Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuron ; 111(16): 2544-2556.e9, 2023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37591201

RESUMO

Information processing and storage in the brain rely on AMPA-receptors (AMPARs) and their context-dependent dynamics in synapses and extra-synaptic sites. We found that distribution and dynamics of AMPARs in the plasma membrane are controlled by Noelins, a three-member family of conserved secreted proteins expressed throughout the brain in a cell-type-specific manner. Noelin tetramers tightly assemble with the extracellular domains of AMPARs and interconnect them in a network-like configuration with a variety of secreted and membrane-anchored proteins including Neurexin1, Neuritin1, and Seizure 6-like. Knock out of Noelins1-3 profoundly reduced AMPARs in synapses onto excitatory and inhibitory (inter)neurons, decreased their density and clustering in dendrites, and abolished activity-dependent synaptic plasticity. Our results uncover an endogenous mechanism for extracellular anchoring of AMPARs and establish Noelin-organized networks as versatile determinants of constitutive and context-dependent neurotransmission.


Assuntos
Encéfalo , Proteínas de Membrana , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico , Proteínas de Membrana/genética , Transporte Biológico , Membrana Celular , Receptores de AMPA
2.
Metabolism ; 129: 155122, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35026233

RESUMO

BACKGROUND AND AIMS: Olfactomedin 2 (OLFM2; also known as noelin 2) is a pleiotropic protein that plays a major role in olfaction and Olfm2 null mice exhibit reduced olfactory sensitivity, as well as abnormal motor coordination and anxiety-related behavior. Here, we investigated the possible metabolic role of OLFM2. METHODS: Olfm2 null mice were metabolically phenotyped. Virogenetic modulation of central OLFM2 was also performed. RESULTS: Our data showed that, the global lack of OLFM2 in mice promoted anorexia and increased energy expenditure due to elevated brown adipose tissue (BAT) thermogenesis and browning of white adipose tissue (WAT). This phenotype led to resistance to high fat diet (HFD)-induced obesity. Notably, virogenetic overexpression of Olfm2 in the lateral hypothalamic area (LHA) induced weight gain associated with decreased BAT thermogenesis. CONCLUSION: Overall, this evidence first identifies central OLFM2 as a new molecular actor in the regulation of whole-body energy homeostasis.


Assuntos
Tecido Adiposo Marrom , Termogênese , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético/genética , Proteínas da Matriz Extracelular , Glicoproteínas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Termogênese/genética
3.
J Neurochem ; 143(6): 635-644, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28975619

RESUMO

The olfm1a and olfm1b genes in zebrafish encode conserved secreted glycoproteins. These genes are preferentially expressed in the brain and retina starting from 16 h post-fertilization until adulthood. Functions of the Olfm1 gene is still unclear. Here, we produced and analyzed a null zebrafish mutant of both olfm1a and olfm1b genes (olfm1 null). olfm1 null fish were born at a normal Mendelian ratio and showed normal body shape and fertility as well as no visible defects from larval stages to adult. Olfm1 proteins were preferentially localized in the synaptosomes of the adult brain. Olfm1 co-immunoprecipitated with GluR2 and soluble NSF attachment protein receptor complexes indicating participation of Olfm1 in both pre- and post-synaptic events. Phosphorylation of GluR2 was not changed while palmitoylation of GluR2 was decreased in the brain synaptosomal membrane fraction of olfm1 null compared with wt fish. The levels of GluR2, SNAP25, flotillin1, and VAMP2 were markedly reduced in the synaptic microdomain of olfm1 null brain compared with wt. The internalization of GluR2 in retinal cells and the localization of VAMP2 in brain synaptosome were modified by olfm1 null mutation. This indicates that Olfm1 may regulate receptor trafficking from the intracellular compartments to the synaptic membrane microdomain, partly through the alteration of post-translational GluR2 modifications such as palmitoylation. Olfm1 may be considered a novel regulator of the composition and function of the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor complex.


