglia. Furthermore, we subjected CXCR2-/- mice to the cuprizone model demonstrating that remyelination was not altered in comparison to wildtype controls. In addition, the number of OPC and the amount of microglial accumulation were similar in both CXCR2-/- and wildtype animals during the whole demyelination and remyelination process. These results suggest that despite expression on OPC and microglia CXCR2 plays only a minor role during remyelination.


Subject(s)
Cuprizone/toxicity , Demyelinating Diseases/metabolism , Myelin Sheath/physiology , Neuroglia/metabolism , Neuroglia/physiology , Receptors, Interleukin-8B/biosynthesis , Animals , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Myelin Sheath/drug effects , Myelin Sheath/pathology , Neuroglia/drug effects , Neuroglia/pathology , Receptors, Interleukin-8B/physiology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...