Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
J Neuroinflammation ; 15(1): 90, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558941

ABSTRACT

BACKGROUND: Traumatic spinal cord injury (SCI) results in upregulation of chondroitin sulfate proteoglycans (CSPGs) by reactive glia that impedes repair and regeneration in the spinal cord. Degradation of CSPGs is known to be beneficial in promoting endogenous repair mechanisms including axonal sprouting/regeneration, oligodendrocyte replacement, and remyelination, and is associated with improvements in functional outcomes after SCI. Recent evidence suggests that CSPGs may regulate secondary injury mechanisms by modulating neuroinflammation after SCI. To date, the role of CSPGs in SCI neuroinflammation remains largely unexplored. The recent discovery of CSPG-specific receptors, leukocyte common antigen-related (LAR) and protein tyrosine phosphatase-sigma (PTPσ), allows unraveling the cellular and molecular mechanisms of CSPGs in SCI. In the present study, we have employed parallel in vivo and in vitro approaches to dissect the role of CSPGs and their receptors LAR and PTPσ in modulating the inflammatory processes in the acute and subacute phases of SCI. METHODS: In a clinically relevant model of compressive SCI in female Sprague Dawley rats, we targeted LAR and PTPσ by two intracellular functionally blocking peptides, termed ILP and ISP, respectively. We delivered ILP and ISP treatment intrathecally to the injured spinal cord in a sustainable manner by osmotic mini-pumps for various time-points post-SCI. We employed flow cytometry, Western blotting, and immunohistochemistry in rat SCI, as well as complementary in vitro studies in primary microglia cultures to address our questions. RESULTS: We provide novel evidence that signifies a key immunomodulatory role for LAR and PTPσ receptors in SCI. We show that blocking LAR and PTPσ reduces the population of classically activated M1 microglia/macrophages, while promoting alternatively activated M2 microglia/macrophages and T regulatory cells. This shift was associated with a remarkable elevation in pro-regenerative immune mediators, interleukin-10 (IL-10), and Arginase-1. Our parallel in vitro studies in microglia identified that while CSPGs do not induce an M1 phenotype per se, they promote a pro-inflammatory phenotype. Interestingly, inhibiting LAR and PTPσ in M1 and M2 microglia positively modulates their inflammatory response in the presence of CSPGs, and harnesses their ability for phagocytosis and mobilization. Interestingly, our findings indicate that CSPGs regulate microglia, at least in part, through the activation of the Rho/ROCK pathway downstream of LAR and PTPσ. CONCLUSIONS: We have unveiled a novel role for LAR and PTPσ in regulating neuroinflammation in traumatic SCI. Our findings provide new insights into the mechanisms by which manipulation of CSPG signaling can promote recovery from SCI. More importantly, this work introduces the potential of ILP/ISP as a viable strategy for modulating the immune response following SCI and other neuroinflammatory conditions of the central nervous system.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Inflammation/etiology , Inflammation/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Spinal Cord Injuries/complications , Animals , Animals, Newborn , Cell Movement/drug effects , Cell Polarity/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Cytokines/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/physiology , Microglia/drug effects , Microglia/metabolism , Neural Stem Cells/drug effects , Peroxidase/metabolism , Phagocytosis/drug effects , Rats , Rats, Sprague-Dawley
2.
Glia ; 66(3): 538-561, 2018 03.
Article in English | MEDLINE | ID: mdl-29148104

ABSTRACT

Oligodendroglial cell death and demyelination are hallmarks of neurotrauma and multiple sclerosis that cause axonal damage and functional impairments. Remyelination remains a challenge as the ability of endogenous precursor cells for oligodendrocyte replacement is hindered in the unfavorable milieu of demyelinating conditions. Here, in a rat model of lysolecithin lysophosphatidyl-choline (LPC)-induced focal demyelination, we report that Neuregulin-1 (Nrg-1), an important factor for oligodendrocytes and myelination, is dysregulated in demyelinating lesions and its bio-availability can promote oligodendrogenesis and remyelination. We delivered recombinant human Nrg-1ß1 (rhNrg-1ß1) intraspinally in the vicinity of LPC demyelinating lesion in a sustained manner using poly lactic-co-glycolic acid microcarriers. Availability of Nrg-1 promoted generation and maturation of new oligodendrocytes, and accelerated endogenous remyelination by both oligodendrocyte and Schwann cell populations in demyelinating foci. Importantly, Nrg-1 enhanced myelin thickness in newly remyelinated spinal cord axons. Our complementary in vitro studies also provided direct evidence that Nrg-1 significantly promotes maturation of new oligodendrocytes and facilitates their transition to a myelinating phenotype. Nrg-1 therapy remarkably attenuated the upregulated expression chondroitin sulfate proteoglycans (CSPGs) specific glycosaminoglycans in the extracellular matrix of demyelinating foci and promoted interleukin-10 (IL-10) production by immune cells. CSPGs and IL-10 are known to negatively and positively regulate remyelination, respectively. We found that Nrg-1 effects are mediated through ErbB2 and ErbB4 receptor activation. Our work provides novel evidence that dysregulated levels of Nrg-1 in demyelinating lesions of the spinal cord pose a challenge to endogenous remyelination, and appear to be an underlying cause of myelin thinning in newly remyelinated axons.


Subject(s)
Demyelinating Diseases/therapy , Immunomodulation , Neuregulin-1/administration & dosage , Neuroprotective Agents/administration & dosage , Remyelination/physiology , Spinal Cord/immunology , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Drug Carriers , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Humans , Lactic Acid , Male , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Oligodendroglia/immunology , Oligodendroglia/pathology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Spinal Cord/pathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/pathology , Spinal Cord Diseases/therapy
3.
Glia ; 65(7): 1152-1175, 2017 07.
Article in English | MEDLINE | ID: mdl-28456012

ABSTRACT

Spinal cord injury (SCI) results in glial activation and neuroinflammation, which play pivotal roles in the secondary injury mechanisms with both pro- and antiregeneration effects. Presently, little is known about the endogenous molecular mechanisms that regulate glial functions in the injured spinal cord. We previously reported that the expression of neuregulin-1 (Nrg-1) is acutely and chronically declined following traumatic SCI. Here, we investigated the potential ramifications of Nrg-1 dysregulation on glial and immune cell reactivity following SCI. Using complementary in vitro approaches and a clinically-relevant model of severe compressive SCI in rats, we demonstrate that immediate delivery of Nrg-1 (500 ng/day) after injury enhances a neuroprotective phenotype in inflammatory cells associated with increased interleukin-10 and arginase-1 expression. We also found a decrease in proinflammatory factors including IL-1ß, TNF-α, matrix metalloproteinases (MMP-2 and 9) and nitric oxide after injury. In addition, Nrg-1 modulates astrogliosis and scar formation by reducing inhibitory chondroitin sulfate proteoglycans after SCI. Mechanistically, Nrg-1 effects on activated glia are mediated through ErbB2 tyrosine phosphorylation in an ErbB2/3 heterodimer complex. Furthermore, Nrg-1 exerts its effects through downregulation of MyD88, a downstream adaptor of Toll-like receptors, and increased phosphorylation of Erk1/2 and STAT3. Nrg-1 treatment with the therapeutic dosage of 1.5 µg/day significantly improves tissue preservation and functional recovery following SCI. Our findings for the first time provide novel insights into the role and mechanisms of Nrg-1 in acute SCI and suggest a positive immunomodulatory role for Nrg-1 that can harness the beneficial properties of activated glia and inflammatory cells in recovery following SCI.


Subject(s)
Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Neuregulin-1/therapeutic use , Neuroglia/physiology , Recovery of Function/physiology , Spinal Cord Injuries/complications , Animals , Animals, Newborn , Arginase/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Interleukin-10/metabolism , Lipopolysaccharides/toxicity , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Neuroglia/drug effects , Rats , Recovery of Function/drug effects , Signal Transduction/drug effects , Spinal Cord Injuries/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL