Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 9(1): 4962, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470740

ABSTRACT

Activation of the Hippo pathway effector Yap underlies many liver cancers, however no germline or somatic mutations have been identified. Autophagy maintains essential metabolic functions of the liver, and autophagy-deficient murine models develop benign adenomas and hepatomegaly, which have been attributed to activation of the p62/Sqstm1-Nrf2 axis. Here, we show that Yap is an autophagy substrate and mediator of tissue remodeling and hepatocarcinogenesis independent of the p62/Sqstm1-Nrf2 axis. Hepatocyte-specific deletion of Atg7 promotes liver size, fibrosis, progenitor cell expansion, and hepatocarcinogenesis, which is rescued by concurrent deletion of Yap. Our results shed new light on mechanisms of Yap degradation and the sequence of events that follow disruption of autophagy, which is impaired in chronic liver disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Hepatocytes/cytology , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Liver/metabolism , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Carcinogenesis , Cell Cycle Proteins , Cell Differentiation , Female , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/pathology , Liver Neoplasms/genetics , Male , Mice , Phosphoproteins/genetics , Proteolysis , Transcription Factors , YAP-Signaling Proteins
3.
J Clin Invest ; 127(8): 3136-3151, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28737509

ABSTRACT

Lesions and neurologic disability in inflammatory CNS diseases such as multiple sclerosis (MS) result from the translocation of leukocytes and humoral factors from the vasculature, first across the endothelial blood-brain barrier (BBB) and then across the astrocytic glia limitans (GL). Factors secreted by reactive astrocytes open the BBB by disrupting endothelial tight junctions (TJs), but the mechanisms that control access across the GL are unknown. Here, we report that in inflammatory lesions, a second barrier composed of reactive astrocyte TJs of claudin 1 (CLDN1), CLDN4, and junctional adhesion molecule A (JAM-A) subunits is induced at the GL. In a human coculture model, CLDN4-deficient astrocytes were unable to control lymphocyte segregation. In models of CNS inflammation and MS, mice with astrocyte-specific Cldn4 deletion displayed exacerbated leukocyte and humoral infiltration, neuropathology, motor disability, and mortality. These findings identify a second inducible barrier to CNS entry at the GL. This barrier may be therapeutically targetable in inflammatory CNS disease.


Subject(s)
Astrocytes/cytology , Central Nervous System/pathology , Inflammation , Nervous System Diseases/pathology , Tight Junctions , Animals , Blood-Brain Barrier/pathology , Cell Adhesion Molecules/metabolism , Claudin-1/metabolism , Claudin-4/metabolism , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Receptors, Cell Surface/metabolism
4.
PLoS One ; 12(1): e0170477, 2017.
Article in English | MEDLINE | ID: mdl-28107514

ABSTRACT

Proper regulation of the coordinated transcriptional program that drives oligodendrocyte (OL) differentiation is essential for central nervous system myelin formation and repair. Nuclear import, mediated in part by a group of karyopherin alpha (Kpna) proteins, regulates transcription factor access to the genome. Understanding how canonical nuclear import functions to control genomic access in OL differentiation may aid in the creation of novel therapeutics to stimulate myelination and remyelination. Here, we show that members of the Kpna family regulate OL differentiation, and may play distinct roles downstream of different pro-myelinating stimuli. Multiple family members are expressed in OLs, and their pharmacologic inactivation dose-dependently decreases the rate of differentiation. Additionally, upon differentiation, the three major Kpna subtypes (P/α2, Q/α3, S/α1) display differential responses to the pro-myelinating cues T3 and CNTF. Most notably, the Q/α3 karyopherin Kpna4 is strongly upregulated by CNTF treatment both compared with T3 treatment and other Kpna responses. Kpna4 inactivation results in inhibition of CNTF-induced OL differentiation, in the absence of changes in proliferation or viability. Collectively, these findings suggest that canonical nuclear import is an integral component of OL differentiation, and that specific Kpnas may serve vital and distinct functions downstream of different pro-myelinating cues.


