Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 15(7)2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-39062605

RESUMEN

FBRSL1, together with FBRS and AUTS2 (Activator of Transcription and Developmental Regulator; OMIM 607270), constitutes a tripartite AUTS2 gene family. AUTS2 and FBRSL1 are evolutionarily more closely related to each other than to FBRS (Fibrosin 1; OMIM 608601). Despite its paralogous relation to AUTS2, FBRSL1's precise role remains unclear, though it likely shares functions in neurogenesis and transcriptional regulation. Herein, we report the clinical presentation with therapeutic approaches and the molecular etiology of a patient harboring a de novo truncating variant (c.371dupC) in FBRSL1, leading to a premature stop codon (p.Cys125Leufs*7). Our study extends previous knowledge by highlighting potential interactions and implications of this variant, alongside maternal and paternal duplications, for the patient's phenotype. Using sequence conservation data and in silico analysis of the truncated protein, we generated a predicted domain structure. Furthermore, our in silico analysis was extended by taking into account SNP array results. The extension of in silico analysis was performed due to the possibility that the coexistence of FBRSL1 truncating variant contemporary with maternal and paternal duplication could be a modifier of proband's phenotype and/or influence the novel syndrome clinical characteristics. FBRSL1 protein may be involved in neurodevelopment due to its homology with AUTS2, together with distinctive neuronal expression profiles, and thus should be considered as a potential modulation of clinical characteristics in a novel syndrome. Finally, considering that FBRSL1 is apparently involved in neurogenesis and in transcriptional regulatory networks that orchestrate gene expression, together with the observation that different genetic syndromes are associated with distinct genomic DNA methylation patterns, the specific episignature has been explored.


Asunto(s)
Proteínas del Citoesqueleto , Discapacidad Intelectual , Factores de Transcripción , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Factores de Transcripción/genética , Proteínas del Citoesqueleto/genética , Masculino , Femenino , Síndrome , Fenotipo , Codón sin Sentido/genética
2.
Dis Model Mech ; 17(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38501224

RESUMEN

De novo truncating variants in fibrosin-like 1 (FBRSL1), a member of the AUTS2 gene family, cause a disability syndrome, including organ malformations such as heart defects. Here, we use Xenopus laevis to investigate whether Fbrsl1 plays a role in heart development. Xenopus laevis fbrsl1 is expressed in tissues relevant for heart development, and morpholino-mediated knockdown of Fbrsl1 results in severely hypoplastic hearts. Our data suggest that Fbrsl1 is required for the development of the first heart field, which contributes to the ventricle and the atria, but not for the second heart field, which gives rise to the outflow tract. The morphant heart phenotype could be rescued using a human N-terminal FBRSL1 isoform that contains an alternative exon, but lacks the AUTS2 domain. N-terminal isoforms carrying patient variants failed to rescue. Interestingly, a long human FBRSL1 isoform, harboring the AUTS2 domain, also did not rescue the morphant heart defects. Thus, our data suggest that different FBRSL1 isoforms may have distinct functions and that only the short N-terminal isoform, appears to be critical for heart development.


Asunto(s)
Cardiopatías Congénitas , Corazón , Proteínas de Xenopus , Xenopus laevis , Animales , Humanos , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Corazón/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Fenotipo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Xenopus laevis/embriología , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética
3.
Front Pharmacol ; 15: 1351536, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38495105