Assuntos
Proteínas da Matriz Extracelular/genética , Glicoproteínas/genética , Transporte Proteico/genética , Receptores de AMPA/metabolismo , Animais , Encéfalo/metabolismo , Técnicas de Inativação de Genes , Peixe-Zebra
4.
Exp Neurol ; 261: 802-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25218043

RESUMO

Olfactomedin 2 (Olfm2) is a secretory glycoprotein belonging to the family of olfactomedin domain-containing proteins. A previous study has shown that a mutation in OLFM2 is associated with primary open angle glaucoma in Japanese patients. In the present study, we generated Olfm2 deficient mice by replacing the Olfm2 gene with the LacZ gene. The loss of Olfm2 resulted in no gross abnormalities. However, Olfm2 null mice showed reduced exploration, locomotion, olfactory sensitivity, abnormal motor coordination, and anxiety related behavior. The pattern of the Olfm2 gene expression was studied in the brain and eye using ß-galactosidase staining. In the brain, Olfm2 was mainly expressed in the olfactory bulb, cortex, piriform cortex, olfactory trabeculae, and inferior and superior colliculus. In the eye expression was detected mainly in retinal ganglion cells. In Olfm2 null mice, the amplitude of the first negative wave in the visual evoked potential test was significantly reduced as compared with wild-type littermates. Olfm2, similar to Olfm1, interacted with the GluR2 subunit of the AMPAR complexes and Olfm2 co-segregated with the AMPA receptor subunit GluR2 and other synaptic proteins in the synaptosomal membrane fraction upon biochemical fractionation of the adult mice cortex and retina. Immunoprecipitation from the synaptosomal membrane fraction of the Olfm2 null mouse brain cortex using the GluR2 antibody showed reduced levels of several components of the AMPAR complex in the immunoprecipitates including Olfm1, PSD95 and CNIH2. These results suggest that heterodimers of Olfm1 and Olfm2 interact with AMPAR more efficiently than Olfm2 homodimers and that Olfm2 plays a role in the organization of the AMPA receptor complexes.


Assuntos
Proteínas da Matriz Extracelular/deficiência , Glicoproteínas/deficiência , Transtornos dos Movimentos/genética , Transtornos do Olfato/genética , Receptores de AMPA/metabolismo , Deleção de Sequência/genética , Transtornos da Visão/genética , Animais , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Potenciais Evocados Visuais/genética , Proteínas da Matriz Extracelular/genética , Glicoproteínas/genética , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Nervo Óptico/patologia , Retina/metabolismo , Transtornos da Visão/complicações
5.
Exp Neurol ; 250: 205-18, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24095980

RESUMO

Olfactomedin 1 (Olfm1) is a secreted glycoprotein that is preferentially expressed in neuronal tissues. Here we show that deletion of exons 4 and 5 from the Olfm1 gene, which encodes a 52 amino acid long region in the N-terminal part of the protein, increased neonatal death and reduced body weight of surviving homozygous mice. Magnetic resonance imaging analyses revealed reduced brain volume and attenuated size of white matter tracts such as the anterior commissure, corpus callosum, and optic nerve. Adult Olfm1 mutant mice demonstrated abnormal behavior in several tests including reduced marble digging, elevated plus maze test, nesting activity and latency on balance beam tests as compared with their wild-type littermates. The olfactory system was both structurally and functionally disturbed by the mutation in the Olfm1 gene as shown by functional magnetic resonance imaging analysis and a smell test. Deficiencies of the olfactory system may contribute to the neonatal death and loss of body weight of Olfm1 mutant. Shotgun proteomics revealed 59 candidate proteins that co-precipitated with wild-type or mutant Olfm1 proteins in postnatal day 1 brain. Olfm1-binding targets included GluR2, Cav2.1, teneurin-4 and Kidins220. Modified interaction of Olfm1 with binding targets led to an increase in intracellular Ca(2+) concentration and activation of ERK1/2, MEK1 and CaMKII in the hippocampus and olfactory bulb of Olfm1 mutant mice compared with their wild-type littermates. Excessive activation of the CaMKII and Ras-ERK pathways in the Olfm1 mutant olfactory bulb and hippocampus by elevated intracellular calcium may contribute to the abnormal behavior and olfactory activity of Olfm1 mutant mice.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Éxons/genética , Proteínas da Matriz Extracelular/genética , Feminino , Glicoproteínas/genética , Imunoprecipitação , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Proteômica/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência
6.
J Biol Chem ; 287(44): 37171-84, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22923615

RESUMO

Olfm1, a secreted highly conserved glycoprotein, is detected in peripheral and central nervous tissues and participates in neural progenitor maintenance, cell death in brain, and optic nerve arborization. In this study, we identified Olfm1 as a molecule promoting axon growth through interaction with the Nogo A receptor (NgR1) complex. Olfm1 is coexpressed with NgR1 in dorsal root ganglia and retinal ganglion cells in embryonic and postnatal mice. Olfm1 specifically binds to NgR1, as judged by alkaline phosphatase assay and coimmunoprecipitation. The addition of Olfm1 inhibited the growth cone collapse of dorsal root ganglia neurons induced by myelin-associated inhibitors, indicating that Olfm1 attenuates the NgR1 receptor functions. Olfm1 caused the inhibition of NgR1 signaling by interfering with interaction between NgR1 and its coreceptors p75NTR or LINGO-1. In zebrafish, inhibition of optic nerve extension by olfm1 morpholino oligonucleotides was partially rescued by dominant negative ngr1 or lingo-1. These data introduce Olfm1 as a novel NgR1 ligand that may modulate the functions of the NgR1 complex in axonal growth.


Assuntos
Axônios/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas da Matriz Extracelular/fisiologia , Glicoproteínas/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Células COS , Chlorocebus aethiops , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Cones de Crescimento/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Mielina/fisiologia , Proteínas Nogo , Nervo Óptico/citologia , Nervo Óptico/embriologia , Especificidade de Órgãos , Células PC12 , Ligação Proteica , Ratos , Receptor de Fator de Crescimento Neural/metabolismo , Peixe-Zebra , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Invest Ophthalmol Vis Sci ; 52(5): 2584-92, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21228389

RESUMO

PURPOSE: Olfactomedin 2 (OLFM2) belongs to the family of olfactomedin domain-containing proteins. Genetic data suggest its association with glaucoma in Japanese patients. However, its functions are still elusive. In this study, the properties of mammalian OLFM2 were investigated. METHODS: Expression of the rat and mouse Olfm2 gene was studied by using real-time PCR and in situ hybridization. Substitutions were introduced into OLFM2 by mutagenesis in vitro. Intracellular localization of OLFM2 was studied by confocal microscopy after transient transfection in HEK293 cells. Interaction of OLFM2 with olfactomedin 1 (Olfm1), olfactomedin 3 (Olfm3), myocilin, and gliomedin was studied by using co-immunoprecipitation. RESULTS: Two major human OLFM2 mRNAs encode secreted proteins with a length of 454 and 478 amino acids. OLFM2 is more closely related to OLFM1 and -3 than to any other family members. Olfm2 showed the most dynamic expression pattern compared with Olfm1 and -3 during mouse eye development and was expressed preferentially in the developing retinal ganglion cell layer. Among three OLFM2 substitutions tested (T86M, R144Q, and L420S), only L420S completely blocked secretion of the protein. OLFM2 interacted with Olfm1 and -3, but not with myocilin and gliomedin. Co-transfection of the L420S mutant with wild-type Olfm1 and -3 significantly inhibited secretion of Olfm1 and -3. CONCLUSIONS: Highly conserved OLFM2 protein may play an important role in the course of retinal and eye development. Severe mutations in one of the closely related olfactomedin domain-containing proteins (Olfm1-3) may block the secretion and probably the activity of all three family members, leading to more pronounced diseases of the retina than the knockout of individual genes.


Assuntos
Proteínas da Matriz Extracelular/genética , Olho/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glicoproteínas/genética , Domínios e Motivos de Interação entre Proteínas/genética , Células Ganglionares da Retina/metabolismo , Sequência de Aminoácidos , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Olho/crescimento & desenvolvimento , Proteínas do Olho/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Glicoproteínas/metabolismo , Células HEK293/metabolismo , Humanos , Imunoprecipitação , Hibridização In Situ , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Mapeamento de Interação de Proteínas , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Transfecção
8.
Mol Vis ; 15: 319-25, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19204788

RESUMO

PURPOSE: To determine the immunophenotypes of macular corneal dystrophy (MCD) in Indian patients and to correlate them with mutations in the carbohydrate 6-sulfotransferase (CHST6) gene. METHODS: Sixty-four patients from 53 families with MCD that were previously screened for mutations in CHST6 were included in an immunophenotype analysis. Antigenic keratan sulfate (AgKS) in serum as well as corneal tissue was evaluated in 31 families. Only cornea was evaluated in 11 families, and only serum was evaluated in 11 families. AgKS was detected in formalin-fixed, paraffin-embedded corneal sections by immunohistochemistry and in serum by ELISA using a monoclonal antibody against sulfated forms of KS in patients with MCD as well as normal controls. RESULTS: Analysis of corneal and/or serum AgKS disclosed MCD type I (27 families), MCD type IA (5 families), and MCD type II (3 families) in the cases studied. An additional 10 families were either MCD type I or MCD type IA since only serum AgKS data were available. Seven families manifested atypical immunophenotypes since the corneal AgKS expression was either of MCD type I or MCD type IA, but serum AgKS levels ranged from 19 ng/ml to 388 ng/ml. More than one immunophenotype was detected amongst siblings in two families. Each immunophenotype was associated with mutational heterogeneity in CHST6. CONCLUSIONS: MCD type I was the predominant immunophenotype in the Indian population studied followed by MCD type IA and then MCD type II. We detected further immunophenotypic heterogeneity by finding atypical patterns of AgKS reactivity in a subset of families. There were no simple correlations between immunophenotypes and specific mutations in CHST6, suggesting that factors other than CHST6 mutations may be contributing to the immunophenotypes in MCD.


Assuntos
Distrofias Hereditárias da Córnea/genética , Sulfato de Queratano/imunologia , Mutação , Sulfotransferases/genética , Córnea/imunologia , Córnea/patologia , Distrofias Hereditárias da Córnea/imunologia , Distrofias Hereditárias da Córnea/metabolismo , Distrofias Hereditárias da Córnea/patologia , Família , Humanos , Imuno-Histoquímica , Índia , Sulfato de Queratano/análise , Sulfato de Queratano/sangue , Fenótipo , Estatística como Assunto , Sulfotransferases/metabolismo , Carboidrato Sulfotransferases
9.
Mol Vis ; 13: 1327-32, 2007 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-17679935

RESUMO

PURPOSE: Autosomal recessive congenital hereditary endothelial dystrophy (AR-CHED or CHED2) is a bilateral corneal disorder manifesting at birth or in early childhood. CHED2 is caused by mutations in the sodium bicarbonate transporter-like solute carrier family 4 member 11 (SLC4A11) gene on chromosome 20p13. We screened 42 unrelated families with CHED2 in order to establish the spectrum of mutations in SLC4A11 and to look for genotype-phenotype correlations. METHODS: Forty-two families (49 affected and 73 unaffected members) with recessive CHED were recruited according to predefined diagnostic criteria. Clinical data including age at onset and presentation, pre- and post-operative visual acuities, and presence of nystagmus were taken from patient records. Histopathologic parameters such as corneal thickness, Descemet membrane thickness, and endothelial cell counts were assessed on corneal sections. DNA from patients was screened for sequence changes by polymerase chain reaction (PCR)-amplification of coding regions of SLC4A11 and single strand conformation polymorphism analysis followed by sequencing. Sequence changes found were tested in 50 unrelated normal controls. RESULTS: Twenty-seven different mutations were identified in 35 unrelated families, 19 of which were not previously reported. The mutations identified consisted of 13 missense, 5 nonsense, 7 deletions, 1 complex (deletion plus insertion) mutation, and 1 splice site mutation. Both mutant alleles were identified in 33 families and only one mutant allele in two families. No correlations were evident between clinical or histopathologic parameters and SLC4A11 mutations. CONCLUSIONS: These data add to the mutational repertoire of SLC4A11 and establish the high degree of mutational heterogeneity in autosomal recessive CHED.


Assuntos
Proteínas de Transporte de Ânions/genética , Antiporters/genética , Distrofias Hereditárias da Córnea/genética , Genes Recessivos , Mutação/genética , Adolescente , Adulto , Criança , Pré-Escolar , Humanos , Lactente , Recém-Nascido , Fenótipo
10.
J Med Genet ; 44(1): 64-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16825429

RESUMO

OBJECTIVE: To map and identify the gene for autosomal recessive congenital hereditary endothelial dystrophy (CHED2, OMIM 217700), a disorder characterised by diffuse bilateral corneal clouding that may lead to visual impairment and requiring corneal transplantation. METHODS: Members of 16 families with autosomal recessive CHED were genotyped for 13 microsatellite markers at the CHED2 locus on chromosome 20p13-12. Two-point linkage analysis was carried out using the FASTLINK version of the MLINK program. Mutation screening was carried out by amplification of exons and flanking regions by polymerase chain reaction, followed by direct automated sequencing. RESULTS: Linkage and haplotype analysis placed the disease locus within a 2.2 cM (1.3 Mb) interval flanked by D20S198 and D20S889, including SLC4A11. The maximum LOD score of 11.1 was obtained with D20S117 at theta = 0. Sequencing of SLC4A11 showed homozygotic mutations in affected members from 12 of 16 families. CONCLUSION: These results confirm that mutations in the SLC4A11 gene cause autosomal recessive CHED.


Assuntos
Proteínas de Transporte de Ânions/genética , Antiporters/genética , Distrofias Hereditárias da Córnea/genética , Genes Recessivos , Criança , Pré-Escolar , Cromossomos Humanos Par 20/genética , Consanguinidade , Éxons , Feminino , Humanos , Lactente , Escore Lod , Masculino , Repetições de Microssatélites , Mutação , Linhagem
11.
Mol Vis ; 9: 730-4, 2003 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-14735064

RESUMO

PURPOSE: Macular corneal dystrophy (MCD) is an autosomal recessive disorder characterized by progressive central haze, confluent punctate opacities and abnormal deposits in the cornea. It is caused by mutations in the carbohydrate sulfotransferase-6 (CHST6) gene, encoding corneal N-acetyl glucosamine-6-O-sulfotransferase (C-GlcNAc-6-ST). We screened the CHST6 gene for mutations in Indian families with MCD, in order to determine the range of pathogenic mutations. METHODS: Genomic DNA was isolated from peripheral blood leukocytes of patients with MCD and normal controls. The coding regions of the CHST6 gene were amplified using three pairs of primers and amplified products were directly sequenced. RESULTS: We identified 22 (5 nonsense, 5 frameshift, 2 insertion, and 10 missense) mutations in 36 patients from 31 families with MCD, supporting the conclusion that loss of function of this gene is responsible for this corneal disease. Seventeen of these mutations are novel. CONCLUSIONS: These data highlight the allelic heterogeneity of macular corneal dystrophy in Indian patients.


Assuntos
Distrofias Hereditárias da Córnea/genética , Mutação , Sulfotransferases/genética , Adolescente , Adulto , Alelos , Distrofias Hereditárias da Córnea/enzimologia , Distrofias Hereditárias da Córnea/epidemiologia , Análise Mutacional de DNA , Heterogeneidade Genética , Humanos , Índia/epidemiologia , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Carboidrato Sulfotransferases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA