Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.334
Filtrar
1.
Genes (Basel) ; 15(6)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38927586

RESUMEN

To date, only 13 studies have described patients with large overlapping deletions of 10p11.2-p12. These individuals shared a common phenotype characterized by intellectual disability, developmental delay, distinct facial dysmorphic features, abnormal behaviour, visual impairment, cardiac malformation, and cryptorchidism in males. Molecular cytogenetic analysis revealed that the deletion in this chromosomal region shares a common smallest region of overlap (SRO) of 80 kb, which contains only the WAC gene (WW-domain-containing adaptor with coiled coil). In this clinical case report, we report a 5-year-old girl, born from non-consanguineous parents, with a 10p11.22p11.21 microdeletion. She presents clinical features that overlap with other patients described in the literature, such as dysmorphic traits, speech delay, and behavioural abnormalities (hyperactivity), even though the WAC gene is not involved in the microdeletion. Our results are the first to highlight that the deletion described here represents a contiguous gene syndrome that is enough to explain the distinct phenotype but partially overlaps with the previous cases reported in the literature, even though the same genes are not involved. In particular, in this study, we speculate about the role of the WAC gene that seems to be associated with normal motor development. In fact, we found that our patient is the only one described in the literature with a large deletion in the 10p11.22p11.21 region without the involvement of the WAC gene deletion, and, interestingly, the patient did not have motor delay.


Asunto(s)
Deleción Cromosómica , Humanos , Femenino , Preescolar , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Síndrome , Fenotipo , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología
2.
Genes (Basel) ; 15(6)2024 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-38927727

RESUMEN

Mutations in the gene SCAPER (S phase Cyclin A-Associated Protein residing in the Endoplasmic Reticulum) have recently been associated with retinitis pigmentosa (RP) and intellectual disability (ID). In 2011, a possible involvement of SCAPER in human diseases was discovered for the first time due to the identification of a homozygous mutation causing ID in an Iranian family. Later, five studies were published in 2019 that described patients with autosomal recessive syndromic retinitis pigmentosa (arRP) accompanied by ID and attention-deficit/hyperactivity disorder (ADHD). This present study describes three patients from an Arab consanguineous family in Israel with similar clinical features of the SCAPER syndrome. In addition, new manifestations of ocular symptoms, nystagmus, glaucoma, and elevator palsy, were observed. Genetic testing of the patients and both parents via whole-exome sequencing revealed the homozygous mutation c.2023-2A>G in SCAPER. Phenotypic and genotypic descriptions for all available cases described in the literature including our current three cases (37 cases) were carried out, in addition to a bioinformatics analysis for all the genetic variants that was undertaken. Our study confirms and extends the clinical manifestations of SCAPER-related disorders.


Asunto(s)
Biología Computacional , Discapacidad Intelectual , Mutación , Linaje , Fenotipo , Retinitis Pigmentosa , Adolescente , Adulto , Femenino , Humanos , Proteínas Portadoras/genética , Biología Computacional/métodos , Consanguinidad , Secuenciación del Exoma , Genes Recesivos , Homocigoto , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Péptidos y Proteínas de Señalización Intercelular , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
3.
Genes (Basel) ; 15(6)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38927725

RESUMEN

This study delves into the diagnostic yield of whole-exome sequencing (WES) in pediatric patients presenting with developmental delay/intellectual disability (DD/ID), while also exploring the utility of Reverse Phenotyping (RP) in refining diagnoses. A cohort of 100 pediatric patients underwent WES, yielding a diagnosis in 66% of cases. Notably, RP played a significant role in cases with negative prior genetic testing, underscoring its significance in complex diagnostic scenarios. The study revealed a spectrum of genetic conditions contributing to DD/ID, illustrating the heterogeneity of etiological factors. Despite challenges, WES demonstrated effectiveness, particularly in cases with metabolic abnormalities. Reverse phenotyping was indicated in half of the patients with positive WES findings. Neural network models exhibited moderate-to-exceptional predictive abilities for aiding in patient selection for WES and RP. These findings emphasize the importance of employing comprehensive genetic approaches and RP in unraveling the genetic underpinnings of DD/ID, thereby facilitating personalized management and genetic counseling for affected individuals and families. This research contributes insights into the genetic landscape of DD/ID, enhancing our understanding and guiding clinical practice in this particular field of clinical genetics.


