Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Exp Med ; 220(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37067791

ABSTRACT

Material transfer is an essential form of intercellular communication to exchange information and resources between cells. Material transfer between neurons and from glia to neurons has been demonstrated to support neuronal survival and activity. Understanding the extent of material transfer in the healthy nervous system is limited. Here we report that in the mouse central nervous system (CNS), neurons receive nuclear and ribosomal material of Sox10-lineage cell (SOL) origin. We show that transfer of SOL-derived material to neurons is region dependent, establishes during postnatal brain maturation, and dynamically responds to LPS-induced neuroinflammation in the adult mouse brain. We identified satellite oligodendrocyte-neuron pairs with loss of plasma membrane integrity between nuclei, suggesting direct material transfer. Together, our findings provide evidence of regionally coordinated transfer of SOL-derived nuclear and ribosomal material to neurons in the mouse CNS, with potential implications for the understanding and modulation of neuronal function and treatment of neurological disorders.


Subject(s)
Neuroglia , Neurons , Animals , Mice , Neurons/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Brain/metabolism , SOXE Transcription Factors/metabolism
2.
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.
Am J Physiol Cell Physiol ; 320(4): C619-C634, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33406028

ABSTRACT

Hyperglycemia exacerbates edema formation and worsens neurological outcome in ischemic stroke. Edema formation in the early hours of stroke involves transport of ions and water across an intact blood-brain barrier (BBB), and swelling of astrocytes. We showed previously that high glucose (HG) exposures of 24 hours to 7 days increase abundance and activity of BBB Na+-K+-2Cl- cotransport (NKCC) and Na+/H+ exchange 1 (NHE1). Further, bumetanide and HOE-642 inhibition of these transporters significantly reduces edema and infarct following middle cerebral artery occlusion in hyperglycemic rats, suggesting that NKCC and NHE1 are effective therapeutic targets for reducing edema in hyperglycemic stroke. The mechanisms underlying hyperglycemia effects on BBB NKCC and NHE1 are not known. In the present study we investigated whether serum-glucocorticoid regulated kinase 1 (SGK1) and protein kinase C beta II (PKCßII) are involved in HG effects on BBB NKCC and NHE1. We found transient increases in phosphorylated SGK1 and PKCßII within the first hour of HG exposure, after 5-60 min for SGK1 and 5 min for PKCßII. However, no changes were observed in cerebral microvascular endothelial cell SGK1 or PKCßII abundance or phosphorylation (activity) after 24 or 48 h HG exposures. Further, we found that HG-induced increases in NKCC and NHE1 abundance were abolished by inhibition of SGK1 but not PKCßII, whereas the increases in NKCC and NHE activity were abolished by inhibition of either kinase. Finally, we found evidence that STE20/SPS1-related proline/alanine-rich kinase and oxidative stress-responsive kinase-1 (SPAK/OSR1) participate in the HG-induced effects on BBB NKCC.


Subject(s)
Blood-Brain Barrier/drug effects , Endothelial Cells/drug effects , Glucose/toxicity , Immediate-Early Proteins/metabolism , Protein Kinase C beta/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium-Hydrogen Exchanger 1/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Cattle , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/pathology , Enzyme Activation , Humans , Phosphorylation , Signal Transduction , Time Factors
4.
Transl Neurodegener ; 9(1): 13, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32345341