Subject(s)
Cell Differentiation/physiology , Oligodendroglia/physiology , alpha Karyopherins/physiology , Animals , Cell Line , Gene Expression Profiling , Gene Expression Regulation, Developmental/physiology , Mice , Stem Cells/physiology
5.
PLoS Biol ; 14(5): e1002467, 2016 05.
Article in English | MEDLINE | ID: mdl-27213272

ABSTRACT

Growth factors of the gp130 family promote oligodendrocyte differentiation, and viability, and myelination, but their mechanisms of action are incompletely understood. Here, we show that these effects are coordinated, in part, by the transcriptional activator Krüppel-like factor-6 (Klf6). Klf6 is rapidly induced in oligodendrocyte progenitors (OLP) by gp130 factors, and promotes differentiation. Conversely, in mice with lineage-selective Klf6 inactivation, OLP undergo maturation arrest followed by apoptosis, and CNS myelination fails. Overlapping transcriptional and chromatin occupancy analyses place Klf6 at the nexus of a novel gp130-Klf-importin axis, which promotes differentiation and viability in part via control of nuclear trafficking. Klf6 acts as a gp130-sensitive transactivator of the nuclear import factor importin-α5 (Impα5), and interfering with this mechanism interrupts step-wise differentiation. Underscoring the significance of this axis in vivo, mice with conditional inactivation of gp130 signaling display defective Klf6 and Impα5 expression, OLP maturation arrest and apoptosis, and failure of CNS myelination.


Subject(s)
Central Nervous System/metabolism , Kruppel-Like Transcription Factors/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Differentiation , Cell Survival/genetics , Chromatin/metabolism , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Stem Cells/metabolism , alpha Karyopherins/metabolism
6.
Cell Rep ; 15(4): 748-760, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27149841

ABSTRACT

Oligodendrocytes derive from progenitors (OPCs) through the interplay of epigenomic and transcriptional events. By integrating high-resolution methylomics, RNA-sequencing, and multiple transgenic lines, this study defines the role of DNMT1 in developmental myelination. We detected hypermethylation of genes related to cell cycle and neurogenesis during differentiation of OPCs, yet genetic ablation of Dnmt1 resulted in inefficient OPC expansion and severe hypomyelination associated with ataxia and tremors in mice. This phenotype was not caused by lineage switch or massive apoptosis but was characterized by a profound defect of differentiation associated with changes in exon-skipping and intron-retention splicing events and by the activation of an endoplasmic reticulum stress response. Therefore, loss of Dnmt1 in OPCs is not sufficient to induce a lineage switch but acts as an important determinant of the coordination between RNA splicing and protein synthesis necessary for myelin formation.

7.
Mult Scler Relat Disord ; 6: 10-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27063617

ABSTRACT

Multiple sclerosis (MS) is a recurrent inflammatory disease of the central nervous system, which ultimately causes substantial disability in many patients. A key clinical feature of this disease is the occurrence of relapses, consisting of episodes of neurological dysfunction followed by periods of remission. This review considers in detail the importance of the occurrence of relapses to the ultimate course of MS and the impact of relap setreatment (both acutely and prophylactically) on the long-term outcome for individuals. The ultimate goal of therapy in MS is the reduction of long-term disability. Clinical trials in MS, however, typically only extend for a very short time period compared to the time it takes for disability to evolve. Consequently, short-term outcome measures that are associated with, and predict, future disability need to be identified. In this regard, not only are relapses a characteristic feature of MS, they have also been proven to be associated with the occurrence of long-term disability. Moreover, treatments that reduce the number and severity of these attacks improve the long-term prognosis.


Subject(s)
Multiple Sclerosis/physiopathology , Disability Evaluation , Humans , Multiple Sclerosis/diagnosis , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
8.
PLoS Biol ; 13(11): e1002300, 2015.
Article in English | MEDLINE | ID: mdl-26558429

ABSTRACT

Alterations in the structure and organization of the aging central nervous system (CNS), and associated functional deficits, result in cognitive decline and increase susceptibility to neurodegeneration. Age-related changes to the neurovascular unit (NVU), and their consequences for cerebrovascular function, are implicated as driving cognitive impairment during aging as well as in neurodegenerative disease. The molecular events underlying these effects are incompletely characterized. Similarly, the mechanisms underlying effects of factors that reduce the impact of aging on the brain, such as physical exercise, are also opaque. A study in this issue of PLOS Biology links the NVU to cognitive decline in the aging brain and suggests a potential underlying molecular mechanism. Notably, the study further links the protective effects of chronic exercise on cognition to neurovascular integrity during aging.


