Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 161: 105556, 2021 12.
Article in English | MEDLINE | ID: mdl-34752925

ABSTRACT

Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease with high variability of clinical symptoms. In most cases MS appears as a relapsing-remitting disease course that at a later stage transitions into irreversible progressive decline of neurologic function. The mechanisms underlying MS progression remain poorly understood. Experimental autoimmune encephalomyelitis (EAE) is an animal model of MS. Here we demonstrate that mice that develop mild EAE after immunization with myelin oligodendrocyte glycoprotein 35-55 are prone to undergo clinical progression around 30 days after EAE induction. EAE progression was associated with reduction in CD11c+ microglia and dispersed coalescent parenchymal infiltration. We found sex-dependent differences mediated by p38α signaling, a key regulator of inflammation. Selective reduction of CD11c+ microglia in female mice with CD11c-promoter driven p38α knockout correlated with increased rate of EAE progression. In protected animals, we found CD11c+ microglia forming contacts with astrocyte processes at the glia limitans and immune cells retained within perivascular spaces. Together, our study identified pathological hallmarks of chronic EAE progression and suggests that CD11c+ microglia may regulate immune cell parenchymal infiltration in autoimmune demyelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Mice, Inbred C57BL , Microglia/pathology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein
3.
PLoS One ; 13(9): e0204536, 2018.
Article in English | MEDLINE | ID: mdl-30226905

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0167573.].

4.
Mol Neurobiol ; 55(12): 9001-9015, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29623612

ABSTRACT

Myelination in the central nervous system takes place predominantly during the postnatal development of humans and rodents by myelinating oligodendrocytes (OLs), which are differentiated from oligodendrocyte progenitor cells (OPCs). We recently reported that Sox2 is essential for developmental myelination in the murine brain and spinal cord. It is still controversial regarding the role of Sox2 in oligodendroglial lineage progression in the postnatal murine spinal cord. Analyses of a series of cell- and stage-specific Sox2 mutants reveal that Sox2 plays a biphasic role in regulating oligodendroglial lineage progression in the postnatal murine spinal cord. Sox2 controls the number of OPCs for subsequent differentiation through regulating their proliferation. In addition, Sox2 regulates the timing of OL differentiation and modulates the rate of oligodendrogenesis. Our experimental data prove that Sox2 is an intrinsic positive timer of oligodendroglial lineage progression and suggest that interventions affecting oligodendroglial Sox2 expression may be therapeutic for overcoming OPC differentiation arrest in dysmyelinating and demyelinating disorders.


Subject(s)
Oligodendroglia/cytology , Oligodendroglia/metabolism , SOXB1 Transcription Factors/metabolism , Spinal Cord/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Movement , Cell Proliferation , Gene Deletion , Gene Expression Regulation , Integrases/metabolism , Mice, Transgenic , Mutation/genetics , Myelin Sheath/metabolism , Spinal Cord/embryology , Tamoxifen/pharmacology , Time Factors
5.
Mol Ther ; 26(3): 793-800, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29456021

ABSTRACT

Canavan disease, a leukodystrophy caused by loss-of-function ASPA mutations, is characterized by brain dysmyelination, vacuolation, and astrogliosis ("spongiform leukodystrophy"). ASPA encodes aspartoacylase, an oligodendroglial enzyme that cleaves the abundant brain amino acid N-acetyl-L-aspartate (NAA) to L-aspartate and acetate. Aspartoacylase deficiency results in a 50% or greater elevation in brain NAA concentration ([NAAB]). Prior studies showed that homozygous constitutive knockout of Nat8l, the gene encoding the neuronal NAA synthesizing enzyme N-acetyltransferase 8-like, prevents aspartoacylase-deficient mice from developing spongiform leukodystrophy. We now report that brain Nat8l knockdown elicited by intracerebroventricular/intracisternal administration of an adeno-associated viral vector carrying a short hairpin Nat8l inhibitory RNA to neonatal aspartoacylase-deficient AspaNur7/Nur7 mice lowers [NAAB] and suppresses development of spongiform leukodystrophy.


