Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Vis Exp ; (138)2018 08 10.
Article in English | MEDLINE | ID: mdl-30148479

ABSTRACT

RNA interference via the endogenous miRNA pathway regulates gene expression by controlling protein synthesis through post-transcriptional gene silencing. In recent years, miRNA-mediated gene regulation has shown potential for treatment of neurological disorders caused by a toxic gain of function mechanism. However, efficient delivery to target tissues has limited its application. Here we used a transgenic mouse model for spinal and bulbar muscular atrophy (SBMA), a neuromuscular disease caused by polyglutamine expansion in the androgen receptor (AR), to test gene silencing by a newly identified AR-targeting miRNA, miR-298. We overexpressed miR-298 using a recombinant adeno-associated virus (rAAV) serotype 9 vector to facilitate transduction of non-dividing cells. A single tail-vein injection in SBMA mice induced sustained and widespread overexpression of miR-298 in skeletal muscle and motor neurons and resulted in amelioration of the neuromuscular phenotype in the mice.


Subject(s)
Gene Expression Regulation/genetics , Genetic Therapy/methods , MicroRNAs/genetics , Neuromuscular Diseases/genetics , Neuromuscular Diseases/therapy , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Neuromuscular Diseases/pathology , Rodentia , Serogroup
2.
Ann Clin Transl Neurol ; 5(3): 369-375, 2018 03.
Article in English | MEDLINE | ID: mdl-29560381

ABSTRACT

Amyotrophic lateral sclerosis 8 (ALS8) is a rare progressive neurodegenerative disease resulting from mutation in the gene for vesicle-associated membrane protein-associated protein B. We evaluated a North American patient using exome sequencing, and identified a P56S mutation. The disease protein had similar subcellular localization and expression levels in the patient and control fibroblasts. Patient fibroblasts showed increased basal endoplasmic reticulum stress and dysfunction of nucleocytoplasmic transport as evidenced by impaired Ran trafficking. This finding extends the identification of ALS8 into North America, and indicates a cellular defect similar to other forms of hereditary motor neuron disease.

3.
Mol Cell ; 69(3): 426-437.e7, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395064

ABSTRACT

R-loops are three-stranded nucleic acid structures found abundantly and yet often viewed as by-products of transcription. Studying cells from patients with a motor neuron disease (amyotrophic lateral sclerosis 4 [ALS4]) caused by a mutation in senataxin, we uncovered how R-loops promote transcription. In ALS4 patients, the senataxin mutation depletes R-loops with a consequent effect on gene expression. With fewer R-loops in ALS4 cells, the expression of BAMBI, a negative regulator of transforming growth factor ß (TGF-ß), is reduced; that then leads to the activation of the TGF-ß pathway. We uncovered that genome-wide R-loops influence promoter methylation of over 1,200 human genes. DNA methyl-transferase 1 favors binding to double-stranded DNA over R-loops. Thus, in forming R-loops, nascent RNA blocks DNA methylation and promotes further transcription. Hence, our results show that nucleic acid structures, in addition to sequences, influence the binding and activity of regulatory proteins.


Subject(s)
Gene Expression Regulation/genetics , Promoter Regions, Genetic , RNA Helicases/genetics , RNA Helicases/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , DNA/genetics , DNA/ultrastructure , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Helicases , DNA Methylation/genetics , Humans , Membrane Proteins/metabolism , Multifunctional Enzymes , Mutation , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational , RNA/genetics , RNA/ultrastructure , RNA-Binding Motifs , Transcriptional Activation/genetics , Transforming Growth Factor beta/metabolism
4.
Ann Clin Transl Neurol ; 4(4): 272-275, 2017 04.
Article in English | MEDLINE | ID: mdl-28382308

ABSTRACT

Hereditary spastic paraplegias (HSPs) are well-characterized disorders but rarely reported in Africa. We evaluated a Malian family in which three individuals had HSP and distal muscle atrophy and sensory loss. HSP panel testing identified a novel heterozygous missense mutation in KIF5A (c.1086G>C, p.Lys362Asn) that segregated with the disease (SPG10). Lys362 is highly conserved across species and Lys362Asn is predicted to be damaging. This study shows that HSPs are present in sub-Saharan Africa, although likely underdiagnosed. Increasing efficiency and decreasing costs of DNA sequencing will make it more feasible to diagnose HSPs in developing countries.

5.
Hum Mol Genet ; 25(10): 1979-1989, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962150

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA, also known as Kennedy's disease) is one of nine neurodegenerative disorders that are caused by expansion of polyglutamine-encoding CAG repeats. Intracellular accumulation of abnormal proteins in these diseases, a pathological hallmark, is associated with defects in protein homeostasis. Enhancement of the cellular proteostasis capacity with small molecules has therefore emerged as a promising approach to treatment. Here, we characterize a novel curcumin analog, ASC-JM17, as an activator of central pathways controlling protein folding, degradation and oxidative stress resistance. ASC-JM17 acts on Nrf1, Nrf2 and Hsf1 to increase the expression of proteasome subunits, antioxidant enzymes and molecular chaperones. We show that ASC-JM17 ameliorates toxicity of the mutant androgen receptor (AR) responsible for SBMA in cell, fly and mouse models. Knockdown of the Drosophila Nrf1 and Nrf2 ortholog cap 'n' collar isoform-C, but not Hsf1, blocks the protective effect of ASC-JM17 on mutant AR-induced eye degeneration in flies. Our observations indicate that activation of the Nrf1/Nrf2 pathway is a viable option for pharmacological intervention in SBMA and potentially other polyglutamine diseases.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/genetics , Curcumin/analogs & derivatives , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Muscular Disorders, Atrophic/genetics , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 2/genetics , Receptors, Androgen/genetics , Transcription Factors/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Bulbo-Spinal Atrophy, X-Linked/drug therapy , Bulbo-Spinal Atrophy, X-Linked/pathology , Curcumin/administration & dosage , Curcumin/chemistry , Disease Models, Animal , Drosophila melanogaster/genetics , Gene Knockdown Techniques , Heat Shock Transcription Factors , Humans , Mice , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/pathology , Oxidative Stress/drug effects , Peptides/genetics , Proteasome Endopeptidase Complex/drug effects , Protein Aggregation, Pathological/genetics , Protein Folding/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage
6.
Mol Ther ; 24(5): 937-45, 2016 05.
Article in English | MEDLINE | ID: mdl-26755334

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a currently untreatable adult-onset neuromuscular disease caused by expansion of a polyglutamine repeat in the androgen receptor (AR). In SBMA, as in other polyglutamine diseases, a toxic gain of function in the mutant protein is an important factor in the disease mechanism; therefore, reducing the mutant protein holds promise as an effective treatment strategy. In this work, we evaluated a microRNA (miRNA) to reduce AR expression. From a list of predicted miRNAs that target human AR, we selected microRNA-298 (miR-298) for its ability to downregulate AR mRNA and protein levels when transfected in cells overexpressing wild-type and mutant AR and in SBMA patient-derived fibroblasts. We showed that miR-298 directly binds to the 3'-untranslated region of the human AR transcript, and counteracts AR toxicity in vitro. Intravenous delivery of miR-298 with adeno-associated virus serotype 9 vector resulted in efficient transduction of muscle and spinal cord and amelioration of the disease phenotype in SBMA mice. Our findings support the development of miRNAs as a therapeutic strategy for SBMA and other neurodegenerative disorders caused by toxic proteins.


Subject(s)
Down-Regulation , Genetic Therapy/methods , MicroRNAs/genetics , Muscular Atrophy, Spinal/therapy , Receptors, Androgen/genetics , 3' Untranslated Regions , Administration, Intravenous , Animals , Cell Line , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Humans , MCF-7 Cells , Mice , Muscular Atrophy, Spinal/genetics
8.
JAMA Neurol ; 72(5): 561-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25751282

ABSTRACT

IMPORTANCE: The family of genes implicated in hereditary spastic paraplegias (HSPs) is quickly expanding, mostly owing to the widespread availability of next-generation DNA sequencing methods. Nevertheless, a genetic diagnosis remains unavailable for many patients. OBJECTIVE: To identify the genetic cause for a novel form of pure autosomal dominant HSP. DESIGN, SETTING, AND PARTICIPANTS: We examined and followed up with a family presenting to a tertiary referral center for evaluation of HSP for a decade until August 2014. Whole-exome sequencing was performed in 4 patients from the same family and was integrated with linkage analysis. Sanger sequencing was used to confirm the presence of the candidate variant in the remaining affected and unaffected members of the family and screen the additional patients with HSP. Five affected and 6 unaffected participants from a 3-generation family with pure adult-onset autosomal dominant HSP of unknown genetic origin were included. Additionally, 163 unrelated participants with pure HSP of unknown genetic cause were screened. MAIN OUTCOME AND MEASURE: Mutation in the neuronal isoform of carnitine palmitoyl-transferase (CPT1C) gene. RESULTS: We identified the nucleotide substitution c.109C>T in exon 3 of CPT1C, which determined the base substitution of an evolutionarily conserved Cys residue for an Arg in the gene product. This variant strictly cosegregated with the disease phenotype and was absent in online single-nucleotide polymorphism databases and in 712 additional exomes of control participants. We showed that CPT1C, which localizes to the endoplasmic reticulum, is expressed in motor neurons and interacts with atlastin-1, an endoplasmic reticulum protein encoded by the ATL1 gene known to be mutated in pure HSPs. The mutation, as indicated by nuclear magnetic resonance spectroscopy studies, alters the protein conformation and reduces the mean (SD) number (213.0 [46.99] vs 81.9 [14.2]; P < .01) and size (0.29 [0.01] vs 0.26 [0.01]; P < .05) of lipid droplets on overexpression in cells. We also observed a reduction of mean (SD) lipid droplets in primary cortical neurons isolated from Cpt1c-/- mice as compared with wild-type mice (1.0 [0.12] vs 0.44 [0.05]; P < .001), suggesting a dominant negative mechanism for the mutation. CONCLUSIONS AND RELEVANCE: This study expands the genetics of autosomal dominant HSP and is the first, to our knowledge, to link mutation in CPT1C with a human disease. The association of the CPT1C mutation with changes in lipid droplet biogenesis supports a role for altered lipid-mediated signal transduction in HSP pathogenesis.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Spastic Paraplegia, Hereditary/genetics , Adult , Animals , Humans , Italy , Mice , Mice, Knockout , Middle Aged , Mutation/genetics , Pedigree
9.
Neuromuscul Disord ; 24(11): 978-81, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25047668

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is an X-linked neuromuscular disease caused by a trinucleotide (CAG) repeat expansion in the androgen receptor gene. Patients with SBMA have weakness, atrophy, and fasciculations in the bulbar and extremity muscles. Individuals with CAG repeat lengths greater than 62 have not previously been reported. We evaluated a 29year old SBMA patient with 68 CAGs who had unusually early onset and findings not seen in others with the disease. Analysis of the androgen receptor gene confirmed the repeat length of 68 CAGs in both peripheral blood and fibroblasts. Evaluation of muscle and sensory function showed deficits typical of SBMA, and in addition the patient had manifestations of autonomic dysfunction and abnormal sexual development. These findings extend the known phenotype associated with SBMA and shed new insight into the effects of the mutated androgen receptor.


Subject(s)
Muscular Disorders, Atrophic/genetics , Receptors, Androgen/genetics , Trinucleotide Repeats/genetics , Adult , Humans , Magnetic Resonance Imaging , Male , Muscle Weakness/etiology , Muscular Disorders, Atrophic/complications , Nerve Fibers/pathology
10.
Neurobiol Dis ; 70: 12-20, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24925468

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA, Kennedy's disease) is a motor neuron disease caused by polyglutamine repeat expansion in the androgen receptor. Although degeneration occurs in the spinal cord and muscle, the exact mechanism is not clear. Induced pluripotent stem cells from spinal and bulbar muscular atrophy patients provide a useful model for understanding the disease mechanism and designing effective therapy. Stem cells were generated from six patients and compared to control lines from three healthy individuals. Motor neurons from four patients were differentiated from stem cells and characterized to understand disease-relevant phenotypes. Stem cells created from patient fibroblasts express less androgen receptor than control cells, but show androgen-dependent stabilization and nuclear translocation. The expanded repeat in several stem cell clones was unstable, with either expansion or contraction. Patient stem cell clones produced a similar number of motor neurons compared to controls, with or without androgen treatment. The stem cell-derived motor neurons had immunoreactivity for HB9, Isl1, ChAT, and SMI-32, and those with the largest repeat expansions were found to have increased acetylated α-tubulin and reduced HDAC6. Reduced HDAC6 was also found in motor neuron cultures from two other patients with shorter repeats. Evaluation of stably transfected mouse cells and SBMA spinal cord showed similar changes in acetylated α-tubulin and HDAC6. Perinuclear lysosomal enrichment, an HDAC6 dependent process, was disrupted in motor neurons from two patients with the longest repeats. SBMA stem cells present new insights into the disease, and the observations of reduced androgen receptor levels, repeat instability, and reduced HDAC6 provide avenues for further investigation of the disease mechanism and development of effective therapy.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/physiopathology , Induced Pluripotent Stem Cells/physiology , Motor Neurons/physiology , Acetylation , Adult , Aged , Bulbo-Spinal Atrophy, X-Linked/genetics , Cells, Cultured , DNA Repeat Expansion , Female , Fibroblasts/physiology , Histone Deacetylase 6 , Histone Deacetylases/deficiency , Humans , Male , Middle Aged , Neurogenesis/physiology , Receptors, Androgen/metabolism , Tubulin/metabolism , Young Adult
11.
Ann Neurol ; 75(4): 525-32, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24515897

ABSTRACT

OBJECTIVE: Spinal muscular atrophy (SMA) is one of the most common severe hereditary diseases of infancy and early childhood in North America, Europe, and Asia. SMA is usually caused by deletions of the survival motor neuron 1 (SMN1) gene. A closely related gene, SMN2, modifies the disease severity. SMA carriers have only 1 copy of SMN1 and are relatively common (1 in 30-50) in populations of European and Asian descent. SMN copy numbers and SMA carrier frequencies have not been reliably estimated in Malians and other sub-Saharan Africans. METHODS: We used a quantitative polymerase chain reaction assay to determine SMN1 and SMN2 copy numbers in 628 Malians, 120 Nigerians, and 120 Kenyans. We also explored possible mechanisms for SMN1 and SMN2 copy number differences in Malians, and investigated their effects on SMN mRNA and protein levels. RESULTS: The SMA carrier frequency in Malians is 1 in 209, lower than in Eurasians. Malians and other sub-Saharan Africans are more likely to have ≥3 copies of SMN1 than Eurasians, and more likely to lack SMN2 than Europeans. There was no evidence of gene conversion, gene locus duplication, or natural selection from malaria resistance to account for the higher SMN1 copy numbers in Malians. High SMN1 copy numbers were not associated with increased SMN mRNA or protein levels in human cell lines. INTERPRETATION: SMA carrier frequencies are much lower in sub-Saharan Africans than in Eurasians. This finding is important to consider in SMA genetic counseling in individuals with black African ancestry.


Subject(s)
DNA Copy Number Variations/genetics , Muscular Atrophy, Spinal/epidemiology , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Africa South of the Sahara/epidemiology , Female , Genetic Predisposition to Disease , Humans , Male , RNA, Messenger/metabolism , Survival of Motor Neuron 2 Protein/genetics
12.
Mol Med ; 18: 1261-8, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22952056

ABSTRACT

Spinal and bulbar muscular atrophy is an X-linked motor neuron disease caused by polyglutamine expansion in the androgen receptor. Patients develop slowly progressive proximal muscle weakness, muscle atrophy and fasciculations. Affected individuals often show gynecomastia, testicular atrophy and reduced fertility as a result of mild androgen insensitivity. No effective disease-modifying therapy is currently available for this disease. Our recent studies have demonstrated that insulinlike growth factor (IGF)-1 reduces the mutant androgen receptor toxicity through activation of Akt in vitro, and spinal and bulbar muscular atrophy transgenic mice that also overexpress a noncirculating muscle isoform of IGF-1 have a less severe phenotype. Here we sought to establish the efficacy of daily intraperitoneal injections of mecasermin rinfabate, recombinant human IGF-1 and IGF-1 binding protein 3, in a transgenic mouse model expressing the mutant androgen receptor with an expanded 97 glutamine tract. The study was done in a controlled, randomized, blinded fashion, and, to reflect the clinical settings, the injections were started after the onset of disease manifestations. The treatment resulted in increased Akt phosphorylation and reduced mutant androgen receptor aggregation in muscle. In comparison to vehicle-treated controls, IGF-1-treated transgenic mice showed improved motor performance, attenuated weight loss and increased survival. Our results suggest that peripheral tissue can be targeted to improve the spinal and bulbar muscular atrophy phenotype and indicate that IGF-1 warrants further investigation in clinical trials as a potential treatment for this disease.


Subject(s)
Insulin-Like Growth Factor I/therapeutic use , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/pathology , Animals , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Muscles/drug effects , Muscles/metabolism , Muscles/pathology , Muscles/physiopathology , Muscular Disorders, Atrophic/enzymology , Muscular Disorders, Atrophic/physiopathology , Mutant Proteins/metabolism , Phosphorylation/drug effects , Protein Structure, Quaternary , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , Weight Loss/drug effects
13.
Mol Cell Neurosci ; 37(1): 178-86, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18029196

ABSTRACT

The Dab1 docking protein is required for the proper organization of brain laminae and for a signal transduction pathway initiated by Reelin binding to the ApoER2 and VLDLR receptors on the cell surface of neurons. Dab1 physically interacts with APP; however, it is not known whether the APP gene influences Dab1 function. Here we demonstrate a genetic interaction between Dab1 and APP. Dab1-hypomorphic animals have neuronal ectopias in the neocortex and reduced cerebellar volume, possibly a consequence of Purkinje cell misplacement. These phenotypes are exacerbated in transgenic animals overexpressing a mutant form of APP, APP(swe), which is characterized by increased processing at the beta-secretase site. The Dab1-hypomorphic phenotype is improved in the cerebellum of animals that are deficient for APP. Together this suggests that APP expression constrains the consequences of Dab1 activity during brain development.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Brain/anatomy & histology , Brain/growth & development , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Gene Expression Regulation , Mice , Mice, Transgenic , Protein Binding/genetics , Reelin Protein
14.
J Cell Sci ; 117(Pt 19): 4527-36, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15316068

ABSTRACT

Reelin-induced Dab1 tyrosine phosphorylation has been implicated in the regulation of neuronal positioning during brain development. The downstream consequences of Dab1 tyrosine phosphorylation are not fully understood, however. Here we identify CrkII, CrkL and Dock1 in complexes bound to tyrosine-phosphorylated Dab1, through mass spectrometry. The CrkII-Dab1 interaction requires tyrosine phosphorylation of Dab1 at residues 220 or 232 and is promoted by Reelin treatment of embryonic forebrain neurons. Unlike other CrkII binding proteins, such as paxillin and p130Cas, expression of Dab1 interfered with CrkII-dependent cell migration of Nara Bladder Tumor II (NBT-II) cells, in a tyrosine phosphorylation-site dependent manner. Overexpression of CrkIIGFP rescued the migration of these cells, suggesting that Dab1 makes Crk a limiting factor for migration. The Dock1-Dab1 association is indirect and requires CrkII. In organisms such as Drosophila melanogaster and Caenorhabditis elegans, signaling complexes, which contain Crk and Dock1 family members are conserved and act through Rac. We show that a rough-eye phenotype in Drosophila caused by exogenous expression of tyrosine-phosphorylated mouse Dab1RFP is partially rescued by a loss-of-function mutation in myoblast city, a Dock1-like gene in Drosophila. We propose a model that tyrosine-phosphorylated Dab1 engages the conserved Crk-Dock1-Rac signaling cassette, but when bound to Dab1 this signaling complex does not support migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/pharmacology , Cell Movement/physiology , Extracellular Matrix Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Cell Movement/drug effects , Cells, Cultured , Collagen/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila/ultrastructure , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Mammalian/metabolism , Embryo, Nonmammalian , Mass Spectrometry , Mice , Microscopy, Electron, Scanning , Molecular Sequence Data , Neurons/metabolism , Paxillin , Phosphoproteins/metabolism , Phosphorylation , Protein Binding/drug effects , Proto-Oncogene Proteins c-crk , Reelin Protein , Serine Endopeptidases , rac GTP-Binding Proteins
15.
Mol Cell Biol ; 23(20): 7210-21, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517291

ABSTRACT

The tyrosine phosphorylation sites of the Disabled 1 (Dab1) docking protein are essential for the transmission of the Reelin signal, which regulates neuronal placement. Here we identify Nck beta as a phosphorylation-dependent, Dab1-interacting protein. The SH2 domain of Nck beta but not Nck alpha binds Dab1 phosphorylated on the Reelin-regulated site, Y220, or on Y232. Nck beta is coexpressed with Dab1 in the developing brain and in cultured neurons, where Reelin stimulation leads to the redistribution of Nck beta from the cell soma into neuronal processes. We found that tyrosine-phosphorylated Dab1 in synergy with Nck beta disrupts the actin cytoskeleton in transfected cells. In Drosophila melanogaster, exogenous expression of mouse Dab1 causes tyrosine phosphorylation site-dependent morphological changes in the compound eye. This phenotype is enhanced by overexpression of the Drosophila Nck protein Dock, suggesting a conserved interaction between the Disabled and Nck family members. We suggest a model in which Dab1 phosphorylation leads to the recruitment of Nck beta to the membrane, where it acts to remodel the actin cytoskeleton.


Subject(s)
Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Tyrosine/metabolism , Actins/metabolism , Animals , Cell Line , Cytoskeleton/metabolism , Drosophila melanogaster/metabolism , Humans , Immunohistochemistry , Mice , Microscopy, Electron, Scanning , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Rats , Reelin Protein , Serine Endopeptidases , Time Factors , Two-Hybrid System Techniques , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...