Subject(s)
Brain/physiology , Cognitive Aging , Exercise , Models, Cardiovascular , Models, Neurological , Neurons/physiology , Neurovascular Coupling , Animals , Brain/physiopathology , Cognition Disorders/physiopathology , Cognition Disorders/prevention & control , Humans , Motor Activity , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/prevention & control
9.
Neurol Neuroimmunol Neuroinflamm ; 2(4): e134, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26236760

ABSTRACT

Neuromyelitis optica (NMO) (and NMO spectrum disorder) is an autoimmune inflammatory disease of the CNS primarily affecting spinal cord and optic nerves. Reliable and sensitive biomarkers for onset, relapse, and progression in NMO are urgently needed because of the heterogeneous clinical presentation, severity of neurologic disability following relapses, and variability of therapeutic response. Detecting aquaporin-4 (AQP4) antibodies (AQP4-IgG or NMO-IgG) in serum supports the diagnosis of seropositive NMO. However, whether AQP4-IgG levels correlate with disease activity, severity, response to therapy, or long-term outcomes is unclear. Moreover, biomarkers for patients with seronegative NMO have yet to be defined and validated. Collaborative international studies hold great promise for establishing and validating biomarkers that are useful in therapeutic trials and clinical management. In this review, we discuss known and potential biomarkers for NMO.

10.
Brain ; 138(Pt 6): 1548-67, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25805644

ABSTRACT

In inflammatory central nervous system conditions such as multiple sclerosis, breakdown of the blood-brain barrier is a key event in lesion pathogenesis, predisposing to oedema, excitotoxicity, and ingress of plasma proteins and inflammatory cells. Recently, we showed that reactive astrocytes drive blood-brain barrier opening, via production of vascular endothelial growth factor A (VEGFA). Here, we now identify thymidine phosphorylase (TYMP; previously known as endothelial cell growth factor 1, ECGF1) as a second key astrocyte-derived permeability factor, which interacts with VEGFA to induce blood-brain barrier disruption. The two are co-induced NFκB1-dependently in human astrocytes by the cytokine interleukin 1 beta (IL1B), and inactivation of Vegfa in vivo potentiates TYMP induction. In human central nervous system microvascular endothelial cells, VEGFA and the TYMP product 2-deoxy-d-ribose cooperatively repress tight junction proteins, driving permeability. Notably, this response represents part of a wider pattern of endothelial plasticity: 2-deoxy-d-ribose and VEGFA produce transcriptional programs encompassing angiogenic and permeability genes, and together regulate a third unique cohort. Functionally, each promotes proliferation and viability, and they cooperatively drive motility and angiogenesis. Importantly, introduction of either into mouse cortex promotes blood-brain barrier breakdown, and together they induce severe barrier disruption. In the multiple sclerosis model experimental autoimmune encephalitis, TYMP and VEGFA co-localize to reactive astrocytes, and correlate with blood-brain barrier permeability. Critically, blockade of either reduces neurologic deficit, blood-brain barrier disruption and pathology, and inhibiting both in combination enhances tissue preservation. Suggesting importance in human disease, TYMP and VEGFA both localize to reactive astrocytes in multiple sclerosis lesion samples. Collectively, these data identify TYMP as an astrocyte-derived permeability factor, and suggest TYMP and VEGFA together promote blood-brain barrier breakdown.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Thymidine Phosphorylase/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Brain Barrier/physiopathology , Cells, Cultured , Cerebral Cortex/drug effects , Deoxyribose/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelium, Vascular/metabolism , Humans , Interleukin-1beta/pharmacology , Mice , Mice, Transgenic , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Thymidine Phosphorylase/antagonists & inhibitors , Thymidine Phosphorylase/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/pharmacology
11.
Development ; 141(12): 2414-28, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24917498