Subject(s)
Acetyltransferases/genetics , Amidohydrolases/deficiency , Canavan Disease/genetics , Canavan Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Canavan Disease/pathology , Canavan Disease/physiopathology , Dependovirus/genetics , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Knockout , Motor Activity , Neurons/metabolism , RNA, Messenger/genetics , Transduction, Genetic
6.
J Neurosci ; 38(7): 1802-1820, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29335358

ABSTRACT

In the CNS, myelination and remyelination depend on the successful progression and maturation of oligodendroglial lineage cells, including proliferation and differentiation of oligodendroglial progenitor cells (OPCs). Previous studies have reported that Sox2 transiently regulates oligodendrocyte (OL) differentiation in the embryonic and perinatal spinal cord and appears dispensable for myelination in the postnatal spinal cord. However, the role of Sox2 in OL development in the brain has yet to be defined. We now report that Sox2 is an essential positive regulator of developmental myelination in the postnatal murine brain of both sexes. Stage-specific paradigms of genetic disruption demonstrated that Sox2 regulated brain myelination by coordinating upstream OPC population supply and downstream OL differentiation. Transcriptomic analyses further supported a crucial role of Sox2 in brain developmental myelination. Consistently, oligodendroglial Sox2-deficient mice developed severe tremors and ataxia, typical phenotypes indicative of hypomyelination, and displayed severe impairment of motor function and prominent deficits of brain OL differentiation and myelination persisting into the later CNS developmental stages. We also found that Sox2 was required for efficient OPC proliferation and expansion and OL regeneration during remyelination in the adult brain and spinal cord. Together, our genetic evidence reveals an essential role of Sox2 in brain myelination and CNS remyelination, and suggests that manipulation of Sox2 and/or Sox2-mediated downstream pathways may be therapeutic in promoting CNS myelin repair.SIGNIFICANCE STATEMENT Promoting myelin formation and repair has translational significance in treating myelin-related neurological disorders, such as periventricular leukomalacia and multiple sclerosis in which brain developmental myelin formation and myelin repair are severely affected, respectively. In this report, analyses of a series of genetic conditional knock-out systems targeting different oligodendrocyte stages reveal a previously unappreciated role of Sox2 in coordinating upstream proliferation and downstream differentiation of oligodendroglial lineage cells in the mouse brain during developmental myelination and CNS remyelination. Our study points to the potential of manipulating Sox2 and its downstream pathways to promote oligodendrocyte regeneration and CNS myelin repair.


Subject(s)
Brain/growth & development , Brain/physiology , Central Nervous System/physiology , Myelin Sheath/physiology , Oligodendroglia/physiology , SOXB1 Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Demyelinating Diseases/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Movement Disorders/physiopathology , Nerve Regeneration/physiology , Spinal Cord/growth & development , Spinal Cord/physiology , Transcriptome
7.
J Neurosci ; 37(2): 413-421, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28077719

ABSTRACT

Canavan disease is a leukodystrophy caused by aspartoacylase (ASPA) deficiency. The lack of functional ASPA, an enzyme enriched in oligodendroglia that cleaves N-acetyl-l-aspartate (NAA) to acetate and l-aspartic acid, elevates brain NAA and causes "spongiform" vacuolation of superficial brain white matter and neighboring gray matter. In children with Canavan disease, neuroimaging shows early-onset dysmyelination and progressive brain atrophy. Neuron loss has been documented at autopsy in some cases. Prior studies have shown that mice homozygous for the Aspa nonsense mutation Nur7 also develop brain vacuolation. We now report that numbers of cerebral cortical and cerebellar neurons are decreased and that cerebral cortex progressively thins in AspaNur7/Nur7 mice. This neuronal pathology is prevented by constitutive disruption of Nat8l, which encodes the neuronal NAA-synthetic enzyme N-acetyltransferase-8-like. SIGNIFICANCE STATEMENT: This is the first demonstration of cortical and cerebellar neuron depletion and progressive cerebral cortical thinning in an animal model of Canavan disease. Genetic suppression of N-acetyl-l-aspartate (NAA) synthesis, previously shown to block brain vacuolation in aspartoacylase-deficient mice, also prevents neuron loss and cerebral cortical atrophy in these mice. These results suggest that lowering the concentration of NAA in the brains of children with Canavan disease would prevent or slow progression of neurological deficits.


Subject(s)
Aspartic Acid/analogs & derivatives , Canavan Disease/metabolism , Disease Models, Animal , Neurons/metabolism , Animals , Aspartic Acid/biosynthesis , Aspartic Acid/deficiency , Aspartic Acid/genetics , Canavan Disease/genetics , Canavan Disease/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology
8.
PLoS One ; 11(12): e0167573, 2016.
Article in English | MEDLINE | ID: mdl-27907123

ABSTRACT

Charcot-Marie-Tooth disease type 2A (CMT2A), the most common axonal form of hereditary sensory motor neuropathy, is caused by mutations of mitofusin-2 (MFN2). Mitofusin-2 is a GTPase required for fusion of mitochondrial outer membranes, repair of damaged mitochondria, efficient mitochondrial energetics, regulation of mitochondrial-endoplasmic reticulum calcium coupling and axonal transport of mitochondria. We knocked T105M MFN2 preceded by a loxP-flanked STOP sequence into the mouse Rosa26 locus to permit cell type-specific expression of this pathogenic allele. Crossing these mice with nestin-Cre transgenic mice elicited T105M MFN2 expression in neuroectoderm, and resulted in diminished numbers of mitochondria in peripheral nerve axons, an alteration in skeletal muscle fiber type distribution, and a gait abnormality.


Subject(s)
Alleles , Charcot-Marie-Tooth Disease/genetics , GTP Phosphohydrolases/genetics , Hemizygote , Muscle Fibers, Skeletal/metabolism , Peripheral Nerves/metabolism , Amino Acid Substitution , Animals , Axonal Transport , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Embryo, Mammalian , GTP Phosphohydrolases/deficiency , Gait , Gene Expression Regulation , Hindlimb/pathology , Humans , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Muscle Fibers, Skeletal/pathology , Nestin/genetics , Nestin/metabolism , Neural Plate/metabolism , Neural Plate/pathology , Peripheral Nerves/pathology , Phenotype , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
9.
Cell Mol Bioeng ; 9(1): 96-106, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-27087859

ABSTRACT

Numerous signaling molecules are altered following nerve injury, serving as a blueprint for drug delivery approaches that promote nerve repair. However, challenges with achieving the appropriate temporal duration of recombinant protein delivery have limited the therapeutic success of this approach. Genetic engineering of mesenchymal stem cells (MSCs) to enhance the secretion of proangiogenic molecules such as vascular endothelial growth factor (VEGF) may provide an alternative. We hypothesized that the administration of VEGF-expressing human MSCs would stimulate neurite outgrowth and proliferation of cell-types involved in neural repair. When cultured with dorsal root ganglion (DRG) explants in vitro, control and VEGF-expressing MSCs (VEGF-MSCs) increased neurite extension and proliferation of Schwann cells (SCs) and endothelial cells, while VEGF-MSCs stimulated significantly greater proliferation of endothelial cells. When embedded within a 3D fibrin matrix, VEGF-MSCs maintained overexpression and expressed detectable levels over 21 days. After transplantation into a murine sciatic nerve injury model, VEGF-MSCs maintained high VEGF levels for 2 weeks. This study provides new insight into the role of VEGF on peripheral nerve injury and the viability of transplanted genetically engineered MSCs. The study aims to provide a framework for future studies with the ultimate goal of developing an improved therapy for nerve repair.

