Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
bioRxiv ; 2024 May 08.
Article in English | MEDLINE | ID: mdl-38766123

ABSTRACT

Although axotomized neurons retain the ability to initiate the formation of growth cones and attempt to regenerate after spinal cord injury, the scar area formed as a result of the lesion in most adult mammals contains a variety of reactive cells that elaborate multiple extracellular matrix and enzyme components that are not suitable for regrowth 1,2 . Newly migrating axons in the vicinity of the scar utilize upregulated LAR family receptor protein tyrosine phosphatases, such as PTPσ, to associate with extracellular chondroitin sulphate proteoglycans (CSPGs), which have been discovered to tightly entrap the regrowing axon tip and transform it into a dystrophic non-growing endball. The scar is comprised of two compartments, one in the lesion penumbra, the glial scar, composed of reactive microglia, astrocytes and OPCs; and the other in the lesion epicenter, the fibrotic scar, which is made up of fibroblasts, pericytes, endothelial cells and inflammatory cells. While the fibrotic scar is known to be strongly inhibitory, even more so than the glial scar, the molecular determinants that curtail axon elongation through the injury core are largely uncharacterized. Here, we show that one sole member of the entire family of collagens, collagen I, creates an especially potent inducer of endball formation and regeneration failure. The inhibitory signaling is mediated by mechanosensitive ion channels and RhoA activation. Staggered systemic administration of two blood-brain barrier permeable-FDA approved drugs, aspirin and pirfenidone, reduced fibroblast incursion into the complete lesion and dramatically decreased collagen I, as well as CSPG deposition which were accompanied by axonal growth and considerable functional recovery. The anatomical substrate for robust axonal regeneration was provided by laminin producing GFAP + and NG2 + bridging cells that spanned the wound. Our results reveal a collagen I-mechanotransduction axis that regulates axonal regrowth in spinal cord injury and raise a promising strategy for rapid clinical application.

2.
J Neurotrauma ; 40(23-24): 2500-2521, 2023 12.
Article in English | MEDLINE | ID: mdl-37606910

ABSTRACT

Spinal cord injuries (SCI), for which there are limited effective treatments, result in enduring paralysis and hypoesthesia, in part because of the inhibitory microenvironment that develops and limits regeneration/sprouting, especially during chronic stages. Recently, we discovered that targeted enzymatic removal of the inhibitory chondroitin sulfate proteoglycan (CSPG) component of the extracellular and perineuronal net (PNN) matrix via Chondroitinase ABC (ChABC) rapidly restored robust respiratory function to the previously paralyzed hemi-diaphragm after remarkably long times post-injury (up to 1.5 years) following a cervical level 2 lateral hemi-transection. Importantly, ChABC treatment at cervical level 4 in this chronic model also elicited improvements in gross upper arm function. In the present study, we focused on arm and hand function, seeking to highlight and optimize crude as well as fine motor control of the forearm and digits at lengthy chronic stages post-injury. However, instead of using ChABC, we utilized a novel and more clinically relevant systemic combinatorial treatment strategy designed to simultaneously reduce and overcome inhibitory CSPGs. Following a 3-month upper cervical spinal hemi-lesion using adult female Sprague Dawley rats, we show that the combined treatment had a profound effect on functional recovery of the chronically paralyzed forelimb and paw, as well as on precision movements of the digits. The regenerative and immune system related events that we describe deepen our basic understanding of the crucial role of CSPG-mediated inhibition via the PTPσ receptor in constraining functional synaptic plasticity at lengthy time points following SCI, hopefully leading to clinically relevant translational benefits.


Subject(s)
Chondroitin Sulfate Proteoglycans , Spinal Cord Injuries , Animals , Female , Rats , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfate Proteoglycans/pharmacology , Nerve Regeneration/physiology , Rats, Sprague-Dawley , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Spinal Cord , Forelimb
3.
Cell Rep ; 40(4): 111137, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35905716

ABSTRACT

In addition to neuroprotective strategies, neuroregenerative processes could provide targets for stroke recovery. However, the upregulation of inhibitory chondroitin sulfate proteoglycans (CSPGs) impedes innate regenerative efforts. Here, we examine the regulatory role of PTPσ (a major proteoglycan receptor) in dampening post-stroke recovery. Use of a receptor modulatory peptide (ISP) or Ptprs gene deletion leads to increased neurite outgrowth and enhanced NSCs migration upon inhibitory CSPG substrates. Post-stroke ISP treatment results in increased axonal sprouting as well as neuroblast migration deeply into the lesion scar with a transcriptional signature reflective of repair. Lastly, peptide treatment post-stroke (initiated acutely or more chronically at 7 days) results in improved behavioral recovery in both motor and cognitive functions. Therefore, we propose that CSPGs induced by stroke play a predominant role in the regulation of neural repair and that blocking CSPG signaling pathways will lead to enhanced neurorepair and functional recovery in stroke.


