Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(6): e0305286, 2024.
Article in English | MEDLINE | ID: mdl-38905198

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0269972.].

2.
PLoS One ; 17(6): e0269972, 2022.
Article in English | MEDLINE | ID: mdl-35763520

ABSTRACT

Proteoglycan glycosaminoglycan (GAG) chains are attached to a serine residue in the protein through a linkage series of sugars, the first of which is xylose. Xylosides are chemicals which compete with the xylose at the enzyme xylosyl transferase to prevent the attachment of GAG chains to proteins. These compounds have been employed at concentrations in the millimolar range as tools to study the role of GAG chains in proteoglycan function. In the course of our studies with xylosides, we conducted a dose-response curve for xyloside actions on neural cells. To our surprise, we found that concentrations of xylosides in the nanomolar to micromolar range had major effects on cell morphology of hippocampal neurons as well as of Neuro2a cells, affecting both actin and tubulin cytoskeletal dynamics. Such effects/morphological changes were not observed with higher xyloside concentrations. We found a dose-dependent alteration of GAG secretion by Neuro2a cells; however, concentrations of xylosides which were effective in altering neuronal morphology did not cause a large change in the rate of GAG chain secretion. In contrast, both low and high concentrations of xylosides altered HS and CS composition. RNAseq of treated cells demonstrated alterations in gene expression only after treatment with millimolar concentration of xylosides that had no effect on cell morphology. These observations support a novel action of xylosides on neuronal cells.


Subject(s)
Glycosides , Xylose , Glycosaminoglycans/metabolism , Glycosides/chemistry , Proteoglycans/metabolism , Xylose/pharmacology
3.
J Neurochem ; 157(3): 494-507, 2021 05.
Article in English | MEDLINE | ID: mdl-33320336

ABSTRACT

Phospholipid Phosphatase-Related Protein Type 1 (PLPPR1) is a member of a family of lipid phosphatase related proteins, integral membrane proteins characterized by six transmembrane domains. This family of proteins is enriched in the brain and recent data indicate potential pleiotropic functions in several different contexts. An inherent ability of this family of proteins is to induce morphological changes, and we have previously reported that members of this family interact with each other and may function co-operatively. However, the function of PLPPR1 is not yet understood. Here we show that the expression of PLPPR1 reduces the inhibition of neurite outgrowth of cultured mouse hippocampal neurons by chondroitin sulfate proteoglycans and the retraction of neurites of Neuro-2a cells by lysophosphatidic acid (LPA). Further, we show that PLPPR1 reduces the activation of Ras homolog family member A (RhoA) by LPA in Neuro-2a cells, and that this is because of an association of PLPPR1with the Rho-specific guanine nucleotide dissociation inhibitor (RhoGDI1). These results establish a novel signaling pathway for the PLPPR1 protein.


Subject(s)
Axons/physiology , Membrane Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , Animals , Cell Line , Cells, Cultured , Chondroitin Sulfate Proteoglycans/pharmacology , Hippocampus/cytology , Immunohistochemistry , Lysophospholipids/pharmacology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Neurites , Proteomics , Transfection , ras Proteins/physiology , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics
4.
J Histochem Cytochem ; 69(1): 61-80, 2021 01.
Article in English | MEDLINE | ID: mdl-32936033

ABSTRACT

The orderly development of the nervous system is characterized by phases of cell proliferation and differentiation, neural migration, axonal outgrowth and synapse formation, and stabilization. Each of these processes is a result of the modulation of genetic programs by extracellular cues. In particular, chondroitin sulfate proteoglycans (CSPGs) have been found to be involved in almost every aspect of this well-orchestrated yet delicate process. The evidence of their involvement is complex, often contradictory, and lacking in mechanistic clarity; however, it remains obvious that CSPGs are key cogs in building a functional brain. This review focuses on current knowledge of the role of CSPGs in each of the major stages of neural development with emphasis on areas requiring further investigation.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Nervous System/growth & development , Neurogenesis , Animals , Brain/cytology , Brain/embryology , Brain/growth & development , Brain/metabolism , Cell Movement , Humans , Nervous System/cytology , Nervous System/embryology , Nervous System/metabolism , Neurons/cytology , Neurons/metabolism , Synapses/metabolism
5.
Glycobiology ; 31(2): 116-125, 2021 02 09.
Article in English | MEDLINE | ID: mdl-32614944

ABSTRACT

Complex glycans play vital roles in many biological processes, ranging from intracellular signaling and organ development to tumor growth. Glycan expression is routinely assessed by the application of glycan-specific antibodies to cells and tissues. However, glycan-specific antibodies quite often show a large number of bands on immunoblots and it is hard to interpret the data when reliable controls are lacking. This limits the scope of glycobiology studies and poses challenges for replication. We sought to resolve this issue by developing a novel strategy that utilizes an immunoreaction enhancing technology to vastly improve the speed and quality of glycan-based immunoblots. As a representative case study, we used chondroitin sulfate glycosaminoglycan (CS-GAG) chains as the carbohydrate target and a monoclonal antibody, CS-56, as the probe. We discovered that preincubation of the antibody with its antigenic CS-GAG chain distinguishes true-positive signals from false-positive ones. We successfully applied this strategy to 10E4, a monoclonal anti heparan sulfate GAGs (HS-GAGs) antibody, where true-positive signals were confirmed by chemical HS-GAG depolymerization on the membrane. This evidence that glycan-specific antibodies can generate clear and convincing data on immunoblot with highly replicable results opens new opportunities for many facets of life science research in glycobiology.


Subject(s)
Chondroitin Sulfates/analysis , Immunoblotting , Animals , Antibodies, Monoclonal/immunology , Chondroitin Sulfates/immunology , HeLa Cells , Humans , Mice , Mice, Inbred C57BL
6.
Nat Commun ; 11(1): 5614, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154382

ABSTRACT

Adult mammalian central nervous system axons have intrinsically poor regenerative capacity, so axonal injury has permanent consequences. One approach to enhancing regeneration is to increase the axonal supply of growth molecules and organelles. We achieved this by expressing the adaptor molecule Protrudin which is normally found at low levels in non-regenerative neurons. Elevated Protrudin expression enabled robust central nervous system regeneration both in vitro in primary cortical neurons and in vivo in the injured adult optic nerve. Protrudin overexpression facilitated the accumulation of endoplasmic reticulum, integrins and Rab11 endosomes in the distal axon, whilst removing Protrudin's endoplasmic reticulum localization, kinesin-binding or phosphoinositide-binding properties abrogated the regenerative effects. These results demonstrate that Protrudin promotes regeneration by functioning as a scaffold to link axonal organelles, motors and membranes, establishing important roles for these cellular components in mediating regeneration in the adult central nervous system.


Subject(s)
Axons/physiology , Central Nervous System/physiology , Endoplasmic Reticulum/metabolism , Nerve Regeneration , Vesicular Transport Proteins/metabolism , Animals , Axons/metabolism , Cells, Cultured , Endoplasmic Reticulum/genetics , Endosomes/metabolism , Female , Humans , Integrins/metabolism , Mice , Mice, Inbred C57BL , Mutation , Nerve Regeneration/drug effects , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/administration & dosage , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Phosphorylation , Protein Domains , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/physiology , Vesicular Transport Proteins/administration & dosage , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
7.
J Vis Exp ; (163)2020 09 26.
Article in English | MEDLINE | ID: mdl-33044451

ABSTRACT

A western blot (also known as an immunoblot) is a canonical method for biomedical research. It is commonly used to determine the relative size and abundance of specific proteins as well as post-translational protein modifications. This technique has a rich history and remains in widespread use due to its simplicity. However, the western blotting procedure famously takes hours, even days, to complete, with a critical bottleneck being the long incubation times that limit its throughput. These incubation steps are required due to the slow diffusion of antibodies from the bulk solution to the immobilized antigens on the membrane: the antibody concentration near the membrane is much lower than the bulk concentration. Here, we present an innovation that dramatically reduces these incubation intervals by improving antigen binding via cyclic draining and replenishing (CDR) of the antibody solution. We also utilized an immunoreaction enhancing technology to preserve the sensitivity of the assay. A combination of the CDR method with a commercial immunoreaction enhancing agent boosted the output signal and substantially reduced the antibody incubation time. The resulting ultra-high-speed western blot can be accomplished in 20 minutes without any loss in sensitivity. This method can be applied to western blots using both chemiluminescent and fluorescent detection. This simple protocol allows researchers to better explore the analysis of protein expression in many samples.


Subject(s)
Blotting, Western/methods , Antibodies/immunology , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Image Processing, Computer-Assisted , Membranes, Artificial , Polyvinyls/chemistry
9.
Front Cell Neurosci ; 14: 208, 2020.
Article in English | MEDLINE | ID: mdl-32848612

ABSTRACT

Traumatic spinal cord injury produces long-term neurological damage, and presents a significant public health problem with nearly 18,000 new cases per year in the U.S. The injury results in both acute and chronic changes in the spinal cord, ultimately resulting in the production of a glial scar, consisting of multiple cells including fibroblasts, macrophages, microglia, and reactive astrocytes. Within the scar, there is an accumulation of extracellular matrix (ECM) molecules-primarily tenascins and chondroitin sulfate proteoglycans (CSPGs)-which are considered to be inhibitory to axonal regeneration. In this review article, we discuss the role of CSPGs in the injury response, especially how sulfated glycosaminoglycan (GAG) chains act to inhibit plasticity and regeneration. This includes how sulfation of GAG chains influences their biological activity and interactions with potential receptors. Comprehending the role of CSPGs in the inhibitory properties of the glial scar provides critical knowledge in the much-needed production of new therapies.

10.
Exp Cell Res ; 389(2): 111911, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32061832

ABSTRACT

Phospholipid Phosphatase-Related Protein Type 1 (PLPPR1) is a six-transmembrane protein that belongs to the family of plasticity-related gene proteins, which is a novel brain-specific subclass of the lipid phosphate phosphatase superfamily. PLPPR1-5 have prominent roles in synapse formation and axonal pathfinding. We found that PLPPR1 overexpression in the mouse neuroblastoma cell line (Neuro2a) results in increase in cell adhesion and reduced cell migration. During migration, these cells leave behind long fibrous looking extensions of the plasma membrane causing a peculiar phenotype. Cells expressing PLPPR1 showed decreased actin turnover and decreased disassembly of focal adhesions. PLPPR1 also reduced active Rac1, and expressing dominant negative Rac1 produced a similar phenotype to overexpression of PLPPR1. The PLPPR1-induced phenotype of long fibers was reversed by introducing constitutively active Rac1. In summary, we show that PLPPR1 decreases active Rac1 levels that leads to cascade of events which increases cell adhesion.


Subject(s)
Cell Adhesion , Focal Adhesions , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neuroblastoma/pathology , Neuropeptides/metabolism , Phosphoric Monoester Hydrolases/metabolism , rac1 GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cell Movement , Membrane Proteins/genetics , Mice , Neuroblastoma/metabolism , Neuropeptides/genetics , Phosphoric Monoester Hydrolases/genetics , Signal Transduction , Tumor Cells, Cultured , rac1 GTP-Binding Protein/genetics
11.
J Biochem ; 168(1): 15-22, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32027361

ABSTRACT

The immunoblotting technique (also known as western blotting) is an essential tool used in biomedical research to determine the relative size and abundance of specific proteins and protein modifications. However, long incubation times severely limit its throughput. We have devised a system that improves antigen binding by cyclic draining and replenishing (CDR) of the antibody solution in conjunction with an immunoreaction enhancing agent. Biochemical analyses revealed that the CDR method reduced the incubation time of the antibodies, and the presence of a commercial immunoreaction enhancing agent altered the affinity of the antibody, respectively. Combination of the CDR method with the immunoreaction enhancing agent considerably enhanced the output signal and further reduced the incubation time of the antibodies. The resulting high-speed immunoblot can be completed in 20 min without any loss in sensitivity. Further, the antibodies are fully reusable. This method is effective for both chemiluminescence and fluorescence detection. Widespread adoption of this technique could dramatically boost efficiency and productivity across the life sciences.


Subject(s)
Antibodies/immunology , Biomedical Enhancement/methods , Blotting, Western/methods , Cells/metabolism , Immunoblotting/methods , Proteins/analysis , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Proteins/metabolism
12.
Biomolecules ; 10(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31935947

ABSTRACT

Sulfation is a common modification of extracellular glycans, tyrosine residues on proteins, and steroid hormones, and is important in a wide variety of signaling pathways. We investigated the role of sulfation on endogenous oxidative stress, such as glutamate-induced oxytosis and erastin-induced ferroptosis, using mouse hippocampal HT22 cells. Sodium chlorate competitively inhibits the formation of 3'-phosphoadenosine 5'-phosphosulfate, the high energy sulfate donor in cellular sulfation reactions. The treatment of HT22 cells with sodium chlorate decreased sulfation of heparan sulfate proteoglycans and chondroitin sulfate proteoglycans. Sodium chlorate and ß-d-xyloside, which prevents proteoglycan glycosaminoglycan chain attachment, exacerbated both glutamate- and erastin-induced cell death, suggesting that extracellular matrix influenced oxytosis and ferroptosis. Moreover, sodium chlorate enhanced the generation of reactive oxygen species and influx of extracellular Ca2+ in the process of oxytosis and ferroptosis. Interestingly, sodium chlorate did not affect antioxidant glutathione levels. Western blot analysis revealed that sodium chlorate enhanced erastin-induced c-Jun N-terminal kinase phosphorylation, which is preferentially activated by cell stress-inducing signals. Collectively, our findings indicate that sulfation is an important modification for neuroprotection against oxytosis and ferroptosis in neuronal hippocampal cells.


Subject(s)
Ferroptosis/physiology , Regulated Cell Death/physiology , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cell Line , Chlorates/pharmacology , Ferroptosis/drug effects , Glutamic Acid/metabolism , Glutathione/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphoadenosine Phosphosulfate/chemistry , Proteoglycans/chemistry , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Somatomedins/metabolism
13.
Exp Eye Res ; 190: 107859, 2020 01.
Article in English | MEDLINE | ID: mdl-31705897

ABSTRACT

The accumulation of chondroitin sulfate proteoglycans (CSPGs) in the glial scar following acute damage to the central nervous system (CNS) limits the regeneration of injured axons. Given the rich diversity of CSPG core proteins and patterns of GAG sulfation, identifying the composition of these CSPGs is essential for understanding their roles in injury and repair. Differential expression of core proteins and sulfation patterns have been characterized in the brain and spinal cord of mice and rats, but a comprehensive study of these changes following optic nerve injury has not yet been performed. Here, we show that the composition of CSPGs in the optic nerve and retina following optic nerve crush (ONC) in mice and rats exhibits an increase in aggrecan, brevican, phosphacan, neurocan and versican, similar to changes following spinal cord injury. We also observe an increase in inhibitory 4-sulfated (4S) GAG chains, which suggests that the persistence of CSPGs in the glial scar opposes the growth of CNS axons, thereby contributing to the failure of regeneration and recovery of function.


Subject(s)
Crush Injuries/metabolism , Optic Nerve Injuries/metabolism , Optic Nerve/metabolism , Retina/metabolism , Aggrecans/metabolism , Animals , Brevican/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Disease Models, Animal , Drug Combinations , Female , Glycosaminoglycans/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neurocan/metabolism , Rats , Rats, Sprague-Dawley , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Sulfamonomethoxine , Trimethoprim , Versicans/metabolism
14.
Glia ; 66(11): 2427-2437, 2018 11.
Article in English | MEDLINE | ID: mdl-30378174

ABSTRACT

Loss of substantia nigra dopaminergic neurons results in Parkinson disease (PD). Degenerative PD usually presents in the seventh decade whereas genetic disorders, including mutations in PARK2, predispose to early onset PD. PARK2 encodes the parkin E3 ubiquitin ligase which confers pleotropic effects on mitochondrial and cellular fidelity and as a mediator of endoplasmic reticulum (ER) stress signaling. Although the majority of studies investigating ameliorative effects of parkin focus on dopaminergic neurons we found that astrocytes are enriched with parkin. Furthermore, astrocytes deficient in parkin display stress-induced elevation of nucleotide-oligomerization domain receptor 2 (NOD2), a cytosolic receptor integrating ER stress and inflammation. Given the neurotropic and immunomodulatory role of astrocytes we reasoned that parkin may regulate astrocyte ER stress and inflammation to control neuronal homeostasis. We show that, in response to ER stress, parkin knockdown astrocytes exhibit exaggerated ER stress, JNK activation and cytokine release, and reduced neurotropic factor expression. In coculture studied we demonstrate that dopaminergic SHSY5Y cells and primary neurons with the presence of parkin depleted astrocytes are more susceptible to ER stress and inflammation-induced apoptosis than wildtype astrocytes. Parkin interacted with, ubiquitylated and diminished NOD2 levels. Additionally, the genetic induction of parkin ameliorated inflammation in NOD2 expressing cells and knockdown of NOD2 in astrocytes suppressed inflammatory defects in parkin deficient astrocytes and concurrently blunted neuronal apoptosis. Collectively these data identify a role for parkin in modulating NOD2 as a regulatory node in astrocytic control of neuronal homeostasis.


Subject(s)
Astrocytes/ultrastructure , Endoplasmic Reticulum Stress/physiology , Inflammation/pathology , Nerve Growth Factors/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Ubiquitin-Protein Ligases/deficiency , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Astrocytes/drug effects , Cells, Cultured , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells , Humans , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/genetics , Oncogene Protein p55(v-myc)/metabolism , Oxidopamine/pharmacology , Transcription Factor CHOP/metabolism , Ubiquitin-Protein Ligases/genetics
16.
J Biol Chem ; 293(29): 11639-11647, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29880643

ABSTRACT

Receptor protein-tyrosine phosphatase RPTPσ has important functions in modulating neural development and regeneration. Compelling evidence suggests that both heparan sulfate (HS) and chondroitin sulfate (CS) glycosaminoglycans (GAGs) bind to a series of Lys residues located in the first Ig domain of RPTPσ. However, HS promotes and CS inhibits axonal growth. Mutation of these Lys residues abolished binding and signal transduction of RPTPσ to CS, whereas HS binding was reduced, and signaling persisted. This activity was mediated through novel heparin-binding sites identified in the juxtamembrane region. Although different functional outcomes of HS and CS have been previously attributed to the differential oligomeric state of RPTPσ upon GAG binding, we found that RPTPσ was clustered by both heparin and CS GAG rich in 4,6-O-disulfated disaccharide units. We propose an additional mechanism by which RPTPσ distinguishes between HS and CS through these novel binding sites.


Subject(s)
Chondroitin Sulfates/metabolism , Heparin/metabolism , Heparitin Sulfate/metabolism , Proteoglycans/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Signal Transduction , Animals , Cattle , HEK293 Cells , Humans , Mice , Recombinant Proteins/metabolism , Swine
17.
Elife ; 72018 05 15.
Article in English | MEDLINE | ID: mdl-29762123

ABSTRACT

The failure of mammalian CNS neurons to regenerate their axons derives from a combination of intrinsic deficits and extrinsic factors. Following injury, chondroitin sulfate proteoglycans (CSPGs) within the glial scar inhibit axonal regeneration, an action mediated by the sulfated glycosaminoglycan (GAG) chains of CSPGs, especially those with 4-sulfated (4S) sugars. Arylsulfatase B (ARSB) selectively cleaves 4S groups from the non-reducing ends of GAG chains without disrupting other, growth-permissive motifs. We demonstrate that ARSB is effective in reducing the inhibitory actions of CSPGs both in in vitro models of the glial scar and after optic nerve crush (ONC) in adult mice. ARSB is clinically approved for replacement therapy in patients with mucopolysaccharidosis VI and therefore represents an attractive candidate for translation to the human CNS.


Subject(s)
Axons/drug effects , Axons/physiology , Chondroitin Sulfate Proteoglycans/chemistry , N-Acetylgalactosamine-4-Sulfatase/metabolism , Optic Nerve Injuries/drug therapy , Regeneration/drug effects , Sulfates/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice , N-Acetylgalactosamine-4-Sulfatase/administration & dosage , Treatment Outcome
18.
Neural Regen Res ; 13(2): 289-297, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29557379

ABSTRACT

As one major component of extracellular matrix (ECM) in the central nervous system, chondroitin sulfate proteoglycans (CSPGs) have long been known as inhibitors enriched in the glial scar that prevent axon regeneration after injury. Although many studies have shown that CSPGs inhibited neurite outgrowth in vitro using different types of neurons, the mechanism by which CSPGs inhibit axonal growth remains poorly understood. Using cerebellar granule neuron (CGN) culture, in this study, we evaluated the effects of different concentrations of both immobilized and soluble CSPGs on neuronal growth, including cell adhesion, spreading and neurite growth. Neurite length decreased while CSPGs concentration arised, meanwhile, a decrease in cell density accompanied by an increase in cell aggregates formation was observed. Soluble CSPGs also showed an inhibition on neurite outgrowth, but it required a higher concentration to induce cell aggregates formation than coated CSPGs. We also found that growth cone size was significantly reduced on CSPGs and neuronal cell spreading was restrained by CSPGs, attributing to an inhibition on lamellipodial extension. The effect of CSPGs on neuron adhesion was further evidenced by interference reflection microscopy (IRM) which directly demonstrated that both CGNs and cerebral cortical neurons were more loosely adherent to a CSPG substrate. These data demonstrate that CSPGs have an effect on cell adhesion and spreading in addition to neurite outgrowth.

19.
J Neurosci Res ; 96(4): 573-588, 2018 04.
Article in English | MEDLINE | ID: mdl-29344975

ABSTRACT

The brain extracellular matrix (ECM) plays a crucial role in both the developing and adult brain by providing structural support and mediating cell-cell interactions. In this review, we focus on the major constituents of the ECM and how they function in both normal and injured brain, and summarize the changes in the composition of the ECM as well as how these changes either promote or inhibit recovery of function following traumatic brain injury (TBI). Modulation of ECM composition to facilitates neuronal survival, regeneration and axonal outgrowth is a potential therapeutic target for TBI treatment.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Extracellular Matrix/physiology , Extracellular Matrix Proteins/physiology , Metalloproteases/physiology , Proteoglycans/physiology , Tenascin/physiology
20.
Trends Neurosci ; 41(1): 47-61, 2018 01.
Article in English | MEDLINE | ID: mdl-29150096

ABSTRACT

Proteoglycans (PGs) in the extracellular matrix (ECM) play vital roles in axon growth and navigation, plasticity, and regeneration of injured neurons. Different classes of PGs may support or inhibit cell growth, and their functions are determined in part by highly specific structural features. Among these, the pattern of sulfation on the PG sugar chains is a paramount determinant of a diverse and flexible set of outcomes. Recent studies of PG sulfation illustrate the challenges of attributing biological actions to specific sulfation patterns, and suggest ways in which highly similar molecules may exert opposing effects on neurons. The receptors for PGs, which have yet to be fully characterized, display a similarly nuanced spectrum of effects. Different classes of PG function via overlapping families of receptors and signaling pathways. This enables them to control axon growth and guidance with remarkable specificity, but it poses challenges for determining the precise binding interactions and downstream effects of different PGs and their assorted sulfated epitopes. This review examines existing and emerging evidence for the roles of PG sulfation and receptor interactions in determining how these complex molecules influence neuronal development, growth, and function.


Subject(s)
Membrane Transport Proteins/metabolism , Neurons/metabolism , Proteoglycans/metabolism , Animals , Neuronal Outgrowth/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...