10.
J Neurosci ; 35(9): 3756-63, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25740506

ABSTRACT

Astrocytes are the most abundant cells in the CNS, and have many essential functions, including maintenance of blood-brain barrier integrity, and CNS water, ion, and glutamate homeostasis. Mammalian astrogliogenesis has generally been considered to be completed soon after birth, and to be reactivated in later life only under pathological circumstances. Here, by using genetic fate-mapping, we demonstrate that new corpus callosum astrocytes are continuously generated from nestin(+) subventricular zone (SVZ) neural progenitor cells (NPCs) in normal adult mice. These nestin fate-mapped corpus callosum astrocytes are uniformly postmitotic, express glutamate receptors, and form aquaporin-4(+) perivascular endfeet. The entry of new astrocytes from the SVZ into the corpus callosum appears to be balanced by astroglial apoptosis, because overall numbers of corpus callosum astrocytes remain constant during normal adulthood. Nestin fate-mapped astrocytes also flow anteriorly from the SVZ in association with the rostral migratory stream, but do not penetrate into the deeper layers of the olfactory bulb. Production of new astrocytes from nestin(+) NPCs is absent in the normal adult cortex, striatum, and spinal cord. Our study is the first to demonstrate ongoing SVZ astrogliogenesis in the normal adult mammalian forebrain.


Subject(s)
Astrocytes/physiology , Corpus Callosum/cytology , Corpus Callosum/physiology , Lateral Ventricles/cytology , Lateral Ventricles/physiology , Animals , Brain/cytology , Brain/growth & development , Cell Movement , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nestin/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Spinal Cord/cytology , Spinal Cord/growth & development , Vesicular Glutamate Transport Proteins/metabolism
11.
Ann Neurol ; 77(5): 884-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25712859

ABSTRACT

Canavan disease is caused by inactivating ASPA (aspartoacylase) mutations that prevent cleavage of N-acetyl-L-aspartate (NAA), resulting in marked elevations in central nervous system (CNS) NAA and progressively worsening leukodystrophy. We now report that ablating NAA synthesis by constitutive genetic disruption of Nat8l (N-acetyltransferase-8 like) permits normal CNS myelination and prevents leukodystrophy in a murine Canavan disease model.


Subject(s)
Aspartic Acid/analogs & derivatives , Canavan Disease/metabolism , Canavan Disease/prevention & control , Disease Models, Animal , Animals , Aspartic Acid/deficiency , Aspartic Acid/genetics , Aspartic Acid/metabolism , Canavan Disease/genetics , Female , Male , Mice , Mice, Knockout
12.
J Neurosci ; 34(32): 10729-42, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25100604

ABSTRACT

Motoneuron death after transection of the axons (axotomy) in neonates is believed to share the same mechanistic bases as naturally occurring programmed cell death during development. The c-Jun N-terminal kinase pathway is activated in both forms of motoneuron death, but it remains unknown to what extent these two forms of motoneuron death depend on this pathway and which upstream kinases are involved. We found that numbers of facial motoneurons are doubled in neonatal mice deficient in either ZPK/DLK (zipper protein kinase, also known as dual leucine zipper kinase), a mitogen-activated protein kinase kinase kinase, or in MKK4/MAP2K4, a mitogen-activated protein kinase kinase directly downstream of ZPK/DLK, and that the facial motoneurons in those mutant mice are completely resistant to axotomy-induced death. Conditional deletion of MKK4/MAP2K4 in neurons further suggested that ZPK/DLK and MKK4/MAP2K4-dependent mechanisms underlying axotomy-induced death are motoneuron autonomous. Nevertheless, quantitative analysis of facial motoneurons during embryogenesis revealed that both ZPK/DLK and MKK4/MAP2K4-dependent and -independent mechanisms contribute to developmental elimination of excess motoneurons. In contrast to MKK4/MAP2K4, mice lacking MKK7/MAP2K7, another mitogen-activated protein kinase kinase directly downstream of ZPK/DLK, conditionally in neurons did not have excess facial motoneurons. However, some MKK7/MAP2K7-deficient facial motoneurons were resistant to axotomy-induced death, indicating a synergistic effect of MKK7/MAP2K7 on axotomy-induced death of these facial motoneurons. Together, our study provides compelling evidence for the pivotal roles of the ZPK/DLK and MKK4/MAP2K4-dependent mechanism in axotomy-induced motoneuron death in neonates and also demonstrates that axotomy-induced motoneuron death is not identical to developmental motoneuron death with respect to the involvement of ZPK/DLK, MKK4/MAP2K4 and MKK7/MAP2K7.