Asunto(s)
Discapacidades del Desarrollo , Secuenciación del Exoma , Discapacidad Intelectual , Fenotipo , Humanos , Secuenciación del Exoma/métodos , Discapacidades del Desarrollo/genética , Niño , Masculino , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Femenino , Preescolar , Lactante , Adolescente , Pruebas Genéticas/métodos
4.
Genes (Basel) ; 15(6)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38927744

RESUMEN

While balanced reciprocal translocations are relatively common, they often remain clinically silent unless they lead to the disruption of functional genes. In this study, we present the case of a boy exhibiting developmental delay and mild intellectual disability. Initial karyotyping revealed a translocation t(5;6)(q13;q23) between chromosomes 5 and 6 with limited resolution. Optical genome mapping (OGM) enabled a more precise depiction of the breakpoint regions involved in the reciprocal translocation. While the breakpoint region on chromosome 6 did not encompass any known gene, OGM revealed the disruption of the RASGRF2 (Ras protein-specific guanine nucleotide releasing factor 2) gene on chromosome 5, implicating RASGRF2 as a potential candidate gene contributing to the observed developmental delay in the patient. Variations in RASGRF2 have so far not been reported in developmental delay, but research on the RASGRF2 gene underscores its significance in various aspects of neurodevelopment, including synaptic plasticity, signaling pathways, and behavioral responses. This study highlights the utility of OGM in identifying breakpoint regions, providing possible insights into the understanding of neurodevelopmental disorders. It also helps affected individuals in gaining more knowledge about potential causes of their conditions.


Asunto(s)
Discapacidades del Desarrollo , Translocación Genética , Humanos , Masculino , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Factores de Intercambio de Guanina Nucleótido ras/genética , Mapeo Cromosómico , Cromosomas Humanos Par 5/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología
6.
Genes (Basel) ; 15(5)2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38790177

RESUMEN

SATB1 (MIM #602075) is a relatively new gene reported only in recent years in association with neurodevelopmental disorders characterized by variable facial dysmorphisms, global developmental delay, poor or absent speech, altered electroencephalogram (EEG), and brain abnormalities on imaging. To date about thirty variants in forty-four patients/children have been described, with a heterogeneous spectrum of clinical manifestations. In the present study, we describe a new patient affected by mild intellectual disability, speech disorder, and non-specific abnormalities on EEG and neuroimaging. Family studies identified a new de novo frameshift variant c.1818delG (p.(Gln606Hisfs*101)) in SATB1. To better define genotype-phenotype associations in the different types of reported SATB1 variants, we reviewed clinical data from our patient and from the literature and compared manifestations (epileptic activity, EEG abnormalities and abnormal brain imaging) due to missense variants versus those attributable to loss-of-function/premature termination variants. Our analyses showed that the latter variants are associated with less severe, non-specific clinical features when compared with the more severe phenotypes due to missense variants. These findings provide new insights into SATB1-related disorders.


Asunto(s)
Encéfalo , Electroencefalografía , Epilepsia , Proteínas de Unión a la Región de Fijación a la Matriz , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Epilepsia/genética , Epilepsia/diagnóstico por imagen , Epilepsia/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Encéfalo/fisiopatología , Masculino , Femenino , Mutación con Pérdida de Función , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/patología , Neuroimagen/métodos , Niño , Mutación del Sistema de Lectura/genética , Fenotipo , Preescolar
7.
Genes (Basel) ; 15(5)2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38790209

RESUMEN

Intellectual disability (ID), which affects around 2% to 3% of the population, accounts for 0.63% of the overall prevalence of neurodevelopmental disorders (NDD). ID is characterized by limitations in a person's intellectual and adaptive functioning, and is caused by pathogenic variants in more than 1000 genes. Here, we report a rare missense variant (c.350T>C; p.(Leu117Ser)) in HACE1 segregating with NDD syndrome with clinical features including ID, epilepsy, spasticity, global developmental delay, and psychomotor impairment in two siblings of a consanguineous Pakistani kindred. HACE1 encodes a HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (HACE1), which is involved in protein ubiquitination, localization, and cell division. HACE1 is also predicted to interact with several proteins that have been previously implicated in the ID phenotype in humans. The p.(Leu117Ser) variant replaces an evolutionarily conserved residue of HACE1 and is predicted to be deleterious by various in silico algorithms. Previously, eleven protein truncating variants of HACE1 have been reported in individuals with NDD. However, to our knowledge, p.(Leu117Ser) is the second missense variant in HACE1 found in an individual with NDD.


Asunto(s)
Epilepsia , Discapacidad Intelectual , Espasticidad Muscular , Mutación Missense , Linaje , Ubiquitina-Proteína Ligasas , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Ubiquitina-Proteína Ligasas/genética , Masculino , Femenino , Epilepsia/genética , Pakistán , Espasticidad Muscular/genética , Trastornos Psicomotores/genética , Trastornos Psicomotores/patología , Niño , Preescolar
8.
Eur J Med Genet ; 69: 104948, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38735569

RESUMEN

Anorectal malformations (ARMs) represent a wide spectrum of congenital anomalies of the anus and rectum, of which more than half are syndromic. Their etiology is highly heterogeneous and still poorly understood. We report a 4-year-old girl who initially presented with an isolated ARM, and subsequently developed a global developmental delay as part of an ARID1B-related Coffin-Siris syndrome (CSS). A co-occurrence of ARMs and CSS in an individual by chance is unexpected since both diseases are very rare. A review of the literature enabled us to identify 10 other individuals with both CSS and ARMs. Among the ten individuals reported in this study, 8 had a variant in ARID1A, 2 in ARID1B, and 1 in SMARCA4. This more frequent than expected association between CSS and ARM indicates that some ARMs are most likely part of the CSS spectrum, especially for ARID1A-related CSS.


Asunto(s)
Anomalías Múltiples , Malformaciones Anorrectales , Proteínas de Unión al ADN , Cara , Deformidades Congénitas de la Mano , Discapacidad Intelectual , Micrognatismo , Cuello , Factores de Transcripción , Humanos , Femenino , Micrognatismo/genética , Micrognatismo/patología , Preescolar , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Factores de Transcripción/genética , Cuello/anomalías , Cuello/patología , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/patología , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Proteínas de Unión al ADN/genética , Malformaciones Anorrectales/genética , Cara/anomalías , Cara/patología , ADN Helicasas/genética , Proteínas Nucleares/genética , Canal Anal/anomalías , Canal Anal/patología , Fenotipo
9.
Cell Death Dis ; 15(5): 379, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38816421

RESUMEN

CSMD1 (Cub and Sushi Multiple Domains 1) is a well-recognized regulator of the complement cascade, an important component of the innate immune response. CSMD1 is highly expressed in the central nervous system (CNS) where emergent functions of the complement pathway modulate neural development and synaptic activity. While a genetic risk factor for neuropsychiatric disorders, the role of CSMD1 in neurodevelopmental disorders is unclear. Through international variant sharing, we identified inherited biallelic CSMD1 variants in eight individuals from six families of diverse ancestry who present with global developmental delay, intellectual disability, microcephaly, and polymicrogyria. We modeled CSMD1 loss-of-function (LOF) pathogenesis in early-stage forebrain organoids differentiated from CSMD1 knockout human embryonic stem cells (hESCs). We show that CSMD1 is necessary for neuroepithelial cytoarchitecture and synchronous differentiation. In summary, we identified a critical role for CSMD1 in brain development and biallelic CSMD1 variants as the molecular basis of a previously undefined neurodevelopmental disorder.