ABSTRACT

In the embryonic CNS, development of myelin-forming oligodendrocytes is limited by bone morphogenetic proteins, which constitute one arm of the transforming growth factor-ß (Tgfß) family and signal canonically via Smads 1/5/8. Tgfß ligands and Activins comprise the other arm and signal via Smads 2/3, but their roles in oligodendrocyte development are incompletely characterized. Here, we report that Tgfß ligands and activin B (ActB) act in concert in the mammalian spinal cord to promote oligodendrocyte generation and myelination. In mouse neural tube, newly specified oligodendrocyte progenitors (OLPs) are first exposed to Tgfß ligands in isolation, then later in combination with ActB during maturation. In primary OLP cultures, Tgfß1 and ActB differentially activate canonical Smad3 and non-canonical MAP kinase signaling. Both ligands enhance viability, and Tgfß1 promotes proliferation while ActB supports maturation. Importantly, co-treatment strongly activates both signaling pathways, producing an additive effect on viability and enhancing both proliferation and differentiation such that mature oligodendrocyte numbers are substantially increased. Co-treatment promotes myelination in OLP-neuron co-cultures, and maturing oligodendrocytes in spinal cord white matter display strong Smad3 and MAP kinase activation. In spinal cords of ActB-deficient Inhbb(-/-) embryos, apoptosis in the oligodendrocyte lineage is increased and OLP numbers transiently reduced, but numbers, maturation and myelination recover during the first postnatal week. Smad3(-/-) mice display a more severe phenotype, including diminished viability and proliferation, persistently reduced mature and immature cell numbers, and delayed myelination. Collectively, these findings suggest that, in mammalian spinal cord, Tgfß ligands and ActB together support oligodendrocyte development and myelin formation.


Subject(s)
Activins/metabolism , Central Nervous System/embryology , Gene Expression Regulation, Developmental , Oligodendroglia/cytology , Transforming Growth Factor beta1/metabolism , Animals , Cell Adhesion , Cell Proliferation , Cell Survival , Cells, Cultured , Coculture Techniques , Gene Expression Profiling , Humans , Ligands , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Signal Transduction , Smad3 Protein/genetics , Spinal Cord/embryology
12.
Glia ; 62(4): 580-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24470341

ABSTRACT

The oligodendrocyte (OL), the myelinating cell of the central nervous system, undergoes dramatic changes in the organization of its cytoskeleton as it differentiates from a precursor (oligodendrocyte precursor cells) to a myelin-forming cell. These changes include an increase in its branching cell processes, a phenomenon necessary for OL to myelinate multiple axon segments. We have previously shown that levels and activity of non-muscle myosin II (NMII), a regulator of cytoskeletal contractility, decrease as a function of differentiation and that inhibition of NMII increases branching and myelination of OL in coculture with neurons. We have also found that mixed glial cell cultures derived from NMIIB knockout mice display an increase in mature myelin basic protein-expressing OL compared with wild-type cultures. We have now extended our studies to investigate the role of NMIIB ablation on myelin repair following focal demyelination by lysolecithin. To this end, we generated an oligodendrocyte-specific inducible knockout model using a Plp-driven promoter in combination with a temporally activated CRE-ER fusion protein. Our data indicate that conditional ablation of NMII in adult mouse brain, expedites lesion resolution and remyelination by Plp+ oligodendrocyte-lineage cells when compared with that observed in control brains. Taken together, these data validate the function of NMII as that of a negative regulator of OL myelination in vivo and provide a novel target for promoting myelin repair in conditions such as multiple sclerosis.


Subject(s)
Demyelinating Autoimmune Diseases, CNS/physiopathology , Nerve Regeneration/physiology , Nonmuscle Myosin Type IIB/deficiency , Animals , Antigens/metabolism , Autophagy-Related Proteins , Basic Helix-Loop-Helix Transcription Factors/metabolism , Corpus Callosum/pathology , Demyelinating Autoimmune Diseases, CNS/genetics , Demyelinating Autoimmune Diseases, CNS/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Lysophosphatidylcholines , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/pathology , Nerve Tissue Proteins/metabolism , Nonmuscle Myosin Type IIB/genetics , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/pathology , Proteoglycans/metabolism
13.
Acta Neuropathol ; 124(3): 411-24, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22766690

ABSTRACT

Laquinimod (LAQ) is a new oral immunomodulatory compound that reduces relapse rate, brain atrophy and disability progression in multiple sclerosis (MS). LAQ has well-documented effects on inflammation in the periphery, but little is known about its direct activity within the central nervous system (CNS). To elucidate the impact of LAQ on CNS-intrinsic inflammation, we investigated the effects of LAQ on cuprizone-induced demyelination in mice in vivo and on primary CNS cells in vitro. Demyelination, inflammation, axonal damage and glial pathology were evaluated in LAQ-treated wild type and Rag-1-deficient mice after cuprizone challenge. Using primary cells we tested for effects of LAQ on oligodendroglial survival as well as on cytokine secretion and NF-κB activation in astrocytes and microglia. LAQ prevented cuprizone-induced demyelination, microglial activation, axonal transections, reactive gliosis and oligodendroglial apoptoses in wild type and Rag-1-deficient mice. LAQ significantly decreased pro-inflammatory factors in stimulated astrocytes, but not in microglia. Oligodendroglial survival was not affected by LAQ in vitro. Astrocytic, but not microglial, NF-κB activation was markedly reduced by LAQ as evidenced by NF-κB reporter assay. LAQ also significantly decreased astrocytic NF-κB activation in cuprizone-treated mice. Our data indicate that LAQ prevents cuprizone-induced demyelination by attenuating astrocytic NF-κB activation. These effects are CNS-intrinsic and not mediated by peripheral immune cells. Therefore, LAQ downregulation of the astrocytic pro-inflammatory response may be an important mechanism underlying its protective effects on myelin, oligodendrocytes and axons. Modulation of astrocyte activation may be an attractive therapeutic target to prevent tissue damage in MS.


Subject(s)
Astrocytes/drug effects , Demyelinating Diseases/prevention & control , NF-kappa B/metabolism , Oligodendroglia/drug effects , Quinolones/pharmacology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/drug effects , Axons/metabolism , Axons/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Male , Mice , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Myelin Sheath/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology
14.
J Clin Invest ; 122(7): 2454-68, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22653056

ABSTRACT

In inflammatory CNS conditions such as multiple sclerosis (MS), current options to treat clinical relapse are limited, and more selective agents are needed. Disruption of the blood-brain barrier (BBB) is an early feature of lesion formation that correlates with clinical exacerbation, leading to edema, excitotoxicity, and entry of serum proteins and inflammatory cells. Here, we identify astrocytic expression of VEGF-A as a key driver of BBB permeability in mice. Inactivation of astrocytic Vegfa expression reduced BBB breakdown, decreased lymphocyte infiltration and neuropathology in inflammatory and demyelinating lesions, and reduced paralysis in a mouse model of MS. Knockdown studies in CNS endothelium indicated activation of the downstream effector eNOS as the principal mechanism underlying the effects of VEGF-A on the BBB. Systemic administration of the selective eNOS inhibitor cavtratin in mice abrogated VEGF-A-induced BBB disruption and pathology and protected against neurologic deficit in the MS model system. Collectively, these data identify blockade of VEGF-A signaling as a protective strategy to treat inflammatory CNS disease.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Multiple Sclerosis/metabolism , Vascular Endothelial Growth Factor A/physiology , Animals , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Cells, Cultured , Cytokines/metabolism , DNA-Binding Proteins , Demyelinating Diseases , Gene Expression Regulation , Humans , Inflammation/metabolism , Interleukin-1beta/physiology , Lymphocytes/pathology , Lysosomal Membrane Proteins , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/pathology , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Nuclear Proteins/metabolism , Occludin , Permeability , Primary Cell Culture , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Methods Mol Biol ; 814: 401-14, 2012.
Article in English | MEDLINE | ID: mdl-22144322

ABSTRACT

Multiple sclerosis (MS) is characterized by CNS demyelination and oligodendrocyte depletion, axonal loss, and reactive astrogliosis. Myelin loss causes conduction block, while remyelination is associated with recovery of conduction and return of function. Reactive astrocytes are a prominent feature of MS plaques, and have been implicated as producing factors regulating oligodendrocyte progenitor differentiation and myelin formation. Understanding their impact on these events may lead to new approaches for oligodendrocyte protection and/or remyelination in MS. Here, we outline protocols for the establishment and analysis of primary monocultures and cocultures of human astrocytes and oligodendrocytes. These approaches are designed to facilitate analysis of mechanisms underlying astrocytic regulation of progenitor survival and myelin repair.


Subject(s)
Astrocytes/physiology , Cell Communication/physiology , Cell Culture Techniques/methods , Multiple Sclerosis/physiopathology , Myelin Sheath/physiology , Oligodendroglia/physiology , Astrocytes/cytology , Blotting, Western , Humans , Immunohistochemistry , Myelin Sheath/pathology , Oligodendroglia/cytology , Polymerase Chain Reaction , Time-Lapse Imaging
16.
J Neuropathol Exp Neurol ; 70(12): 1138-50, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22082665

ABSTRACT

Central nervous system hypomyelination is a feature common to a number of transgenic (Tg) mouse lines that express a variety of unrelated exogenous (i.e. non-central nervous system) transgenes. In this report, we document hypomyelination structurally by immunocytochemistry and functionally in the Tg line MBP-JE, which over expresses the chemokine CCL2 (MCP-1) within oligodendrocytes targeted by a myelin basic protein (MBP) promoter. Analysis of hypomyelinated optic nerves of Tg mice revealed progressive decrease in oligodendrocyte numbers with age (p < 0.01). Although molecular mechanisms underlying hypomyelination in this and other Tg models remain largely unknown, we present preliminary findings on oligodendrocyte progenitor cell (OPC) cultures in which, although OPC expressed CCR2, the receptor for CCL2, treatment with CCL2 had no significant effect on OPC proliferation, differentiation, or apoptosis. We suggest that hypomyelination in the MBP-JE model might not be due to CCL2 expression but rather the result of transcriptional dysfunction related to random insertion of the MBP promoter that disrupts myelinogenesis and leads to oligodendrocyte demise. Because an MBP promoter is a common denominator in most Tg lines displaying hypomyelination, we hypothesize that use of myelin gene sequences in the regulator region of Tg constructs might underlie this perturbation of myelination in such models.


Subject(s)
Demyelinating Diseases/genetics , Myelin Basic Protein/genetics , Myelin Sheath/genetics , Oligodendroglia/metabolism , Promoter Regions, Genetic , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Neural Stem Cells
17.
FEBS Lett ; 585(23): 3813-20, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21864535

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS. Conduction block in demyelinated axons underlies early neurological symptoms, but axonal transection and neuronal loss are believed to be responsible for more permanent chronic deficits. Several therapies are approved for treatment of relapsing-remitting MS, all of which are immunoregulatory and clinically proven to reduce the rate of lesion formation and exacerbation. However, existing approaches are only partially effective in preventing the onset of disability in MS patients, and novel treatments to protect myelin-producing oligodendrocytes and enhance myelin repair may improve long-term outcomes. Studies in vivo in genetically modified mice have assisted in the characterization of mechanisms underlying the generation of neuropathology in MS patients, and have identified potential avenues for oligodendrocyte protection and myelin repair. However, no treatments are yet approved that target these areas directly, and in addition, the relationship between demyelination and axonal transection in the lesions of the disease remains unclear. Here, we review translational research targeting oligodendrocyte protection and myelin repair in models of autoimmune demyelination, and their potential relevance as therapies in MS.


Subject(s)
Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/pathology , Wound Healing , Animals , Humans , Models, Immunological , Oligodendroglia/pathology , Signal Transduction
18.
J Immunol ; 187(3): 1129-41, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21709156

ABSTRACT

Current therapies for multiple sclerosis target inflammation but do not directly address oligodendrocyte protection or myelin repair. The gp130 family cytokines ciliary neurotrophic factor, leukemia inhibitory factor, and IL-11 have been identified as oligodendrocyte growth factors, and IL-11 is also strongly immunoregulatory, but their underlying mechanisms of action are incompletely characterized. In this study, we demonstrate that these effects of IL-11 are mediated via differential regulation of apoptosis in oligodendrocytes versus Ag-presenting dendritic cells (DCs), and are dependent on lineage-specific activity of the transcription factors Stat1 versus Stat3. Focal demyelinating lesions induced in cerebral cortices of IL-11Rα(-/-) mice using stereotactic microinjection of lysolecithin were larger than in controls, and remyelination was delayed. In IL-11Rα(-/-) mice, lesions displayed extensive oligodendrocyte loss and axonal transection, and increased infiltration by inflammatory cells including CD11c(+) DCs, CD3(+) lymphocytes, and CD11b(+) phagocytes. In oligodendrocyte progenitor cell (OPC) cultures, IL-11 restricted caspase 9 activation and apoptosis, and it increased myelination in OPC-neuron cocultures. Importantly, siRNA inhibition of Stat1 enhanced the antiapoptotic effects of IL-11 on OPCs, but IL-11 induced apoptosis in the presence of Stat3 silencing. In contrast, IL-11 augmented caspase activation and apoptosis in cultures of CD11c(+) DCs, but not in CD11b(+) or CD3(+) cells. Inhibition of Stat3 exacerbated the proapoptotic effects of IL-11 on DCs, whereas they were ablated in Stat1(-/-) cultures. Collectively, these findings reveal novel mechanisms underlying the actions of a neuroprotective and immunoregulatory member of the gp130 cytokine family, suggesting avenues to enhance oligodendrocyte viability and restrict CNS inflammation in multiple sclerosis.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Interleukin-11/therapeutic use , Neuroprotective Agents/therapeutic use , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Demyelinating Diseases/therapy , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Gene Targeting/methods , Interleukin-11/deficiency , Interleukin-11/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Oligodendroglia/immunology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Stem Cells/immunology , Stem Cells/metabolism , Stem Cells/pathology
19.
Mt Sinai J Med ; 78(2): 244-57, 2011.
Article in English | MEDLINE | ID: mdl-21425268