Subject(s)
Central Nervous System/pathology , Facial Nerve Injuries/pathology , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinases/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Axotomy/adverse effects , Calcium-Binding Proteins/metabolism , Cell Death/physiology , Central Nervous System/growth & development , Central Nervous System/metabolism , Choline O-Acetyltransferase/metabolism , Dextrans , Disease Models, Animal , Facial Nerve Injuries/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Phosphopyruvate Hydratase/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rhodamines , Signal Transduction/genetics
13.
J Neurosci ; 34(24): 8175-85, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24920622

ABSTRACT

Current multiple sclerosis (MS) therapies only partially prevent chronically worsening neurological deficits, which are largely attributable to progressive loss of CNS axons. Prior studies of experimental autoimmune encephalomyelitis (EAE) induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG peptide), a model of MS, documented continued axon loss for months after acute CNS inflammatory infiltrates had subsided, and massive astroglial induction of CCL2 (MCP-1), a chemokine for CCR2(+) monocytes. We now report that conditional deletion of astroglial CCL2 significantly decreases CNS accumulation of classically activated (M1) monocyte-derived macrophages and microglial expression of M1 markers during the initial CNS inflammatory phase of MOG peptide EAE, reduces the acute and long-term severity of clinical deficits and slows the progression of spinal cord axon loss. In addition, lack of astroglial-derived CCL2 results in increased accumulation of Th17 cells within the CNS in these mice, but also in greater confinement of CD4(+) lymphocytes to CNS perivascular spaces. These findings suggest that therapies designed to inhibit astroglial CCL2-driven trafficking of monocyte-derived macrophages to the CNS during acute MS exacerbations have the potential to significantly reduce CNS axon loss and slow progression of neurological deficits.


Subject(s)
Astrocytes/metabolism , Axons/pathology , Central Nervous System/pathology , Chemokine CCL2/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Macrophages/metabolism , Analysis of Variance , Animals , Axons/ultrastructure , Bacterial Proteins/genetics , Central Nervous System/ultrastructure , Chemokine CCL2/genetics , Flow Cytometry , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Luminescent Proteins/genetics , Macrophages/immunology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Proteins/genetics
14.
J Neuroinflammation ; 11: 105, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24924222

ABSTRACT

Multiple sclerosis (MS) is characterized by central nervous system (CNS) inflammation, demyelination, and axonal degeneration. CXCL10 (IP-10), a chemokine for CXCR3+ T cells, is known to regulate T cell differentiation and migration in the periphery, but effects of CXCL10 produced endogenously in the CNS on immune cell trafficking are unknown. We created floxed cxcl10 mice and crossed them with mice carrying an astrocyte-specific Cre transgene (mGFAPcre) to ablate astroglial CXCL10 synthesis. These mice, and littermate controls, were immunized with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG peptide) to induce experimental autoimmune encephalomyelitis (EAE). In comparison to the control mice, spinal cord CXCL10 mRNA and protein were sharply diminished in the mGFAPcre/CXCL10fl/fl EAE mice, confirming that astroglia are chiefly responsible for EAE-induced CNS CXCL10 synthesis. Astroglial CXCL10 deletion did not significantly alter the overall composition of CD4+ lymphocytes and CD11b+ cells in the acutely inflamed CNS, but did diminish accumulation of CD4+ lymphocytes in the spinal cord perivascular spaces. Furthermore, IBA1+ microglia/macrophage accumulation within the lesions was not affected by CXCL10 deletion. Clinical deficits were milder and acute demyelination was substantially reduced in the astroglial CXCL10-deleted EAE mice, but long-term axon loss was equally severe in the two groups. We concluded that astroglial CXCL10 enhances spinal cord perivascular CD4+ lymphocyte accumulation and acute spinal cord demyelination in MOG peptide EAE, but does not play an important role in progressive axon loss in this MS model.