Asunto(s)
Discapacidad Intelectual , Proteínas de la Membrana , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Femenino , Masculino , Trastornos del Neurodesarrollo/genética , Alelos , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/patología , Niño , Preescolar , Diferenciación Celular/genética , Proteínas Supresoras de Tumor
10.
Stem Cell Res ; 77: 103442, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38739972

RESUMEN

Intellectual disability (ID) is a diverse neurodevelopmental condition and almost half of the cases have a genetic etiology. SGIP1 acts as an endocytic protein that influences the signaling of receptors in neuronal systems related to energy homeostasis through its interaction with endophilins. This study focuses on the generation and characterization of induced pluripotent stem cells (iPSC) from two unrelated patients due to a frameshift variant (c.764dupA, NM_032291.4) and a splice donor site variant (c.74 + 1G > A, NM_032291.4) in the SGIP1 gene.


Asunto(s)
Homocigoto , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Femenino , Línea Celular , Niño
11.
Eur J Med Genet ; 69: 104945, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697389

RESUMEN

To date 11 patients with Coffin-Siris syndrome type 7 (OMIM 618027) have been described since the first literature report. All reported patients carried de novo variants with presumed dominant negative effect, which localized in the PHD1/PHD2 domains of DPF2. Here we report on the first familial case of Coffin-Siris syndrome type 7. The index patient presented during the 1st year of life with failure to thrive and ectodermal anomalies. The genetic analysis using whole exome sequencing showed a likely pathogenic missense variant in the PHD1 region. The family analysis showed that the mother as well as the older brother of the index patient also carried the detected DPF2 variant in heterozygous state. The mother had a history of school difficulties but no history of failure to thrive and was overall mildly affected. The brother showed developmental delay with autistic features, ectodermal anomalies and overlapping morphologic features but did not have a history of growth failure problems. To our knowledge this is the first report of an inherited likely pathogenic variant in DPF2, underlining the variability of the associated phenotype as well as the importance of considering inherited DPF2 variants during the variant filtering strategy of whole exome data.


Asunto(s)
Anomalías Múltiples , Cara , Deformidades Congénitas de la Mano , Discapacidad Intelectual , Micrognatismo , Cuello , Linaje , Factores de Transcripción , Humanos , Masculino , Micrognatismo/genética , Micrognatismo/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/patología , Cara/anomalías , Cara/patología , Factores de Transcripción/genética , Femenino , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Cuello/anomalías , Cuello/patología , Mutación Missense , Proteínas de Unión al ADN/genética , Fenotipo , Lactante , Adulto
12.
Mol Genet Metab ; 142(2): 108492, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759397

RESUMEN

Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , N-Acetilglucosaminiltransferasas , Placa Neural , Fenotipo , Humanos , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Placa Neural/metabolismo , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/patología , Trastornos Congénitos de Glicosilación/metabolismo , Sistemas CRISPR-Cas , Glicosilación , Edición Génica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología
13.
Mol Genet Genomic Med ; 12(5): e2420, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38773911

