Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 9(10): 1366-1378, 2017 10.
Article in English | MEDLINE | ID: mdl-28855301

ABSTRACT

We have characterised the proteolytic cleavage events responsible for the shedding of triggering receptor expressed on myeloid cells 2 (TREM2) from primary cultures of human macrophages, murine microglia and TREM2-expressing human embryonic kidney (HEK293) cells. In all cell types, a soluble 17 kDa N-terminal cleavage fragment was shed into the conditioned media in a constitutive process that is inhibited by G1254023X and metalloprotease inhibitors and siRNA targeting ADAM10. Inhibitors of serine proteases and matrix metalloproteinases 2/9, and ADAM17 siRNA did not block TREM2 shedding. Peptidomimetic protease inhibitors highlighted a possible cleavage site, and mass spectrometry confirmed that shedding occurred predominantly at the H157-S158 peptide bond for both wild-type and H157Y human TREM2 and for the wild-type murine orthologue. Crucially, we also show that the Alzheimer's disease-associated H157Y TREM2 variant was shed more rapidly than wild type from HEK293 cells, possibly by a novel, batimastat- and ADAM10-siRNA-independent, sheddase activity. These insights offer new therapeutic targets for modulating the innate immune response in Alzheimer's and other neurological diseases.


Subject(s)
Alzheimer Disease/genetics , Membrane Glycoproteins/metabolism , Proteolysis , Receptors, Immunologic/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Animals, Newborn , Culture Media, Conditioned , HEK293 Cells , Humans , Ketocholesterols/pharmacology , Macrophages/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Immunologic/genetics
2.
J Neurosci ; 35(21): 8359-75, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26019348

ABSTRACT

Integrins are adhesion and survival molecules involved in axon growth during CNS development, as well as axon regeneration after injury in the peripheral nervous system (PNS). Adult CNS axons do not regenerate after injury, partly due to a low intrinsic growth capacity. We have previously studied the role of integrins in axon growth in PNS axons; in the present study, we investigate whether integrin mechanisms involved in PNS regeneration may be altered or lacking from mature CNS axons by studying maturing CNS neurons in vitro. In rat cortical neurons, we find that integrins are present in axons during initial growth but later become restricted to the somato-dendritic domain. We investigated how this occurs and whether it can be altered to enhance axonal growth potential. We find a developmental change in integrin trafficking; transport becomes predominantly retrograde throughout axons, but not dendrites, as neurons mature. The directionality of transport is controlled through the activation state of ARF6, with developmental upregulation of the ARF6 GEF ARNO enhancing retrograde transport. Lowering ARF6 activity in mature neurons restores anterograde integrin flow, allows transport into axons, and increases axon growth. In addition, we found that the axon initial segment is partly responsible for exclusion of integrins and removal of this structure allows integrins into axons. Changing posttranslational modifications of tubulin with taxol also allows integrins into the proximal axon. The experiments suggest that the developmental loss of regenerative ability in CNS axons is due to exclusion of growth-related molecules due to changes in trafficking.


Subject(s)
ADP-Ribosylation Factors/metabolism , Axons/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Integrins/deficiency , Regeneration/physiology , ADP-Ribosylation Factor 6 , Animals , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/metabolism , Cerebral Cortex/embryology , Rats
3.
PLoS One ; 9(10): e109401, 2014.
Article in English | MEDLINE | ID: mdl-25314276

ABSTRACT

In their natural habitat, the peripheral nerve, Schwann cells (SCs) form nicely aligned pathways (also known as the bands of Büngner) that guide regenerating axons to their targets. Schwann cells that are implanted in the lesioned spinal cord fail to align in pathways that could support axon growth but form cellular clusters that exhibit only limited intermingling with the astrocytes and meningeal cells (MCs) that are present in the neural scar. The formation of cell clusters can be studied in co-cultures of SCs and MCs. In these co-cultures SCs form cluster-like non-overlapping cell aggregates with well-defined boundaries. There are several indications that neuropilins (NRPs) play an important role in MC-induced SC aggregation. Both SCs and MCs express NRP1 and NRP2 and SCs express the NRP ligands Sema3B, C and E while MCs express Sema3A, C, E and F. We now demonstrate that in SC-MC co-cultures, siRNA mediated knockdown of NRP2 in SCs decreased the formation of SC clusters while these SCs maintained their capacity to align in bands of Büngner-like columnar arrays. Unexpectedly, knockdown of NRP1 expression resulted in a significant increase in SC aggregation. These results suggest that a reduction in NRP2 expression may enhance the capacity of implanted SCs to interact with MCs that invade a neural scar formed after a lesion of the spinal cord.


Subject(s)
Meninges/cytology , Neuropilin-1/metabolism , Neuropilin-2/metabolism , Schwann Cells/cytology , Animals , Cell Communication , Cells, Cultured , Coculture Techniques , Female , Meninges/metabolism , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/genetics , Neuropilin-2/antagonists & inhibitors , Neuropilin-2/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Inbred F344 , Schwann Cells/metabolism , Transfection
4.
J Neurosci ; 33(27): 11116-35, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23825416

ABSTRACT

Olfactory ensheathing cells (OECs) have neuro-restorative properties in animal models for spinal cord injury, stroke, and amyotrophic lateral sclerosis. Here we used a multistep screening approach to discover genes specifically contributing to the regeneration-promoting properties of OECs. Microarray screening of the injured olfactory pathway and of cultured OECs identified 102 genes that were subsequently functionally characterized in cocultures of OECs and primary dorsal root ganglion (DRG) neurons. Selective siRNA-mediated knockdown of 16 genes in OECs (ADAMTS1, BM385941, FZD1, GFRA1, LEPRE1, NCAM1, NID2, NRP1, MSLN, RND1, S100A9, SCARB2, SERPINI1, SERPINF1, TGFB2, and VAV1) significantly reduced outgrowth of cocultured DRG neurons, indicating that endogenous expression of these genes in OECs supports neurite extension of DRG neurons. In a gain-of-function screen for 18 genes, six (CX3CL1, FZD1, LEPRE1, S100A9, SCARB2, and SERPINI1) enhanced and one (TIMP2) inhibited neurite growth. The most potent hit in both the loss- and gain-of-function screens was SCARB2, a protein that promotes cholesterol secretion. Transplants of fibroblasts that were genetically modified to overexpress SCARB2 significantly increased the number of regenerating DRG axons that grew toward the center of a spinal cord lesion in rats. We conclude that expression of SCARB2 enhances regenerative sprouting and that SCARB2 contributes to OEC-mediated neuronal repair.


Subject(s)
Axons/physiology , Lysosomal Membrane Proteins/biosynthesis , Molecular Imprinting/methods , Nerve Regeneration/physiology , Olfactory Mucosa/physiology , Receptors, Scavenger/biosynthesis , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Female , Genetic Testing/methods , HEK293 Cells , Humans , Lysosomal Membrane Proteins/genetics , Mesothelin , Olfactory Bulb/physiology , Olfactory Mucosa/cytology , Pregnancy , Rats , Rats, Inbred F344 , Rats, Wistar , Receptors, Scavenger/genetics , Sensory Receptor Cells/cytology
5.
Int Rev Neurobiol ; 106: 75-104, 2012.
Article in English | MEDLINE | ID: mdl-23211460

ABSTRACT

Adult central nervous system (CNS) axons fail to regenerate after injury because of inhibitory factors in the surrounding environment and a low intrinsic regenerative capacity. Axons in the adult peripheral nervous system have a higher regenerative capacity, due in part to the presence of certain integrins-receptors for the extracellular matrix. Integrins are critical for axon growth during the development of the nervous system but are absent from some adult CNS axons. Here, we discuss the intrinsic mechanisms that regulate axon regeneration and examine the role of integrins. As correct localization is paramount to integrin function, we further discuss the mechanisms that regulate integrin traffic toward the axonal growth cone.


Subject(s)
Axons/metabolism , Integrins/biosynthesis , Nerve Regeneration/physiology , Animals , Axons/pathology , Growth Cones/metabolism , Growth Cones/pathology , Humans , Neurons/metabolism , Neurons/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
6.
Exp Neurol ; 229(1): 10-45, 2011 May.
Article in English | MEDLINE | ID: mdl-21396936

ABSTRACT

Genome wide transcriptional profiling and large scale proteomics have emerged as two powerful methods to dissect the molecular properties of specific neural tissues or cell types on a global scale. Several genome-wide transcriptional profiling and proteomics studies have been published on cultured olfactory ensheathing cells (OEC). In this article we present a meta-analysis of all five published and publicly available micro-array gene expression datasets of cultured early-passage-OB-OEC with other cell types (Schwann cells, late-passage-OB-OEC, mucosa-OEC, an OEC cell line, and acutely dissected OEC). The aim of this meta-analysis is to identify genes and molecular pathways that are found in multiple instead of one isolated study. 454 Genes were detected in at least three out of five microarray datasets. In this "Top-list", genes involved in the biological processes "growth of neurites", "blood vessel development", "migration of cells" and "immune response" were strongly overrepresented. By applying network analysis tools, molecular networks were constructed and Hub-genes were identified that may function as key genes in the above mentioned interrelated processes. We also identified 7 genes (ENTPD2, MATN2, CTSC, PTHLH, GLRX1, COL27A1 and ID2) with uniformly higher or lower expression in early-passage-OB-OEC in all five microarray comparisons. These genes have diverse but intriguing roles in neuroprotection, neurite extension and/or tissue repair. Our meta-analysis provides novel insights into the molecular basis of OB-OEC-mediated neural repair and can serve as a repository for investigators interested in the molecular biology of OEC. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.


Subject(s)
Gene Expression Regulation , Neuroglia/physiology , Olfactory Bulb/physiology , Protein Array Analysis/methods , Animals , Gene Regulatory Networks/genetics , Humans , Neuroglia/cytology , Olfactory Bulb/cytology
7.
J Neurosci Res ; 87(7): 1556-64, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19140223

ABSTRACT

Schwann cells (SCs) and olfactory ensheathing glia (OEG) have both been used as cellular transplants to promote spinal cord repair. Both cell types support axonal regeneration and have beneficial effects on functional recovery. A significant difference between SCs and OEG is the effect of these cell types on astrocytes (ACs) present in the neural scar. In contrast to OEG, which associate and intermingle with ACs, SCs and ACs form separate cellular territories. Here, we show that OEG and SCs also interact differently with meningeal cells (MCs), another major cellular component of the neural scar. Whereas OEG intermingle with MCs in cocultures, SCs aggregate into well-defined cell clusters. Our data suggest that (a) soluble factor(s) as well as direct cellular contact are involved in the MC-induced SC clustering. Furthermore, the cluster formation of SCs in coculture with MCs is different from the previously reported segregation of SCs and ACs in coculture. The present results help to understand the differential behavior of both cell types after transplantation in the injured spinal cord and will be important to either determine which cell has optimal capacities to render the scar more permissive for regeneration, or to exploit the transplantation of both cell types in combination.


Subject(s)
Meninges/physiology , Neuroglia/physiology , Schwann Cells/physiology , Animals , Astrocytes/physiology , Cell Aggregation/physiology , Coculture Techniques , Culture Media, Conditioned , Female , Immunohistochemistry , Meninges/cytology , Meninges/metabolism , Olfactory Bulb/physiology , Rats , Rats, Inbred F344 , Sciatic Nerve/physiology
8.
Glia ; 56(12): 1285-98, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18615567

ABSTRACT

Olfactory ensheathing glia (OEG) are a specialized type of glia that support the growth of primary olfactory axons from the neuroepithelium in the nasal cavity to the brain. Transplantation of OEG in the injured spinal cord promotes sprouting of injured axons and results in reduced cavity formation, enhanced axonal and tissue sparing, remyelination, and angiogenesis. Gene expression analysis may help to identify the molecular mechanisms underlying the ability of OEG to recreate an environment that supports regeneration in the central nervous system. Here, we compared the transcriptome of cultured OEG (cOEG) with the transcriptomes of cultured Schwann cells (cSCs) and of OEG directly obtained from their natural environment (nOEG), the olfactory nerve layer of adult rats. Functional data mining by Gene Ontology (GO)-analysis revealed a number of overrepresented GO-classes associated with tissue repair. These classes include "response to wounding," "blood vessel development," "cell adhesion," and GO-classes related to the extracellular matrix and were overrepresented in the set of differentially expressed genes between both comparisons. The current screening approach combined with GO-analysis has identified distinct molecular properties of OEG that may underlie their efficacy and interaction with host tissue after implantation in the injured spinal cord. These observations can form the basis for studies on the function of novel target molecules for therapeutic intervention after neurotrauma.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation/physiology , Myelin Sheath/metabolism , Neuroglia/physiology , Olfactory Bulb/growth & development , Schwann Cells/physiology , Animals , Cells, Cultured , Female , Myelin Sheath/genetics , Nerve Regeneration/physiology , Neuroglia/cytology , Olfactory Bulb/cytology , Rats , Rats, Inbred F344 , Schwann Cells/cytology
9.
Brain Res Rev ; 56(1): 236-58, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17884174

ABSTRACT

Olfactory ensheathing glia (OEG) are a specialized type of glia that guide primary olfactory axons from the neuroepithelium in the nasal cavity to the brain. The primary olfactory system is able to regenerate after a lesion and OEG contribute to this process by providing a growth-supportive environment for newly formed axons. In the spinal cord, axons are not able to restore connections after an injury. The effects of OEG transplants on the regeneration of the injured spinal cord have been studied for over a decade. To date, of all the studies using only OEG as a transplant, 41 showed positive effects, while 13 studies showed limited or no effects. There are several contradictory reports on the migratory and axon growth-supporting properties of transplanted OEG. Hence, the regenerative potential of OEG has become the subject of intense discussion. In this review, we first provide an overview of the molecular and cellular characteristics of OEG in their natural environment, the primary olfactory nervous system. Second, their potential to stimulate regeneration in the injured spinal cord is discussed. OEG influence scar formation by their ability to interact with astrocytes, they are able to remyelinate axons and promote angiogenesis. The ability of OEG to interact with scar tissue cells is an important difference with Schwann cells and may be a unique characteristic of OEG. Because of these effects after transplantation and because of their role in primary olfactory system regeneration, the OEG can be considered as a source of neuroregeneration-promoting molecules. To identify these molecules, more insight into the molecular biology of OEG is required. We believe that genome-wide gene expression studies of OEG in their native environment, in culture and after transplantation will ultimately reveal unique combinations of molecules involved in the regeneration-promoting potential of OEG.


Subject(s)
Brain Tissue Transplantation/methods , Nerve Regeneration/physiology , Neuroglia/transplantation , Olfactory Bulb/cytology , Olfactory Bulb/transplantation , Spinal Cord Injuries/therapy , Animals , Brain Tissue Transplantation/trends , Cell Communication/physiology , Cicatrix/physiopathology , Humans , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Myelinated/ultrastructure , Neuroglia/physiology , Olfactory Bulb/physiology , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Spinal Cord Injuries/physiopathology
10.
Mol Cell Neurosci ; 24(4): 902-12, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14697657

ABSTRACT

The neural scar that forms after injury to the mammalian central nervous system is a barrier to sprouting and regenerating axons. In addition to reactive astrocytes that are present throughout the lesion site, leptomeningeal fibroblasts invade the lesion core. When isolated in vitro, these cells form a very poor substrate for growing neurites, even more so than reactive astrocytes. Nevertheless the molecular mechanisms involved in this growth inhibition are not well understood. Semaphorins have been reported to be upregulated in meningeal cells (MCs) on mechanical injury to the brain and spinal cord. In the present study, we show that Sema3A mRNA and active protein are produced by cultured meningeal cells. A protein extract from these cells induces the collapse of embryonic dorsal root ganglion (DRG) growth cones. This collapsing activity is partially blocked by neuropilin-1 antibodies and is absent in meningeal cells derived from Sema3A-knockout mice. In addition to growth cone collapse, recombinant Sema3A but not Sema3C inhibits neurite outgrowth of embryonic DRGs. Consistent with this result we find that the inhibitory effect of meningeal cells on neurite outgrowth is partially overcome on Sema3A-deficient MCs. Furthermore we show that the inhibitory effect of MC-derived Sema3A on neurite outgrowth is modulated by nerve growth factor. Our results show that Sema3A, a chemorepellent during nervous system development, is a major neurite growth-inhibitory molecule in meningeal fibroblasts and is therefore likely to contribute to the inhibitory properties of the neural scar.


Subject(s)
Meninges/metabolism , Neural Inhibition/physiology , Neurites/metabolism , Semaphorin-3A/biosynthesis , Animals , Cells, Cultured , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Growth Cones/metabolism , Growth Inhibitors/biosynthesis , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Meninges/cytology , Meninges/embryology , Meninges/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Semaphorin-3A/deficiency , Semaphorin-3A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...