Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell Commun Signal ; 22(1): 155, 2024 02 29.
Article in English | MEDLINE | ID: mdl-38424563

ABSTRACT

BACKGROUND: Vascular endothelial cells are pivotal in the pathophysiological progression following spinal cord injury (SCI). The UTX (Ubiquitously Transcribed Tetratripeptide Repeat on Chromosome X) serves as a significant regulator of endothelial cell phenotype. The manipulation of endogenous neural stem cells (NSCs) offers a compelling strategy for the amelioration of SCI. METHODS: Two mouse models were used to investigate SCI: NSCs lineage-traced mice and mice with conditional UTX knockout (UTX KO) in endothelial cells. To study the effects of UTX KO on neural differentiation, we harvested extracellular vesicles (EVs) from both UTX KO spinal cord microvascular endothelial cells (SCMECs) and negative control SCMECs. These EVs were then employed to modulate the differentiation trajectory of endogenous NSCs in the SCI model. RESULTS: In our NSCs lineage-traced mice model of SCI, a marked decrease in neurogenesis was observed post-injury. Notably, NSCs in UTX KO SCMECs mice showed enhanced neuronal differentiation compared to controls. RNA sequencing and western blot analyses revealed an upregulation of L1 cell adhesion molecule (L1CAM), a gene associated with neurogenesis, in UTX KO SCMECs and their secreted EVs. This aligns with the observed promotion of neurogenesis in UTX KO conditions. In vivo administration of L1CAM-rich EVs from UTX KO SCMECs (KO EVs) to the mice significantly enhanced neural differentiation. Similarly, in vitro exposure of NSCs to KO EVs resulted in increased activation of the Akt signaling pathway, further promoting neural differentiation. Conversely, inhibiting Akt phosphorylation or knocking down L1CAM negated the beneficial effects of KO EVs on NSC neuronal differentiation. CONCLUSIONS: In conclusion, our findings substantiate that EVs derived from UTX KO SCMECs can act as facilitators of neural differentiation following SCI. This study not only elucidates a novel mechanism but also opens new horizons for therapeutic interventions in the treatment of SCI. Video Abstract.


Subject(s)
Extracellular Vesicles , Neural Cell Adhesion Molecule L1 , Neural Stem Cells , Spinal Cord Injuries , Animals , Mice , Cell Differentiation , Disease Models, Animal , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/pharmacology , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy
2.
Stem Cell Reports ; 18(4): 899-914, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36963389

ABSTRACT

Cell replacement therapy is expected as a new and more radical treatment against brain damage. We previously reported that transplanted human cerebral organoids extend their axons along the corticospinal tract in rodent brains. The axons reached the spinal cord but were still sparse. Therefore, this study optimized the host brain environment by the adeno-associated virus (AAV)-mediated expression of axon guidance proteins in mouse brain. Among netrin-1, SEMA3, and L1CAM, only L1CAM significantly promoted the axonal extension of mouse embryonic brain tissue-derived grafts. L1CAM was also expressed by donor neurons, and this promotion was exerted in a haptotactic manner by their homophilic binding. Primary cortical neurons cocultured on L1CAM-expressing HEK-293 cells supported this mechanism. These results suggest that optimizing the host environment by the AAV-mediated expression of axon guidance molecules enhances the effect of cell replacement therapy.


Subject(s)
Neural Cell Adhesion Molecule L1 , Animals , Mice , Humans , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/pharmacology , HEK293 Cells , Axons/metabolism , Pyramidal Tracts , Brain/metabolism , Netrin-1/metabolism , Netrin-1/pharmacology
3.
Acta Biomater ; 149: 273-286, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35764240

ABSTRACT

Microelectrode arrays for neural recording suffer from low yield and stability partly due to the inflammatory host responses. A neuronal cell adhesion molecule L1 coating has been shown to promote electrode-neuron integration, reduce microglia activation and improve recording. Coupling L1 to surface via a nanoparticle (NP) base layer further increased the protein surface density and stability. However, the exact L1-microglia interaction in these coatings has not been studied. Here we cultured primary microglia on L1 modified surfaces (with and without NP) and characterized microglia activation upon phorbol myristate acetate (PMA) and lipopolysaccharide (LPS) stimulation. Results showed L1 coatings reduced microglia's superoxide production in response to PMA and presented intrinsic antioxidant properties. Meanwhile, L1 decreased iNOS, NO, and pro-inflammatory cytokines (TNF alpha, IL-6, IL-1 beta), while increased anti-inflammatory cytokines (TGF beta 1, IL-10) in LPS stimulated microglia. Furthermore, L1 increased Arg-1 expression and phagocytosis upon LPS stimulation. Rougher NP surface showed lower number of microglia attached per area than their smooth counterpart, lower IL-6 release and superoxide production, and higher intrinsic reducing potential. Finally, we examined the effect of L1 and nanoparticle modifications on microglia response in vivo over 8 weeks with 2-photon imaging. Microglial coverage on the implant surface was found to be lower on the L1 modified substrates relative to unmodified, consistent with the in vitro observation. Our results indicate L1 significantly reduces superoxide production and inflammatory response of microglia and promotes wound healing, while L1 immobilization via a nanoparticle base layer brings added benefit without adverse effects. STATEMENT OF SIGNIFICANCE: Surface modification of microelectrode arrays with L1 has been shown to reduce microglia coverage on neural probe surface in vivo and improves neural recording, but the specific mechanism of action is not fully understood. The results in this study show that surface bound L1 reduces superoxide production from cultured microglia via direct reduction reaction and signaling pathways, increases anti-inflammatory cytokine release and phagocytosis in response to PMA or LPS stimulation. Additionally, roughening the surface with nanoparticles prior to L1 immobilization further increased the benefit of L1 in reducing microglia activation and oxidative stress. Together, our findings shed light on the mechanisms of action of nanotextured and neuroadhesive neural implant coatings and guide future development of seamless tissue interface.


Subject(s)
Nanoparticles , Neural Cell Adhesion Molecule L1 , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cytokines/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/pharmacology , Neurons , Superoxides
4.
Neurotoxicology ; 82: 69-81, 2021 01.
Article in English | MEDLINE | ID: mdl-33197482

ABSTRACT

PSA-NCAM is a molecule of therapeutic interest for its key role in promoting neuritogenesis and synaptic plasticity. The current study was aimed to investigate the neuroregenerative potential of 5-nonyloxytryptamine (5-NOT) as PSA mimetic compound against glutamate induced excitotoxicity. 2D and 3D cultures of cerebellar neurons challenged with glutamate were used to ascertain the effect of 5-NOT on neurite outgrowth, migration and expression of neuronal plasticity markers. Glutamate excitotoxicity is one of the major underlying pathological factor responsible for neurodegeneration in various neurological disorders such as trauma, stroke, ischemia, epilepsy and neurodegenerative diseases.5-NOT treatment was observed to promote axonal growth and defasiculation in glutamate challenged neurons as well as promoted the migration of cerebellar neurons in both wound scratched area and cerebellar explant cultures. Further, 5-NOT treatment upregulated the expression of synaptic plasticity and cell survival pathway proteins which showed reduced expression after glutamate induced excitotoxicity. Thus, this preliminary data reveals thatPSA-mimetic,5-NOT may prove to be a potential neuroprotective candidate for neurodegenerative diseases.


Subject(s)
Cerebellum/drug effects , Glutamic Acid/toxicity , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sialic Acids/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Neuroglia/drug effects , Neuronal Plasticity/drug effects , Rats , Rats, Wistar
5.
Sci Rep ; 8(1): 8957, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895898

ABSTRACT

NCAM1 and NCAM2 have ectodomains consisting of 5 Ig domains followed by 2 membrane-proximal FnIII domains. In this study we investigate and compare the structures and functions of these FnIII domains. The NCAM1 and -2 FnIII2 domains both contain a Walker A motif. In NCAM1 binding of ATP to this motif interferes with NCAM1 binding to FGFR. We obtained a structural model of the NCAM2 FnIII2 domain by NMR spectroscopy, and by titration with an ATP analogue we show that the NCAM2 Walker A motif does not bind ATP. Small angle X-ray scattering (SAXS) data revealed that the NCAM2 FnIII1-2 double domain exhibits a very low degree of flexibility. Moreover, recombinant NCAM2 FnIII domains bind FGFR in vitro, and the FnIII1-2 double domain induces neurite outgrowth in a concentration-dependent manner through activation of FGFR. Several synthetic NCAM1-derived peptides induce neurite outgrowth via FGFR. Only 2 of 5 peptides derived from similar regions in NCAM2 induce neurite outgrowth, but the most potent of these peptides stimulates neurite outgrowth through FGFR-dependent activation of the Ras-MAPK pathway. These results reveal that the NCAM2 FnIII domains form a rigid structure that binds and activates FGFR in a manner related to, but different from NCAM1.


Subject(s)
MAP Kinase Signaling System/drug effects , Neural Cell Adhesion Molecule L1 , Neurites/metabolism , Peptides , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Motifs , Animals , Humans , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/pharmacology , Neural Cell Adhesion Molecules , Peptides/chemistry , Peptides/pharmacology , Protein Domains , Rats , Rats, Wistar
6.
Pharmazie ; 69(6): 461-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24974583

ABSTRACT

L1 cell adhesion molecule (L1CAM) is highly expressed in various types of cancer cells and has been implicated in the control of cell proliferation and motility. Recently, L1CAM was reported to induce the motility of melanoma cells, but the mechanism of this induction remains poorly understood. In this study, we investigated the molecular mechanisms by which L1CAM induces the motility of melanoma cells. Unlike other types of cancer cells, B16F10 melanoma cells highly expressed L1CAM at both the RNA and protein levels, and the expression of L1CAM induced AP-1 activity. In accordance to AP-1 activation, MAPK signaling pathways were activated by L1CAM. Inhibition of L1CAM expression by L1CAM-specific siRNA suppressed the activation of MAPKs such as ERK and p38. However, no significant change was observed in JNK activation. As expected, upstream MAP2K, MKK3/6, MAP3K, and TAK1 were also deactivated by the inhibition of L1CAM expression. L1CAM induced the motility of B16F10 cells. Inhibition of L1CAM expression suppressed migration and invasion of B16F10 cells, but no suppressive effect was observed on their proliferation and anti-apoptotic resistance. Treatment of B16F10 cells with U0126, an ERK inhibitor, or SB203580, a p38 inhibitor, suppressed the migration and invasion abilities of B16F10 cells. Taken together, our results suggest that L1CAM induces the motility of B16F10 melanoma cells via the activation of MAPK pathways. This finding provides a more detailed molecular mechanism of L1CAM-mediated induction of melanoma cell motility.


Subject(s)
Cell Movement/drug effects , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Neural Cell Adhesion Molecule L1/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Proliferation/drug effects , Flow Cytometry , Humans , Luciferases/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Transcription Factor AP-1/genetics , Transfection
7.
Acta Biomater ; 10(10): 4113-26, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24914828

ABSTRACT

We investigated the design of neurotrophic biomaterial constructs for human neural stem cells, guided by neural developmental cues of N-cadherin and L1 adhesion molecules. Polymer substrates fabricated either as two-dimensional (2-D) films or three-dimensional (3-D) microfibrous scaffolds were functionalized with fusion chimeras of N-cadherin-Fc alone and in combination with L1-Fc, and the effects on differentiation, neurite extension and survival of H9 human-embryonic-stem-cell-derived neural stem cells (H9-NSCs) were quantified. Combinations of N-cadherin and L1-Fc co-operatively enhanced neuronal differentiation profiles, indicating the critical nature of the two complementary developmental cues. Notably, substrates presenting low levels of N-cadherin-Fc concentrations, combined with proportionately higher L1-Fc concentration, most enhanced neurite outgrowth and the degree of MAP2+ and neurofilament-M+ H9-NSCs. Low N-cadherin-Fc alone promoted improved cell survival following oxidative stress, compared to higher concentrations of N-cadherin-Fc alone or combinations with L1-Fc. Pharmacological and antibody blockage studies revealed that substrates presenting low levels of N-cadherin are functionally competent so long as they elicit a threshold signal mediated by homophilic N-cadherin and fibroblast growth factor signaling. Overall, these studies highlight the ability of optimal combinations of N-cadherin and L1 to recapitulate a "neurotrophic" microenvironment that enhances human neural stem cell differentiation and neurite outgrowth. Additionally, 3-D fibrous scaffolds presenting low N-cadherin-Fc further enhanced the survival of H9-NSCs compared to equivalent 2-D films. This indicates that similar biofunctionalization approaches based on N-cadherin and L1 can be translated to 3-D "transplantable" scaffolds with enhanced neurotrophic behaviors. Thus, the insights from this study have fundamental and translational impacts for neural-stem-cell-based regenerative medicine.


Subject(s)
Antigens, CD/pharmacology , Biomimetic Materials/pharmacology , Cadherins/pharmacology , Cell Differentiation/drug effects , Immunoglobulin Fc Fragments/pharmacology , Neural Cell Adhesion Molecule L1/pharmacology , Neural Stem Cells/metabolism , Neurites/metabolism , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Neural Stem Cells/cytology , Oxidative Stress/drug effects , Recombinant Fusion Proteins/pharmacology , Regenerative Medicine , Stem Cell Niche , Tissue Scaffolds/chemistry
8.
J Neurochem ; 123(4): 602-12, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22924694

ABSTRACT

Pre-natal alcohol exposure causes fetal alcohol spectrum disorders (FASD), the most common, preventable cause of developmental disability. The developing cerebellum is particularly vulnerable to the effects of ethanol. We reported that ethanol inhibits the stimulation of axon outgrowth in cerebellar granule neurons (CGN) by NAP, an active motif of activity-dependent neuroprotective protein (ADNP), by blocking NAP activation of Fyn kinase and its downstream signaling molecule, the scaffolding protein Cas. Here, we asked whether ethanol inhibits the stimulation of axon outgrowth by diverse axon guidance molecules through a common action on the Src family kinases (SFK). We first demonstrated that netrin-1, glial cell line-derived neurotrophic factor (GDNF), and neural cell adhesion molecule L1 stimulate axon outgrowth in CGNs by activating SFK, Cas, and extracellular signal-regulated kinase 1 and 2 (ERK1/2). The specific SFK inhibitor, PP2, blocked the stimulation of axon outgrowth and the activation of the SFK-Cas-ERK1/2 signaling pathway by each of these axon-guidance molecules. In contrast, brain-derived neurotrophic factor (BDNF) stimulated axon outgrowth and activated ERK1/2 without first activating SFK or Cas. Clinically relevant concentrations of ethanol inhibited axon outgrowth and the activation of the SFK-Cas-ERK1/2 pathway by netrin-1, GDNF, and L1, but did not disrupt BDNF-induced axon outgrowth or ERK1/2 activation. These results indicate that SFK, but not ERK1/2, is a primary target for ethanol inhibition of axon outgrowth. The ability of ethanol to block the convergent activation of the SFK-Cas-ERK1/2 pathway by netrin-1, GDNF, L1, and ADNP could contribute significantly to the pathogenesis of FASD.


Subject(s)
Axons/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Nerve Growth Factors/pharmacology , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/cytology , Tumor Suppressor Proteins/pharmacology , src-Family Kinases/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebellum/cytology , Chickens , Crk-Associated Substrate Protein/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Netrin-1 , Neurons/drug effects , Pyrimidines/pharmacology , Rats , Signal Transduction/drug effects
9.
Acta Biomater ; 8(10): 3561-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22750248

ABSTRACT

The spinal cord (SC) and dorsal root ganglion (DRG) are target implantation regions for neural prosthetics, but the tissue-electrode interface in these regions is not well-studied. To improve understanding of these locations, the tissue reactions around implanted electrodes were characterized. L1, an adhesion molecule shown to maintain neuronal density and reduce gliosis in brain tissue, was then evaluated in SC and DRG implants. Following L1 immobilization onto neural electrodes, the bioactivities of the coatings were verified in vitro using neuron, astrocyte and microglia cultures. Non-modified and L1-coated electrodes were implanted into adult rats for 1 or 4 weeks. Hematoxylin and eosin staining along with cell-type specific antibodies were used to characterize the tissue response. In the SC and DRG, cells aggregated at the electrode-tissue interface. Microglia staining was more intense around the implant site and decreased with distance from the interface. Neurofilament staining in both locations decreased or was absent around the implant, compared with surrounding tissue. With L1, neurofilament staining was significantly increased while neuronal cell death decreased. These results indicate that L1-modified electrodes may result in an improved chronic neural interface and will be evaluated in recording and stimulation studies.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Electrodes, Implanted , Ganglia, Spinal/pathology , Inflammation/pathology , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/pathology , Spinal Cord/pathology , Animals , Antigens, Nuclear/metabolism , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Cell Adhesion/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/enzymology , Glial Fibrillary Acidic Protein/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Staining and Labeling , Surface Properties/drug effects , Vimentin/metabolism
10.
Biomaterials ; 32(3): 681-92, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20933270

ABSTRACT

Brain tissue inflammatory responses, including neuronal loss and gliosis at the neural electrode/tissue interface, limit the recording stability and longevity of neural probes. The neural adhesion molecule L1 specifically promotes neurite outgrowth and neuronal survival. In this study, we covalently immobilized L1 on the surface of silicon-based neural probes and compared the tissue response between L1 modified and non-modified probes implanted in the rat cortex after 1, 4, and 8 weeks. The effect of L1 on neuronal health and survival, and glial cell reactions were evaluated with immunohistochemistry and quantitative image analysis. Similar to previous findings, persistent glial activation and significant decreases of neuronal and axonal densities were found at the vicinity of the non-modified probes. In contrast, the immediate area (100 µm) around the L1 modified probe showed no loss of neuronal bodies and a significantly increased axonal density relative to background. In this same region, immunohistochemistry analyses show a significantly lower activation of microglia and reaction of astrocytes around the L1 modified probes when compared to the control probes. These improvements in tissue reaction induced by the L1 coating are likely to lead to improved functionality of the implanted neural electrodes during chronic recordings.


Subject(s)
Gliosis/metabolism , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/cytology , Neurons/drug effects , Animals , Astrocytes/drug effects , Gliosis/pathology , Immunohistochemistry , Male , Microglia/drug effects , Models, Biological , Neural Cell Adhesion Molecule L1/chemistry , Rats , Rats, Sprague-Dawley
11.
Eur J Neurosci ; 32(1): 89-98, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20597970

ABSTRACT

Polysialylated neuronal cell adhesion molecule (PSA-NCAM), a polysialylated protein constitutively expressed in the hippocampus, is involved in neuronal growth, synaptic plasticity and neurotrophin signaling. In particular, PSA-NCAM mediates Ret-independent glial-derived neurotrophic factor (GDNF) signaling, leading to downstream FAK activation. GDNF has potent seizure-suppressant action, whereas PSA-NCAM is upregulated by seizure activity. However, the involvement of Ret-independent GDNF signaling in temporal lobe epilepsy (TLE) is not established. We tested the effects of PSA-NCAM inactivation on neurodegeneration and epileptogenesis in a mouse model of TLE. In this model, unilateral intrahippocampal kainic acid (KA) injection induced degeneration of CA1, CA3c and hilar neurons, followed by spontaneous recurrent focal seizures. In the contralateral, morphologically preserved hippocampus, a long-lasting increase of PSA-NCAM immunoreactivity was observed. Inactivation of PSA-NCAM by endoneuraminidase (EndoN) administration into the contralateral ventricle of KA-treated mice caused severe degeneration of CA3a,b neurons and dentate gyrus granule cells in the epileptic focus, and led to early onset of focal seizures. This striking trans-hemispheric alteration suggested that PSA-NCAM mediates GDNF signaling, leading to transport of neuroprotective signals into the lesioned hippocampus. This hypothesis was confirmed by injecting GDNF antibodies into the contralateral hippocampus of KA-treated mice, thereby reproducing the enhanced neurodegeneration seen after PSA-NCAM inactivation. Furthermore, contralateral EndoN and anti-GDNF treatment decreased GDNF family receptor alpha1 immunoreactivity and FAK phosphorylation in the epileptic focus. Thus, Ret-independent GDNF signaling across the commissural projection might protect CA3a,b neurons and delay seizure onset. These findings implicate GDNF in the control of epileptogenesis and offer a possible mechanism explaining lesion asymmetry in mesial TLE.


Subject(s)
Epilepsy, Temporal Lobe , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Nerve Degeneration , Neural Cell Adhesion Molecule L1/pharmacology , Sialic Acids/pharmacology , Signal Transduction/physiology , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/physiopathology , Excitatory Amino Acid Agonists/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glycoside Hydrolases/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Kainic Acid/pharmacology , Male , Mice , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neuroprotective Agents/pharmacology
12.
Mol Cell Neurosci ; 44(2): 118-28, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20298788

ABSTRACT

We have investigated the role of Vav2, a reported Rac1/Cdc42 GEF, on the development of Xenopus spinal neurons in vitro and in vivo. Both gain and loss of Vav2 function inhibited the rate neurite extension on laminin (LN), while only GFP-Vav2 over-expression enhanced process formation and branching. Vav2 over-expression protected neurons from RhoA-mediated growth cone collapse, similar to constitutively active Rac1, suggesting that Vav2 activates Rac1 in spinal neurons. Enhanced branching on LN required both Vav2 GEF activity and N-terminal tyrosine residues, but protection from RhoA-mediated collapse only required GEF activity. Interestingly, wild-type spinal neurons exhibited increased branching on the cell adhesion molecule L1, which required Vav2 GEF function, but not N-terminal tyrosine residues. Finally, we find that Vav2 differentially affects the Rohon-Beard peripheral and central process extension but promotes neurite branching of commissural interneurons near the ventral midline. Together, we suggest that balanced Vav2 activity is necessary for optimal neurite outgrowth and promotes branching by targeting GEF activity to branch points.


Subject(s)
Cell Differentiation/physiology , Neurites/metabolism , Neurogenesis/physiology , Proto-Oncogene Proteins c-vav/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Cells, Cultured , Growth Cones/drug effects , Growth Cones/metabolism , Growth Cones/ultrastructure , Humans , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/pharmacology , Neurites/ultrastructure , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Spinal Cord/cytology , Tyrosine/chemistry , Tyrosine/metabolism , Xenopus laevis , rac1 GTP-Binding Protein/metabolism
13.
Biomaterials ; 30(27): 4503-12, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19523683

ABSTRACT

Concentration gradients of matrix-bound guidance cues in the extracellular matrix direct cell growth in native tissues and are of great interest for the design of biomedical scaffolds and on implant surfaces. This study describes effects of covalently immobilized gradients of the 6th Ig-like domain of cell adhesion molecule L1 (TG-L1Ig6) within 3D-fibrin matrices on cell alignment. Linear gradients of TG-L1Ig6 were established and shown to be stable for at least 24 h whereas soluble gradients disappeared completely. Fibroblast alignment along the gradients was observed when cultured on top and within TG-L1Ig6-gradient matrices. Fibroblasts responded to an increase of 0.2 microg TG-L1Ig6/ml per mm matrix, corresponding to a concentration change of <1% per cell. Significant differences were observed when fibroblasts were cultured within the TG-L1Ig6-gradient matrices as the number of aligned cells decreased by 20-30% in the middle of the gradient when compared to cells cultivated on top of the gradient. This finding might be explained by approximately 13% reduction in the average cell length of fibroblasts within compared to fibroblasts cultured on top of the gradient matrix. In contrast to fibroblasts endothelial cells did not show any alignment with TG-L1Ig6-gradient matrices. The study indicates that cells exposed to gradients of matrix-bound TG-L1Ig6 are able to respond differentially to 2D- or 3D-environments suggesting the use of gradients for cell guidance within 3D-scaffolds and on implant surfaces to improve their biomedical functions.


Subject(s)
Cell Movement/drug effects , Extracellular Matrix/metabolism , Fibrin/pharmacology , Immobilized Proteins/pharmacology , Immunoglobulins/chemistry , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/pharmacology , Animals , Cattle , Cell Line , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Protein Stability/drug effects , Protein Structure, Tertiary , Recombinant Fusion Proteins/pharmacology , Serum Albumin, Bovine/metabolism , Solubility/drug effects , Transglutaminases/metabolism
14.
Neurosci Res ; 63(3): 224-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19110015

ABSTRACT

During development, asymmetric Ca(2+) signals across the growth cone mediate bidirectional axon guidance depending on intracellular levels of cyclic AMP: Ca(2+) signals trigger attractive or repulsive turning when cyclic AMP levels are high or low, respectively. Here, we report that the cell adhesion molecule L1 elevates cyclic AMP levels in neurons via ankyrin(B), a protein that links the L1 cytoplasmic tail with the spectrin network. We also show that the loss of ankyrin(B) expression converts Ca(2+)-triggered attraction to repulsion when the growth cone migrates via an L1-dependent mechanism. These results indicate that ankyrin(B) regulates axon guidance via cyclic AMP.


Subject(s)
Ankyrins/physiology , Cyclic AMP/metabolism , Growth Cones/drug effects , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/cytology , Animals , Animals, Newborn , Ankyrins/deficiency , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/metabolism , Ganglia, Spinal/cytology , Mice , Mice, Knockout , Neurons/drug effects , Protein Kinase Inhibitors/pharmacology , Thionucleotides/pharmacology
15.
Acta Biomater ; 4(5): 1208-17, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18420473

ABSTRACT

Silicon-based implantable neural electrode arrays are known to experience failure during long-term recording, partially due to host tissue responses. Surface modification and immobilization of biomolecules may provide a means to improve their biocompatibility and integration within the host brain tissue. Previously, the laminin biomolecule or laminin fragments have been used to modify the neural probe's silicon surface to promote neuronal attachment and growth. Here we report the successful immobilization of the L1 biomolecule on a silicon surface. L1 is a neuronal adhesion molecule that can specifically promote neurite outgrowth and neuronal survival. Silane chemistry and the heterobifunctional coupling agent 4-maleimidobutyric acid N-hydroxysuccinimide ester (GMBS) were used to covalently bind these two biomolecules onto the surface of silicon dioxide wafers, which mimic the surface of silicon-based implantable neural probes. After covalent binding of the biomolecules, polyethylene glycol (PEG)-NH(2) was used to cap the unreacted GMBS groups. Surface immobilization was verified by goniometry, dual polarization interferometry, and immunostaining techniques. Primary murine neurons or astrocytes were used to evaluate the modified silicon surfaces. Both L1- and laminin-modified surfaces promoted neuronal attachment, while the L1-modified surface demonstrated significantly enhanced levels of neurite outgrowth (p<0.05). In addition, the laminin-modified surface promoted astrocyte attachment, while the L1-modified surface showed significantly reduced levels of astrocyte attachment relative to the laminin-modified surface and other controls (p<0.05). These results demonstrate the ability of the L1-immobilized surface to specifically promote neuronal growth and neurite extension, while inhibiting the attachment of astrocytes, one of the main cellular components of the glial sheath. Such unique properties present vast potentials to improve the biocompatibility and chronic recording performance of neural probes.


Subject(s)
Astrocytes/physiology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Microelectrodes , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/cytology , Neurons/physiology , Adsorption , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Materials Testing , Neural Cell Adhesion Molecule L1/chemistry , Neurons/drug effects , Polyethylene Glycols/chemistry , Protein Binding , Rats , Rats, Sprague-Dawley , Silicon Dioxide/chemistry , Surface Properties
16.
Brain ; 130(Pt 4): 954-69, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17438016

ABSTRACT

Paucity of permissive molecules and abundance of inhibitory molecules in the injured spinal cord of adult mammals prevent axons from successful regeneration and, thus, contribute to the failure of functional recovery. Using an adeno-associated viral (AAV) vector, we expressed the regeneration-promoting cell adhesion molecule L1 in both neurons and glia in the lesioned spinal cord of adult mice. Exogenous L1, detectable already 1 week after thoracic spinal cord compression and immediate vector injection, was expressed at high levels up to 5 weeks, the longest time-period studied. Dissemination of L1-transduced cells throughout the spinal cord was wide, spanning over more than 10 mm rostral and 10 mm caudal to the lesion scar. L1 was not detectable in the fibronectin-positive lesion core. L1 overexpression led to improved stepping abilities and muscle coordination during ground locomotion over a 5-week observation period. Superior functional improvement was associated with enhanced reinnervation of the lumbar spinal cord by 5-HT axons. Corticospinal tract axons did not regrow beyond the lesion scar but extended distally into closer proximity to the injury site in AAV-L1-treated compared with control mice. The expression of the neurite outgrowth-inhibitory chondroitin sulphate proteoglycan NG2 was decreased in AAV-L1-treated spinal cords, along with reduction of the reactive astroglial marker GFAP. In vitro experiments confirmed that L1 inhibits astrocyte proliferation, migration, process extension and GFAP expression. Analyses of intracellular signalling indicated that exogenous L1 activates diverse cascades in neurons and glia. Thus, AAV-mediated L1 overexpression appears to be a potent means to favourably modify the local environment in the injured spinal cord and promote regeneration. Our study demonstrates a clinically feasible approach of promising potential.


Subject(s)
Adenoviridae/genetics , Nerve Regeneration/physiology , Neural Cell Adhesion Molecule L1/physiology , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Animals , Astrocytes/physiology , Axons/physiology , Cell Division/physiology , Cell Movement/physiology , Female , Gene Expression/genetics , Genetic Vectors , Mice , Mice, Inbred C57BL , Movement/physiology , Neural Cell Adhesion Molecule L1/analysis , Neural Cell Adhesion Molecule L1/pharmacology , Neuroglia/chemistry , Neuroglia/physiology , Neurons/chemistry , Neurons/physiology , Serotonin/immunology , Serotonin Agents/immunology , Signal Transduction/genetics , Signal Transduction/physiology , Spinal Cord/chemistry , Spinal Cord/physiopathology , Spinal Cord Injuries/genetics
17.
Neurotoxicology ; 28(3): 457-62, 2007 May.
Article in English | MEDLINE | ID: mdl-17267039

ABSTRACT

L1 cell adhesion molecule (L1), a protein critical for appropriate development of the central nervous system, is a target for ethanol teratogenicity. Ethanol inhibits both L1 mediated cell adhesion as well as L1 mediated neurite outgrowth. L1 has been shown to increase cell survival in cerebellar granule cells while ethanol has been shown to increase cell death. We sought to determine if L1 protected cells from ethanol induced cell death. Cerebellar granule cells from postnatal day 6 rat pups were cultured on either poly l-lysine with or without an L1 substratum. Alcohol was added at 2h post-plating and cell survival was measured at various times. L1 substratum significantly increased cell survival at 72 and 120 h. Ethanol significantly reduced cell survival at 48 h, with no effect at 72 or 120 h, both in the presence and absence of L1. At 48 h, L1 significantly increased cell survival in the presence of ethanol. We conclude that ethanol interferes with processes other than L1-L1 interactions in causing cell death, and that ethanol effects would be more severe in the absence of L1.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/drug effects , Neuroprotective Agents , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cerebellum/cytology , Cerebellum/drug effects , Humans , Mice , NIH 3T3 Cells , Neural Cell Adhesion Molecule L1/isolation & purification , Neurites/drug effects , Neurites/ultrastructure , Plasmids/genetics , Polylysine/pharmacology , Potassium Deficiency/pathology , Rats , Rats, Sprague-Dawley
18.
J Neurosci ; 26(42): 10888-109898, 2006 Oct 18.
Article in English | MEDLINE | ID: mdl-17050727

ABSTRACT

Polysialic acid (PSA) regulates functions of the neural cell adhesion molecule (NCAM) during development and in neuroplasticity in the adult; the underlying mechanisms at different phases of learning and memory consolidation are, however, unknown. To investigate the contributions of PSA versus the extracellular domain of the NCAM glycoprotein backbone to synaptic plasticity, we applied NCAM, PSA-NCAM, and PSA to acute slices of the hippocampal CA1 region of NCAM-deficient mice and measured their effects on long-term potentiation (LTP). Remarkably, only PSA and PSA-NCAM, but not NCAM restored normal LTP. Application of these molecules to the dorsal hippocampus of wild-type mice showed that PSA-NCAM and PSA, but not NCAM, injected before fear conditioning, impaired formation of hippocampus-dependent contextual memory. Consolidation of contextual memory was affected by PSA-NCAM only when injected during its late, but not early phases. None of the tested compounds disturbed extrahippocampal-cued memory. Mice lacking the polysialyltransferase (ST8SialV/PST) responsible for attachment of PSA to NCAM in adulthood showed a mild deficit only in hippocampal contextual learning, when compared with NCAM-deficient mice that were disturbed in both contextual and cued memories. Contextual and tone memory in NCAM-deficient mice could be partially restored by injection of PSA-NCAM, but not of NCAM, into the hippocampus, suggesting that the impact of PSA-NCAM in synaptic plasticity and learning is not mediated by modulation of NCAM-NCAM homophilic interactions. In conclusion, our data support the view that polysialylated NCAM is involved in both formation and late consolidation of contextual memory.


Subject(s)
Conditioning, Psychological/physiology , Fear/physiology , Long-Term Potentiation/physiology , Memory/physiology , Neural Cell Adhesion Molecule L1/physiology , Sialic Acids/physiology , Animals , Conditioning, Psychological/drug effects , Fear/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/pharmacology , Sialic Acids/deficiency , Sialic Acids/pharmacology
19.
J Biol Chem ; 281(46): 34859-69, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16987814

ABSTRACT

The neural cell adhesion molecule (NCAM) and its associated glycan polysialic acid play important roles in the development of the nervous system and N-methyl-D-aspartate(NMDA)receptor-dependent synaptic plasticity in the adult. Here, we investigated the influence of polysialic acid on NMDA receptor activity. We found that glutamate-elicited NMDA receptor currents in cultured hippocampal neurons were reduced by approximately 30% with the application of polysialic acid or polysialylated NCAM but not by the sialic acid monomer, chondroitin sulfate, or non-polysialylated NCAM. Polysialic acid inhibited NMDA receptor currents elicited by 3 microm glutamate but not by 30 microm glutamate, suggesting that polysialic acid acts as a competitive antagonist, possibly at the glutamate binding site. The polysialic acid induced effects were mimicked and fully occluded by the NR2B subunit specific antagonist, ifenprodil. Recordings from single synaptosomal NMDA receptors reconstituted in lipid bilayers revealed that polysialic acid reduced open probability but not the conductance of NR2B-containing NMDA receptors in a polysialic acid and glutamate concentration-dependent manner. The activity of single NR2B-lacking synaptosomal NMDA receptors was not affected by polysialic acid. Application of polysialic acid to hippocampal cultures reduced excitotoxic cell death induced by low micromolar concentration of glutamate via activation of NR2B-containing NMDA receptors, whereas enzymatic removal of polysialic acid resulted in increased cell death that occluded glutamate-induced excitotoxicity. These observations indicate that the cell adhesion molecule-associated glycan polysialic acid is able to prevent excitotoxicity via inhibition of NR2B subunit-containing NMDA receptors.


Subject(s)
Glutamic Acid/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/pharmacology , Neurons/cytology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sialic Acids/metabolism , Sialic Acids/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Hippocampus/cytology , Mice , Neural Cell Adhesion Molecule L1/genetics , Polysaccharides/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Sialic Acids/genetics
20.
Exp Neurol ; 202(1): 238-49, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16860320

ABSTRACT

Overexpression of nerve growth factor (NGF) using adenoviruses (Adts) after spinal cord injury induces extensive regeneration and sprouting of calcitonin-gene-related peptide immunoreactive (CGRP-IR) fibers, whereas overexpression of cell adhesion molecules (CAMs) has no effect on the normal distribution of these fibers. Interestingly, co-expression of cell adhesion molecule L1 and NGF significantly decreases (p<0.0001) CGRP-IR fiber sprouting within the spinal cord, when compared to NGF alone. Co-expression of cell adhesion molecules NCAM or N-cadherin had no effect on NGF-induced CGRP-IR fiber sprouting. These data demonstrate that reduced sprouting is specific to L1 co-expression and not other cell adhesion molecules. In vitro studies carried out to address potential mechanisms show that neurite outgrowth over astrocytes overexpressing L1 in the presence of NGF is comparable to controls, indicating that other factors present in vivo might be involved in the L1-mediated reduction in sprouting. One potential factor is semaphorin 3A (sema3A), which mediates growth cone collapse of CGRP-positive axons. Recent studies have shown that L1 is important in sema3A receptor signaling for cortical neurons. In our study, co-expression of sema3A indeed reduces neurite outgrowth from DRG neurons by about 40% on L1-expressing astrocytes. Based on these results, we hypothesize that overexpression of L1 potentiates sema3A signaling resulting in reduced sprouting.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Gene Expression Regulation/drug effects , Nerve Fibers/drug effects , Nerve Growth Factor/pharmacology , Neural Cell Adhesion Molecule L1/pharmacology , Spinal Cord Injuries , Adenoviridae/physiology , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Contactin 2 , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Ganglia, Spinal/pathology , Gene Expression Regulation/physiology , Nerve Fibers/physiology , Neurites/drug effects , Neurites/physiology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...