Subject(s)
Astrocytes/metabolism , Axons/pathology , Chemokine CXCL10/deficiency , Encephalomyelitis, Autoimmune, Experimental/pathology , Animals , Calcium-Binding Proteins/metabolism , Central Nervous System/pathology , Chemokine CXCL10/genetics , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Glial Fibrillary Acidic Protein/metabolism , Leukocytes/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Myelin Basic Protein/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Spinal Cord/pathology , Spleen/pathology , Time Factors
15.
J Neuropathol Exp Neurol ; 73(4): 335-44, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24607968

ABSTRACT

We provide evidence of cortical neuronopathy in myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis, an established model of chronic multiple sclerosis. To investigate phenotypic perturbations in neurons in this model, we used apoptotic markers and immunohistochemistry with antibodies to NeuN and other surrogate markers known to be expressed by adult pyramidal Layer V somas, including annexin V, encephalopsin, and Emx1. We found no consistent evidence of chronic loss of Layer V neurons but detected both reversible and chronic decreases in the expression of these markers in conjunction with evidence of cortical demyelination and presynaptic loss. These phenotypic perturbations were present in, but not restricted to, the neocortical Layer V. We also investigated inflammatory responses in the cortex and subcortical white matter of the corpus callosum and spinal dorsal funiculus and found that those in the cortex and corpus callosum were delayed compared with those in the spinal cord. Inflammatory infiltrates initially included T cells, neutrophils, and Iba1-positive microglia/macrophages in the corpus callosum, whereas only Iba1-positive cells were present in the cortex. These data indicate that we have identified a new temporal pattern of subtle phenotypic perturbations in neocortical neurons in this chronic multiple sclerosis model.


Subject(s)
Motor Neurons/pathology , Multiple Sclerosis/pathology , Neocortex/pathology , Animals , Caspase 3/metabolism , Cell Death/drug effects , Disease Models, Animal , Encephalitis/etiology , Freund's Adjuvant/toxicity , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Motor Neurons/drug effects , Multiple Sclerosis/chemically induced , Multiple Sclerosis/complications , Myelin Basic Protein/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Neocortex/drug effects , Peptide Fragments/toxicity , Phosphopyruvate Hydratase/metabolism , Synaptophysin/metabolism , Time Factors
16.
Ann Biomed Eng ; 42(6): 1271-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24570390

ABSTRACT

Upon nerve injury, the body creates an environment consisting of permissive and non-permissive cues that instruct the function of cells involved in nerve repair. Among other roles, the developing extracellular matrix (ECM) acts as an underlying substrate to guide the union of neurites extending from the proximal stump for bridging the nerve gap. Chondroitin sulfate proteoglycans (CSPGs) are present in the nerve ECM and inhibit axon growth, potentially providing molecular cues to prevent aberrant growth and direct regeneration. In this study, we examined the potential of CSPGs to guide dorsal root ganglia (DRG) neurite outgrowth when freely available in the media or presented from a polymeric membrane. Soluble CSPGs added to the media of DRG explant cultures inhibited neurite outgrowth without spatial bias, caused retraction of axons, and decreased neurite extension in a dose-dependent manner. Poly-L-lactic acid membranes were chemically treated to enhance adsorption of CSPGs to the surface. CSPGs bound to 1,6-hexanediamine-treated membranes directed the orientation of neurite outgrowth, as neurites avoided bound CSPGs and a higher number and percentage grew on treated membranes lacking CSPGs. DRG explants cultured on CSPG-coated membranes without 1,6-hexanediamine-treatment had a smaller number of neurites and decreased neurite outgrowth, suggesting CSPGs were not retained on the membrane and were released into the culture medium. Taken together, these data demonstrate the potential of CSPG presentation to guide axonal growth. This approach offers a strategy to improve upon existing nerve guidance conduits by incorporating axon guidance molecules to direct nerve regeneration.


Subject(s)
Chondroitin Sulfate Proteoglycans/chemistry , Coated Materials, Biocompatible/chemistry , Ganglia, Spinal/metabolism , Membranes, Artificial , Neurites/metabolism , Animals , Cells, Cultured , Ganglia, Spinal/cytology , Mice
17.
J Neurosci ; 33(7): 3113-30, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23407966

ABSTRACT

The expression of the gut tumor suppressor gene adenomatous polyposis coli (Apc) and its role in the oligodendroglial lineage are poorly understood. We found that immunoreactive APC is transiently induced in the oligodendroglial lineage during both normal myelination and remyelination following toxin-induced, genetic, or autoimmune demyelination murine models. Using the Cre/loxP system to conditionally ablate APC from the oligodendroglial lineage, we determined that APC enhances proliferation of oligodendroglial progenitor cells (OPCs) and is essential for oligodendrocyte differentiation in a cell-autonomous manner. Biallelic Apc disruption caused translocation of ß-catenin into the nucleus and upregulated ß-catenin-mediated Wnt signaling in early postnatal but not adult oligodendroglial lineage cells. The results of conditional ablation of Apc or Ctnnb1 (the gene encoding ß-catenin) and of simultaneous conditional ablation of Apc and Ctnnb1 revealed that ß-catenin is dispensable for postnatal oligodendroglial differentiation, that Apc one-allele deficiency is not sufficient to dysregulate ß-catenin-mediated Wnt signaling in oligodendroglial lineage cells, and that APC regulates oligodendrocyte differentiation through ß-catenin-independent, as well as ß-catenin-dependent, mechanisms. Gene ontology analysis of microarray data suggested that the ß-catenin-independent mechanism involves APC regulation of the cytoskeleton, a result compatible with established APC functions in neural precursors and with our observation that Apc-deleted OPCs develop fewer, shorter processes in vivo. Together, our data support the hypothesis that APC regulates oligodendrocyte differentiation through both ß-catenin-dependent and additional ß-catenin-independent mechanisms.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Oligodendroglia/physiology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/immunology , Animals , Antibodies , Blotting, Western , Cell Differentiation/physiology , Cell Lineage/physiology , Cells, Cultured , Estrogen Antagonists/pharmacology , Immunoprecipitation , In Situ Hybridization , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Confocal , Myelin-Associated Glycoprotein/biosynthesis , Nerve Regeneration/physiology , RNA/biosynthesis , RNA/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Stem Cells/physiology , Tamoxifen/pharmacology , beta Catenin/physiology
18.
J Neurol Sci ; 333(1-2): 55-9, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23294494

ABSTRACT

Astroglia, the most abundant cells in the human CNS, and even more prominent in multiple sclerosis patients, participate in CNS innate and adaptive immunity, and have been hypothesized to play an important role in multiple sclerosis progression. Experimental autoimmune encephalomyelitis elicited in mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55 provides a means by which to explore the genesis and disease significance of astrogliosis during a chronic immune-mediated CNS inflammatory/demyelinative disorder that, in its' pathological features, strongly resembles multiple sclerosis.


Subject(s)
Astrocytes/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gliosis/pathology , Models, Biological , Multiple Sclerosis/pathology , Animals , Astrocytes/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gliosis/chemically induced , Gliosis/immunology , Humans , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments
19.
Tissue Eng Part A ; 17(23-24): 2931-42, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21882895

ABSTRACT

Fibrin is a promising matrix for use in promoting nerve repair given its natural occurrence in peripheral nerve injuries, and the biophysical properties of this matrix can be regulated to modulate tissue regeneration. In this study, we examined the effect of physical and mechanical properties of fibrin gels on dorsal root ganglia (DRG) neurite extension. Increases in fibrinogen concentration increased the number of fibrin strands, resulting in decreased pore size and increased stiffness. Neurite extension was reduced when DRG explants were cultured within fibrin gels of increasing fibrinogen concentrations (from 9.5 to 141 mg/mL). The addition of NaCl also increased the number of fibrin strands, reducing fiber diameter and porosity, while increasing mechanical strength, and reductions in neurite extension correlated with increases in NaCl content. We determined that neurite extension within fibrin gels is dependent on fibrinolysis and is mediated by the secretion of serine proteases and matrix metalloproteinases by entrapped DRGs, as confirmed by culturing cells in the presence of inhibitors against these enzymes and real-time-polymerase chain reaction. Taken together, the results of this study provide new insight into the effect of fibrin gel biophysical properties on neurite extension and suggest new opportunities to improve the efficacy of these materials when used as nerve guidance conduits.


Subject(s)
Fibrin/pharmacology , Gels/pharmacology , Mechanical Phenomena/drug effects , Neurites/drug effects , Neurites/metabolism , Animals , Fibrin/ultrastructure , Fibrinogen/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/growth & development , Humans , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Protease Inhibitors/pharmacology , Proteolysis/drug effects , Serine Proteases/genetics , Serine Proteases/metabolism , Sodium Chloride/pharmacology , Time Factors
20.
J Neurosci ; 31(33): 11914-28, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849552

ABSTRACT

Accumulations of hypertrophic, intensely glial fibrillary acidic protein-positive (GFAP(+)) astroglia, which also express immunoreactive nestin and vimentin, are prominent features of multiple sclerosis lesions. The issues of the cellular origin of hypertrophic GFAP(+)/vimentin(+)/nestin(+) "reactive" astroglia and also the plasticities and lineage relationships among three macroglial progenitor populations-oligodendrocyte progenitor cells (OPCs), astrocytes and ependymal cells-during multiple sclerosis and other CNS diseases remain controversial. We used genetic fate-mappings with a battery of inducible Cre drivers (Olig2-Cre-ER(T2), GFAP-Cre-ER(T2), FoxJ1-Cre-ER(T2) and Nestin-Cre-ER(T2)) to explore these issues in adult mice with myelin oligodendrocyte glycoprotein peptide-induced experimental autoimmune encephalomyelitis (EAE). The proliferative rate of spinal cord OPCs rose fivefold above control levels during EAE, and numbers of oligodendroglia increased as well, but astrogenesis from OPCs was rare. Spinal cord ependymal cells, previously reported to be multipotent, did not augment their low proliferative rate, nor give rise to astroglia or OPCs. Instead, the hypertrophic, vimentin(+)/nestin(+), reactive astroglia that accumulated in spinal cord in this multiple sclerosis model were derived by proliferation and phenotypic transformation of fibrous astroglia in white matter, and solely by phenotypic transformation of protoplasmic astroglia in gray matter. This comprehensive analysis of macroglial plasticity in EAE helps to clarify the origins of astrogliosis in CNS inflammatory demyelinative disorders.


Subject(s)
Astrocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Neuronal Plasticity/physiology , Oligodendroglia/pathology , Animals , Astrocytes/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendroglia/physiology , Spinal Cord/cytology , Spinal Cord/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...