Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cereb Cortex ; 33(7): 3866-3881, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35989311

RESUMEN

Protein quality control (PQC) is essential for maintaining protein homeostasis and guarding the accuracy of neurodevelopment. Previously, we found that a conserved EBAX-type CRL regulates the protein quality of SAX-3/ROBO guidance receptors in Caenorhabditis elegans. Here, we report that ZSWIM8, the mammalian homolog of EBAX-1, is essential for developmental stability of mammalian brains. Conditional deletion of Zswim8 in the embryonic nervous system causes global cellular stress, partial perinatal lethality and defective migration of neural progenitor cells. CRISPR-mediated knockout of ZSWIM8 impairs spine formation and synaptogenesis in hippocampal neurons. Mechanistic studies reveal that ZSWIM8 controls protein quality of Disabled 1 (Dab1), a key signal molecule for brain development, thus protecting the signaling strength of Dab1. As a ubiquitin ligase enriched with intrinsically disordered regions (IDRs), ZSWIM8 specifically recognizes IDRs of Dab1 through a "disorder targets misorder" mechanism and eliminates misfolded Dab1 that cannot be properly phosphorylated. Adult survivors of ZSWIM8 CKO show permanent hippocampal abnormality and display severely impaired learning and memory behaviors. Altogether, our results demonstrate that ZSWIM8-mediated PQC is critical for the stability of mammalian brain development.


Asunto(s)
Proteína Reelina , Ubiquitina , Animales , Femenino , Embarazo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Ligasas , Mamíferos/metabolismo , Serina Endopeptidasas/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Tejido Nervioso/metabolismo
2.
Biochem Biophys Res Commun ; 664: 117-127, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37146559

RESUMEN

Diabetic retinopathy (DR) is a common microvascular complication of diabetes mellitus. Reelin, an extracellular matrix protein, and its effector protein Disabled1 (DAB1) have been linked to cellular events and retinal development. However, whether and how Reelin/DAB1 signaling causes DR remains to be investigated. In our study, significantly increased expression of Reelin, very low density lipoprotein receptor (VLDLR), ApoE receptor 2 (ApoER2) and phosphorylated DAB1 in retinas of streptozotocin (STZ)-induced DR mouse model was observed, along with enhanced expression of proinflammatory factors. Similar results are confirmed in high glucose (HG)-treated human retinal pigment epithelium cell line ARPE-19. Surprisingly, dysregulated tripartite motif-containing 40 (TRIM40), an E3 ubiquitin ligase, is found to be involved in DR progression by bioinformatic analysis. We observe a negative correlation between TRIM40 and p-DAB1 protein expression levels under HG conditions. Importantly, we find that TRIM40 over-expression markedly ameliorates HG-induced p-DAB1, PI3K, p-protein B kinase (AKT) and inflammatory response in HG-treated cells, but dose not affect Reelin expression. Of note, Co-IP and double immunofluorescence identify an interaction between TRIM40 and DAB1. Furthermore, we show that TRIM40 enhances K48-linked polyubiquitination of DAB1, thereby promoting DAB1 degradation. Finally, promoting TRIM40 expression by intravenous injection of the constructed adeno-associated virus (AAV-TRIM40) markedly ameliorates DR phenotypes in STZ-treated mice, as indicated by the decreased blood glucose and glycosylated hemoglobin (HbAlc) levels, and increased hemoglobin contents. Additionally, diabetes-related elevation of acellular capillaries was also meliorated in mice over-expressing TRIM40. The electroretinogram (ERG) deficits were strongly rescued in mice receiving AAV-TRIM40 injection. Moreover, AAV-TRIM40 attenuates the inflammation and p-DAB1 expression in retinal tissues of STZ-treated mice. Collectively, our findings disclose a mechanism through which TRIM40 limits DAB1 stability under physiological conditions and reveals TRIM40 as a potential therapeutic target for the intervention of Reelin/DAB1 signaling, contributing to DR treatment.


Asunto(s)
Retinopatía Diabética , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Inflamación , Proteínas del Tejido Nervioso/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transducción de Señal
3.
Int J Neurosci ; : 1-15, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38060511