Subject(s)
Neural Stem Cells , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Chondroitin Sulfate Proteoglycans/metabolism , Nerve Regeneration/physiology , Neural Stem Cells/metabolism , Peptides , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism
4.
Cell Rep ; 39(3): 110711, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35443166

ABSTRACT

Neurons must function for decades of life, but how these non-dividing cells are preserved is poorly understood. Using mouse serotonin (5-HT) neurons as a model, we report an adult-stage transcriptional program specialized to ensure the preservation of neuronal connectivity. We uncover a switch in Lmx1b and Pet1 transcription factor function from controlling embryonic axonal growth to sustaining a transcriptomic signature of 5-HT connectivity comprising functionally diverse synaptic and axonal genes. Adult-stage deficiency of Lmx1b and Pet1 causes slowly progressing degeneration of 5-HT synapses and axons, increased susceptibility of 5-HT axons to neurotoxic injury, and abnormal stress responses. Axon degeneration occurs in a die back pattern and is accompanied by accumulation of α-synuclein and amyloid precursor protein in spheroids and mitochondrial fragmentation without cell body loss. Our findings suggest that neuronal connectivity is transcriptionally protected by maintenance of connectivity transcriptomes; progressive decay of such transcriptomes may contribute to age-related diseases of brain circuitry.


Subject(s)
Serotonin , Transcription Factors , Animals , Axons/metabolism , Mice , Neurons/metabolism , Serotonin/metabolism , Synapses/metabolism , Transcription Factors/metabolism
5.
Cell Tissue Res ; 387(3): 319-336, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34076775

ABSTRACT

Severe spinal cord injury causes permanent loss of function and sensation throughout the body. The trauma causes a multifaceted torrent of pathophysiological processes which ultimately act to form a complex structure, permanently remodeling the cellular architecture and extracellular matrix. This structure is traditionally termed the glial/fibrotic scar. Similar cellular formations occur following stroke, infection, and neurodegenerative diseases of the central nervous system (CNS) signifying their fundamental importance to preservation of function. It is increasingly recognized that the scar performs multiple roles affecting recovery following traumatic injury. Innovative research into the properties of this structure is imperative to the development of treatment strategies to recover motor function and sensation following CNS trauma. In this review, we summarize how the regeneration potential of the CNS alters across phyla and age through formation of scar-like structures. We describe how new insights from next-generation sequencing technologies have yielded a more complex portrait of the molecular mechanisms governing the astrocyte, microglial, and neuronal responses to injury and development, especially of the glial component of the scar. Finally, we discuss possible combinatorial therapeutic approaches centering on scar modulation to restore function after severe CNS injury.


Subject(s)
Gliosis , Spinal Cord Injuries , Astrocytes/pathology , Cicatrix/pathology , Gliosis/pathology , Humans , Neuroglia/pathology , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy
6.
J Surg Res ; 268: 354-362, 2021 12.
Article in English | MEDLINE | ID: mdl-34403855

ABSTRACT

BACKGROUND: Histomorphometry quantitatively evaluates nerve regeneration. Total myelinated fiber count (TMFC) is most accurately obtained manually across full nerve cross-sections, but most researchers opt for automated, sampled analysis. Few of the numerous techniques available have been validated. The goal of this study was to compare common histomorphometric methods (full manual [FM], sampled manual [SM], and sampled automatic [SA]) to determine their reliability and consistency. MATERIAL AND METHODS: Twenty-four rats underwent sciatic nerve (SN) repair with 20mm isografts; SNs distal to the graft were analyzed. TMFC was manually determined in each full cross-section. Counts were also extrapolated from sampled fields, both manually and automatically with ImageJ software. Myelinated fiber diameter, axon diameter, and myelin sheath thickness were measured manually in full and sampled fields; G-ratio was calculated. Repeated-measures MANOVA, Spearman correlation, and Wilcoxon signed-rank tests were performed. A systematic review of histomorphometry in rat SN repair was performed to analyze the variability of techniques in the literature. RESULTS: FM TMFC was 13,506 ± 4,217. Both sampled methods yielded significantly different TMFCs (SM:14.4 ± 13.4%, P< 0.001; SA:21.8 ± 44.7%, P = 0.037). All three methods strongly correlated with each other, especially FM and SM (rs = 0.912, P< 0.001). FM fiber diameter, axon diameter, and myelin sheath thickness did not differ from SM (P = 0.493, 0.209, and 0.331, respectively). 65% of papers used sampling; 78% utilized automated or semi-automated analysis. Software, sampling, and histomorphometric parameters varied widely. CONCLUSION: SM and SA analysis are reliable with standardized, systematic sampling. Transparency is essential to allow comparison of data; meanwhile, researchers must be cognizant of the wide variety of methodologies in the literature.