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is one of the neurodegeneration diseases characterized by the gradual loss of dopaminergic (DA) neurons in the substantia nigra region of the brain. Substantial evidence indicates that at the cellular level mitochondrial dysfunction is a key factor leading to pathological features such as neuronal death and accumulation of misfolded α-synuclein aggregations. Autologous transplantation of healthy purified mitochondria has shown to attenuate phenotypes in vitro and in vivo models of PD. However, there are significant technical difficulties in obtaining large amounts of purified mitochondria with normal function. In addition, the half-life of mitochondria varies between days to a few weeks. Thus, identifying a continuous source of healthy mitochondria via intercellular mitochondrial transfer is an attractive option for therapeutic purposes. In this study, we asked whether iPSCs derived astrocytes can serve as a donor to provide functional mitochondria and rescue injured DA neurons after rotenone exposure in an in vitro model of PD. METHODS: We generated DA neurons and astrocytes from human iPSCs and hESCs. We established an astroglial-neuronal co-culture system to investigate the intercellular mitochondrial transfer, as well as the neuroprotective effect of mitochondrial transfer. We employed immunocytochemistry and FACS analysis to track mitochondria. RESULTS: We showed evidence that iPSCs-derived astrocytes or astrocytic conditioned media (ACM) can rescue DA neurons degeneration via intercellular mitochondrial transfer in a rotenone induced in vitro PD model. Specifically, we showed that iPSCs-derived astrocytes from health spontaneously release functional mitochondria into the media. Mito-Tracker Green tagged astrocytic mitochondria were detected in the ACM and were shown to be internalized by the injured neurons via a phospho-p38 depended pathway. Transferred mitochondria were able to significantly reverse DA neurodegeneration and axonal pruning following exposure to rotenone. When rotenone injured neurons were cultured in presence of ACM depleted of mitochondria (by ultrafiltration), the neuroprotective effects were abolished. CONCLUSIONS: Our studies provide the proof of principle that iPSCs-derived astrocytes can act as mitochondria donor to the injured DA neurons and attenuate pathology. Using iPSCs derived astrocytes as a donor can provide a novel strategy that can be further developed for cellular therapy for PD.


Subject(s)
Astrocytes/physiology , Dopaminergic Neurons/physiology , Induced Pluripotent Stem Cells/physiology , Insecticides/toxicity , Mitochondria/physiology , Rotenone/toxicity , Astrocytes/drug effects , Cell Survival , Coculture Techniques , Dopaminergic Neurons/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Mitochondria/drug effects
6.
Stem Cell Res Ther ; 10(1): 272, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455382

ABSTRACT

BACKGROUND: Oligodendrocytes are a type of glial cells that synthesize the myelin sheath around the axons and are critical for the nerve conduction in the CNS. Oligodendrocyte death and defects are the leading causes of several myelin disorders such as multiple sclerosis, progressive multifocal leukoencephalopathy, periventricular leukomalacia, and several leukodystrophies. Temporal activation of the Sonic Hedgehog (SHH) pathway is critical for the generation of oligodendrocyte progenitors, and their differentiation and maturation in the brain and spinal cord during embryonic development in mammals. METHODS: Our protocol utilized adherent cultures of human induced pluripotent stem cells (iPSC) and human embryonic stem cells (hESCs) with a green fluorescent protein (GFP) reporter knocked into one allele of the OLIG2 gene locus, dual SMAD inhibition, and transient partial inhibition of glioma-associated oncogene 1 (GLI1) by the small molecule GANT61 during the formation of the SOX2/PAX6-positive neural stem cells (NSCs). The SHH pathway was later restimulated by a Smoothened agonist purmorphamine to induce the generation of OLIG2 glial precursors. One hundred ninety-two individual oligodendrocyte precursor cells (OPCs) from GANT61 and control group were analyzed by single-cell RNA sequencing (RNA-Seq). RESULTS: We demonstrate here that transient and partial inhibition of the SHH pathway transcription factor GLI1 in NSCs by a small molecule inhibitor GANT61 was found to generate OPCs that were more migratory and could differentiate earlier toward myelin-producing oligodendrocytes. Single-cell transcriptomic analysis (RNA-Seq) showed that GANT61-NSC-derived oligodendrocyte precursor cells (OPCs) had differential activation of some of the genes in the cytoskeleton rearrangement pathways that are involved in OPC motility and induction of maturation. At the protein level, this was also associated with higher levels of myelin-specific genes in the GANT61 group compared to controls. GANT61-NSC-derived OPCs were functional and could generate compact myelin in vitro and in vivo after transplantation in myelin-deficient shiverer mice. CONCLUSIONS: This is a small molecule-based in vitro protocol that leads to the faster generation of functional oligodendrocytes. The development of protocols that lead to efficient and faster differentiation of oligodendrocytes from progenitors provides important advances toward the development of autologous neural stem cell-based therapies using human iPSCs.


Subject(s)
Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Zinc Finger Protein GLI1/metabolism , Animals , Axons/drug effects , Axons/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Female , Hedgehog Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Neural Stem Cells/drug effects , Neurons/drug effects , Neurons/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Rats , Signal Transduction/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism
7.
J Cereb Blood Flow Metab ; 39(9): 1678-1692, 2019 09.
Article in English | MEDLINE | ID: mdl-29739261

ABSTRACT

Cerebral edema is exacerbated in diabetic ischemic stroke through poorly understood mechanisms. We showed previously that blood-brain barrier (BBB) Na-K-Cl cotransport (NKCC) and Na/H exchange (NHE) are major contributors to edema formation in normoglycemic ischemic stroke. Here, we investigated whether hyperglycemia-exacerbated edema involves changes in BBB NKCC and NHE expression and/or activity and whether inhibition of NKCC or NHE effectively reduces edema and injury in a type I diabetic model of hyperglycemic stroke. Cerebral microvascular endothelial cell (CMEC) NKCC and NHE abundances and activities were determined by Western blot, radioisotopic flux and microspectrofluorometric methods. Cerebral edema and Na in rats subjected to middle cerebral artery occlusion (MCAO) were assessed by nuclear magnetic resonance methods. Hyperglycemia exposures of 1-7d significantly increased CMEC NKCC and NHE abundance and activity. Subsequent exposure to ischemic factors caused more robust increases in NKCC and NHE activities than in normoglycemic CMEC. MCAO-induced edema and brain Na uptake were greater in hyperglycemic rats. Intravenous bumetanide and HOE-642 significantly attenuated edema, brain Na uptake and ischemic injury. Our findings provide evidence that BBB NKCC and NHE contribute to increased edema in hyperglycemic stroke, suggesting that these Na transporters are promising therapeutic targets for reducing damage in diabetic stroke.


Subject(s)
Brain Edema/complications , Hyperglycemia/complications , Infarction, Middle Cerebral Artery/complications , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Edema/metabolism , Brain Edema/pathology , Cattle , Cell Line , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Hyperglycemia/pathology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchangers/analysis , Sodium-Potassium-Chloride Symporters/analysis , Streptozocin
8.
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
10.
Sci Rep ; 6: 22556, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26925573

ABSTRACT

The mitochondrial translocator protein (TSPO) has been implicated in CNS diseases. Here, we sought to determine the specific role of TSPO in experimental autoimmune encephalomyelitis (EAE), the most studied animal model of multiple sclerosis (MS). To fundamentally elucidate the functions of TSPO, we first developed a viable TSPO knockout mouse. A conditional TSPO knockout mouse was generated by utilizing the Cre-Lox system. We generated a TSPO floxed mouse, and then crossed this mouse with a Cre recombinase expressing mouse driven by the human glial fibrillary acidic protein (hGFAP) promoter. The resultant mouse was a neural linage line specific TSPO knockout. The loss of TSPO in the CNS did not result in overt developmental defects or phenotypes. The TSPO-/- mouse showed a decrease in GFAP expression, correlating with a decrease in astrogliosis in response to neural injury during EAE. This decrease in astrogliosis was also witnessed in the lessening of severity of EAE clinical scoring, indicating an in vivo functional role for TSPO in suppressing EAE. The TSPO-/- mouse could be a useful tool in better understanding the role of TSPO in CNS disease, and our results implicate TSPO as a potential therapeutic target in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Glial Fibrillary Acidic Protein/genetics , Receptors, GABA/genetics , Animals , Central Nervous System/pathology , Chemokine CXCL10/genetics , Disease Models, Animal , Female , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/genetics
11.
12.
Nat Commun ; 4: 2196, 2013.
Article in English | MEDLINE | ID: mdl-23880652

ABSTRACT

Human pluripotent stem cells (hPSCs) have been differentiated to astroglia, but the utilization of hPSC-derived astroglia as cell therapy for neurological diseases has not been well studied. Astroglia are heterogeneous, and not all astroglia are equivalent in promoting neural repair. A prerequisite for cell therapy is to derive defined cell populations with superior therapeutic effects. Here we use an Olig2-GFP human embryonic stem cell (hESC) reporter to demonstrate that hESC-derived Olig2(+) progenitors generate a subtype of previously uncharacterized astroglia (Olig2PC-Astros). These Olig2PC-Astros differ substantially from astroglia differentiated from Olig2-negative hESC-derived neural progenitor cells (NPC-Astros), particularly in their neuroprotective properties. When grafted into brains subjected to global ischaemia, Olig2PC-Astros exhibit superior neuroprotective effects and improved behavioural outcome compared to NPC-Astros. Thus, this new paradigm of human astroglial differentiation is useful for studying the heterogeneity of human astroglia, and the unique Olig2PC-Astros may constitute a new cell therapy for treating cerebral ischaemia and other neurological diseases.


Subject(s)
Astrocytes/cytology , Astrocytes/transplantation , Brain Ischemia/therapy , Embryonic Stem Cells/cytology , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Expression , Genes, Reporter , Green Fluorescent Proteins , Hippocampus/metabolism , Hippocampus/pathology , Humans , Injections, Intraventricular , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Stereotaxic Techniques , Transplantation, Heterologous
13.
EMBO Mol Med ; 5(6): 891-903, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23681668

ABSTRACT

Local production of neurosteroids such as progesterone and allopregnanolone confers neuroprotection in central nervous system (CNS) inflammatory diseases. The mitochondrial translocator protein (TSPO) performs a rate-limiting step in the conversion of cholesterol to pregnenolone and its steroid derivatives. Previous studies have shown that TSPO is upregulated in microglia and astroglia during neural inflammation, and radiolabelled TSPO ligands such as PK11195 have been used to image and localize injury in the CNS. Recent studies have shown that modulating TSPO activity with pharmacological ligands such as etifoxine can initiate the production of neurosteroids locally in the injured CNS. In this study, we examined the effects of etifoxine, a clinically available anxiolytic drug, in the development and progression of mouse experimental autoimmune encephalomyelitis (EAE), an experimental model for multiple sclerosis (MS). Our results showed that etifoxine attenuated EAE severity when administered before the development of clinical signs and also improved symptomatic recovery when administered at the peak of the disease. In both cases, recovery was correlated with diminished inflammatory pathology in the lumbar spinal cord. Modulation of TSPO activity by etifoxine led to less peripheral immune cell infiltration of the spinal cord, and increased oligodendroglial regeneration after inflammatory demyelination in EAE. Our results suggest that a TSPO ligand, e.g. etifoxine, could be a potential new therapeutic option for MS with benefits that could be comparable to the administration of systemic steroids but potentially avoiding the detrimental side effects of long-term direct use of steroids.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Oxazines/therapeutic use , Receptors, GABA/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Ligands , Mice , Mice, Inbred C57BL , Microglia/cytology , Receptors, GABA/chemistry , Spinal Cord/metabolism
14.
Front Biosci (Landmark Ed) ; 17(1): 65-89, 2012 01 01.
Article in English | MEDLINE | ID: mdl-22201733

ABSTRACT

Research on the biology of adult stem cells, embryonic stem cells and induced pluripotent stem cells, as well as cell-based strategies for treating nervous system disorders has begun to create the hope that these cells may be used for therapy in humans after injury or disease. In animal models of neurological diseases, transplantation of stem cells or their derivatives can improve function not only due to direct replacement of lost neurons or glia, but also by providing trophic support. Despite intense research efforts to translate these studies from the bench to bedside, critical problems remain at several steps in this process. Recent technological advancements in both the derivation of stem cells and their directed differentiation to lineage-committed progenitors have brought us closer to therapeutic applications. Several preclinical studies have already explored the behavior of transplanted cells with respect to proliferation, migration, differentiation and survival, especially in complex pathological disease environments. In this review, we examine the current status, progress, pitfalls, and potential of these stem cell technologies, focusing on directed differentiation of human stem cells into various neural lineages, including dopaminergic neurons, motor neurons, oligodendroglia, microglia, and astroglia, and on advancements in cell-based regenerative strategies for neural repair and criteria for successful therapeutic applications.


Subject(s)
Nervous System Diseases/therapy , Neural Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology , Adult , Adult Stem Cells/cytology , Astrocytes/cytology , Cell Differentiation , Dopaminergic Neurons/cytology , Embryonic Stem Cells/cytology , Humans , Microglia/cytology , Motor Neurons/cytology , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Oligodendroglia/cytology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation
15.
Neurobiol Dis ; 44(1): 63-72, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704706

ABSTRACT

Dextromethorphan (DM) is a dextrorotary morphinan and a widely used component of cough medicine. Relatively high doses of DM in combination with quinidine are used for the treatment of mood disorders for patients with multiple sclerosis (MS). However, at lower doses, morphinans exert anti-inflammatory activities through the inhibition of NOX2-dependent superoxide production in activated microglia. Here we investigated the effects of high (10 mg/kg, i.p., "DM-10") and low (0.1 mg/kg, i.p., "DM-0.1") doses of DM on the development and progression of mouse experimental autoimmune encephalomyelitis (EAE), an animal model of MS. We found no protection by high dose DM treatment. Interestingly, a minor late attenuation by low dose DM treatment was seen in severe EAE that was characterized by a chronic disease course and a massive spinal cord infiltration of CD45(+) cells including T-lymphocytes, macrophages and neutrophils. Furthermore, in a less severe form of EAE, where lower levels of CD4(+) and CD8(+) T-cells, Iba1(+) microglia/macrophages and no significant infiltration of neutrophils were seen in the spinal cord, the treatment with DM-0.1 was remarkably more beneficial. The effect was the most significant at the peak of disease and was associated with an inhibition of NOX2 expression and a decrease in infiltration of monocytes and lymphocytes into the spinal cord. In addition, chronic treatment with low dose DM resulted in decreased demyelination and reduced axonal loss in the lumbar spinal cord. Our study is the first report to show that low dose DM is effective in treating EAE of moderate severity. Our findings reveal that low dose morphinan DM treatment may represent a new promising protective strategy for treating MS.


Subject(s)
Dextromethorphan/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/enzymology , Excitatory Amino Acid Antagonists/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Neuroprotective Agents , Spinal Cord/pathology , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/drug effects , Demyelinating Diseases/pathology , Dextromethorphan/administration & dosage , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/pathology , Excitatory Amino Acid Antagonists/administration & dosage , Glycoproteins/biosynthesis , Immunohistochemistry , Lymphocyte Count , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Myelin-Oligodendrocyte Glycoprotein , NADPH Oxidase 2 , Neutrophil Infiltration/drug effects , Peptide Fragments/biosynthesis , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects , Spinal Cord/metabolism , Superoxides/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
16.
J Biol Chem ; 284(38): 26070-84, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19628872

ABSTRACT

Poly(ADP-ribose) polymerase-1 (PARP-1) has been implicated in the pathogenesis of several central nervous system (CNS) disorders. However, the role of PARP-1 in autoimmune CNS injury remains poorly understood. Therefore, we studied experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis in mice with a targeted deletion of PARP-1. We identified inherent physiological abnormalities in the circulating and splenic immune composition between PARP-1(-/-) and wild type (WT) mice. Upon EAE induction, PARP-1(-/-) mice had an earlier onset and developed a more severe EAE compared with WT cohorts. Splenic response was significantly higher in PARP-1(-/-) mice largely because of B cell expansion. Although formation of Th1 and Th17 effector T lymphocytes was unaffected, PARP-1(-/-) mice had significantly earlier CD4+ T lymphocyte and macrophage infiltration into the CNS during EAE. However, we did not detect significant differences in cytokine profiles between PARP-1(-/-) and WT spinal cords at the peak of EAE. Expression analysis of different PARP isozymes in EAE spinal cords showed that PARP-1 was down-regulated in WT mice and that PARP-3 but not PARP-2 was dramatically up-regulated in both PARP-1(-/-) and WT mice, suggesting that these PARP isozymes could have distinct roles in different CNS pathologies. Together, our results indicate that PARP-1 plays an important role in regulating the physiological immune composition and in immune modulation during EAE; our finding identifies a new aspect of immune regulation by PARPs in autoimmune CNS pathology.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Multiple Sclerosis/enzymology , Poly(ADP-ribose) Polymerases , Spinal Cord/enzymology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/immunology , Macrophages/enzymology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Th1 Cells/enzymology , Th1 Cells/immunology , Th1 Cells/pathology , Up-Regulation/genetics , Up-Regulation/immunology
17.
PLoS Med ; 4(4): e124, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17425404

ABSTRACT

BACKGROUND: In multiple sclerosis, inflammation can successfully be prevented, while promoting repair is still a major challenge. Microglial cells, the resident phagocytes of the central nervous system (CNS), are hematopoietic-derived myeloid cells and express the triggering receptor expressed on myeloid cells 2 (TREM2), an innate immune receptor. Myeloid cells are an accessible source for ex vivo gene therapy. We investigated whether myeloid precursor cells genetically modified to express TREM2 affect the disease course of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. METHODS AND FINDINGS: EAE was induced in mice by immunization with a myelin autoantigen. Intravenous application of TREM2-transduced bone marrow-derived myeloid precursor cells at the EAE peak led to an amelioration of clinical symptoms, reduction in axonal damage, and prevention of further demyelination. TREM2-transduced myeloid cells applied intravenously migrated into the inflammatory spinal cord lesions of EAE-diseased mice, showed increased lysosomal and phagocytic activity, cleared degenerated myelin, and created an anti-inflammatory cytokine milieu within the CNS. CONCLUSIONS: Intravenously applied bone marrow-derived and TREM2-tranduced myeloid precursor cells limit tissue destruction and facilitate repair within the murine CNS by clearance of cellular debris during EAE. TREM2 is a new attractive target for promotion of repair and resolution of inflammation in multiple sclerosis and other neuroinflammatory diseases.


Subject(s)
Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Membrane Glycoproteins/physiology , Microglia/physiology , Myeloid Cells/transplantation , Phagocytosis , Receptors, Immunologic/physiology , Animals , Apoptosis , Cell Differentiation/drug effects , Cell Lineage , Cell Movement , Cells, Cultured/physiology , Cells, Cultured/transplantation , Cytokines/genetics , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/surgery , Female , Flow Cytometry , Gene Expression Regulation , Genes, Reporter , Genes, Synthetic , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Lentivirus/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Monocytes/physiology , Multiple Sclerosis , Myeloid Cells/drug effects , Myeloid Cells/physiology , Neurons/pathology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Promoter Regions, Genetic , Receptors, Immunologic/genetics , Transduction, Genetic
18.
Neurobiol Dis ; 23(2): 247-59, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16733089

ABSTRACT

Increased neurogenesis in response to brain injury is considered a mechanism of regeneration after neuronal loss. Using organotypic hippocampal cultures (OHC), we investigated the interplay between neuronal damage (propidium iodide uptake), microglia activation (OX-42 immunohistochemistry), cell proliferation (bromodeoxyuridine incorporation), and neurogenesis (double labeling of bromodeoxyuridine with doublecortin or beta-III tubulin) after oxygen-glucose deprivation (OGD). We observed that microglia activation and upregulation of pro-inflammatory cytokines mRNA preceded neuronal loss and was followed by increased cell proliferation. Neurogenesis was inhibited 3 days after OGD in both neurogenic zones of the slice, the dentate gyrus and the posterior periventricle (pPV). After 6 days, neurogenesis was restored and significantly increased in the pPV. Indomethacin or minocycline reduced the OGD-induced damage, proliferation, and increase of microglia. Both agents did not interfere with OGD-induced pPV neurogenesis. Our study shows for the first time that neuroprotection against OGD-induced damage in OHC by anti-inflammatory treatment is associated with intact neurogenesis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Ischemia/physiopathology , Hippocampus/physiology , Neuroprotective Agents/pharmacology , Animals , Cell Division/drug effects , Cytokines/genetics , DNA Primers , Disease Models, Animal , Doublecortin Protein , Glucose/deficiency , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Hypoxia , Indomethacin/pharmacology , Microglia/drug effects , Microglia/physiology , Organ Culture Techniques , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...