ABSTRACT

Multiple sclerosis is an inflammatory demyelinating disease of the brain and spinal cord with a presumed autoimmune etiology. Conduction block in demyelinated axons underlies early neurological symptoms, whereas axonal transection is believed responsible for more permanent later deficits. Approved treatments for the disease are immunoregulatory and reduce the rate of lesion formation and clinical exacerbation, but are only partially effective in preventing the onset of disability in multiple sclerosis patients. Approaches that directly protect myelin-producing oligodendrocytes and enhance remyelination may improve long-term outcomes and reduce the rate of axonal transection. Studies in genetically modified animals have improved our understanding of mechanisms underlying central nervous system pathology in multiple sclerosis models, and have identified pathways that regulate oligodendrocyte viability and myelin repair. However, although clinical trials are ongoing, many have been unsuccessful, and no treatments are yet approved that target these areas in multiple sclerosis. In this review, we examine avenues for oligodendrocyte protection and endogenous myelin repair in animal models of demyelination and remyelination, and their relevance as therapeutics in human patients.


Subject(s)
Gene Regulatory Networks/immunology , Immunologic Factors , Multiple Sclerosis , Myelin Sheath , Oligodendroglia , Spinal Cord Regeneration/drug effects , Animals , Autoimmunity/drug effects , Axons/drug effects , Axons/metabolism , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental , Humans , Immunologic Factors/metabolism , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Molecular Targeted Therapy , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuroprotective Agents/therapeutic use , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Spinal Cord Regeneration/immunology , Therapies, Investigational
20.
Glia ; 58(8): 964-74, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20169621

ABSTRACT

Notch1 receptor signaling regulates oligodendrocyte progenitor differentiation and myelin formation in development, and during remyelination in the adult CNS. In active multiple sclerosis lesions, Notch1 localizes to oligodendrocyte lineage cells, and its ligand Jagged1 is expressed by reactive astrocytes. Here, we examined induction of Jagged1 in human astrocytes, and its impact on oligodendrocyte differentiation. In human astrocyte cultures, the cytokine TGFbeta1 induced Jagged1 expression and blockade of the TGFbeta1 receptor kinase ALK5 abrogated Jagged1 induction. TGFbeta2 and beta3 had similar effects, but induction was not observed in response to the TGFbeta family member activin A or other cytokines. Downstream, TGFbeta1 activated Smad-dependent signaling, and Smad-independent pathways that included PI3 kinase, p38, and JNK MAP kinase, but only inhibition of the Smad-dependent pathway blocked Jagged1 expression. SiRNA inhibition of Smad3 downregulated induction of Jagged1, and this was potentiated by Smad2 siRNA. Purified oligodendrocyte progenitor cells (OPCs) nucleofected with Notch1 intracellular signaling domain displayed a shift towards proliferation at the expense of differentiation, demonstrating functional relevance of Notch1 signaling in OPCs. Furthermore, human OPCs plated onto Jagged1-expressing astrocytes exhibited restricted differentiation. Collectively, these data illustrate the mechanisms underlying Jagged1 induction in human astrocytes, and suggest that TGFbeta1-induced activation of Jagged1-Notch1 signaling may impact the size and differentiation of the OPC pool in the human CNS.


Subject(s)
Astrocytes/drug effects , Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation , Extracellular Matrix Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Oligodendroglia/physiology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Smad3 Protein/metabolism , Stem Cells/physiology , Transforming Growth Factor beta/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/metabolism , Cerebral Cortex/cytology , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/metabolism , Humans , Jagged-1 Protein , RNA, Small Interfering/pharmacology , Rats , Receptor, Transforming Growth Factor-beta Type I , Serrate-Jagged Proteins , Transfection/methods , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...