Subject(s)
Axons , Nerve Regeneration , Animals , Axons/physiology , Myelin Sheath/physiology , Rats , Reproducibility of Results , Sciatic Nerve/surgery
7.
Microsurgery ; 41(3): 286-295, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33511636

ABSTRACT

INTRODUCTION: Rat sciatic nerve injury (PNR) is the most utilized model in studies on peripheral nerve regeneration. However, large animal models are increasingly favored based on the assumption that nerve regeneration in rodents achieves more favorable outcomes than in humans. The purpose of this meta-analysis was to investigate which rat PNR models are more stringent and should be used before utilizing large animal experimentation. METHODS: A PRISMA-guided meta-analysis of the English literature regarding functional outcomes in rat peripheral nerve injury models was conducted. Outcomes of five basic scenarios: (1) transected nerve/negative control, (2) transection with primary microsurgical repair, (3) isogenic/autologous grafts, (4) acellular-allogenic grafts, and (5) limb transplantation were compared to sciatic nerves without any intervention/positive control. Outcomes were compared using Sciatic Functional Index (SFI). Log-based projections were generated and evaluated using mean squared error (MSE), one-way-ANOVA, and Tukey-HSD post-hoc analysis. RESULTS: In total, 167 articles met the inclusion criteria. The earliest manifestations of motor recovery were encountered in the transection and primary repair group (p <.0005). There was a significant difference in recovery time and degree of recovery between all surgical models (p <.0005). At 24 weeks, the SFI in hindlimb transplantation group was significantly worse than all other groups (-74.07 ± 2.74, p <.0005). Autografts smaller than 10 mm recovered sooner than autografts longer than 10 mm (p = .021) and autografts recovered faster than allografts. CONCLUSION: This meta-analysis does not support the belief that neuro-regeneration is exceptional in transection models. These models remain adequate to provide translatable information and should initially be used in investigational studies.


Subject(s)
Peripheral Nerve Injuries , Animals , Autografts , Hindlimb , Nerve Regeneration , Rats , Recovery of Function , Sciatic Nerve
8.
Neural Regen Res ; 16(1): 26-35, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32788444

ABSTRACT

Proteases comprise a variety of enzymes defined by their ability to catalytically hydrolyze the peptide bonds of other proteins, resulting in protein lysis. Cathepsins, specifically, encompass a class of at least twenty proteases with potent endopeptidase activity. They are located subcellularly in lysosomes, organelles responsible for the cell's degradative and autophagic processes, and are vital for normal lysosomal function. Although cathepsins are involved in a multitude of cell signaling activities, this chapter will focus on the role of cathepsins (with a special emphasis on Cathepsin B) in neuronal plasticity. We will broadly define what is known about regulation of cathepsins in the central nervous system and compare this with their dysregulation after injury or disease. Importantly, we will delineate what is currently known about the role of cathepsins in axon regeneration and plasticity after spinal cord injury. It is well established that normal cathepsin activity is integral to the function of lysosomes. Without normal lysosomal function, autophagy and other homeostatic cellular processes become dysregulated resulting in axon dystrophy. Furthermore, controlled activation of cathepsins at specialized neuronal structures such as axonal growth cones and dendritic spines have been positively implicated in their plasticity. This chapter will end with a perspective on the consequences of cathepsin dysregulation versus controlled, localized regulation to clarify how cathepsins can contribute to both neuronal plasticity and neurodegeneration.

9.
Exp Neurol ; 328: 113276, 2020 06.
Article in English | MEDLINE | ID: mdl-32145250

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs), extracellular matrix molecules that increase dramatically following a variety of CNS injuries or diseases, have long been known for their potent capacity to curtail cell migrations as well as axon regeneration and sprouting. The inhibition can be conferred through binding to their major cognate receptor, Protein Tyrosine Phosphatase Sigma (PTPσ). However, the precise mechanisms downstream of receptor binding that mediate growth inhibition have remained elusive. Recently, CSPGs/PTPσ interactions were found to regulate autophagic flux at the axon growth cone by dampening the autophagosome-lysosomal fusion step. Because of the intense interest in autophagic phenomena in the regulation of a wide variety of critical cellular functions, we summarize here what is currently known about dysregulation of autophagy following spinal cord injury, and highlight this critical new mechanism underlying axon regeneration failure. Furthermore, we review how CSPGs/PTPσ interactions influence plasticity through autophagic regulation and how PTPσ serves as a switch to execute either axon outgrowth or synaptogenesis. This has exciting implications for the role CSPGs play not only in axon regeneration failure after spinal cord injury, but also in neurodegenerative diseases where, again, inhibitory CSPGs are upregulated.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Spinal Cord Injuries/metabolism , Animals , Autophagy/physiology , Humans , Spinal Cord Injuries/pathology
10.
Elife ; 82019 07 29.
Article in English | MEDLINE | ID: mdl-31355748

ABSTRACT

Formation of long-range axons occurs over multiple stages of morphological maturation. However, the intrinsic transcriptional mechanisms that temporally control different stages of axon projection development are unknown. Here, we addressed this question by studying the formation of mouse serotonin (5-HT) axons, the exemplar of long-range profusely arborized axon architectures. We report that LIM homeodomain factor 1b (Lmx1b)-deficient 5-HT neurons fail to generate axonal projections to the forebrain and spinal cord. Stage-specific targeting demonstrates that Lmx1b is required at successive stages to control 5-HT axon primary outgrowth, selective routing, and terminal arborization. We show a Lmx1b→Pet1 regulatory cascade is temporally required for 5-HT arborization and upregulation of the 5-HT axon arborization gene, Protocadherin-alphac2, during postnatal development of forebrain 5-HT axons. Our findings identify a temporal regulatory mechanism in which a single continuously expressed transcription factor functions at successive stages to orchestrate the progressive development of long-range axon architectures enabling expansive neuromodulation.


Subject(s)
Axons/physiology , LIM-Homeodomain Proteins/metabolism , Serotonergic Neurons/physiology , Transcription Factors/metabolism , Animals , Gene Expression Profiling , LIM-Homeodomain Proteins/deficiency , Mice , Prosencephalon/cytology , Spinal Cord/cytology , Transcription Factors/deficiency
11.
Exp Neurol ; 320: 112972, 2019 10.
Article in English | MEDLINE | ID: mdl-31234058

ABSTRACT

Intravital imaging of the immune system is a powerful technique for studying biology of the immune response in the spinal cord using a variety of disease models ranging from traumatic injury to autoimmune disorders. Here, we will discuss specific technical aspects as well as many intriguing biological phenomena that have been revealed with the use of intravital imaging for investigation of the immune system in the spinal cord. We will discuss surgical techniques for exposing and stabilizing the spine that are critical for obtaining images, visualizing immune and CNS cells with genetically expressed fluorescent proteins, fluorescent labeling techniques and briefly discuss some of the challenges of image analysis.


Subject(s)
Intravital Microscopy/methods , Neuroimaging/methods , Spinal Cord , Animals , Humans
12.
J Med Chem ; 62(10): 4902-4914, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31042384

ABSTRACT

Myelin is the protective sheath that surrounds nerves in vertebrates to protect axons, which thereby facilitates impulse conduction. Damage to myelin is associated with many neurodegenerative diseases such as multiple sclerosis and also includes spinal cord injury (SCI). The small size of the spinal cord poses formidable challenges to in vivo monitoring of myelination, which we investigated via conducting a structure-activity relationship study to determine the optimum positron-emitting agent to use for imaging myelin using positron emission tomography (PET). From these studies, [18F]PENDAS was identified as the lead agent to use in conjunction with PET imaging to delineate the integrity of spinal cord myelin. A subsequent in vivo PET imaging study of [18F]PENDAS in rats with SCI showed promising pharmacokinetic results that justify further development of imaging markers for diagnosing myelin-related diseases. Additionally, [18F]PENDAS could be valuable in determining the efficacy of therapies that are currently under development.


Subject(s)
Brain/diagnostic imaging , Fluorine Radioisotopes , Myelin Sheath/pathology , Positron-Emission Tomography/methods , Small Molecule Libraries/chemical synthesis , Spinal Cord/diagnostic imaging , Animals , Brain/metabolism , Ligands , Mice , Molecular Structure , Myelin Sheath/metabolism , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Spinal Cord/metabolism , Structure-Activity Relationship
13.
Glia ; 67(1): 125-145, 2019 01.
Article in English | MEDLINE | ID: mdl-30394599

ABSTRACT

Following spinal cord injury (SCI), the population of mature oligodendrocytes undergoes substantial cell death; promoting their preservation and replacement is a viable strategy for preserving axonal integrity and white matter repair in the injured spinal cord. Dramatic upregulation of matrix chondroitin sulfate proteoglycans (CSPGs) is shown to pose an obstacle to endogenous repair processes, and targeting CSPGs improves functional recovery after SCI. However, the cellular and molecular mechanisms underlying the inhibitory effects of CSPGs remain largely undefined. Modulation of CSPGs specific signaling receptors, leukocyte common antigen-related (LAR), and protein tyrosine phosphatase-sigma (PTPσ) allows us to uncover the role and mechanisms of CSPGs in regulating oligodendrocytes in SCI. Here, utilizing specific functionally blocking peptides in a clinically relevant model of contusive/compressive SCI in the rat, we demonstrate that inhibition of PTPσ and LAR receptors promotes oligodendrogenesis by endogenous precursor cells, attenuates caspase 3-mediated cell death in mature oligodendrocytes, and preserves myelin. In parallel in vitro systems, we have unraveled that CSPGs directly induce apoptosis in populations of neural precursor cells and oligodendrocyte progenitor cells and limit their ability for oligodendrocyte differentiation, maturation, and myelination. These negative effects of CSPGs are mediated through the activation of both LAR and PTPσ receptors and the downstream Rho/ROCK pathway. Thus, we have identified a novel inhibitory role for PTPσ and LAR in regulating oligodendrocyte differentiation and apoptosis in the injured adult spinal cord and a new feasible therapeutic strategy for optimizing endogenous cell replacement following SCI.


Subject(s)
Oligodendroglia/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Spinal Cord Injuries/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Chondroitin Sulfate Proteoglycans/biosynthesis , Female , Mice , Mice, Inbred C57BL , Oligodendroglia/drug effects , Oligodendroglia/pathology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology
14.
Nat Commun ; 9(1): 4843, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30482901

ABSTRACT

There exists an abundance of barriers that hinder functional recovery following spinal cord injury, especially at chronic stages. Here, we examine the rescue of breathing up to 1.5 years following cervical hemisection in the rat. In spite of complete hemidiaphragm paralysis, a single injection of chondroitinase ABC in the phrenic motor pool restored robust and persistent diaphragm function while improving neuromuscular junction anatomy. This treatment strategy was more effective when applied chronically than when assessed acutely after injury. The addition of intermittent hypoxia conditioning further strengthened the ventilatory response. However, in a sub-population of animals, this combination treatment caused excess serotonergic (5HT) axon sprouting leading to aberrant tonic activity in the diaphragm that could be mitigated via 5HT2 receptor blockade. Through unmasking of the continuing neuroplasticity that develops after injury, our treatment strategy ensured rapid and robust patterned respiratory recovery after a near lifetime of paralysis.


Subject(s)
Respiration , Spinal Cord Injuries/physiopathology , Animals , Chondroitin Sulfates/metabolism , Diaphragm/physiopathology , Extracellular Matrix/metabolism , Female , Neuronal Plasticity , Paralysis/physiopathology , Rats, Sprague-Dawley , Receptors, Serotonin/metabolism , Serotonin/metabolism
15.
Nat Commun ; 9(1): 4126, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297691

ABSTRACT

Multiple Sclerosis (MS) is characterized by focal CNS inflammation leading to the death of oligodendrocytes (OLs) with subsequent demyelination, neuronal degeneration, and severe functional deficits. Inhibitory chondroitin sulfate proteoglycans (CSPGs) are increased in the extracellular matrix in the vicinity of MS lesions and are thought to play a critical role in myelin regeneration failure. We here show that CSPGs curtail remyelination through binding with their cognate receptor, protein tyrosine phosphatase σ (PTPσ) on oligodendrocyte progenitor cells (OPCs). We report that inhibition of CSPG/PTPσ signaling by systemically deliverable Intracellular Sigma Peptide (ISP), promotes OPC migration, maturation, remyelination, and functional recovery in animal models of MS. Furthermore, we report a downstream molecular target of PTPσ modulation in OPCs involving upregulation of the protease MMP-2 that allows OPCs to enzymatically digest their way through CSPGs. In total, we demonstrate a critical role of PTPσ/CSPG interactions in OPC remyelination in MS.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Matrix Metalloproteinase 2/metabolism , Multiple Sclerosis/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cell Movement/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Humans , Mice, Inbred C57BL , Multiple Sclerosis/prevention & control , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Peptides/pharmacology , Protein Binding/drug effects , Stem Cells/drug effects , Stem Cells/metabolism
17.
J Neurosci ; 38(23): 5399-5414, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29760175