RESUMEN

Neuroblastoma (NB) is the most common extracranial pediatric solid tumor originating from the abnormal development of cells of the sympathoadrenal lineage of the neural crest. Targeting GD2 ganglioside (GD2), a glycolipid expressed on neuroblastoma cells, with GD2 ganglioside-recognizing antibodies affects several pivotal signaling routes that drive or influence the malignant phenotype of the cells. Previously performed gene expression profiling helped us to identify the PHLDA1 (pleckstrin homology-like domain family A member 1) gene as the most upregulated gene in the IMR-32 human neuroblastoma cells treated with the mouse 14G2a monoclonal antibody. Mass spectrometry-based proteomic analyses were applied to better characterize a role of PHLDA1 protein in the response of neuroblastoma cells to chimeric ch14.18/CHO antibody. Additionally, global protein expression profile analysis in the IMR-32 cell line with PHLDA1 silencing revealed the increase in biological functions of mitochondria, accompanied by differentiation-like phenotype of the cells. Moreover, mass spectrometry analysis of the proteins co-immunoprecipitated using anti-PHLDA1-specific antibody, selected a group of possible PHLDA1 binding partners. Also, a more detailed analysis suggested that PHLDA1 interacts with the DCAF7/AUTS2 complex, a key component of neuronal differentiation in vitro. Importantly, our results indicate that PHLDA1 silencing enhances the EGF receptor signaling pathway and combinatory treatment of gefitinib and ch14.18/CHO antibodies might be beneficial for neuroblastoma patients. Data are available via ProteomeXchange with the identifier PXD044319.

4.
Genetics ; 226(1)2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37816306

RESUMEN

Rearrangements within the AUTS2 region are associated with a rare syndromic disorder with intellectual disability, developmental delay, and behavioral abnormalities as core features. In addition, smaller regional variants are linked to wide range of neuropsychiatric disorders, underscoring the gene's essential role in brain development. Like many essential neurodevelopmental genes, AUTS2 is large and complex, generating distinct long (AUTS2-l) and short (AUTS2-s) protein isoforms from alternative promoters. Although evidence suggests unique isoform functions, the contributions of each isoform to specific AUTS2-linked phenotypes have not been clearly resolved. Furthermore, Auts2 is widely expressed across the developing brain, but cell populations most central to disease presentation have not been determined. In this study, we focused on the specific roles of AUTS2-l in brain development, behavior, and postnatal brain gene expression, showing that brain-wide AUTS2-l ablation leads to specific subsets of the recessive pathologies associated with mutations in 3' exons (exons 8-19) that disrupt both major isoforms. We identify downstream genes that could explain expressed phenotypes including hundreds of putative direct AUTS2-l target genes. Furthermore, in contrast to 3' Auts2 mutations which lead to dominant hypoactivity, AUTS2-l loss-of-function is associated with dominant hyperactivity and repetitive behaviors, phenotypes exhibited by many human patients. Finally, we show that AUTS2-l ablation in Calbindin 1-expressing cell lineages is sufficient to yield learning/memory deficits and hyperactivity with abnormal dentate gyrus granule cell maturation, but not other phenotypic effects. These data provide new clues to in vivo AUTS2-l functions and novel information relevant to genotype-phenotype correlations in the human AUTS2 region.


Asunto(s)
Proteínas del Citoesqueleto , Factores de Transcripción , Humanos , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Factores de Transcripción/genética , Calbindinas/metabolismo , Patología Molecular , Encéfalo/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
Mol Neurobiol ; 60(6): 2973-2985, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36754912

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder of unknown cause, although one hypothesis suggests a potential imbalance between excitation and inhibition that leads to changes in neuronal activity and a disturbance in the brain network. However, the mechanisms through which neuronal activity contributes to the development of ASD remain largely unexplained. In this study, we described that neuronal activity at the transcriptional and translational levels regulated the expression of Auts2 isoforms. The prolonged stimulation of cultured cortical neurons significantly reduced the auts2 transcripts, accompanied by the decrease of FL-Auts2 protein, as well as one of the short isoforms (S-Auts2 var.1). Blocking neuronal activity increased the number of auts2 transcripts but not protein levels. Furthermore, blocking the NMDA receptors during stimulation could partially restore the FL-Auts2 and S-Auts2 var.1 at protein level, but not at mRNA level. Finally, Auts2 expression in the hippocampus was reduced in mice exposed to an enriched environment, a behavior paradigm designed to increase the brain activity through abundant sensory and social stimulations. Thus, our study revealed a novel regulatory effect of neuronal activity on the transcription and translation of ASD-risk gene auts2.