RESUMEN

OBJECTIVE: This study aims to report a severe phenotype of Arboleda-Tham syndrome in a 20-month-old girl, characterized by global developmental delay, distinct facial features, intellectual disability. Arboleda-Tham syndrome is known for its wide phenotypic spectrum and is associated with truncating variants in the KAT6A gene. METHODS: To diagnose this case, a combination of clinical phenotype assessment and whole-exome sequencing technology was employed. The genetic analysis involved whole-exome sequencing, followed by confirmation of the identified variant through Sanger sequencing. RESULTS: The whole-exome sequencing revealed a novel de novo frameshift mutation c.3048del (p.Leu1017Serfs*17) in the KAT6A gene, which is classified as likely pathogenic. This mutation was not found in the ClinVar and HGMD databases and was not present in her parents. The mutation leads to protein truncation or activation of nonsense-mediated mRNA degradation. The mutation is located within exon 16, potentially leading to protein truncation or activation of nonsense-mediated mRNA degradation. Protein modeling suggested that the de novo KAT6A mutation might alter hydrogen bonding and reduce protein stability, potentially damaging the protein structure and function. CONCLUSION: This study expands the understanding of the genetic basis of Arboleda-Tham syndrome, highlighting the importance of whole-exome sequencing in diagnosing cases with varied clinical presentations. The discovery of the novel KAT6A mutation adds to the spectrum of known pathogenic variants and underscores the significance of this gene in the syndrome's pathology.


Asunto(s)
Discapacidades del Desarrollo , Secuenciación del Exoma , Humanos , Femenino , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Discapacidades del Desarrollo/diagnóstico , Lactante , Mutación del Sistema de Lectura , Histona Acetiltransferasas/genética , Fenotipo , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Discapacidad Intelectual/diagnóstico
14.
Genes (Basel) ; 15(4)2024 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-38674358

RESUMEN

Pathogenic ASH1L variants have been reported in probands with broad phenotypic presentations, including intellectual disability, autism spectrum disorder, attention deficit hyperactivity disorder, seizures, congenital anomalies, and other skeletal, muscular, and sleep differences. Here, we review previously published individuals with pathogenic ASH1L variants and report three further probands with novel ASH1L variants and previously unreported phenotypic features, including mixed receptive language disorder and gait disturbances. These novel data from the Brain Gene Registry, an accessible repository of clinically derived genotypic and phenotypic data, have allowed for the expansion of the phenotypic and genotypic spectrum of this condition.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Trastornos del Neurodesarrollo , Fenotipo , Humanos , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Masculino , N-Metiltransferasa de Histona-Lisina/genética , Femenino , Niño , Genotipo , Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Factores de Transcripción/genética , Preescolar , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Mutación , Adolescente
15.
Eur J Med Genet ; 69: 104944, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679370

RESUMEN

Here we report the case of a young boy with developmental delay, thin sparse hair, early closure of the anterior fontanel, bilateral choanal atresia, brachyturicephaly; and dysmorphic features closely resembling those seen in trichorhinophalangeal syndrome (TRPS). These features include sparse hair, sparse lateral eyebrows, a bulbous pear shaped nose, a long philtrum, thin lips, small/hypoplastic nails, pes planovalgus; bilateral cone-shaped epiphyses at the proximal 5th phalanx, slender long bones, coxa valga, mild scoliosis, and delayed bone age. Given that TRPS had been excluded by a thorough genetic analysis, whole exome sequencing was performed and a heterozygous likely pathogenic variant was identified in the FBXO11 gene (NM_001190274.2: c.1781A > G; p. His594Arg), confirming the diagnosis of the newly individualized IDDFBA syndrome: Intellectual Developmental Disorder, dysmorphic Facies, and Behavioral Abnormalities (OMIM# 618,089). Our findings further delineate the clinical spectrum linked to FBXO11 and highlight the importance of investigating further cases with mutations in this gene to establish a potential genotype-phenotype correlation.


Asunto(s)
Proteínas F-Box , Fenotipo , Humanos , Masculino , Proteínas F-Box/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Síndrome de Langer-Giedion/genética , Síndrome de Langer-Giedion/patología , Nariz/anomalías , Nariz/patología , Dedos/anomalías , Dedos/patología , Niño , Atresia de las Coanas/genética , Atresia de las Coanas/patología , Mutación , Enfermedades del Cabello , Proteína-Arginina N-Metiltransferasas
16.
Genes (Basel) ; 15(4)2024 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-38674365

RESUMEN

O'Donnell-Luria-Rodan (ODLURO) syndrome is an autosomal dominant disorder caused by mutations in the KMT2E gene. The clinical phonotype of the affected individuals is typically characterized by global developmental delay, autism, epilepsy, hypotonia, macrocephaly, and very mild dysmorphic facial features. In this report, we describe the case of a 6-year-old boy with ODLURO syndrome who is a carrier of the synonymous mutation c.186G>A (p.Ala62=) in the KMT2E gene, predicted to alter splicing by in silico tools. Given the lack of functional studies on the c.186G>A variant, in order to assess its potential functional effect, we sequenced the patient's cDNA demonstrating its impact on the mechanism of splicing. To the best of our knowledge, our patient is the second to date reported carrying this synonymous mutation, but he is the first whose functional investigation has confirmed the deleterious consequence of the variant, resulting in exon 4 skipping. Additionally, we suggest a potential etiological mechanism that could be responsible for the aberrant splicing mechanism in KMT2E.


Asunto(s)
Proteínas de Unión al ADN , Discapacidades del Desarrollo , Niño , Humanos , Masculino , Trastorno Autístico/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Megalencefalia/genética , Fenotipo , Empalme del ARN/genética , Mutación Silenciosa
17.
Genes (Basel) ; 15(4)2024 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-38674371

RESUMEN

The adaptor protein 4 (AP-4) constitutes a conserved hetero-tetrameric complex within the family of adaptor protein (AP) complex, crucial for the signal-mediated trafficking of integral membrane proteins. Mutations affecting all subunits of the AP-4 complex have been linked to autosomal-recessive cerebral palsy and a complex hereditary spastic paraparesis (HSP) phenotype. Our report details the case of a 14-year-old boy born to consanguineous parents, presenting psychomotor delay, severe intellectual disability, microcephaly, and trigonocephaly. Despite a history of febrile seizures, subsequent years were devoid of seizures, with normal EEG. Exome sequencing revealed pathogenic variants in both the AP4B1 and ERF genes. Significantly, the patient exhibited features associated with AP4B1 mutations, including distinctive traits such as cranial malformations. The ERF gene variant, linked to craniosynostosis, likely contributes to the observed trigonocephaly. This case represents the initial documentation of a concurrent mutation in the AP4B1 and ERF genes, underscoring the critical role of exome analysis in unraveling complex phenotypes. Understanding these complex genotypes offers valuable insights into broader syndromic conditions, facilitating comprehensive patient management.


Asunto(s)
Complejo 4 de Proteína Adaptadora , Mutación , Factores de Terminación de Péptidos , Fenotipo , Proteínas Represoras , Humanos , Masculino , Adolescente , Factores de Terminación de Péptidos/genética , Complejo 4 de Proteína Adaptadora/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Secuenciación del Exoma , Microcefalia/genética , Microcefalia/patología , Craneosinostosis/genética , Craneosinostosis/patología
18.
Clin Genet ; 106(1): 109-113, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38665048

RESUMEN

Usmani-Riazuddin syndrome (USRISR, MIM# 619548; USRISD, MIM#619467) is a very rare genetic condition. recently associated with deleterious variants in AP1G1 (MIM* 603533). It is characterized by multisystemic involvement including intellectual disability, speech and developmental delay, behavioral anomalies, muscular tone disorders, seizures, limb defects, and unspecified facial gestalt. In this report, we describe this syndrome for the second time, in association to a novel AP1G1 variant identified in a toddler with multisystemic involvement including intellectual disability, speech and developmental delay, behavioral anomalies, arrhythmias, hearing loss, skin changes, and limb defects. Next generation sequencing (NGS) analysis through clinical exome disclosed AP1G1: c.1969C>G (p.Leu657Val), de novo, likely pathogenic variant, according to ACMG classification criteria. Proband's facial features resembled the spectrum of chromatinopathies. Clinical pictures were analyzed and a clinical overlap was supported by DeepGestalt analysis (www.face2gene.com). The system identified 6 chromatin disorders out of 30 possible diagnoses. The remaining 24 included 9 miscellaneous cryptic chromosomal abnormalities (excluded due to normal microarray study). To the best of our knowledge, this is the first description of likely distinctive facial features in a patient with Usmani-Riazuddin syndrome. Further multicentric analyses are needed for a better definition of this aspect.


Asunto(s)
Discapacidad Intelectual , Fenotipo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación/genética , Preescolar , Femenino , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167178, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636614

RESUMEN

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by haploinsufficiency of transcription factor 4 (TCF4). In this work, we focused on the cerebral cortex and investigated in detail the progenitor cell dynamics and the outcome of neurogenesis in a PTHS mouse model. Labeling and quantification of progenitors and newly generated neurons at various time points during embryonic development revealed alterations affecting the dynamic of cortical progenitors since the earliest stages of cortex formation in PTHS mice. Consequently, establishment of neuronal populations and layering of the cortex were found to be altered in heterozygotes subjects at birth. Interestingly, defective layering process of pyramidal neurons was partially rescued by reintroducing TCF4 expression using focal in utero electroporation in the cerebral cortex. Coincidentally with a defective dorsal neurogenesis, we found that ventral generation of interneurons was also defective in this model, which may lead to an excitation/inhibition imbalance in PTHS. Overall, sex-dependent differences were detected with more marked effects evidenced in males compared with females. All of this contributes to expand our understanding of PTHS, paralleling the advances of research in autism spectrum disorder and further validating the PTHS mouse model as an important tool to advance preclinical studies.


Asunto(s)
Corteza Cerebral , Modelos Animales de Enfermedad , Hiperventilación , Discapacidad Intelectual , Neurogénesis , Factor de Transcripción 4 , Animales , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Femenino , Masculino , Ratones , Hiperventilación/metabolismo , Hiperventilación/genética , Hiperventilación/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Discapacidad Intelectual/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Facies , Caracteres Sexuales , Interneuronas/metabolismo , Interneuronas/patología , Células Piramidales/metabolismo , Células Piramidales/patología , Haploinsuficiencia
20.
Eur J Med Genet ; 69: 104939, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38614309

RESUMEN

ADH5/ALDH2 deficiency is a rare inherited syndrome characterized by short stature, microcephaly, delayed mental development, and hematopoietic dysfunction and has recently been proposed as a disease paradigm. Acute and severe presentations include aplastic anemia, myelodysplastic syndrome, or leukemia, requiring bone marrow transplantation during childhood. Conversely, non-hematological manifestations may exhibit a prolonged and nonspecific clinical trajectory, with growth failure and developmental delay, most of which are often overlooked, particularly in patients with milder symptoms. Here, we describe the clinical course of a girl with a wide spectrum of clinical presentations, including nonspecific hematopoietic disorders, growth retardation, mild developmental delay, amblyopia, hemophagocytic lymphohistiocytosis, and verruca vulgaris, culminating in a genetic diagnosis of AMeD syndrome at 12 years of age. We also summarized the clinical manifestations of previously reported cases of AMeD syndrome. Cumulatively, 13 females and 5 males have been documented, with a cardinal triad of symptoms, aplastic anemia, short stature, and intellectual disability. Additional characteristic observations included pigmentary deposition in approximately half of the cases and skeletal difficulties in one-quarter. We propose that early diagnosis of patients who exhibit relatively mild phenotypes of skin or skeletal lesions is important for managing and improving the quality of life of patients with AMeD syndrome.


Asunto(s)
Fenotipo , Humanos , Femenino , Niño , Aldehído Deshidrogenasa Mitocondrial/genética , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Masculino , Microcefalia/genética , Microcefalia/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Anemia Aplásica/genética , Anemia Aplásica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...