ABSTRACT

Severed axon tips reform growth cones following spinal cord injury that fail to regenerate, in part, because they become embedded within an inhibitory extracellular matrix. Chondroitin sulfate proteoglycans (CSPGs) are the major axon inhibitory matrix component that is increased within the lesion scar and in perineuronal nets around deafferented neurons. We have recently developed a novel peptide modulator (intracellular sigma peptide) of the cognate receptor of CSPGs, protein tyrosine phosphatase σ (RPTPσ), which has been shown to markedly improve sensorimotor function, micturition, and coordinated locomotor behavior in spinal cord contused rats. However, the mechanism(s) underlying how modulation of RPTPσ mediates axon outgrowth through inhibitory CSPGs remain unclear. Here, we describe how intracellular sigma peptide modulation of RPTPσ induces enhanced protease Cathepsin B activity. Using DRG neurons from female Sprague Dawley rats cultured on an aggrecan/laminin spot assay and a combination of biochemical techniques, we provide evidence suggesting that modulation of RPTPσ regulates secretion of proteases that, in turn, relieves CSPG inhibition through its digestion to allow axon migration though proteoglycan barriers. Understanding the mechanisms underlying RPTPσ modulation elucidates how axon regeneration is impaired by proteoglycans but can then be facilitated following injury.SIGNIFICANCE STATEMENT Following spinal cord injury, chondroitin sulfate proteoglycans (CSPGs) upregulate and potently inhibit axon regeneration and functional recovery. Protein tyrosine phosphatase σ (RPTPσ) has been identified as a critical cognate receptor of CSPGs. We have previously characterized a synthetic peptide (intracellular sigma peptide) that targets the regulatory intracellular domain of the receptor to allow axons to regenerate despite the presence of CSPGs. Here, we have found that one important mechanism by which peptide modulation of the receptor enhances axon outgrowth is through secretion of a protease, Cathepsin B, which enables digestion of CSPGs. This work links protease secretion to the CSPG receptor RPTPσ for the first time with implications for understanding the molecular mechanisms underlying neural regeneration and plasticity.


Subject(s)
Cathepsin B/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Nerve Regeneration/physiology , Neuronal Outgrowth/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Cells, Cultured , Female , Rats , Rats, Sprague-Dawley
19.
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
20.
Physiol Rev ; 98(2): 881-917, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29513146

ABSTRACT

Since no approved therapies to restore mobility and sensation following spinal cord injury (SCI) currently exist, a better understanding of the cellular and molecular mechanisms following SCI that compromise regeneration or neuroplasticity is needed to develop new strategies to promote axonal regrowth and restore function. Physical trauma to the spinal cord results in vascular disruption that, in turn, causes blood-spinal cord barrier rupture leading to hemorrhage and ischemia, followed by rampant local cell death. As subsequent edema and inflammation occur, neuronal and glial necrosis and apoptosis spread well beyond the initial site of impact, ultimately resolving into a cavity surrounded by glial/fibrotic scarring. The glial scar, which stabilizes the spread of secondary injury, also acts as a chronic, physical, and chemo-entrapping barrier that prevents axonal regeneration. Understanding the formative events in glial scarring helps guide strategies towards the development of potential therapies to enhance axon regeneration and functional recovery at both acute and chronic stages following SCI. This review will also discuss the perineuronal net and how chondroitin sulfate proteoglycans (CSPGs) deposited in both the glial scar and net impede axonal outgrowth at the level of the growth cone. We will end the review with a summary of current CSPG-targeting strategies that help to foster axonal regeneration, neuroplasticity/sprouting, and functional recovery following SCI.


Subject(s)
Axons/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Nerve Regeneration/physiology , Neurons/metabolism , Spinal Cord Injuries/metabolism , Animals , Humans , Neuroglia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...