Asunto(s)
Trastorno del Espectro Autista , Proteínas del Citoesqueleto , Ratones , Animales , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Factores de Transcripción/metabolismo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Encéfalo/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
6.
Exp Neurol ; 361: 114298, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36525998

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by abnormal social behavior and communication. The autism susceptibility candidate 2 (AUTS2) gene has been associated with multiple neurological diseases, including ASD. Glucose metabolism plays an important role in social behaviors associated with ASD, but the potential role of AUTS2 in glucose metabolism has not been studied. Here, we generated Auts2flox/flox; Emx1Cre+ conditional knockout mice with Auts2 deletion specifically in Exm1-positive neurons in the brain (Auts2-cKO mice) to evaluate the effects of Auts2 knockdown on social behaviors and metabolic pathways. Auts2-cKO mice exhibited ASD-like behaviors, including impaired social interactions and repetitive grooming behaviors. At the molecular level, we found that Auts2 knockdown reduced brain glucose uptake and inhibited the pentose phosphate pathway. Auts2 knockdown also resulted in signs of oxidative stress, and we documented increased levels of reactive oxygen species and malondialdehyde as well as decreased levels of antioxidant molecules, including glutathione and superoxide dismutases in Auts2-cKO mouse brains compared to controls. Finally, Auts2 knockdown significantly disrupted mitochondrial homeostasis and inhibited activity of the SIRT1-SIRT3 axis. Taken together, our findings indicate that loss of AUTS2 expression in Emx1-expressing cells induces multiple changes in metabolic pathways that have been linked to the pathology of ASD. Further characterization of the role of AUTS2 in Emx1-expressing cells in regulating the metabolism of brain neurons may identify opportunities to treat ASD and AUTS2-deficiency disorders with metabolism-targeted therapies.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Ratones , Animales , Trastorno Autístico/genética , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Encéfalo/metabolismo , Conducta Social , Estrés Oxidativo , Glucosa , Proteínas del Citoesqueleto/metabolismo , Factores de Transcripción/metabolismo
7.
Brain ; 146(1): 387-404, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-35802027

RESUMEN

Variants in the AUTS2 gene are associated with a broad spectrum of neurological conditions characterized by intellectual disability, microcephaly, and congenital brain malformations. Here, we use a human cerebral organoid model to investigate the pathophysiology of a heterozygous de novo missense AUTS2 variant identified in a patient with multiple neurological impairments including primary microcephaly and profound intellectual disability. Proband cerebral organoids exhibit reduced growth, deficits in neural progenitor cell (NPC) proliferation and disrupted NPC polarity within ventricular zone-like regions compared to control cerebral organoids. We used CRISPR-Cas9-mediated gene editing to correct this variant and demonstrate rescue of impaired organoid growth and NPC proliferative deficits. Single-cell RNA sequencing revealed a marked reduction of G1/S transition gene expression and alterations in WNT-ß-catenin signalling within proband NPCs, uncovering a novel role for AUTS2 in NPCs during human cortical development. Collectively, these results underscore the value of cerebral organoids to investigate molecular mechanisms underlying AUTS2 syndrome.


Asunto(s)
Trastorno Autístico , Discapacidad Intelectual , Microcefalia , Células-Madre Neurales , Humanos , Microcefalia/genética , Microcefalia/metabolismo , Discapacidad Intelectual/genética , Organoides/metabolismo , Proteínas del Citoesqueleto , Factores de Transcripción/metabolismo
8.
Stem Cell Rev Rep ; 19(2): 531-549, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36258139

RESUMEN

Despite a prominent risk factor for Neurodevelopmental disorders (NDD), it remains unclear how Autism Susceptibility Candidate 2 (AUTS2) controls the neurodevelopmental program. Our studies investigated the role of AUTS2 in neuronal differentiation and discovered that AUTS2, together with WDR68 and SKI, forms a novel protein complex (AWS) specifically in neuronal progenitors and promotes neuronal differentiation through inhibiting BMP signaling. Genomic and biochemical analyses demonstrated that the AWS complex achieves this effect by recruiting the CUL4 E3 ubiquitin ligase complex to mediate poly-ubiquitination and subsequent proteasomal degradation of phosphorylated SMAD1/5/9. Furthermore, using primary cortical neurons, we observed aberrant BMP signaling and dysregulated expression of neuronal genes upon manipulating the AWS complex, indicating that the AWS-CUL4-BMP axis plays a role in regulating neuronal lineage specification in vivo. Thus, our findings uncover a sophisticated cellular signaling network mobilized by a prominent NDD risk factor, presenting multiple potential therapeutic targets for NDD.


Asunto(s)
Proteínas del Citoesqueleto , Trastornos del Neurodesarrollo , Neuronas , Transducción de Señal , Factores de Transcripción , Trastornos del Neurodesarrollo/genética , Proteínas del Citoesqueleto/genética , Factores de Transcripción/genética
9.
Gene ; 848: 146864, 2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36067863

RESUMEN

Duck meat is starting to receive more attention due to its unique meaty characteristics. Pekin duck is an important breed resource of meat duck, which has been used in meat production and product research. However, the study about whole genome resequencing analysis of ducks for meat production has not been reported and the underlying mechanisms of meat production remain undefined. Here, lines with high lean meat percentage (S, n = 30) and low lean meat percentage (Z, n = 30) were used to analyze. The values of body weight, breast meat weight and leg meat weight in S line (body weight: 3,071 ± 26.83 g), breast meat: 391.3 ± 6.670 g; leg meat: 121.1 ± 2.184 g) were significantly higher than those in Z line (body weight: 2,584 ± 38.53 g, breast meat: 263.9 ± 6.984 g; leg meat: 110.1 ± 3.645 g). The values of body size in Z line (26.47 ± 0.1571 cm) were significantly higher than that in S line (25.38 ± 0.2475 cm). A total of 14,220,037 SNPs were obtained from 19 individuals by whole-genome resequencing, and the separate analyses of FST (range from 0.30 to 0.52) and log2θπ ratio (range from 5.8 to 8.1) revealed 50 and 124 candidate genes in the top 0.1% regions respectively, which involved in 209 and 298 candidate regions. The integration of two approaches resulted in 7 overlapping genes. Notably, AUTS2 gene is related to activator of developmental regulator. As expected, we found that in the chr29:2.29-2.30 Mb region of AUTS2, the FST value is 0.32, and the S line (π = 7.3 × 10 -5) shows a very low level of π value compared with Z line (π = 8.8 × 10 -3). Genotyping and GWAS analysis showed that 2 candidate SNPs (chr29:2,296,787 and chr29:2,296,832) were associated with high meat percentage, which were verified by Sanger sequencing. Taken together, lean meat percentage was much higher in S line individuals by comparing with Z line. The integration of FST and θπ resulted in only 7 overlapping genes that in the top 0.1% candidate regions of them. The chr29:2,296,787 and chr29:2,296,832 in the AUTS2 gene could be important molecular markers for high lean meat adaptation selection in S line.


Asunto(s)
Patos , Carne , Animales , Tamaño Corporal , Peso Corporal/genética , Cruzamiento , Proteínas del Citoesqueleto/genética , Patos/genética , Factores de Transcripción/genética
10.
Comput Struct Biotechnol J ; 20: 2759-2777, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35685361

RESUMEN

Tick-borne encephalitis virus (TBEV), the most medically relevant tick-transmitted flavivirus in Eurasia, targets the host central nervous system and frequently causes severe encephalitis. The severity of TBEV-induced neuropathogenesis is highly cell-type specific and the exact mechanism responsible for such differences has not been fully described yet. Thus, we performed a comprehensive analysis of alterations in host poly-(A)/miRNA/lncRNA expression upon TBEV infection in vitro in human primary neurons (high cytopathic effect) and astrocytes (low cytopathic effect). Infection with severe but not mild TBEV strain resulted in a high neuronal death rate. In comparison, infection with either of TBEV strains in human astrocytes did not. Differential expression and splicing analyses with an in silico prediction of miRNA/mRNA/lncRNA/vd-sRNA networks found significant changes in inflammatory and immune response pathways, nervous system development and regulation of mitosis in TBEV Hypr-infected neurons. Candidate mechanisms responsible for the aforementioned phenomena include specific regulation of host mRNA levels via differentially expressed miRNAs/lncRNAs or vd-sRNAs mimicking endogenous miRNAs and virus-driven modulation of host pre-mRNA splicing. We suggest that these factors are responsible for the observed differences in the virulence manifestation of both TBEV strains in different cell lines. This work brings the first complex overview of alterations in the transcriptome of human astrocytes and neurons during the infection by two TBEV strains of different virulence. The resulting data could serve as a starting point for further studies dealing with the mechanism of TBEV-host interactions and the related processes of TBEV pathogenesis.

11.
Front Mol Neurosci ; 15: 858582, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35431798

RESUMEN

AUTS2 syndrome is a genetic disorder that causes intellectual disability, microcephaly, and other phenotypes. Syndrome severity is worse when mutations involve 3' regions (exons 9-19) of the AUTS2 gene. Human AUTS2 protein has two major isoforms, full-length (1259 aa) and C-terminal (711 aa), the latter produced from an alternative transcription start site in exon 9. Structurally, AUTS2 contains the putative "AUTS2 domain" (∼200 aa) conserved among AUTS2 and its ohnologs, fibrosin, and fibrosin-like-1. Also, AUTS2 contains extensive low-complexity sequences and intrinsically disordered regions, features typical of RNA-binding proteins. During development, AUTS2 is expressed by specific progenitor cell and neuron types, including pyramidal neurons and Purkinje cells. AUTS2 localizes mainly in cell nuclei, where it regulates transcription and RNA metabolism. Some studies have detected AUTS2 in neurites, where it may regulate cytoskeletal dynamics. Neurodevelopmental functions of AUTS2 have been studied in diverse model systems. In zebrafish, auts2a morphants displayed microcephaly. In mice, excision of different Auts2 exons (7, 8, or 15) caused distinct phenotypes, variously including neonatal breathing abnormalities, cerebellar hypoplasia, dentate gyrus hypoplasia, EEG abnormalities, and behavioral changes. In mouse embryonic stem cells, AUTS2 could promote or delay neuronal differentiation. Cerebral organoids, derived from an AUTS2 syndrome patient containing a pathogenic missense variant in exon 9, exhibited neocortical growth defects. Emerging technologies for analysis of human cerebral organoids will be increasingly useful for understanding mechanisms underlying AUTS2 syndrome. Questions for future research include whether AUTS2 binds RNA directly, how AUTS2 regulates neurogenesis, and how AUTS2 modulates neural circuit formation.

12.
J Integr Bioinform ; 19(1)2022 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-35254744

RESUMEN

Spliced alignments are a key step in the construction of high-quality homology-based annotations of protein sequences. The exon/intron structure, which is computed as part of spliced alignment procedures, often conveys important information for the distinguishing paralogous members of gene families. Here we present an exon-centric pipeline for spliced alignment that is intended in particular for applications that involve exon-by-exon comparisons of coding sequences. We show that the simple, blat-based approach has advantages over established tools in particular for genes with very large introns and applications to fragmented genome assemblies.


Asunto(s)
Alineación de Secuencia , Exones , Intrones/genética
13.
Front Cell Dev Biol ; 9: 779009, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805182

RESUMEN

Truncating variants in specific exons of Fibrosin-like protein 1 (FBRSL1) were recently reported to cause a novel malformation and intellectual disability syndrome. The clinical spectrum includes microcephaly, facial dysmorphism, cleft palate, skin creases, skeletal anomalies and contractures, postnatal growth retardation, global developmental delay as well as respiratory problems, hearing impairment and heart defects. The function of FBRSL1 is largely unknown, but pathogenic variants in the FBRSL1 paralog Autism Susceptibility Candidate 2 (AUTS2) are causative for an intellectual disability syndrome with microcephaly (AUTS2 syndrome). Some patients with AUTS2 syndrome also show additional symptoms like heart defects and contractures overlapping with the phenotype presented by patients with FBRSL1 mutations. For AUTS2, a dual function, depending on different isoforms, was described and suggested for FBRSL1. Both, nuclear FBRSL1 and AUTS2 are components of the Polycomb subcomplexes PRC1.3 and PRC1.5. These complexes have essential roles in developmental processes, cellular differentiation and proliferation by regulating gene expression via histone modification. In addition, cytoplasmic AUTS2 controls neural development, neuronal migration and neurite extension by regulating the cytoskeleton. Here, we review recent data on FBRSL1 in respect to previously published data on AUTS2 to gain further insights into its molecular function, its role in development as well as its impact on human genetics.

14.
Mol Cell ; 81(22): 4663-4676.e8, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34637754

RESUMEN

The heterogeneous family of complexes comprising Polycomb repressive complex 1 (PRC1) is instrumental for establishing facultative heterochromatin that is repressive to transcription. However, two PRC1 species, ncPRC1.3 and ncPRC1.5, are known to comprise novel components, AUTS2, P300, and CK2, that convert this repressive function to that of transcription activation. Here, we report that individuals harboring mutations in the HX repeat domain of AUTS2 exhibit defects in AUTS2 and P300 interaction as well as a developmental disorder reflective of Rubinstein-Taybi syndrome, which is mainly associated with a heterozygous pathogenic variant in CREBBP/EP300. Moreover, the absence of AUTS2 or mutation in its HX repeat domain gives rise to misregulation of a subset of developmental genes and curtails motor neuron differentiation of mouse embryonic stem cells. The transcription factor nuclear respiratory factor 1 (NRF1) has a novel and integral role in this neurodevelopmental process, being required for ncPRC1.3 recruitment to chromatin.


Asunto(s)
Encéfalo/metabolismo , Proteína de Unión a CREB/genética , Proteínas del Citoesqueleto/metabolismo , Proteína p300 Asociada a E1A/genética , Células Madre Embrionarias/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Cromatina/química , Femenino , Genómica , Células HEK293 , Heterocigoto , Humanos , Masculino , Ratones , Neuronas/metabolismo , Unión Proteica , Dominios Proteicos , Proteómica , Activación Transcripcional
15.
Genes (Basel) ; 12(9)2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34573342

RESUMEN

Haploinsufficiency of AUTS2 has been associated with a syndromic form of neurodevelopmental delay characterized by intellectual disability, autistic features, and microcephaly, also known as AUTS2 syndrome. While the phenotype associated with large deletions and duplications of AUTS2 is well established, clinical features of patients harboring AUTS2 sequence variants have not been extensively described. In this study, we describe the phenotype of five new patients with AUTS2 pathogenic variants, three of them harboring loss-of-function sequence variants. The phenotype of the patients was characterized by attention deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) or autistic features and mild global developmental delay (GDD) or intellectual disability (ID), all in 4/5 patients (80%), a frequency higher than previously reported for ADHD and autistic features. Microcephaly and short stature were found in 60% of the patients; and feeding difficulties, generalized hypotonia, and ptosis, were each found in 40%. We also provide the aggregated frequency of the 32 items included in the AUTS2 syndrome severity score (ASSS) in patients currently reported in the literature. The main characteristics of the syndrome are GDD/ID in 98% of patients, microcephaly in 65%, feeding difficulties in 62%, ADHD or hyperactivity in 54%, and autistic traits in 52%. Finally, using the location of 31 variants from the literature together with variants from the five patients, we found significantly higher ASSS values in patients with pathogenic variants affecting the 3' end of the gene, confirming the genotype-phenotype correlation initially described.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/etiología , Trastorno del Espectro Autista/etiología , Proteínas del Citoesqueleto/genética , Factores de Transcripción/genética , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno del Espectro Autista/genética , Niño , Preescolar , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/genética , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Discapacidad Intelectual/genética , Mutación con Pérdida de Función , Masculino , Síndrome
16.
BMC Psychiatry ; 21(1): 360, 2021 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-34273950

RESUMEN

BACKGROUND: Pathogenic variants of the AUTS2 (Autism Susceptibility candidate 2) gene predispose to intellectual disability, autism spectrum disorder, attention deficit hyperactivity disorder, facial dysmorphism and short stature. This phenotype is therefore associated with neurocognitive disturbances and social cognition, indicating potential functional maladjustment in the affected subjects, and a potentially significant impact on quality of life. Although many isolated cases have been reported in the literature, to date no families have been described. This case reports on a family (three generations) with a frameshift variant in the AUTS2 gene. CASE PRESENTATION: The proband is 13 years old with short stature, dysmorphic features, moderate intellectual disability and autism spectrum disorder. His mother is 49 years old and also has short stature and similar dysmorphic features. She does not have autism disorder but presents an erotomaniac delusion. Her cognitive performance is heterogeneous. The two aunts are also of short stature. The 50-year-old aunt has isolated social cognition disorders. The 45-year-old aunt has severe cognitive impairment and autism spectrum disorder. The molecular analysis of the three sisters and the proband shows the same AUTS2 heterozygous duplication leading to a frame shift expected to produce a premature stop codon, p.(Met593Tyrfs*85). Previously reported isolated cases revealed phenotypic and cognitive impairment variability. In this case report, these variabilities are present within the same family, presenting the same variant. CONCLUSIONS: The possibility of a phenotypic spectrum within the same family highlights the need for joint psychiatry and genetics research.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Adolescente , Trastorno del Espectro Autista/genética , Proteínas del Citoesqueleto/genética , Femenino , Humanos , Discapacidad Intelectual/genética , Persona de Mediana Edad , Fenotipo , Calidad de Vida , Factores de Transcripción/genética
17.
Front Psychiatry ; 12: 580433, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33967843

RESUMEN

Neurodevelopmental disorders are psychiatric diseases that are usually first diagnosed in infancy, childhood and adolescence. Autism spectrum disorder (ASD) is a neurodevelopmental disorder, characterized by core symptoms including impaired social communication, cognitive rigidity and repetitive behavior, accompanied by a wide range of comorbidities such as intellectual disability (ID) and dysmorphisms. While the cause remains largely unknown, genetic, epigenetic, and environmental factors are believed to contribute toward the onset of the disease. Autism Susceptibility Candidate 2 (Auts2) is a gene highly associated with ID and ASD. Therefore, understanding the function of Auts2 gene can provide a unique entry point to untangle the complex neuronal phenotypes of neurodevelpmental disorders. In this review, we discuss the recent discoveries regarding the molecular and cellular functions of Auts2. Auts2 was shown to be a key-regulator of transcriptional network and a mediator of epigenetic regulation in neurodevelopment, the latter potentially providing a link for the neuronal changes of ASD upon environmental risk-factor exposure. In addition, Auts2 could synchronize the balance between excitation and inhibition through regulating the number of excitatory synapses. Cytoplasmic Auts2 could join the fine-tuning of actin dynamics during neuronal migration and neuritogenesis. Furthermore, Auts2 was expressed in developing mouse and human brain regions such as the frontal cortex, dorsal thalamus, and hippocampus, which have been implicated in the impaired cognitive and social function of ASD. Taken together, a comprehensive understanding of Auts2 functions can give deep insights into the cause of the heterogenous manifestation of neurodevelopmental disorders such as ASD.

18.
Genes (Basel) ; 12(2)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562463

RESUMEN

Neurodevelopmental disorders (NDDs) are a group of highly prevalent, clinically and genetically heterogeneous pediatric disorders comprising, according to the Diagnostic and Statistical Manual of Mental Disorders 5th edition (DSM-V), intellectual disability, developmental delay, autism spectrum disorders, and other neurological and cognitive disorders manifesting in the developmental age. To date, more than 1000 genes have been implicated in the etiopathogenesis of NNDs. Among them, AUTS2 (OMIM # 607270) encodes a protein involved in neural migration and neuritogenesis, and causes NNDs with different molecular mechanisms including copy number variations, single or multiple exonic deletion and single nucleotide variants. We describes a 9-year-old boy with global developmental delay, absent speech, minor craniofacial anomalies, hypoplasia of the cerebellar vermis and thinning of the corpus callosum, resulted carrier of the de novo AUTS2 c.1603_1626del deletion at whole exome sequencing (WES) predicted to cause the loss of eight amino acids [p.(His535_Thr542del)]. Notably, our patient is the first reported so far in medical literature carrying an in-frame deletion and the first in which absent language, hypoplasia of the cerebellar vermis and thinning of the corpus callosum has been observed thus useful to expand the molecular spectrum of AUTS2 pathogenic variants and to broaden our knowledge on the clinical phenotype associated.


Asunto(s)
Trastorno Dismórfico Corporal/genética , Proteínas del Citoesqueleto/genética , Discapacidades del Desarrollo/genética , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Trastorno Dismórfico Corporal/patología , Corteza Cerebral/anomalías , Corteza Cerebral/patología , Niño , Discapacidades del Desarrollo/patología , Exones , Humanos , Discapacidad Intelectual/patología , Masculino , Fenotipo , Eliminación de Secuencia/genética , Trastornos del Habla/genética , Trastornos del Habla/patología , Secuenciación del Exoma
19.
Am J Med Genet A ; 185(4): 1261-1265, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33577136

RESUMEN

Haploinsufficiency of AUTS2 has been associated with neurodevelopmental disorders and dysmorphic features (MIM # 615834). More than 50 patients have been described, mostly carrying de novo deletions of one or more exons, including eight patients with exon 6 deletions. We report on two siblings, a girl and a boy aged 11 and 13 years, in whom the same pathogenic 85 kb deletion on 7q11.22 encompassing exon 6 of AUTS2 by SNP array analysis was identified. Both children had typical symptoms of AUTS2 syndrome such as intellectual impairment and behavioral problems, but with markedly different expression. SNP array analysis excluded the deletion in blood samples of both parents and a healthy brother. Conventional karyotyping of both parents and additional FISH analyses, marking the flanking regions of the deletion, did not show any structural rearrangements involving 7q11.22. A germ cell mosaicism was suggested as the most probable explanation for occurrence of the same deletion in these two siblings. To our knowledge this is the first report of germ cell mosaicism for AUTS2 syndrome. It additionally provides further evidence of intrafamilial phenotypic variability in AUTS2 syndrome and adds clinical information to the phenotypic spectrum of patients with AUTS2 exon 6 deletions.


Asunto(s)
Anomalías Múltiples/genética , Proteínas del Citoesqueleto/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Adolescente , Niño , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/patología , Exones/genética , Femenino , Células Germinativas/metabolismo , Células Germinativas/patología , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Mosaicismo , Eliminación de Secuencia/genética
20.
Cells ; 11(1)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-35011572

RESUMEN

Neurodevelopmental disorders (NDDs), including autism spectrum disorders (ASD) and intellectual disability (ID), are a large group of neuropsychiatric illnesses that occur during early brain development, resulting in a broad spectrum of syndromes affecting cognition, sociability, and sensory and motor functions. Despite progress in the discovery of various genetic risk factors thanks to the development of novel genomics technologies, the precise pathological mechanisms underlying the onset of NDDs remain elusive owing to the profound genetic and phenotypic heterogeneity of these conditions. Autism susceptibility candidate 2 (AUTS2) has emerged as a crucial gene associated with a wide range of neuropsychological disorders, such as ASD, ID, schizophrenia, and epilepsy. AUTS2 has been shown to be involved in multiple neurodevelopmental processes; in cell nuclei, it acts as a key transcriptional regulator in neurodevelopment, whereas in the cytoplasm, it participates in cerebral corticogenesis, including neuronal migration and neuritogenesis, through the control of cytoskeletal rearrangements. Postnatally, AUTS2 regulates the number of excitatory synapses to maintain the balance between excitation and inhibition in neural circuits. In this review, we summarize the knowledge regarding AUTS2, including its molecular and cellular functions in neurodevelopment, its genetics, and its role in behaviors.


Asunto(s)
Trastornos del Neurodesarrollo/genética , Factores de Transcripción/genética , Animales , Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Neurogénesis/genética , Factores de Transcripción/química , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...