RESUMEN

INTRODUCTION: Granule cell dispersion (GCD) is pathognomonic of hippocampal sclerosis seen in the mesial temporal lobe epilepsy (MTLE). Current animal studies indicate deficiency of Reelin is associated with abnormal granule cell migration leading to GCD. The present study aimed to evaluate complete Reelin signalling pathway to assess whether Reelin deficiency is related to MTLE. MATERIALS AND METHODS: Hippocampal sclerosis was confirmed by H and E stain. To explore the amount and cellular location of the Reelin cascade molecules, the hippocampal tissues from MTLE surgery and controls (n = 15 each) were studied using Immuno-histochemistry (IHC). Additionally, confocal imaging was used to validate the IHC findings by co-localization of different proteins. Quantification of IHC images was performed using histo-score and confocal images by Image J software. RESULTS: Immune expression of active Reelin was significantly reduced in patients. Reelin receptors were deranged, apolipoprotein E receptor 2 was increased while very low-density lipoprotein receptor was reduced. Disabled-1, a downstream molecule was significantly reduced in MTLE. Its ultimate target, cofilin was thus disinhibited and expressed more in MTLE. Reelin cleaving protease, matrix metalloprotease-9 (MMP-9) and MMP-9 inhibitor, tissue inhibitor of matrix protease-1, showed reduced expression in extracellular matrix. Semi-quantification of immunohistochemistry was done using Histo (H) score. H score of Reelin in diseased patients was 15 against 125 for control patients. These results were validated by confocal fluorescence microscopy. CONCLUSIONS: Reelin signalling cascade was deranged in chronic MTLE. Pharmacological manipulation of Reelin cascade can be done at various levels and it may provide novel treatment options for MTLE.

4.
Toxicol Ind Health ; 39(2): 104-114, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36617730

RESUMEN

Aluminum (Al) is a common neurotoxic element that can exacerbate intracellular ß-amyloid (Aß) deposition. Reelin is a highly conserved extracellular glycoprotein that is involved in intracellular Aß deposition. However, the action of Reelin on aluminum-induced Aß deposition is not fully understood. Here, we investigated the effects of the Reelin-Dab1 signaling pathway on Aß deposition in aluminum maltol (Al(mal)3) exposure in rat pheochromocytoma-derived cells (PC12). Our results showed that Al(mal)3 exposure decreased activity of PC12, increased expression of Aß42, and decreased expression of Aß40. Moreover, Al(mal)3 exposure in PC12 induced Reelin-Dab1 signaling pathway-associated proteins changed, decreased expression of Reelin and Dab1, and increased expression of pdab1. Moreover, the expression of Reelin, Dab1, and Aß40 was found to be elevated in PC12 exposed to Al(mal)3 and corticosterone compared to those exposed to Al(mal)3. Also, the expression of Reelin, Dab1, and Aß40 was found to be depressed in PC12 exposed to Al(mal)3 and streptozotocin compared with cells exposed to Al(mal)3 alone. These results suggested that Al(mal)3 inhibits the expression of the Reelin-Dab1 signaling pathway, promoting Aß deposition. Thus, our findings provided important evidence to better understand how the Reelin-Dab1 signaling pathway may be a potential mechanism of Aß deposition induced by aluminum.


Asunto(s)
Aluminio , Proteínas de la Matriz Extracelular , Animales , Ratas , Aluminio/toxicidad , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transducción de Señal , Péptidos beta-Amiloides/metabolismo
5.
Mov Disord ; 37(12): 2427-2439, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36148898

RESUMEN

BACKGROUND: Coding and noncoding repeat expansions are an important cause of neurodegenerative diseases. OBJECTIVE: This study determined the clinical and genetic features of a large German family that has been followed for almost 2 decades with an autosomal dominantly inherited spinocerebellar ataxia (SCA) and independent co-occurrence of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). METHODS: We carried out clinical examinations and telephone interviews, reviewed medical records, and performed magnetic resonance imaging and positron emission tomography scans of all available family members. Comprehensive genetic investigations included linkage analysis, short-read genome sequencing, long-read sequencing, repeat-primed polymerase chain reaction, and Southern blotting. RESULTS: The family comprises 118 members across seven generations, 30 of whom were definitely and five possibly affected. In this family, two different pathogenic mutations were found, a heterozygous repeat expansion in C9ORF72 in four patients with ALS/FTD and a heterozygous repeat expansion in DAB1 in at least nine patients with SCA, leading to a diagnosis of DAB1-related ataxia (ATX-DAB1; SCA37). One patient was affected by ALS and SCA and carried both repeat expansions. The repeat in DAB1 had the same configuration but was larger than those previously described ([ATTTT]≈75 [ATTTC]≈40-100 [ATTTT]≈415 ). Clinical features in patients with SCA included spinocerebellar symptoms, sometimes accompanied by additional ophthalmoplegia, vertical nystagmus, tremor, sensory deficits, and dystonia. After several decades, some of these patients suffered from cognitive decline and one from additional nonprogressive lower motor neuron affection. CONCLUSION: We demonstrate genetic and clinical findings during an 18-year period in a unique family carrying two different pathogenic repeat expansions, providing novel insights into their genotypic and phenotypic spectrums. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ataxia Cerebelosa , Demencia Frontotemporal , Ataxias Espinocerebelosas , Humanos , Demencia Frontotemporal/diagnóstico por imagen , Demencia Frontotemporal/genética , Esclerosis Amiotrófica Lateral/diagnóstico por imagen , Esclerosis Amiotrófica Lateral/genética , Proteína C9orf72/genética , Expansión de las Repeticiones de ADN/genética , Ataxia Cerebelosa/genética , Ataxias Espinocerebelosas/genética , Proteínas del Tejido Nervioso/genética , Proteínas Adaptadoras Transductoras de Señales/genética
6.
Pharmacol Res ; 182: 106355, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35842183

RESUMEN

Obesity-prone (OP) individuals have a significant predisposition to obesity and diabetes. Previously, we have found that OP individuals, despite being normal in weight and BMI, have already exhibited diabetes-related DNA methylation signatures. However, the underlying mechanisms remain obscure. Here we determined the effects of gut microbiota on DNA methylation and investigated the underlying mechanism from microbial-derived short-chain fatty acids (SCFAs). Diabetes-related DNA methylation loci were screened and validated in a new OP cohort. Moreover, the OP group was revealed to have distinct gut microbiota compositions, and fecal microbiota transplantation (FMT) demonstrated the role of gut microbiota in inducing diabetes-related DNA methylations and glucolipid disorders. UPLC-ESI-MS/MS analysis indicated a significantly lower level of total fecal SCFAs in the OP group. The gut microbiota from OP subjects yielded markedly decreased total SCFAs, while notably enriched propionate. Additionally, propionate was also identified by variable importance in projection (VIP) score as the most symbolic SCFAs of the OP group. Further cellular experiments verified that propionate could induce hypermethylation at locus cg26345888 and subsequently inhibit the expression of the target gene DAB1, which was crucially associated with clinical vitamin D deficiency and thus may affect the development and progression of diabetes. In conclusion, our study revealed that gut microbiota-derived propionate induces specific DNA methylation, thus predisposing OP individuals to diabetes. The findings partially illuminate the mechanisms of diabetes susceptibility in OP populations, implying gut microbiota and SCFAs may serve as promising targets both for clinical treatment and medication development of diabetes.


Asunto(s)
Diabetes Mellitus , Microbioma Gastrointestinal , Metilación de ADN , Ácidos Grasos Volátiles/metabolismo , Humanos , Obesidad/genética , Obesidad/metabolismo , Propionatos/farmacología , Espectrometría de Masas en Tándem
7.
J Neurosci ; 40(40): 7625-7636, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32913108

RESUMEN

Oligodendrocyte (OL) progenitor cells (OPCs) are generated, proliferate, migrate, and differentiate in the developing brain. Although the development of OPCs is prerequisite for normal brain function, the molecular mechanisms regulating their development in the neocortex are not fully understood. Several molecules regulate the tangential distribution of OPCs in the developing neocortex, but the cue molecule(s) that regulate their radial distribution remains unknown. Here, we demonstrate that the secreted glycoprotein Reelin suppresses the proliferation of OPCs and acts as a repellent for their migration in vitro These functions rely on the binding of Reelin to its receptors and on the signal transduction involving the intracellular protein Dab1. In the late embryonic neocortex of mice with attenuated Reelin signaling [i.e., Reelin heterozygote-deficient, Dab1 heterozygote-deficient mutant, or very low-density lipoprotein receptor (VLDLR)-deficient mice], the number of OPCs increased and their distribution shifted toward the superficial layers. In contrast, the number of OPCs decreased and they tended to distribute in the deep layers in the neocortex of mice with abrogated inactivation of Reelin by proteolytic cleavage, namely a disintegrin and metalloproteinase with thrombospondin type 1 motifs 3 (ADAMTS-3)-deficient mice and cleavage-resistant Reelin knock-in mice. Both male and female animals were used. These data indicate that Reelin-Dab1 signaling regulates the proliferation and radial distribution of OPCs in the late embryonic neocortex and that the regulation of Reelin function by its specific proteolysis is required for the normal development of OPCs.SIGNIFICANCE STATEMENT Here, we report that Reelin-Dab1 signaling regulates the proliferation and radial distribution of OPCs in the late embryonic mouse neocortex. Oligodendrocyte (OL) progenitor cells (OPCs) express Reelin signaling molecules and respond to Reelin stimulation. Reelin-Dab1 signaling suppresses the proliferation of OPCs both in vitro and in vivo Reelin repels OPCs in vitro, and the radial distribution of OPCs is altered in mice with either attenuated or augmented Reelin-Dab1 signaling. This is the first report identifying the secreted molecule that plays a role in the radial distribution of OPCs in the late embryonic neocortex. Our results also show that the regulation of Reelin function by its specific proteolysis is important for the normal development of OPCs.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Oligodendroglía/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas ADAMTS/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/citología , Neocórtex/embriología , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Oligodendroglía/citología , Unión Proteica , Receptores de LDL/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética
8.
Development ; 145(3)2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29361558

RESUMEN

The laminated structure of the retina is fundamental for the organization of the synaptic circuitry that translates light input into patterns of action potentials. However, the molecular mechanisms underlying cell migration and layering of the retina are poorly understood. Here, we show that RBX2, a core component of the E3 ubiquitin ligase CRL5, is essential for retinal layering and function. RBX2 regulates the final cell position of rod bipolar cells, cone photoreceptors and Muller glia. Our data indicate that sustained RELN/DAB1 signaling, triggered by depletion of RBX2 or SOCS7 - a CRL5 substrate adaptor known to recruit DAB1 - causes rod bipolar cell misposition. Moreover, whereas SOCS7 also controls Muller glia cell lamination, it is not responsible for cone photoreceptor positioning, suggesting that RBX2, most likely through CRL5 activity, controls other signaling pathways required for proper cone localization. Furthermore, RBX2 depletion reduces the number of ribbon synapses and disrupts cone photoreceptor function. Together, these results uncover RBX2 as a crucial molecular regulator of retina morphogenesis and cone photoreceptor function.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Retina/embriología , Retina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Deleción Cromosómica , Cromosomas Humanos Par 3 , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Anomalías del Ojo/embriología , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Embarazo , Proteína Reelina , Retina/citología , Células Bipolares de la Retina/citología , Células Bipolares de la Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética
9.
Int J Mol Sci ; 22(4)2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572344

RESUMEN

Disabled 1 (Dab1) is an adapter protein for very low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2) and an integral component of the Reelin pathway which orchestrates neuronal layering during embryonic brain development. Activation of Dab1 is induced by binding of Reelin to ApoER2 and VLDLR and phosphorylation of Dab1 mediated by Src family kinases. Here we show that Dab1 also acts as an adaptor for epidermal growth factor receptor (EGFR) and can be phosphorylated by epidermal growth factor (EGF) binding to EGFR. Phosphorylation of Dab1 depends on the kinase activity of EGFR constituting a signal pathway independent of Reelin and its receptors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Proliferación Celular , Medios de Cultivo Condicionados/metabolismo , Embrión de Mamíferos , Factor de Crecimiento Epidérmico/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Células HEK293 , Humanos , Mucosa Intestinal/citología , Masculino , Ratones , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Neuronas , Fosforilación , Cultivo Primario de Células , Proteína Reelina , Serina Endopeptidasas/metabolismo , Transducción de Señal
10.
Eur J Neurosci ; 52(5): 3322-3338, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32492253

RESUMEN

Mice missing either Reelin or Disabled-1 (Dab1) exhibit dorsal horn neuronal positioning errors and display heat hypersensitivity and mechanical insensitivity. Reelin binds its receptors, apolipoprotein E receptor 2 and very low-density lipoprotein receptor, leading to the recruitment and phosphorylation of Dab1 and activation of downstream pathways that regulate neuronal migration. Previously, we reported that 70% of Dab1 laminae I-II neurons co-expressed LIM-homeobox transcription factor 1-beta (Lmx1b). Here, we asked whether Reelin-expressing dorsal horn neurons co-express Lmx1b, are mispositioned in dab1 mutants, and contribute to nociceptive abnormalities. About 90% of Reelin-labeled neurons are Lmx1b-positive in laminae I-II, confirming that most Reelin and Dab1 neurons are glutamatergic. We determined that Reelin-Lmx1b and Dab1-Lmx1b dorsal horn neurons are separate populations, and together, comprise 37% of Lmx1b-positive cells within and above the Isolectin B4 (IB4) layer in wild-type mice. Compared to wild-type mice, dab1 mutants have a reduced area of laminae I-II outer (above the IB4 layer), more Reelin-Lmx1b neurons within the IB4 layer, and fewer Reelin-Lmx1b neurons within the lateral reticulated area of lamina V and lateral spinal nucleus. Interestingly, both Reelin- and Dab1-labeled dorsal horn neurons sustain similar positioning errors in mutant mice. After noxious thermal and mechanical stimulation, Reelin, Lmx1b, and Reelin-Lmx1b neurons expressed Fos in laminae I-II and the lateral reticulated area in wild-type mice and, therefore, participate in nociceptive circuits. Together, our data suggest that disruption of the Reelin-signaling pathway results in neuroanatomical abnormalities that contribute to the nociceptive changes that characterize these mutant mice.


Asunto(s)
Moléculas de Adhesión Celular Neuronal , Proteínas de la Matriz Extracelular , Animales , Moléculas de Adhesión Celular Neuronal/genética , Movimiento Celular , Proteínas de la Matriz Extracelular/genética , Ratones , Proteínas del Tejido Nervioso/genética , Neuronas , Células del Asta Posterior , Proteína Reelina , Serina Endopeptidasas , Transducción de Señal , Asta Dorsal de la Médula Espinal
11.
Am J Hum Genet ; 101(1): 87-103, 2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28686858

RESUMEN

Advances in human genetics in recent years have largely been driven by next-generation sequencing (NGS); however, the discovery of disease-related gene mutations has been biased toward the exome because the large and very repetitive regions that characterize the non-coding genome remain difficult to reach by that technology. For autosomal-dominant spinocerebellar ataxias (SCAs), 28 genes have been identified, but only five SCAs originate from non-coding mutations. Over half of SCA-affected families, however, remain without a genetic diagnosis. We used genome-wide linkage analysis, NGS, and repeat analysis to identify an (ATTTC)n insertion in a polymorphic ATTTT repeat in DAB1 in chromosomal region 1p32.2 as the cause of autosomal-dominant SCA; this region has been previously linked to SCA37. The non-pathogenic and pathogenic alleles have the configurations [(ATTTT)7-400] and [(ATTTT)60-79(ATTTC)31-75(ATTTT)58-90], respectively. (ATTTC)n insertions are present on a distinct haplotype and show an inverse correlation between size and age of onset. In the DAB1-oriented strand, (ATTTC)n is located in 5' UTR introns of cerebellar-specific transcripts arising mostly during human fetal brain development from the usage of alternative promoters, but it is maintained in the adult cerebellum. Overexpression of the transfected (ATTTC)58 insertion, but not (ATTTT)n, leads to abnormal nuclear RNA accumulation. Zebrafish embryos injected with RNA of the (AUUUC)58 insertion, but not (AUUUU)n, showed lethal developmental malformations. Together, these results establish an unstable repeat insertion in DAB1 as a cause of cerebellar degeneration; on the basis of the genetic and phenotypic evidence, we propose this mutation as the molecular basis for SCA37.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , ADN Intergénico/genética , Predisposición Genética a la Enfermedad , Repeticiones de Microsatélite/genética , Proteínas del Tejido Nervioso/genética , Mapeo Físico de Cromosoma , Ataxias Espinocerebelosas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Adulto , Edad de Inicio , Alelos , Secuencia de Bases , Cerebelo/metabolismo , Segregación Cromosómica/genética , Cromosomas Humanos Par 1/genética , Análisis Mutacional de ADN , Desarrollo Embrionario/genética , Femenino , Células HEK293 , Haplotipos/genética , Humanos , Intrones/genética , Masculino , Persona de Mediana Edad , Mutagénesis Insercional/genética , Proteínas del Tejido Nervioso/metabolismo , Linaje , ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Reelina , Adulto Joven
12.
J Neurosci ; 38(1): 137-148, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29138282

RESUMEN

Reelin controls neuronal migration and layer formation. Previous studies in reeler mice deficient in Reelin focused on the result of the developmental process in fixed tissue sections. It has remained unclear whether Reelin affects the migratory process, migration directionality, or migrating neurons guided by the radial glial scaffold. Moreover, Reelin has been regarded as an attractive signal because newly generated neurons migrate toward the Reelin-containing marginal zone. Conversely, Reelin might be a stop signal because migrating neurons in reeler, but not in wild-type mice, invade the marginal zone. Here, we monitored the migration of newly generated proopiomelanocortin-EGFP-expressing dentate granule cells in slice cultures from reeler, reeler-like mutants and wild-type mice of either sex using real-time microscopy. We discovered that not the actual migratory process and migratory speed, but migration directionality of the granule cells is controlled by Reelin. While wild-type granule cells migrated toward the marginal zone of the dentate gyrus, neurons in cultures from reeler and reeler-like mutants migrated randomly in all directions as revealed by vector analyses of migratory trajectories. Moreover, live imaging of granule cells in reeler slices cocultured to wild-type dentate gyrus showed that the reeler neurons changed their directions and migrated toward the Reelin-containing marginal zone of the wild-type culture, thus forming a compact granule cell layer. In contrast, directed migration was not observed when Reelin was ubiquitously present in the medium of reeler slices. These results indicate that topographically administered Reelin controls the formation of a granule cell layer.SIGNIFICANCE STATEMENT Neuronal migration and the various factors controlling its onset, speed, directionality, and arrest are poorly understood. Slice cultures offer a unique model to study the migration of individual neurons in an almost natural environment. In the present study, we took advantage of the expression of proopiomelanocortin-EGFP by newly generated, migrating granule cells to analyze their migratory trajectories in hippocampal slice cultures from wild-type mice and mutants deficient in Reelin signaling. We show that the compartmentalized presence of Reelin is essential for the directionality, but not the actual migratory process or speed, of migrating granule cells leading to their characteristic lamination in the dentate gyrus.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Movimiento Celular/fisiología , Giro Dentado/citología , Proteínas de la Matriz Extracelular/fisiología , Proteínas del Tejido Nervioso/fisiología , Serina Endopeptidasas/fisiología , Animales , Movimiento Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Gránulos Citoplasmáticos/fisiología , Células Ependimogliales , Femenino , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Mutación , Neuronas/fisiología , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Proteína Reelina
13.
Hum Mutat ; 40(4): 404-412, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30588707

RESUMEN

Dynamic mutations by microsatellite instability are the molecular basis of a growing number of neuromuscular and neurodegenerative diseases. Repetitive stretches in the human genome may drive pathogenicity, either by expansion above a given threshold, or by insertion of abnormal tracts in nonpathogenic polymorphic repetitive regions, as is the case in spinocerebellar ataxia type 37 (SCA37). We have recently established that this neurodegenerative disease is caused by an (ATTTC)n insertion within an (ATTTT)n in a noncoding region of DAB1. We now investigated the mutational mechanism that originated the (ATTTC)n insertion within an ancestral (ATTTT)n . Approximately 3% of nonpathogenic (ATTTT)n alleles are interspersed by AT-rich motifs, contrarily to mutant alleles that are composed of pure (ATTTT)n and (ATTTC)n stretches. Haplotype studies in unaffected chromosomes suggested that the primary mutational mechanism, leading to the (ATTTC)n insertion, was likely one or more T>C substitutions in an (ATTTT)n pure allele of approximately 200 repeats. Then, the (ATTTC)n expanded in size, originating a deleterious allele in DAB1 that leads to SCA37. This is likely the mutational mechanism in three similar (TTTCA)n insertions responsible for familial myoclonic epilepsy. Because (ATTTT)n tracts are frequent in the human genome, many loci could be at risk for this mutational process.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Ataxinas/genética , Mutagénesis Insercional , Proteínas del Tejido Nervioso/genética , Secuencias Repetitivas de Ácidos Nucleicos , Alelos , Animales , Secuencia de Bases , Estudios de Casos y Controles , Cromosomas , Secuencia Conservada , Evolución Molecular , Haplotipos , Humanos , Filogenia , Portugal , Primates , Proteína Reelina
14.
Arterioscler Thromb Vasc Biol ; 38(4): 964-975, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29472232

RESUMEN

OBJECTIVE: The genetic contribution to coronary artery disease (CAD) remains largely unclear. We combined genetic screening with functional characterizations to identify novel loci and candidate genes for CAD. APPROACH AND RESULTS: We performed genome-wide screening followed by multicenter validation in 8 cohorts consisting of 21 828 participants of Han ethnicity and identified 3 novel intragenic SNPs (single nucleotide polymorphisms), rs9486729 (SCML4 [Scm polycomb group protein-like 4]; odds ratio, 1.25; 95% CI, 1.17-1.34; P=3.51×10-11), rs17165136 (THSD7A [thrombospondin type 1 domain-containing 7A]; odds ratio 1.28; 95% CI, 1.21-1.35; P<1.00×10-25), and rs852787 (DAB1 [disabled-1]; odds ratio, 1.29; 95% CI, 1.21-1.38; P=2.02×10-14), associated with CAD with genome-wide significance. The risk allele of rs9486729 and protective allele of rs17165136 were associated with the decreased expression of their host genes, SCML4 and THSD7A, respectively, whereas rs852787 did not have transcriptional effects on any gene. Knockdown of SCML4 activated endothelial cells by increasing the expression of IL-6, E-selectin, and ICAM and weakened their antiapoptotic activity, whereas the knockdown of THSD7A had little effect on these endothelial cell functions but attenuated monocyte adhesion via decreasing the expression of ICAM, L-selectin, and ITGB2. We further showed that inhibiting the expression of SCML4 exacerbated endothelial dysfunction and vascular remodeling in a rat model with partial carotid ligation. CONCLUSIONS: We identify 3 novel loci associated with CAD and show that 2 genes, SCML4 and THSD7A, make functional contributions to atherosclerosis. How rs852787 and its host gene DAB1 are linked to CAD needs further studies.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Proteínas del Grupo Polycomb/genética , Polimorfismo de Nucleótido Simple , Trombospondinas/genética , Adulto , Anciano , Animales , Pueblo Asiatico/genética , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Estenosis Carotídea/genética , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Células Cultivadas , China/epidemiología , Enfermedad de la Arteria Coronaria/diagnóstico , Enfermedad de la Arteria Coronaria/etnología , Enfermedad de la Arteria Coronaria/metabolismo , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Proteínas del Grupo Polycomb/metabolismo , Ratas Sprague-Dawley , Factores de Riesgo , Trombospondinas/metabolismo , Remodelación Vascular
15.
Hum Mutat ; 39(10): 1372-1383, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29969175

RESUMEN

The Reelin-DAB1 signaling pathway plays a crucial role in regulating neuronal migration and synapse function. Although many rare heterozygous variants in the Reelin gene (RELN) have been identified in patients with autism spectrum disorder (ASD), most variants are still of unknown clinical significance. Also, genetic data suggest that heterozygous variants in RELN alone appear to be insufficient to cause ASD. Here, we describe the identification and functional characterization of rare compound heterozygous missense variants in RELN in a patient with ASD in whom we have previously reported hyperfunctional mTORC1 signaling of yet unknown etiology. Using iPSC-derived neural progenitor cells (NPCs) from this patient, we provide experimental evidence that the identified variants are deleterious and lead to diminished Reelin secretion and impaired Reelin-DAB1 signal transduction. Also, our results suggest that mTORC1 pathway overactivation may function as a second hit event contributing to downregulation of the Reelin-DAB1 cascade in patient-derived NPCs, and that inhibition of mTORC1 by rapamycin attenuates Reelin-DAB1 signaling impairment. Taken together, our findings point to an abnormal interplay between Reelin-DAB1 and mTORC1 networks in nonsyndromic ASD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Variación Genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/química , Alelos , Trastorno del Espectro Autista/diagnóstico , Biomarcadores , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal/química , Niño , Preescolar , Proteínas de la Matriz Extracelular/química , Femenino , Expresión Génica , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Conformación Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Reelina , Serina Endopeptidasas/química , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/metabolismo
16.
Biochim Biophys Acta Biomembr ; 1860(5): 1231-1241, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29470947

RESUMEN

Disabled-1 (Dab1) is an essential intracellular adaptor protein in the reelin pathway. Our previous studies in mice intestine showed that Dab1 transmits the reelin signal to cytosolic signalling pathways. Here, we determine the Dab1 isoform expressed in rodent small and large intestine, its subcellular location and co-localization with clathrin, caveolin-1 and N-Wasp. PCR and sequencing analysis reveal that rodent small and large intestine express a Dab1 isoform that misses three (Y198, Y200 and Y220) of the five tyrosine phosphorylation sites present in brain Dab1 isoform (canonical) and contains nuclear localization and export signals. Western blot assays show that both, crypts, which shelter progenitor cells, and enterocytes express the same Dab1 isoform, suggesting that epithelial cell differentiation does not regulate intestinal generation of alternatively spliced Dab1 variants. They also reveal that the canonical and the intestinal Dab1 isoforms differ in their total degree of phosphorylation. Immunostaining assays show that in enterocytes Dab1 localizes at the apical and lateral membranes, apical vesicles, close to adherens junctions and desmosomes, as well as in the nucleus; co-localizes with clathrin and with N-Wasp but not with caveolin-1, and in Caco-2 cells Dab1 localizes at cell-to-cell junctions by a Ca2+-dependent process. In conclusion, the results indicate that in rodent intestine a truncated Dab1 variant transmits the reelin signal and may play a role in clathrin-mediated apical endocytosis and in the control of cell-to-cell junction assembly. A function of intestinal Dab1 variant as a nucleocytoplasmic shuttling protein is also inferred from its sequence and nuclear location.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endocitosis , Uniones Intercelulares/metabolismo , Intestino Grueso/metabolismo , Intestino Delgado/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células CACO-2 , Comunicación Celular/genética , Células Cultivadas , Endocitosis/genética , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Unión Proteica , Isoformas de Proteínas , Ratas , Ratas Wistar , Proteína Reelina , Distribución Tisular
17.
Cereb Cortex ; 27(3): 2052-2063, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965907

RESUMEN

Irregular neuronal migration plays a causal role in mental illnesses such as schizophrenia and autism, but the very nature of the migration deficits necessary to evoke adult behavioral changes is unknown. Here, we used in utero electroporation (IUE) in rats to induce a locally restricted, cortical migration deficit by knockdown of disabled-1 (Dab1), an intracellular converging point of the reelin pathway. After birth, selection of successfully electroporated rats by detection of in vivo bioluminescence of a simultaneously electroporated luciferase gene correlated to and was thus predictive to the number of electroporated neurons in postmortem histochemistry at 6 months of age. Rat neurons silenced for Dab1 did not migrate properly and their number surprisingly decreased after E22. Behavioral tests at adult ages (P180) revealed increased sensitivity to amphetamine as well as decreased habituation, but no deficits in memory tasks or motor functions. The data suggest that even subtle migration deficits involving only ten-thousands of cortical neurons during neurodevelopment can lead to lasting behavioral and neuronal changes into adulthood in some very specific behavioral domains. On the other hand, the lack of effects on various memory-related tasks may indicate resilience and plasticity of cognitive functions critical for survival under these specific conditions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Trastornos del Neurodesarrollo/fisiopatología , Neuronas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Anfetamina/farmacología , Animales , Línea Celular Tumoral , Estimulantes del Sistema Nervioso Central/farmacología , Modelos Animales de Enfermedad , Electroporación , Técnicas de Silenciamiento del Gen , Humanos , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/genética , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/fisiopatología , Trastornos del Neurodesarrollo/etiología , Ratas Sprague-Dawley , Proteína Reelina , Resiliencia Psicológica
18.
J Neurochem ; 142(1): 89-102, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28419454

RESUMEN

Despite the recent identification of over 40 missense heterozygous Reelin gene (RELN) mutations in autism spectrum disorder (ASD), none of these has been functionally characterized. Reelin is an integral signaling ligand for proper brain development and post-natal synapse function - properties likely disrupted in ASD patients. We find that the R2290C mutation, which arose de novo in an affected ASD proband, and other analogous mutations in arginine-amino acid-arginine domains reduce protein secretion. Closer analysis of RELN R2290C heterozygous neurospheres reveals up-regulation of Protein Disulfide Isomerase A1, best known as an endoplasmic reticulum-chaperone protein, which has been linked to neuronal pathology. This effect is recapitulated in a heterozygous RELN mouse mutant that is characterized by defective Reelin secretion. These findings suggest that both a deficiency in Reelin signaling and pathologic impairment of Reelin secretion may contribute to ASD risk.


Asunto(s)
Trastorno del Espectro Autista/genética , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína Disulfuro Isomerasas/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Animales , Trastorno del Espectro Autista/metabolismo , Diferenciación Celular/genética , Cerebelo/metabolismo , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Proteína Disulfuro Isomerasas/biosíntesis , Edición de ARN , Proteína Reelina , Receptores X Retinoide/biosíntesis , Receptores X Retinoide/genética
19.
Dev Growth Differ ; 59(8): 657-673, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28945921

RESUMEN

The hippocampal formation (HF) is morphologically and functionally distinguishable into the subdivisions, such as the dentate gyrus (DG), subiculum, and Ammon's horn. The Ammon's horn is further divided into the CA (Cornu Ammonis)1, CA2, and CA3. The Reelin-Dab1 signal is essential for the morphogenesis of the mammalian brain. In the neocortex of Reelin-Dab1 signal mutants the laminar pattern of the neurons is disrupted along the radial axis. Morphological abnormalities in the HF of the Reelin-Dab1 mutants were known, but how these abnormalities appear during development had not been extensively studied. We examined the morphology of the well-developed Dab1 deficient HF by staining with a silver impregnation method in this report, and found that disruption of lamination in the CA1, CA3, and DG was different. Abnormalities observed in the development of Dab1 deficient CA1 were similar to those reported in the neocortical development, while Dab1 deficient CA3 neuronal progenitors radially spreaded beyond presumptive pyramidal layer. The intermediate progenitor cells ectopically located in the Dab1 deficient DG, but neurogenesis was normal in the CA1 and CA3. These observations suggest that the morphogenesis in these HF subdivisions employs different developmental mechanisms involving Dab1 function.


Asunto(s)
Región CA1 Hipocampal/embriología , Región CA3 Hipocampal/embriología , Embrión de Mamíferos/embriología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Región CA1 Hipocampal/citología , Región CA3 Hipocampal/citología , Embrión de Mamíferos/citología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/citología , Proteína Reelina
20.
Cereb Cortex ; 26(7): 3219-36, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26209842

RESUMEN

Disabled-1 (Dab1) is an essential intracellular protein in the Reelin pathway. It has a nuclear localization signal (NLS; hereafter referred to as "NLS1") and 2 nuclear export signals, and shuttles between the nucleus and the cytoplasm. In this study, we found that Dab1 has an additional unidentified NLS, and that the Dab1 NLS1 mutant could translocate to the nucleus in an unconventional ATP/temperature-dependent and cytoplasmic factor/RanGTP gradient-independent manner. Additional mutations in the NLS1 mutant revealed that K(67) and K(69) are important for the nuclear transport. Furthermore, an excess of the intracellular domain of the Reelin receptors inhibited the nuclear translocation of Dab1. An in utero electroporation study showed that a large amount of Dab1 in the cytoplasm in migrating neurons inhibited the migration, and that forced transport of Dab1 into the nucleus attenuated this inhibitory effect. In addition, rescue experiments using yotari, an autosomal recessive mutant of dab1, revealed that cells expressing Dab1 NLS1 mutant tend to distribute at more superficial positions than those expressing wild-type Dab1. Taken together, these findings suggest that Dab1 has at least 2 NLSs, and that the regulation of the subcellular localization of Dab1 is important for the proper migration of excitatory neurons.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Movimiento Celular/fisiología , Corteza Cerebral/metabolismo , Citoplasma/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Corteza Cerebral/embriología , Corteza Cerebral/patología , Citoplasma/patología , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ratones Endogámicos ICR , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Receptores de Superficie Celular/metabolismo , Receptores de LDL/metabolismo , Receptores Notch/metabolismo , Proteína Reelina , Temperatura , Proteína de Unión al GTP ran/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA