Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell ; 174(6): 1450-1464.e23, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100184

ABSTRACT

Synapses are fundamental units of communication in the brain. The prototypical synapse-organizing complex neurexin-neuroligin mediates synapse development and function and is central to a shared genetic risk pathway in autism and schizophrenia. Neurexin's role in synapse development is thought to be mediated purely by its protein domains, but we reveal a requirement for a rare glycan modification. Mice lacking heparan sulfate (HS) on neurexin-1 show reduced survival, as well as structural and functional deficits at central synapses. HS directly binds postsynaptic partners neuroligins and LRRTMs, revealing a dual binding mode involving intrinsic glycan and protein domains for canonical synapse-organizing complexes. Neurexin HS chains also bind novel ligands, potentially expanding the neurexin interactome to hundreds of HS-binding proteins. Because HS structure is heterogeneous, our findings indicate an additional dimension to neurexin diversity, provide a molecular basis for fine-tuning synaptic function, and open therapeutic directions targeting glycan-binding motifs critical for brain development.


Subject(s)
Heparitin Sulfate/metabolism , Neural Cell Adhesion Molecules/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Glycopeptides/analysis , Heparitin Sulfate/chemistry , Humans , Membrane Proteins , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Neurons/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Rats , Sequence Alignment
2.
Anal Biochem ; 587: 113463, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31574254

ABSTRACT

Synaptic adhesion molecules, including presynaptic neurexins (NRXNs) and post-synaptic leucine-rich repeat transmembrane (LRRTM) proteins are important for development and maintenance of brain neuronal networks. NRXNs are probably the best characterized synaptic adhesion molecules, and one of the major presynaptic organizer proteins. The LRRTMs were found as ligands for NRXNs. Many of the synaptic adhesion proteins have been linked to neurological cognitive disorders, such as schizophrenia and autism spectrum disorders, making them targets of interest for both biological studies, and towards drug development. Therefore, we decided to develop a screening method to target the adhesion proteins, here the LRRTM-NRXN interaction, to find small molecule probes for further studies in cellular settings. To our knowledge, no potent small molecule compounds against the neuronal synaptic adhesion proteins are available. We utilized the AlphaScreen technology, and developed an assay targeting the NRXN-LRRTM2 interaction. We carried out screening of 2000 compounds and identified hits with moderate IC50-values. We also established an orthogonal in-cell Western blot assay to validate hits. This paves way for future development of specific high affinity compounds by further high throughput screening of larger compound libraries using the methods established here. The method could also be applied to screening other NRXN-ligand interactions.


Subject(s)
Calcium-Binding Proteins/antagonists & inhibitors , Nervous System Diseases/metabolism , Neural Cell Adhesion Molecules/antagonists & inhibitors , Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Drosophila , Leucine-Rich Repeat Proteins , Mice , Models, Molecular , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Proteins/chemistry , Proteins/metabolism
3.
J Biol Chem ; 289(29): 19958-75, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24904058

ABSTRACT

The Amigo protein family consists of three transmembrane proteins characterized by six leucine-rich repeat domains and one immunoglobulin-like domain in their extracellular moieties. Previous in vitro studies have suggested a role as homophilic adhesion molecules in brain neurons, but the in vivo functions remain unknown. Here we have cloned all three zebrafish amigos and show that amigo1 is the predominant family member expressed during nervous system development in zebrafish. Knockdown of amigo1 expression using morpholino oligonucleotides impairs the formation of fasciculated tracts in early fiber scaffolds of brain. A similar defect in fiber tract development is caused by mRNA-mediated expression of the Amigo1 ectodomain that inhibits adhesion mediated by the full-length protein. Analysis of differentiated neural circuits reveals defects in the catecholaminergic system. At the behavioral level, the disturbed formation of neural circuitry is reflected in enhanced locomotor activity and in the inability of the larvae to perform normal escape responses. We suggest that Amigo1 is essential for the development of neural circuits of zebrafish, where its mechanism involves homophilic interactions within the developing fiber tracts and regulation of the Kv2.1 potassium channel to form functional neural circuitry that controls locomotion.


Subject(s)
Brain/growth & development , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Zebrafish/metabolism , Animals , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/genetics , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Larva/growth & development , Larva/metabolism , Male , Nerve Net/growth & development , Nerve Net/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
4.
Nucleic Acids Res ; 40(20): 10356-65, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22917588

ABSTRACT

The regulation of Schwann cell (SC) responses to injury stimuli by microRNAs (miRNAs) remains to be explored. Here, we identified 17 miRNAs that showed dynamic expression alterations at five early time points following rat sciatic nerve resection. Then we analyzed the expression pattern of 17 miRNAs, and integrated their putative targets with differentially expressed mRNAs. The resulting 222 potential targets were mainly involved in cell phenotype modulation, including immune response, cell death and cell locomotion. Among 17 miRNAs, miR-182 expression was up-regulated. The enhanced expression of miR-182 was correlated with nerve injury-induced phenotype modulation of SCs. Further investigation revealed that fibroblast growth factor 9 (FGF9) and neurotrimin (NTM) were two direct targets of miR-182 in SCs, with miR-182 binding to the 3'-untranslated region of FGF9 and NTM. Silencing of FGF9 and NTM recapitulated the inhibiting effect of miR-182 mimics on SC proliferation and migration, respectively, whereas enforced knockdown of FGF9 and NTM reversed the promoting effect of miR-182 inhibitor on SC proliferation and migration, respectively. Our data indicate that nerve injury inhibits SC proliferation and migration through rapid regulation of miR-182 by targeting FGF9 and NTM, providing novel insights into the roles of miRNAs in nerve injury and repair.


Subject(s)
Fibroblast Growth Factor 9/genetics , MicroRNAs/metabolism , Neural Cell Adhesion Molecules/genetics , Schwann Cells/metabolism , Sciatic Nerve/injuries , 3' Untranslated Regions , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Down-Regulation , Fibroblast Growth Factor 9/antagonists & inhibitors , Fibroblast Growth Factor 9/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Profiling , Male , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/metabolism , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Schwann Cells/physiology
5.
J Neurochem ; 125(5): 766-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23495921

ABSTRACT

Multiple sclerosis (MS) is a CNS disorder characterized by demyelination and neurodegeneration. Although hallmarks of recovery (remyelination and repair) have been documented in early MS, the regenerative capacity of the adult CNS per se remains uncertain with the wide held belief that it is either limited or non-existent. The neural cell adhesion molecule (NCAM) is a cell adhesion molecule that has been widely implicated in axonal outgrowth, guidance and fasciculation. Here, we used in vitro and in vivo of MS to investigate the role of NCAM in disease progression. We show that in health NCAM levels decrease over time, but this occurs acutely after demyelination and remains reduced in chronic disease. Our findings suggest that depletion of NCAM is one of the factors associated with or possibly responsible for disease progression in MS.


Subject(s)
Disease Progression , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/cerebrospinal fluid , Adult , Amino Acid Sequence , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , Multiple Sclerosis/cerebrospinal fluid , Neural Cell Adhesion Molecules/biosynthesis , Pregnancy , Rats , Rats, Sprague-Dawley , Young Adult
6.
Methods ; 56(3): 338-50, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22037249

ABSTRACT

Intrabodies are recombinantly expressed intracellular antibody fragments that can be used to specifically bind and inhibit the function of cellular proteins of interest. Intrabodies can be targeted to various cell compartments by attaching an appropriate localization peptide sequence to them. An efficient strategy with a high success rate is to anchor intrabodies in the endoplasmatic reticulum where they can inhibit transitory target proteins by binding and preventing them to reach their site of action. Intrabodies can be assembled from antibody gene fragments from various sources into dedicated expression vectors. Conventionally, antibody cDNA sequences are derived from selected hybridoma cell clones that express antibodies with the desired specificity. Alternatively, appropriate clones can be isolated by affinity selection from an antibody in vitro display library. Here an evaluation of endoplasmatic reticulum targeted intrabodies with respect to other knockdown approaches is given and the characteristics of various intrabody expression vectors are discussed. A step by step protocol is provided that was repeatedly used to construct intrabodies derived from diverse antibody isotypes producing hybridoma cell clones. The inactivation of the cell surface receptor neural cell adhesion molecule (NCAM) by a highly efficacious novel endoplasmatic reticulum-anchored intrabody is demonstrated.


Subject(s)
Antibodies/metabolism , Endoplasmic Reticulum/metabolism , Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Base Sequence , Gene Transfer Techniques , Genetic Vectors , Homologous Recombination , Humans , Molecular Sequence Data , Mutation , Neural Cell Adhesion Molecules/antagonists & inhibitors , Peptide Library , Proteins/physiology , RNA, Antisense
7.
Anticancer Drugs ; 23(9): 970-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22713592

ABSTRACT

Zoledronic acid (ZOL), a nitrogen-containing compound, is effective in the treatment of skeletal disorders, but its long-term use in high doses gives rise to complications such as osteonecrosis. We aimed to investigate the effect of low-dose ZOL on the expression of the neural cell adhesion molecule (NCAM), which may be correlated with tumor growth and spinal cord metastasis in lung adenocarcinoma with neuroendocrine differentiation. First, we used the small hairpin RNA technique to directly knock down NCAM expression in cells of a murine lung adenocarcinoma line, line 1 cells, and found that the tumor cells generated showed lower invasive capacity, slower tumor growth, and lesser tendency for spinal cord metastasis than control cells. Further, ZOL decreased NCAM expression and invasiveness in line 1 tumor cells in vitro. Line 1/lacZ cells, a stable clone tagged with the lacZ gene, were introduced into mice, followed by ZOL treatment (1 µg/kg/weekly). Low-dose ZOL significantly reduced spinal cord metastasis probably through reduced NCAM expression in vivo. These findings indicated that NCAM is involved in tumor growth and spinal cord metastasis of lung adenocarcinoma with neuroendocrine differentiation. Treatment with low-dose ZOL can reduce NCAM expression that may contribute toward reduced spinal cord metastasis, suggesting that NCAM is an alternative therapeutic target and that the low-dose ZOL treatment protocol is a reasonable approach for its treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Lung Neoplasms/drug therapy , Neural Cell Adhesion Molecules/antagonists & inhibitors , Spinal Cord Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/secondary , Cell Differentiation , Cell Line, Tumor , Cloning, Molecular , Diphosphonates/administration & dosage , Diphosphonates/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Imidazoles/administration & dosage , Imidazoles/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , RNA, Small Interfering/genetics , Spinal Cord Neoplasms/metabolism , Spinal Cord Neoplasms/secondary , Transfection , Zoledronic Acid
8.
Cereb Cortex ; 21(10): 2217-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21339376

ABSTRACT

Mechanisms inducing perforation of the postsynaptic density (PSD) are poorly understood. We show that neural cell adhesion molecule- deficient (NCAM-/-) hippocampal neurons have an abnormally high percentage of synapses with perforated PSDs. The percentage of synapses with perforated PSDs is also increased in wild-type (NCAM+/+) neurons after the disruption of the NCAM/spectrin complex indicating that the NCAM-assembled spectrin cytoskeleton maintains the structural integrity of PSDs. We demonstrate that PSD perforations contain endocytic zones involved in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) internalization. Induction of long-term potentiation in NCAM+/+ neurons accompanied by insertion of AMPAR into the neuronal cell surface is subsequently followed by formation of perforated synapses and AMPAR endocytosis suggesting that perforation of PSDs is important for membrane homeostasis in activated synapses. In NCAM-/- or NCAM+/+ neurons with dissociated spectrin meshwork, AMPAR endocytosis is enhanced under conditions of basal activity. An abnormally high rate of postsynaptic membrane endocytosis may thus contribute to brain pathologies associated with mutations in NCAM or spectrin.


Subject(s)
Endocytosis , Neural Cell Adhesion Molecules/antagonists & inhibitors , Post-Synaptic Density/pathology , Spectrin/antagonists & inhibitors , Synapses/pathology , Animals , Cells, Cultured , Endocytosis/physiology , Hippocampus/pathology , Hippocampus/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/physiology , Post-Synaptic Density/ultrastructure , Protein Multimerization/physiology , Spectrin/physiology , Synapses/ultrastructure , Synaptic Potentials/physiology
9.
Medicina (Kaunas) ; 47(10): 552-9, 2011.
Article in English | MEDLINE | ID: mdl-22186119

ABSTRACT

BACKGROUND: Mildronate (3-[2,2,2-trimethylhydrazinium] propionate dihydrate) traditionally is a well-known cardioprotective drug. However, our recent studies convincingly demonstrated its neuroprotective properties. The aim of the present study was to evaluate the influence of mildronate on the expression of proteins that are involved in the differentiation and survival of the nigrostriatal dopaminergic neurons in the rat model of Parkinson's disease (PD). The following biomarkers were used: heat shock protein 70 (Hsp70, a molecular chaperone), glial cell line-derived nerve growth factor (GDNF, a growth factor promoting neuronal differentiation, regeneration, and survival), and neural cell adhesion molecule (NCAM). MATERIAL AND METHODS: PD was modeled by 6-hydroxydopamine (6-OHDA) unilateral intrastriatal injection in rats. Mildronate was administered at doses of 10, 20, and 50 mg/kg for 2 weeks intraperitoneally before 6-OHDA injection. Rat brains were dissected on day 28 after discontinuation of mildronate injections. The expression of biomarkers was assessed immunohistochemically and by western blot assay. RESULTS: 6-OHDA decreased the expression of Hsp70 and GDNF in the lesioned striatum and substantia nigra, whereas in mildronate-pretreated (20 and 50 mg/kg) rats, the expression of Hsp70 and GDNF was close to the control group values. NCAM expression also was decreased by 6-OHDA in the striatum and it was totally protected by mildronate at a dose of 50 mg/kg. In contrast, in the substantia nigra, 6-OHDA increased the expression of NCAM, while mildronate pretreatment (20 and 50 mg/kg) reversed the 6-OHDA-induced overexpression of NCAM close to the control values. CONCLUSION: The obtained data showed that mildronate was capable to regulate the expression of proteins that play a role in the homeostasis of neuro-glial processes.


Subject(s)
Cardiovascular Agents/administration & dosage , Methylhydrazines/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease, Secondary/drug therapy , Protein Biosynthesis/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , Male , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/biosynthesis , Oxidopamine/antagonists & inhibitors , Oxidopamine/pharmacology , Parkinson Disease, Secondary/metabolism , Rats , Rats, Wistar , Substantia Nigra/drug effects , Substantia Nigra/metabolism
10.
Comput Math Methods Med ; 2021: 4856820, 2021.
Article in English | MEDLINE | ID: mdl-34158819

ABSTRACT

Thyroid cancer (TC) is the most common endocrine malignant disease with a rising morbidity year by year. Accumulating studies have shown that microRNAs (miRNAs) play a regulatory role in the progression of various tumors, but the molecular regulatory mechanism of miR-196a-2 in TC is still unknown. qRT-PCR was employed to measure the expression of miR-196a-2 and NRXN1 mRNA in TC cells, while western blot was used to detect the protein expression of NRXN1. CCK-8, colony formation and flow cytometry assays were used to measure cell proliferation and apoptosis of TC cells. Dual-luciferase reporter gene assay was used to predict and verify the targeted binding relationship between miR-196a-2 and NRXN1. Our study results manifested that miR-196a-2 was dramatically overexpressed in cells of TC, while NRXN1 was lowly expressed. miR-196a-2 could promote cell proliferation and inhibit cell apoptosis of TC. Additionally, miR-196a-2 could also target and inhibit the expression of NRXN1. Silencing NRXN1 could reverse the inhibitory effect of miR-196a-2 downregulation on cell proliferation of TC, as well as the promoting effect on cell apoptosis. In a conclusion, we found that miR-196a-2 could promote cell proliferation and inhibit cell apoptosis of TC by targeting NRXN1. Therefore, miR-196a-2/NRXN1 is potential to be a molecular therapeutic target for TC.


Subject(s)
Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , MicroRNAs/genetics , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , Thyroid Neoplasms/genetics , Apoptosis/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Computational Biology , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , MicroRNAs/antagonists & inhibitors , Neural Cell Adhesion Molecules/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Stem Cell Assay , Up-Regulation
11.
J Cell Biol ; 155(4): 661-73, 2001 Nov 12.
Article in English | MEDLINE | ID: mdl-11706054

ABSTRACT

The L1 adhesion molecule plays an important role in axon guidance and cell migration in the nervous system. L1 is also expressed by many human carcinomas. In addition to cell surface expression, the L1 ectodomain can be released by a metalloproteinase, but the biological function of this process is unknown. Here we demonstrate that membrane-proximal cleavage of L1 can be detected in tumors and in the developing mouse brain. The shedding of L1 involved a disintegrin and metalloproteinase (ADAM)10, as transfection with dominant-negative ADAM10 completely abolishes L1 release. L1-transfected CHO cells (L1-CHO) showed enhanced haptotactic migration on fibronectin and laminin, which was blocked by antibodies to alpha v beta 5 and L1. Migration of L1-CHO cells, but not the basal migration of CHO cells, was blocked by a metalloproteinase inhibitor, indicating a role for L1 shedding in the migration process. CHO and metalloproteinase-inhibited L1-CHO cells were stimulated to migrate by soluble L1-Fc protein. The induction of migration was blocked by alpha v beta 5-specific antibodies and required Arg-Gly-Asp sites in L1. A 150-kD L1 fragment released by plasmin could also stimulate CHO cell migration. We propose that ectodomain-released L1 promotes migration by autocrine/paracrine stimulation via alpha v beta 5. This regulatory loop could be relevant for migratory processes under physiological and pathophysiological conditions.


Subject(s)
Cell Movement/physiology , Integrins/metabolism , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Receptors, Vitronectin , Amyloid Precursor Protein Secretases , Animals , Antibodies, Monoclonal/immunology , Aspartic Acid Endopeptidases , Autocrine Communication , Binding Sites , Biological Transport , Brain/embryology , Brain/metabolism , CHO Cells , Cell Membrane/metabolism , Cricetinae , Cytoplasm/metabolism , Endopeptidases/metabolism , Fibrinolysin/metabolism , Gene Expression , Humans , Integrins/immunology , Leukocyte L1 Antigen Complex , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , Oligopeptides/metabolism , Peptide Fragments/metabolism , Solubility , Tumor Cells, Cultured
12.
J Neurochem ; 103(4): 1396-407, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17854387

ABSTRACT

A combinatorial library of undecapeptides was produced and utilized for the isolation of peptide binding to the fibronectin type 3 modules (F3I-F3II) of the neural cell adhesion molecule (NCAM). The isolated peptides were sequenced and produced as dendrimers. Two of the peptides (denoted ENFIN2 and ENFIN11) were confirmed to bind to F3I-F3II of NCAM by surface plasmon resonance. The peptides induced neurite outgrowth in primary cerebellar neurons and PC12E2 cells, but had no apparent neuroprotective properties. NCAM is known to activate different intracellular pathways, including signaling through the fibroblast growth factor receptor, the Src-related non-receptor tyrosine kinase Fyn, and heterotrimeric G-proteins. Interestingly, neurite outgrowth stimulated by ENFIN2 and ENFIN11 was independent of signaling through fibroblast growth factor receptor and Fyn, but could be inhibited with pertussis toxin, an inhibitor of certain heterotrimeric G-proteins. Neurite outgrowth induced by trans-homophilic NCAM was unaffected by the peptides, whereas knockdown of NCAM completely abrogated ENFIN2- and ENFIN11-induced neuritogenesis. These observations suggest that ENFIN2 and ENFIN11 induce neurite outgrowth in an NCAM-dependent manner through G-protein-coupled signal transduction pathways. Thus, ENFIN2 and ENFIN11 may be valuable for exploring this particular type of NCAM-mediated signaling.


Subject(s)
Neural Cell Adhesion Molecules/metabolism , Neurites/physiology , Peptides/physiology , Signal Transduction/physiology , Animals , Cell Proliferation , Cells, Cultured , Cerebellum/growth & development , Cerebellum/metabolism , Cerebellum/physiology , Humans , Mice , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , Neurites/metabolism , PC12 Cells , Peptides/genetics , Peptides/metabolism , Protein Binding/physiology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/physiology , Toxoids/pharmacology
13.
PLoS One ; 11(5): e0155384, 2016.
Article in English | MEDLINE | ID: mdl-27163287

ABSTRACT

In the Drosophila olfactory system, odorant information is sensed by olfactory sensory neurons and relayed from the primary olfactory center, the antennal lobe (AL), to higher olfactory centers via olfactory projection neurons (PNs). A major portion of the AL is constituted with dendrites of four groups of PNs, anterodorsal PNs (adPNs), lateral PNs (lPNs), lateroventral PNs (lvPNs) and ventral PNs (vPNs). Previous studies have been focused on the development and function of adPNs and lPNs, while the investigation on those of lvPNs and vPNs received less attention. Here, we study the molecular and cellular mechanisms underlying the morphogenesis of a putative male-pheromone responding vPN, the DA1 vPN. Using an intersection strategy to remove background neurons labeled within a DA1 vPN-containing GAL4 line, we depicted morphological changes of the DA1 vPN that occurs at the pupal stage. We then conducted a pilot screen using RNA interference knock-down approach to identify cell surface molecules, including Down syndrome cell adhesion molecule 1 and Semaphorin-1a, that might play essential roles for the DA1 vPN morphogenesis. Taken together, by revealing molecular and cellular basis of the DA1 vPN morphogenesis, we should provide insights into future comprehension of how vPNs are assembled into the olfactory neural circuitry.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Morphogenesis/genetics , Olfactory Pathways/metabolism , Olfactory Receptor Neurons/metabolism , Sex Attractants/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion Molecules , Dendrites/metabolism , Dendrites/ultrastructure , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Male , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Olfactory Pathways/ultrastructure , Olfactory Receptor Neurons/ultrastructure , Pupa/anatomy & histology , Pupa/genetics , Pupa/growth & development , Pupa/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Semaphorins/antagonists & inhibitors , Semaphorins/genetics , Semaphorins/metabolism , Sex Attractants/genetics , Signal Transduction , Smell/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Neuroimmunol ; 170(1-2): 41-8, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16213598

ABSTRACT

MG is an antibody-mediated disease that is often treated with corticosteroids. Antibodies to the muscle specific tyrosine kinase (MuSK) have been identified in a proportion of patients with myasthenia gravis (MG) without acetylcholine receptor (AChR) antibodies. MuSK-MG patients often suffer from marked facial muscle weakness, and some patients develop facial and tongue muscle atrophy. MuSK is a receptor tyrosine kinase that plays an essential role during development and is thought to play a trophic role in mature muscle. It is possible, therefore, that the muscle atrophy results from the action of the MuSK antibodies themselves, but effects of corticosteroids on muscle might also be involved. Muscle atrophy in vivo is associated with upregulation of striated Muscle RING-Finger protein-1 (MURF-1), and MURF-1 is also upregulated in C2C12 myotubes exposed to the corticosteroid, dexamethasone (Dex). Here we investigated the effects of MuSK antibodies or Dex on MURF-1 expression in C2C12 cultures and in mouse muscles after treatment in vivo, using quantitative Western blotting. We also looked at expression of neural cell adhesion molecule (NCAM, CD56) that is upregulated after denervation in vivo. MuSK-MG plasma and purified IgG from a patient with marked muscle atrophy modestly increased MURF-1 expression in C2C12 cells in culture, and MURF-1 expression in mouse masseter (facial) muscle, but not in gastrocnemius (leg). Dex had a more marked effect on MURF-1 expression in C2C12 cells, but did not affect MURF-1 expression in either muscle. However, both in C2C12 cells and in vivo, Dex substantially reduced NCAM expression. These results provide the first evidence that MuSK-MG plasma can influence expression of an atrophy-related protein, and preliminary evidence that a facial muscle, the masseter, is more susceptible to this effect. They indicate the need for further studies on muscle atrophy, MuSK-MG antibodies, the effects of steroids, and the intracellular pathways involved.


Subject(s)
Autoantibodies/blood , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myasthenia Gravis/blood , Myasthenia Gravis/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , Cell Line , Dexamethasone/pharmacology , Humans , Immunoglobulin G/pharmacology , Masseter Muscle/metabolism , Mice , Mice, Inbred C57BL , Muscular Atrophy/immunology , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/metabolism , Tripartite Motif Proteins
15.
Dev Cell ; 35(1): 93-106, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26460947

ABSTRACT

Precise positioning of dendritic branches is a critical step in the establishment of neuronal circuitry. However, there is limited knowledge on how environmental cues translate into dendrite initiation or branching at a specific position. Here, through a combination of mutation, RNAi, and imaging experiments, we found that a Dscam-Dock-Pak1 hierarchical interaction defines the stereotypical dendrite growth site in the Drosophila aCC motoneuron. This interaction localizes the Cdc42 effector Pak1 to the plasma membrane at the dendrite initiation site before the activation of Cdc42. Ectopic expression of membrane-anchored Pak1 overrides this spatial specification of dendritogenesis, confirming its function in guiding Cdc42 signaling. We further discovered that Dscam1 localization in aCC occurs through an inter-neuronal contact that involves Dscam1 in the partner MP1 neuron. These findings elucidate a mechanism by which Dscam1 controls neuronal morphogenesis through spatial regulation of Cdc42 signaling and, subsequently, cytoskeletal remodeling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dendrites/physiology , Drosophila Proteins/metabolism , GTP-Binding Proteins/metabolism , Motor Neurons/cytology , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , p21-Activated Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Cell Adhesion Molecules , Cell Differentiation , Cell Membrane/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , GTP-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Immunoenzyme Techniques , Interneurons/cytology , Interneurons/metabolism , Morphogenesis/physiology , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , RNA, Small Interfering/genetics , p21-Activated Kinases/genetics
16.
Neuroscience ; 108(1): 7-15, 2001.
Article in English | MEDLINE | ID: mdl-11738127

ABSTRACT

We have investigated the mechanisms regulating the expression of the mu-opioid receptor, using P19 mouse embryonal carcinoma cells, which normally lack this receptor, but which can be induced to express it in aggregated cells by retinoic acid treatment. The expression level of mu-opioid receptor mRNA was found to be closely correlated with aggregation status, and more specifically by cell to cell interaction requiring neural cell adhesion molecules (NCAM). We showed that NCAM activates the mu-opioid receptor gene through a pathway involving phospholipase C-arachidonic acid-calcium channel-calcium/calmodulin kinase II. A similar pathway was previously shown to promote neurite outgrowth, however, with distinct specificity, including the role of calcium channels. Activation of L-type calcium channels elevated mu-opioid receptor expression, while N-type-channel activity had the opposite effect. The effect of anti-NCAM-antibody treatment was not due to retardation of general neural development and was specific to the mu-opioid receptor gene. Our results indicate that the P19 system is an useful model to study the expression of the mu-opioid receptor gene.


Subject(s)
Gene Expression/physiology , Neural Cell Adhesion Molecules/physiology , Receptors, Opioid, mu/genetics , Animals , Arachidonic Acids , Calcium Channels, L-Type/physiology , Calcium Channels, N-Type/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Aggregation/physiology , Glutamate Decarboxylase/genetics , Mice , Neural Cell Adhesion Molecules/antagonists & inhibitors , Time Factors , Tumor Cells, Cultured , Type C Phospholipases/metabolism
17.
Neurotoxicology ; 22(5): 625-33, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11770884

ABSTRACT

A major mechanism guiding neural development is through cell-cell and cell-matrix adhesions and signaling mediated by cell adhesion molecules (CAMs). The majority of CAMs have been grouped into three families: the cadherins, the integrins and the members of the immunoglobulin superfamily including L1. While the elucidation of new receptors and matrix components has become a frequent occurrence, the elucidation of the mechanisms by which they operate, and the function of those mechanisms in complex developmental events remains rudimentary. Members of all three families participate in differential adhesion, signal transduction and physical/mechanical effects. Each of these modes of action is a potential target for developmental neurotoxicants. In this brief review, the role of L1 in normal and abnormal neurodevelopment will be summarized. L1 is a cell surface transmembrane glycoprotein with a single copy gene on the X chromosome. There are two alternatively spliced exons, with the RSLE containing form found only on axons and growth cones of post-mitotic neurons. L1 mediates the following functions: adhesion, neurite extension, neuronal migration, and axon fasciculation. L1 is critical for normal neural development; humans with genetic defects in L1, termed corpus callosum hypoplasia, mental retardation, adducted thumbs, spasticity and hydrocephalus (CRASH) syndrome, and mice lacking expression of L1 have extensive neuropathologic and aberrant behaviors. The observation that patients with fetal alcohol syndrome share similar features to patients with CRASH has lead to the investigation of the effects of ethanol on L1. Physiologic concentrations of ethanol have been shown to inhibit L1 mediated neurite outgrowth in cerebellar granule neurons. Such inhibition may result from decreased expression, altered cell surface distribution, impaired signal transduction, or impaired interaction with the cytoskeleton. These data indicate that L1 and its associated signaling pathways are potentially targets for developmental neurotoxicants.


Subject(s)
Central Nervous System Depressants/toxicity , Developmental Disabilities/chemically induced , Ethanol/toxicity , Fetal Alcohol Spectrum Disorders/metabolism , Membrane Glycoproteins/physiology , Nervous System/drug effects , Nervous System/growth & development , Neural Cell Adhesion Molecules/physiology , Signal Transduction/drug effects , Animals , Child , Developmental Disabilities/metabolism , Female , Humans , Leukocyte L1 Antigen Complex , Membrane Glycoproteins/antagonists & inhibitors , Neural Cell Adhesion Molecules/antagonists & inhibitors , Pregnancy , Signal Transduction/physiology
18.
J Neural Eng ; 8(4): 046004, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21628769

ABSTRACT

In this study neural (N)-cadherin, neural cell adhesion molecule (N-CAM) and L1 proteins and their antibody equivalents were covalently immobilized on a polyethylene-imine (PEI)-coated glass surface to form neuron-adhesive coatings. Impedance sensing and (supplementary) image analysis were used to monitor the effects of these CAMs. Immobilization of high concentrations of both N-cadherin protein and antibody led to good adhesion of neurons to the modified surface, better than surfaces treated with 30.0 and 100.0 µg ml(-1) N-CAM protein and antibody. L1 antibody and protein coating revealed no significant effect on neuronal cell-substrate adhesion. In a second series of combinatorial experiments, we used the same antibodies and proteins as medium-additives to inhibit cell-cell adhesion between neurons. Adhesion of neurons cultured on N-cadherin protein or antibody-modified surfaces was lowered by the addition of a soluble N-cadherin protein and antibody to the culturing medium, accelerating neuronal aggregation. The presence of a soluble N-CAM antibody or protein had no effect on the adhesion of neuronal cells on a N-cadherin protein-modified surface. On a N-cadherin antibody-coated surface, the addition of a soluble N-CAM protein led to cell death of neurons after 48 h, while a N-CAM antibody had no effect. In the presence of a soluble N-cadherin protein and antibody the aggregation of neurons was inhibited, both on N-CAM protein and N-CAM antibody-modified surfaces. Neurons cultured on immobilized antibodies were less affected by the addition of soluble CAM blockers than neurons cultured on immobilized proteins, indicating that antibody-protein bonds are more stable compared to protein-protein bonds.


Subject(s)
Cadherins/physiology , Cell Adhesion/physiology , Leukocyte L1 Antigen Complex/physiology , Neural Cell Adhesion Molecules/physiology , Neurons/physiology , Animals , Animals, Newborn , Antibodies, Blocking/pharmacology , Cadherins/antagonists & inhibitors , Cadherins/immunology , Cell Adhesion/drug effects , Cells, Cultured , Electric Impedance , Electrodes , Leukocyte L1 Antigen Complex/immunology , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/immunology , Neurons/drug effects , Rats , Surface Properties
19.
Neurotoxicology ; 32(2): 255-60, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21182862

ABSTRACT

Toxic lead (Pb) exposure poses serious risks to human health, especially to children at developmental stages, even at low exposure levels. Neural cell adhesion molecule (NCAM) is considered to be a potential early target in the neurotoxicity of Pb due to its role in cell adhesion, neuronal migration, synaptic plasticity, and learning and memory. However, the effect of low-level Pb exposure on the specific expression of NCAM isoforms has not been reported. In the present study, we found that Pb could concentration-dependently (1-100 nM) inhibit the expression of three major NCAM isoforms (NCAM-180, -140, and -120) in primary cultured hippocampal neurons. Furthermore, it was verified that levels of all three major isoforms of NCAM were reduced by Pb exposure in human embryonic kidney (HEK)-293 cells transiently transfected with NCAM-120, -140, or -180 isoform cDNA constructs. In addition, low-level Pb exposure delayed the neurite outgrowth and reduced the survival rate of cultured hippocampal neurons at different time-points. Together, our results demonstrate that developmental low-level Pb exposure can attenuate the expression of all three major NCAM isoforms, which may contribute to the observed Pb-mediated neurotoxicity.


Subject(s)
Gene Expression Regulation/drug effects , Lead/administration & dosage , Lead/toxicity , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/biosynthesis , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Gene Expression Regulation/physiology , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Neurons/drug effects , Neurons/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/biosynthesis , Rats , Rats, Wistar
20.
Stem Cells ; 23(9): 1389-99, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16051987

ABSTRACT

To clarify mechanisms underlying cell-to-cell interactions between hemopoietic stem cells (HSCs) and stromal cells, we established a stromal cell line (FMS/PA6-P) from day-16 fetal bone marrow (BM) adherent cells using an anti-PA6 monoclonal antibody (mAb) specific for BM stromal cells. Importantly, this FMS/PA6-P cell line, showing homogenous fibroblastic morphology, is absent from hematolymphoid and endothelial lineage markers and maintains a high level of expression of PA6 molecule, recognized by the anti-PA6 mAb, for approximately 20 passages. Further, the cell line expressing a high level of PA6 molecule has a better hemopoiesis-supporting capacity in vitro than other stromal cell lines such as PA6 and MS-5. In fact, the PA6 molecule is closely related to the hemopoiesis-supporting capacity of the stromal cells because the proliferation of HSCs was suppressed to a great extent by the anti-PA6 mAb. Affinity chromatography and mass peptide fingerprinting revealed that the protein reacting with the anti-PA6 mAb is neural cell adhesion molecule (NCAM). The frequencies of long-term cobblestone area-forming cells and long-term culture-initiating cells were significantly suppressed by repression of NCAM in the FMS/PA6-P cells using NCAM small interfering RNA. Our findings clearly indicate that NCAM functions on the maintenance of HSCs.


Subject(s)
Cell Communication/physiology , Hematopoiesis/physiology , Neural Cell Adhesion Molecules/physiology , Stromal Cells/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cell Growth Processes/physiology , Cell Line , Cytokines/biosynthesis , Female , Hematopoiesis/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Pregnancy , RNA, Small Interfering/genetics , Stromal Cells/cytology , Stromal Cells/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL