Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Mol Neurosci ; 68(2): 171-180, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30888622

RESUMO

Synaptic cell adhesion molecules, including neurexins and neuroligins, mediate the formation and maintenance of connections between neuronal cells. Although neurexins and neuroligins are known to interact with each other in a calcium-dependent manner and several neuropeptides have been shown to act through G protein-coupled receptors to increase intracellular calcium levels, no studies have examined the role of the neuropeptide oxytocin in association with adhesion molecules. Given that oxytocin receptors are located on presynaptic and postsynaptic membranes and that oxytocin exerts direct effects on neuronal excitability, it could be hypothesized that oxytocin affects the expression of cell surface adhesion molecules. In the present study, we show that incubation in the presence of oxytocin (1 µM, 48 h) exerted cell-specific effects on the levels of neurexin 2α, neurexin 2ß, and neuroligin 3. Oxytocin significantly increased the mRNA expression levels of neurexin 2α, neurexin 2ß, and neuroligin 3 in SH-SY5Y, U-87MG, and primary cerebellar cells. The effect of inhibiting oxytocin receptors on the expression of neurexin 2ß was more dramatic in U-87MG cells than in SH-SY5Y cells. Oxytocin did not exert effects in primary corticohippocampal cells. Additionally, we measured the expression of selected GTPases to determine whether they could mediate the effects of oxytocin. Oxytocin induced a decrease in the mRNA level of Rac1 in U-87MG and primary cerebellar cells and exerted a stimulatory effect on the expression of RhoB at the gene and protein level in SH-SY5Y cells. These results suggest that the regulation of neurexins and neuroligins involves the activation of oxytocin receptors. These effects are likely mediated by the stimulation of RhoB GTPase, at least in certain types of cells.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores de Ocitocina/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Cerebelo/citologia , Humanos , Hipotálamo/citologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Ocitocina/farmacologia , Ratos , Ratos Wistar , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoB de Ligação ao GTP/genética , Proteína rhoB de Ligação ao GTP/metabolismo
2.
Neuromolecular Med ; 19(1): 136-146, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27567921

RESUMO

Reelin is an extracellular glycoprotein which contributes to synaptic plasticity and function of memory in the adult brain. It has been indicated that the Reelin signaling cascade participates in Alzheimer's disease (AD). Besides the neurons, glial cells such as astrocytes also express Reelin protein. While functional loss of astrocytes has been reported to be associated with AD, dysfunction of astrocytic Reelin signaling pathway has not received much attention. Therefore, we investigated the effects of α-boswellic acid (ABA) as one of the major component of Boswellia serrata resin on primary fetal human astrocytes under a stress paradigm as a possible model for AD through study on Reelin cascade. For this aim, we used streptozotocin (STZ), in which from an outlook generates Alzheimer's hallmarks in astrocytes, and assayed Reelin expression, Tau and Akt phosphorylation as well as reactive oxygen species (ROS) generation and apoptosis in the presences of ABA. Our results indicated that while STZ (100 µM) down-regulated the expression of Reelin, ABA (25 µM) up-regulated its expression (p < 0.01) for 24 h. ABA efficiently reduced hyperphosphorylated Tau (Ser404) in STZ-treated astrocytes (p < 0.01). Furthermore, STZ-induced apoptosis by increasing cleaved caspase three (p < 0.01) and ROS generation (p < 0.01), a further pathological hallmark of Tauopathy. On the other hand, ABA decreased ROS generation and promoted proliferation of astrocytes through elevating Survivin expression (p < 0.01). These results showed that ABA could be considered as a potent therapeutic agent for prevention and decreasing the progression of Alzheimer's hallmarks in astrocytes; however, more in vivo studies would be needed.


Assuntos
Astrócitos/efeitos dos fármacos , Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas da Matriz Extracelular/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Triterpenos Pentacíclicos/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Serina Endopeptidases/biossíntese , Proteínas tau/metabolismo , Apoptose/efeitos dos fármacos , Astrócitos/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hipotálamo/citologia , Hipotálamo/embriologia , Proteínas Inibidoras de Apoptose/biossíntese , Proteínas Inibidoras de Apoptose/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Reelina , Serina Endopeptidases/genética , Estreptozocina/farmacologia , Survivina
3.
Transl Psychiatry ; 6: e820, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27219346

RESUMO

Post-traumatic stress disorder (PTSD) develops in only some people following trauma exposure, but the mechanisms differentially explaining risk versus resilience remain largely unknown. PTSD is heritable but candidate gene studies and genome-wide association studies (GWAS) have identified only a modest number of genes that reliably contribute to PTSD. New gene-based methods may help identify additional genes that increase risk for PTSD development or severity. We applied gene-based testing to GWAS data from the Grady Trauma Project (GTP), a primarily African American cohort, and identified two genes (NLGN1 and ZNRD1-AS1) that associate with PTSD after multiple test correction. Although the top SNP from NLGN1 did not replicate, we observed gene-based replication of NLGN1 with PTSD in the Drakenstein Child Health Study (DCHS) cohort from Cape Town. NLGN1 has previously been associated with autism, and it encodes neuroligin 1, a protein involved in synaptogenesis, learning, and memory. Within the GTP dataset, a single nucleotide polymorphism (SNP), rs6779753, underlying the gene-based association, associated with the intermediate phenotypes of higher startle response and greater functional magnetic resonance imaging activation of the amygdala, orbitofrontal cortex, right thalamus and right fusiform gyrus in response to fearful faces. These findings support a contribution of the NLGN1 gene pathway to the neurobiological underpinnings of PTSD.


Assuntos
Encéfalo/fisiopatologia , Moléculas de Adesão Celular Neuronais/genética , Transtornos de Estresse Pós-Traumáticos/genética , Adulto , Negro ou Afro-Americano/genética , Tonsila do Cerebelo/diagnóstico por imagem , Tonsila do Cerebelo/fisiopatologia , Encéfalo/diagnóstico por imagem , Expressão Facial , Reconhecimento Facial , Medo , Feminino , Neuroimagem Funcional , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Córtex Pré-Frontal/diagnóstico por imagem , Córtex Pré-Frontal/fisiopatologia , Reflexo de Sobressalto/fisiologia , Transtornos de Estresse Pós-Traumáticos/diagnóstico por imagem , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Transtornos de Estresse Pós-Traumáticos/psicologia , Lobo Temporal/diagnóstico por imagem , Lobo Temporal/fisiopatologia , Tálamo/diagnóstico por imagem , Tálamo/fisiopatologia , População Branca/genética , Adulto Jovem
4.
Mol Psychiatry ; 21(3): 411-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26055424

RESUMO

Phenotypic and genetic heterogeneity is predominant in autism spectrum disorders (ASD), for which the molecular and pathophysiological bases are still unclear. Significant comorbidity and genetic overlap between ASD and other neurodevelopmental disorders are also well established. However, little is understood regarding the frequent observation of a wide phenotypic spectrum associated with deleterious mutations affecting a single gene even within multiplex families. We performed a clinical, neurophysiological (in vivo electroencephalography-auditory-evoked related potentials) and genetic (whole-exome sequencing) follow-up analysis of two families with known deleterious NLGN4X gene mutations (either truncating or overexpressing) present in individuals with ASD and/or with intellectual disability (ID). Complete phenotypic evaluation of the pedigrees in the ASD individuals showed common specific autistic behavioural features and neurophysiological patterns (abnormal MisMatch Negativity in response to auditory change) that were absent in healthy parents as well as in family members with isolated ID. Whole-exome sequencing in ASD patients from each family identified a second rare inherited genetic variant, affecting either the GLRB or the ANK3 genes encoding NLGN4X interacting proteins expressed in inhibitory or in excitatory synapses, respectively. The GRLB and ANK3 mutations were absent in relatives with ID as well as in control databases. In summary, our findings provide evidence of a double-hit genetic model focused on excitatory/inhibitory synapses in ASD, that is not found in isolated ID, associated with an atypical in vivo neurophysiological pattern linked to predictive coding.


Assuntos
Transtorno Autístico/complicações , Transtorno Autístico/genética , Moléculas de Adesão Celular Neuronais/genética , Potenciais Evocados Auditivos/fisiologia , Genômica , Deficiência Intelectual/etiologia , Estimulação Acústica , Pré-Escolar , Eletroencefalografia , Potenciais Evocados Auditivos/genética , Saúde da Família , Feminino , Seguimentos , Predisposição Genética para Doença , Ácido Glutâmico , Humanos , Masculino , Índice de Gravidade de Doença , Transdução de Sinais/genética , Ácido gama-Aminobutírico
5.
J Comp Neurol ; 523(9): 1359-78, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25565602

RESUMO

We studied the effect of clonal overexpression of neuroligin 3 (NL3) or neuroligin 2 (NL2) in the adult rat cerebral cortex following in utero electroporation (IUEP) at embryonic stage E14. Overexpression of NL3 leads to a large increase in vesicular gamma-aminobutyric acid (GABA) transporter (vGAT) and glutamic acid decarboxylase (GAD)65 in the GABAergic contacts that the overexpressing neurons receive. Overexpression of NL2 produced a similar effect but to a lesser extent. In contrast, overexpression of NL3 or NL2 after IUEP does not affect vesicular glutamate transporter 1 (vGlut1) in the glutamatergic contacts that the NL3 or NL2-overexpressing neurons receive. The NL3 or NL2-overexpressing neurons do not show increased innervation by parvalbumin-containing GABAergic terminals or increased parvalbumin in the same terminals that show increased vGAT. These results indicate that the observed increase in vGAT and GAD65 is not due to increased GABAergic innervation but to increased expression of vGAT and GAD65 in the GABAergic contacts that NL3 or NL2-overexpressing neurons receive. The majority of bright vGAT puncta contacting the NL3-overexpressing neurons have no gephyrin juxtaposed to them, indicating that many of these contacts are nonsynaptic. This contrasts with the majority of the NL2-overexpressing neurons, which show plenty of synaptic gephyrin clusters juxtaposed to vGAT. Besides having an effect on GABAergic contacts, overexpression of NL3 interferes with the neuronal radial migration, in the cerebral cortex, of the neurons overexpressing NL3.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular/fisiologia , Córtex Cerebral/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Adjuvantes Imunológicos , Animais , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Eletroporação , Glutamato Descarboxilase/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Parvalbuminas/metabolismo , Ratos Sprague-Dawley , Ratos Wistar , Sinapses/metabolismo , Transfecção , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
6.
Eur Neuropsychopharmacol ; 24(8): 1324-36, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24946696

RESUMO

Both genetic and environmental factors play important roles in the pathophysiology of schizophrenia. Although prenatal hypoxia is a potential environmental factor implicated in schizophrenia, very little is known about the consequences of combining models of genetic risk factor with prenatal hypoxia. Heterozygous reeler (haploinsufficient for reelin; HRM) and wild-type (WT) mice were exposed to prenatal hypoxia (9% oxygen for two hour) or normoxia at embryonic day 17 (E17). Behavioral (Prepulse inhibition, Y-maze and Open field) and functional (regional volume in frontal cortex and hippocampus as well as hippocampal blood flow) tests were performed at 3 months of age. The levels of hypoxia and stress-related molecules such as hypoxia-inducible factor-1 α (HIF-1α), vascular endothelial factor (VEGF), VEGF receptor-2 (VEGFR2/Flk1) and glucocorticoid receptor (GR) were examined in frontal cortex and hippocampus at E18, 1 month and 3 months of age. In addition, serum VEGF and corticosterone levels were also examined. Prenatal hypoxia induced anxiety-like behavior in both HRM and WT mice. A significant reduction in hippocampal blood flow, but no change in brain regional volume was observed following prenatal hypoxia. Significant age and region-dependent changes in HIF-1α, VEGF, Flk1 and GR were found following prenatal hypoxia. Serum VEGF and corticosterone levels were found decreased following prenatal hypoxia. None of the above prenatal hypoxia-induced changes were either diminished or exacerbated due to reelin deficiency. These results argue against any gene-environment interaction between hypoxia and reelin deficiency.


Assuntos
Interação Gene-Ambiente , Hipóxia/complicações , Hipóxia/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Esquizofrenia/etiologia , Esquizofrenia/genética , Estimulação Acústica , Análise de Variância , Animais , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Corticosterona/sangue , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Lobo Frontal/patologia , Regulação da Expressão Gênica , Técnicas de Genotipagem , Hipocampo/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inibição Psicológica , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Mutantes Neurológicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Psicoacústica , Proteína Reelina , Fluxo Sanguíneo Regional , Esquizofrenia/sangue , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Fator A de Crescimento do Endotélio Vascular/sangue , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
Anim Genet ; 45(1): 59-66, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24033492

RESUMO

Obesity has reached epidemic proportions globally and has become the cause of several major health risks worldwide. Presently, more than 100 loci have been related to obesity and metabolic traits in humans by genome-wide association studies. The complex genetic architecture behind obesity has triggered a need for the development of better animal models than rodents. The pig has emerged as a very promising biomedical model to study human obesity traits. In this study, we have characterized the expression patterns of six obesity-related genes, leptin (LEP), leptin receptor (LEPR), melanocortin 4 receptor (MC4R), fat mass and obesity associated (FTO), neuronal growth regulator 1 (NEGR)1 and adiponectin (ADIPOQ), in seven obesity-relevant tissues (liver; muscle; pancreas; hypothalamus; and retroperitoneal, subcutaneous and mesenteric adipose tissues) in two pig breeds (production pigs and Göttingen minipigs) that deviate phenotypically and genetically from each other with respect to obesity traits. We observe significant differential expression for LEP, LEPR and ADIPOQ in muscle and in all three adipose tissues. Interestingly, in pancreas, LEP expression is only detected in the fat minipigs. FTO shows significant differential expression in all tissues analyzed, and NEGR1 shows significant differential expression in muscle, pancreas, hypothalamus and subcutaneous adipose tissue. The MC4R transcript can be detected only in hypothalamus. In general, the expression profiles of the investigated genes are in accordance with those observed in human studies. Our study shows that both the differences between the investigated breeds and the phenotypic state with respect to obesity/leanness play a large role for differential expression of the obesity-related genes.


Assuntos
Obesidade/genética , Sus scrofa/genética , Transcriptoma , Adiponectina/genética , Tecido Adiposo/metabolismo , Animais , Cruzamento , Moléculas de Adesão Celular Neuronais/genética , Feminino , Humanos , Hipotálamo/metabolismo , Leptina/genética , Músculos/metabolismo , Pâncreas/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Receptores para Leptina/genética
8.
Am J Hum Genet ; 92(3): 375-86, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23472757

RESUMO

NRXN1 microdeletions occur at a relatively high frequency and confer increased risk for neurodevelopmental and neurobehavioral abnormalities. The mechanism that makes NRXN1 a deletion hotspot is unknown. Here, we identified deletions of the NRXN1 region in affected cohorts, confirming a strong association with the autism spectrum and other neurodevelopmental disorders. Interestingly, deletions in both affected and control individuals were clustered in the 5' portion of NRXN1 and its immediate upstream region. To explore the mechanism of deletion, we mapped and analyzed the breakpoints of 32 deletions. At the deletion breakpoints, frequent microhomology (68.8%, 2-19 bp) suggested predominant mechanisms of DNA replication error and/or microhomology-mediated end-joining. Long terminal repeat (LTR) elements, unique non-B-DNA structures, and MEME-defined sequence motifs were significantly enriched, but Alu and LINE sequences were not. Importantly, small-size inverted repeats (minus self chains, minus sequence motifs, and partial complementary sequences) were significantly overrepresented in the vicinity of NRXN1 region deletion breakpoints, suggesting that, although they are not interrupted by the deletion process, such inverted repeats can predispose a region to genomic instability by mediating single-strand DNA looping via the annealing of partially reverse complementary strands and the promoting of DNA replication fork stalling and DNA replication error. Our observations highlight the potential importance of inverted repeats of variable sizes in generating a rearrangement hotspot in which individual breakpoints are not recurrent. Mechanisms that involve short inverted repeats in initiating deletion may also apply to other deletion hotspots in the human genome.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Variações do Número de Cópias de DNA , Sequências Repetidas Invertidas , Transtornos Mentais/genética , Proteínas do Tecido Nervoso/genética , Deleção de Sequência , Proteínas de Ligação ao Cálcio , Estudos de Coortes , Replicação do DNA/genética , DNA de Forma B/genética , DNA de Cadeia Simples/genética , Éxons , Predisposição Genética para Doença , Instabilidade Genômica , Humanos , Moléculas de Adesão de Célula Nervosa , Sequências Repetidas Terminais
9.
Neuroscience ; 238: 114-24, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23438760

RESUMO

Neuronal network remodeling during critical periods of sensory development might be accompanied by alterations in hypothalamic cell populations. MicroRNAs play a central role in regulating neuronal function, including neural stem cell proliferation, and neuronal migration, maturation and integration into viable circuits by modulating different mRNA targets. Here we investigated the role of miR-138 in cell proliferation and migration in a neuron-enriched hypothalamic cell culture prepared from chicks on embryonic day 16. Ectopic expression of miR-138 enhanced hypothalamic cell migration, but did not affect cell proliferation. As a potential mechanism for miR-138's effect on cell migration, we investigated reelin (Reln) as a direct target of miR-138. Luciferase reporter assay and Ago2-immunoprecipitation experiments confirmed direct binding of miR-138 to the Reln 3'-untranslated region. Ectopic miR-138 abolished Reln levels in hypothalamic cells and enhanced their migration, similar to Reln-antisense DNA. Furthermore, inhibition of Reln expression by miR-138 led to decreased phosphorylation level of the key component of Reln-regulated signaling cascades, Disabled 1. These findings describe miR-138 as a novel regulator of hypothalamic cell migration, acting at least in part via inhibition of Reln expression and leading to the inactivation of Reln signals.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular/genética , Proteínas da Matriz Extracelular/metabolismo , Hipotálamo/metabolismo , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Serina Endopeptidases/metabolismo , Regiões 3' não Traduzidas , Animais , Moléculas de Adesão Celular Neuronais/genética , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Proteínas da Matriz Extracelular/genética , Hipotálamo/citologia , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Proteína Reelina , Serina Endopeptidases/genética , Transdução de Sinais/genética
10.
Neuropsychobiology ; 66(1): 57-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22797278

RESUMO

BACKGROUND: Reelin is under epigenetic control and has been reported to be decreased in cortical regions in schizophrenia. METHODS: To establish if expression of reelin is altered in specific cortical, hippocampal or thalamic regions of schizophrenia patients, we measured gene expression of reelin in a postmortem study of elderly patients with schizophrenia and non-affected controls in both hemispheres differentiating between gray and white matter. We compared cerebral postmortem samples (dorsolateral prefrontal cortex BA9 and BA46, superior temporal cortex BA22, entorhinal cortex BA28, sensoric cortex BA1-3, hippocampus, CA4, mediodorsal nucleus of the thalamus) from 12 schizophrenia patients with 13 normal subjects investigating gene expression of reelin in the gray and white matter of both hemispheres by in situ-hybridization. RESULTS: The left prefrontal area (BA9) of schizophrenia patients revealed a decreased expression of reelin-mRNA of 29.1% in the white (p = 0.022) and 13.6% in the gray matter (p = 0.007) compared to the control group. None of the other regions examined showed any statistically significant differences. CONCLUSION: Since reelin is responsible for migration and synapse formation, the decreased gene expression of reelin in the left prefrontal area of schizophrenia patients points to neurodevelopmental deficits in neuronal migration and synaptic plasticity. However, our study group was small, and results should be verified using larger samples.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/análise , Esquizofrenia/metabolismo , Serina Endopeptidases/metabolismo , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Moléculas de Adesão Celular Neuronais/genética , Movimento Celular/genética , Movimento Celular/fisiologia , Córtex Cerebral/metabolismo , Proteínas da Matriz Extracelular/genética , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Humanos , Hibridização In Situ , Masculino , Núcleo Mediodorsal do Tálamo/metabolismo , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal , Neurônios/metabolismo , Proteína Reelina , Esquizofrenia/genética , Serina Endopeptidases/genética , Sinapses/genética , Sinapses/metabolismo , Tálamo/metabolismo
11.
PLoS One ; 6(6): e20982, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21687627

RESUMO

BACKGROUND: Structural variation in the neurexin-1 (NRXN1) gene increases risk for both autism spectrum disorders (ASD) and schizophrenia. However, the manner in which NRXN1 gene variation may be related to brain morphology to confer risk for ASD or schizophrenia is unknown. METHOD/PRINCIPAL FINDINGS: 53 healthy individuals between 18-59 years of age were genotyped at 11 single nucleotide polymorphisms of the NRXN1 gene. All subjects received structural MRI scans, which were processed to determine cortical gray and white matter lobar volumes, and volumes of striatal and thalamic structures. Each subject's sensorimotor function was also assessed. The general linear model was used to calculate the influence of genetic variation on neural and cognitive phenotypes. Finally, in silico analysis was conducted to assess potential functional relevance of any polymorphisms associated with brain measures. A polymorphism located in the 3' untranslated region of NRXN1 significantly influenced white matter volumes in whole brain and frontal lobes after correcting for total brain volume, age and multiple comparisons. Follow-up in silico analysis revealed that this SNP is a putative microRNA binding site that may be of functional significance in regulating NRXN1 expression. This variant also influenced sensorimotor performance, a neurocognitive function impaired in both ASD and schizophrenia. CONCLUSIONS: Our findings demonstrate that the NRXN1 gene, a vulnerability gene for SCZ and ASD, influences brain structure and cognitive function susceptible in both disorders. In conjunction with our in silico results, our findings provide evidence for a neural and cognitive susceptibility mechanism by which the NRXN1 gene confers risk for both schizophrenia and ASD.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Transtornos Globais do Desenvolvimento Infantil/genética , Lobo Frontal/metabolismo , Predisposição Genética para Doença/genética , Proteínas do Tecido Nervoso/genética , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética , Adulto , Proteínas de Ligação ao Cálcio , Criança , Transtornos Globais do Desenvolvimento Infantil/fisiopatologia , Cognição , Biologia Computacional , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Moléculas de Adesão de Célula Nervosa , Esquizofrenia/fisiopatologia , Adulto Jovem
12.
Neural Dev ; 6: 3, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21214893

RESUMO

BACKGROUND: The dorsal thalamus acts as a gateway and modulator for information going to and from the cerebral cortex. This activity requires the formation of reciprocal topographic axon connections between thalamus and cortex. The axons grow along a complex multistep pathway, making sharp turns, crossing expression boundaries, and encountering intermediate targets. However, the cellular and molecular components mediating these steps remain poorly understood. RESULTS: To further elucidate the development of the thalamocortical system, we first created a thalamocortical axon reporter line to use as a genetic tool for sensitive analysis of mutant mouse phenotypes. The TCA-tau-lacZ reporter mouse shows specific, robust, and reproducible labeling of thalamocortical axons (TCAs), but not the overlapping corticothalamic axons, during development. Moreover, it readily reveals TCA pathfinding abnormalities in known cortical mutants such as reeler. Next, we performed an unbiased screen for genes involved in thalamocortical development using random mutagenesis with the TCA reporter. Six independent mutant lines show aberrant TCA phenotypes at different steps of the pathway. These include ventral misrouting, overfasciculation, stalling at the corticostriatal boundary, and invasion of ectopic cortical cell clusters. An outcross breeding strategy coupled with a genomic panel of single nucleotide polymorphisms facilitated genetic mapping with small numbers of mutant mice. We mapped a ventral misrouting mutant to the Emx2 gene, and discovered that some TCAs extend to the olfactory bulbs in this mutant. Mapping data suggest that other lines carry mutations in genes not previously known for roles in thalamocortical development. CONCLUSIONS: These data demonstrate the feasibility of a forward genetic approach to understanding mammalian brain morphogenesis and wiring. A robust axonal reporter enabled sensitive analysis of a specific axon tract inside the mouse brain, identifying mutant phenotypes at multiple steps of the pathway, and revealing a new aspect of the Emx2 mutant. The phenotypes highlight vulnerable choice points and latent tendencies of TCAs, and will lead to a refined understanding of the elements and interactions required to form the thalamocortical system.


Assuntos
Axônios/fisiologia , Córtex Cerebral , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Mutação/genética , Fenótipo , Tálamo , Fatores de Transcrição/genética , Alquilantes/farmacologia , Animais , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Embrião de Mamíferos , Etilnitrosoureia/farmacologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Testes Genéticos/métodos , Óperon Lac/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/anormalidades , Vias Neurais/embriologia , Vias Neurais/crescimento & desenvolvimento , Proteína Reelina , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Tálamo/embriologia , Tálamo/crescimento & desenvolvimento , Tálamo/metabolismo , beta-Galactosidase/metabolismo
13.
Proc Natl Acad Sci U S A ; 107(10): 4710-5, 2010 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-20176955

RESUMO

Despite considerable evidence for a critical role of neuroligin-1 in the specification of excitatory synapses, the cellular mechanisms and physiological roles of neuroligin-1 in mature neural circuits are poorly understood. In mutant mice deficient in neuroligin-1, or adult rats in which neuroligin-1 was depleted, we have found that neuroligin-1 stabilizes the NMDA receptors residing in the postsynaptic membrane of amygdala principal neurons, which allows for a normal range of NMDA receptor-mediated synaptic transmission. We observed marked decreases in NMDA receptor-mediated synaptic currents at afferent inputs to the amygdala of neuroligin-1 knockout mice. However, the knockout mice exhibited a significant impairment in spike-timing-dependent long-term potentiation (STD-LTP) at the thalamic but not the cortical inputs to the amygdala. Subsequent electrophysiological analyses indicated that STD-LTP in the cortical pathway is largely independent of activation of postsynaptic NMDA receptors. These findings suggest that neuroligin-1 can modulate, in a pathway-specific manner, synaptic plasticity in the amygdala circuits of adult animals, likely by regulating the abundance of postsynaptic NMDA receptors.


Assuntos
Tonsila do Cerebelo/fisiologia , Moléculas de Adesão Celular Neuronais/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Transmissão Sináptica/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Potenciais de Ação , Tonsila do Cerebelo/metabolismo , Animais , Western Blotting , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores , Humanos , Potenciação de Longa Duração , Camundongos , Camundongos Knockout , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Tálamo/metabolismo , Tálamo/fisiologia
14.
Psychopharmacology (Berl) ; 204(3): 511-21, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19277610

RESUMO

RATIONALE: Reelin, a large extracellular matrix glycoprotein, is down-regulated in the brain of schizophrenic patients and of heterozygous reeler mice (rl/+). The behavioral phenotype of rl/- mice, however, matches only partially the schizophrenia hallmarks. OBJECTIVES: We recently reported (Marrone et al., Eur J Neurosci 24:20062-22070, 2006) that homozygous reeler mutants (rl/rl) exhibit reduced density of parvalbumin-positive (PV+) GABAergic interneurons in anatomically circumscribed regions of the neostriatum. Assuming that in rl/+ mice may also show regional reduction of striatal GABAergic interneurons, behavioral impairments should selectively emerge in tasks depending on specifically altered striatal circuits. MATERIALS AND METHODS: We mapped the density of striatal PV+ interneurons in rl/+ and wild-type (+/+) mice and measured their performance in tasks depending on distinct striatal subregions. RESULTS: Our findings show that, contrary to what would be expected on the basis of gene dosage criteria, the striatal regions in which rl/rl mice exhibited decreased density of PV+ interneurons were either unaltered (rostral striatum) or equally altered (dorsomedial and ventromedial intermediate striatum, caudal striatum) in rl/+ mice. The anatomical findings were paralleled by behavioral deficits in fear extinction and latent inhibition, respectively, requiring the dorsomedial and ventromedial striatal regions. Conversely, active avoidance performance, which requires the dorsolateral region, was unaffected. CONCLUSIONS: Reelin haploinsufficiency alters the density of PV+ neurons in circumscribed regions of the striatum and selectively disrupts behaviors sensitive to dysfunction of these targeted regions. This aspect should be considered when designing experiments aimed at evaluating the impact of reelin haploinsufficiency in schizophrenia-associated cognitive disturbances in rl/+ mutants.


Assuntos
Comportamento Animal/fisiologia , Moléculas de Adesão Celular Neuronais/genética , Proteínas da Matriz Extracelular/genética , Neostriado/citologia , Neostriado/fisiologia , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Parvalbuminas/genética , Serina Endopeptidases/genética , Estimulação Acústica , Animais , Aprendizagem da Esquiva/fisiologia , Contagem de Células , Extinção Psicológica/fisiologia , Medo/fisiologia , Feminino , Haplótipos , Heterozigoto , Imuno-Histoquímica , Interneurônios/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Rede Nervosa/citologia , Rede Nervosa/fisiologia , Desempenho Psicomotor/fisiologia , Proteína Reelina , Ácido gama-Aminobutírico/fisiologia
15.
Eur J Neurosci ; 27(10): 2568-74, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18547243

RESUMO

Post mortem, reduced brain reelin is noted in schizophrenia. Accordingly, the reelin-haploinsufficient heterozygous reeler mouse (HRM) has been posited as a murine model of the illness. One study reported that HRM exhibit deficits in prepulse inhibition (PPI) of the acoustic startle reflex, a sensorimotor-gating behavior that is disrupted in schizophrenia, although this finding has not been reproduced. To extend the characterization of putative sensorimotor-gating deficits in HRM, these mice were subjected to a sophisticated series of PPI tests. Mice were tested in a cross-modal PPI protocol that combined an acoustic prepulse with a tactile startle stimulus and also in a protocol that included varying prepulse-pulse intervals and varying acoustic startle pulse intensities. Levels of acoustic startle habituation and cross-modal PPI were significantly lower in HRM, although unimodal PPI did not differ. The HRM also exhibited increased PPI compared to wildtypes at short interstimulus intervals between prepulse and pulse stimuli when the interval between the acoustic prepulse and pulse were varied, and were more reactive to higher intensity startle stimuli. Some of these deficits in sensorimotor gating parallel those of schizophrenia, a disease characterized by alterations in synaptic protein expression. Therefore, levels of presynaptic proteins were measured in multiple brain regions using ELISA in HRM. No significant alterations in presynaptic protein expression were found. Thus, HRM exhibit a complex pattern of changes in startle reactivity and sensorimotor gating, with both similarities to and differences from schizophrenia. However, it is unlikely that these behavioral differences may be accounted for by altered regional levels of presynaptic proteins.


Assuntos
Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Inibição Neural/genética , Terminações Pré-Sinápticas/metabolismo , Reflexo Anormal/genética , Transtornos de Sensação/genética , Estimulação Acústica , Animais , Encéfalo/fisiopatologia , Química Encefálica/genética , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Predisposição Genética para Doença/genética , Heterozigoto , Masculino , Camundongos , Camundongos Mutantes Neurológicos , Proteínas do Tecido Nervoso/genética , Estimulação Física , Proteína Reelina , Reflexo de Sobressalto/genética , Esquizofrenia/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Transmissão Sináptica/genética
16.
Cereb Cortex ; 18(5): 1125-38, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17728262

RESUMO

The rat granular retrosplenial cortex (GRS) is a simplified cortex, with distinct stratification and, in the uppermost layers, distinct modularity. Thalamic and cortical inputs are segregated by layers and in layer 1 colocalize, respectively, with apical dendritic bundles originating from neurons in layers 2 or 5. To further investigate this organization, we turned to reelin-deficient reeler mouse and Shaking rat Kawasaki. We found that the disrupted lamination, evident in Nissl stains in these rodents, is in fact a patch-matrix mosaic of segregated afferents and dendrites. Patches consist of thalamocortical connections, visualized by vesicular glutamate transporter 2 (VGluT2) or AChE. The surrounding matrix consists of corticocortical terminations, visualized by VGluT1 or zinc. Dendrites concentrate in the matrix or patches, depending on whether they are OCAM positive (matrix) or negative (patches). In wild-type rodents and, presumably, mutants, OCAM(+) structures originate from layer 5 neurons. By double labeling for dendrites (filled by Lucifer yellow in fixed slice) and OCAM immunofluorescence, we ascertained 2 populations in reeler: dendritic branches either preferred (putative layer 5 neurons) or avoided (putative supragranular neurons) the OCAM(+) matrix. We conclude that input-target relationships are largely preserved in the mutant GRS and that dendrite-dendrite interactions involving OCAM influence the formation of the mosaic configuration.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Córtex Cerebral/patologia , Dendritos/patologia , Proteínas da Matriz Extracelular/genética , Camundongos Mutantes Neurológicos/anatomia & histologia , Proteínas do Tecido Nervoso/genética , Serina Endopeptidases/genética , Tálamo/patologia , Acetilcolinesterase/metabolismo , Animais , Agonistas de Aminoácidos Excitatórios , Feminino , Ácido Ibotênico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Vias Neurais , Células Piramidais/metabolismo , Células Piramidais/patologia , Células Piramidais/ultraestrutura , Ratos , Ratos Mutantes , Ratos Wistar , Proteína Reelina , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
17.
J Neuroendocrinol ; 19(11): 847-59, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17927663

RESUMO

Although it is well established that gonadotrophin-releasing hormone (GnRH) neurones and astrocytes maintain an intimate contact throughout development and adult life, the cell-surface molecules that may contribute to this adhesiveness remain largely unknown. In the peripheral nervous system, the glycosylphosphatidyl inositol (GPI)-anchored protein contactin is a cell-surface neuronal protein required for axonal-glial adhesiveness. A glial transmembrane protein recognised by neuronal contactin is receptor-like protein tyrosine phosphatase beta (RPTP beta), a phosphatase with structural similarities to cell adhesion molecules. In the present study, we show that contactin, and its preferred in cis partner Caspr1, are expressed in GnRH neurones. We also show that the RPTP beta mRNA predominantly expressed in hypothalamic astrocytes encodes an RPTP beta isoform (short RPTP beta) that uses its carbonic anhydrase (CAH) extracellular subdomain to interact with neuronal contactin. Immunoreactive contactin is most abundant in GnRH nerve terminals projecting to both the organum vasculosum of the lamina terminalis and median eminence, implying GnRH axons as an important site of contactin-dependent cell adhesiveness. GT1-7 immortalised GnRH neurones adhere to the CAH domain of RPTPbeta, and this adhesiveness is blocked when contactin GPI anchoring is disrupted or contactin binding capacity is immunoneutralised, suggesting that astrocytic RPTP beta interacts with neuronal contactin to mediate glial-GnRH neurone adhesiveness. Because the abundance of short RPTP beta mRNA increases in the female mouse hypothalamus (but not in the cerebral cortex) before puberty, it appears that an increased interaction between GnRH axons and astrocytes mediated by RPTP beta-contactin is a dynamic mechanism of neurone-glia communication during female sexual development.


Assuntos
Astrócitos/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Neurônios/metabolismo , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/metabolismo , Animais , Astrócitos/citologia , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Contactinas , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Hipotálamo/fisiologia , Camundongos , Neurônios/citologia , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
18.
Physiol Behav ; 92(1-2): 278-82, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17560618

RESUMO

Eating disorders constitute major medical health problems in the western world. Even though little is known about the molecular mechanisms behind abnormal eating behavior, it has become clear that the central nervous system (CNS), particularly the hypothalamus, plays a significant role. The anorexic anx/anx mouse is a unique model for studying food intake and energy expenditure. The anx mutation is linked to marked alterations in hypothalamic distributions of signal substances known to have potent regulatory roles in the control of food intake. Another mouse model that displays an anorectic phenotype similar to the anx/anx mouse is the Contactin KO mouse. This model displays very similar hypothalamic alterations as seen in the anx/anx mouse, arguing for a role of these specific hypothalamic changes in an anorectic phenotype. In human eating disorders, hypothalamic systems corresponding to those defective in mouse models could be compromised since autoantibodies against melanocortin peptides have been detected in anorectic and bulimic patients. These findings represent research avenues that may lead to a better understanding of eating disorders and development of targeted therapeutic approaches.


Assuntos
Anorexia/fisiopatologia , Regulação do Apetite/fisiologia , Moléculas de Adesão Celular Neuronais/fisiologia , Hipotálamo/fisiopatologia , Animais , Anorexia/imunologia , Autoanticorpos/imunologia , Moléculas de Adesão Celular Neuronais/genética , Contactinas , Modelos Animais de Doenças , Comportamento Alimentar/fisiologia , Humanos , Hipotálamo/fisiologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , alfa-MSH/imunologia
19.
J Neurophysiol ; 97(3): 2312-21, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17229826

RESUMO

Reelin signaling through the low-density lipoprotein receptor family members, apoliproprotein E receptor 2 (apoER2) and very-low-density lipoprotein receptor (VLDLR), plays a pivotal role in dictating neuronal lamination during embryonic brain development. Recent evidence suggests that this signaling system also plays a role in the postnatal brain to modulate synaptic transmission, plasticity, and cognitive behavior, mostly likely due to a functional coupling with N-methyl-d-aspartate (NMDA) receptors. In this study, we investigated the effects of reelin on the maturation of CA1 glutamatergic function using electrophysiological and biochemical approaches. In cultured hippocampal slices, reelin treatment increased the amplitude of AMPAR-mediated miniature excitatory postsynaptic currents and the evoked AMPA/NMDA receptor current ratios. In addition, reelin treatment also reduced the number of silent synapses, facilitated a developmental switch from NR2B to NR2A of NMDARs, and increased surface expression of AMPARs in CA1 tissue. In cultured hippocampal neurons from reeler embryos, reduced numbers of AMPAR subunit GluR1 and NMDAR subunit NR1 clustering were observed compared with those obtained from wild-type embryos. Supplementing reelin in the reeler culture obliterated these genotypic differences. These results demonstrate that reelin- and lipoprotein receptor-mediated signaling may operate during developmental maturation of hippocampal glutamatergic function and thus represent a potential important mechanism for controlling synaptic strength and plasticity in the postnatal hippocampus.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo , Proteínas do Tecido Nervoso/metabolismo , Serina Endopeptidases/metabolismo , Transdução de Sinais/fisiologia , Sinapses/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Relação Dose-Resposta à Radiação , Estimulação Elétrica/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Células Piramidais/efeitos da radiação , Proteína Reelina , Serina Endopeptidases/genética , Sinapses/efeitos dos fármacos , Transfecção/métodos
20.
Cancer Res ; 65(18): 8317-23, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16166308

RESUMO

We previously reported that during mouse embryogenesis, plexin D1 (plxnD1) is expressed on neuronal and endothelial cells. Endothelial cells gradually loose plxnD1 expression during development. Here we describe, using in situ hybridization, that endothelial plxnD1 expression is regained during tumor angiogenesis in a mouse model of brain metastasis. Importantly, we found PLXND1 expression also in a number of human brain tumors, both of primary and metastatic origin. Apart from the tumor vasculature, abundant expression was also found on tumor cells. Via panning of a phage display library, we isolated two phages that carry single-domain antibodies with specific affinity towards a PLXND1-specific peptide. Immunohistochemistry with these single-domain antibodies on the same tumors that were used for in situ hybridization confirmed PLXND1 expression on the protein level. Furthermore, both these phages and the derived antibodies specifically homed to vessels in brain lesions of angiogenic melanoma in mice after i.v. injection. These results show that PLXND1 is a clinically relevant marker of tumor vasculature that can be targeted via i.v. injections.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/genética , Moléculas de Adesão Celular Neuronais/biossíntese , Glicoproteínas de Membrana/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Sequência de Aminoácidos , Animais , Anticorpos/genética , Anticorpos/imunologia , Anticorpos/isolamento & purificação , Especificidade de Anticorpos , Bacteriófagos/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Camelídeos Americanos , Moléculas de Adesão Celular Neuronais/genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/terapia , Proteínas do Tecido Nervoso/genética , RNA/biossíntese , RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA