Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.086
Filtrar
1.
Cell Mol Life Sci ; 81(1): 334, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39115595

RESUMEN

Mutations in the IER3IP1 (Immediate Early Response-3 Interacting Protein 1) gene can give rise to MEDS1 (Microcephaly with Simplified Gyral Pattern, Epilepsy, and Permanent Neonatal Diabetes Syndrome-1), a severe condition leading to early childhood mortality. The small endoplasmic reticulum (ER)-membrane protein IER3IP1 plays a non-essential role in ER-Golgi transport. Here, we employed secretome and cell-surface proteomics to demonstrate that the absence of IER3IP1 results in the mistrafficking of proteins crucial for neuronal development and survival, including FGFR3, UNC5B and SEMA4D. This phenomenon correlates with the distension of ER membranes and increased lysosomal activity. Notably, the trafficking of cargo receptor ERGIC53 and KDEL-receptor 2 are compromised, with the latter leading to the anomalous secretion of ER-localized chaperones. Our investigation extended to in-utero knock-down of Ier3ip1 in mouse embryo brains, revealing a morphological phenotype in newborn neurons. In summary, our findings provide insights into how the loss or mutation of a 10 kDa small ER-membrane protein can cause a fatal syndrome.


Asunto(s)
Retículo Endoplásmico , Aparato de Golgi , Microcefalia , Retículo Endoplásmico/metabolismo , Animales , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/patología , Ratones , Aparato de Golgi/metabolismo , Humanos , Mutación , Transporte de Proteínas , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Neuronas/metabolismo , Neuronas/patología
2.
Commun Biol ; 7(1): 831, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977784

RESUMEN

Microtubule associated proteins (MAPs) are widely expressed in the central nervous system, and have established roles in cell proliferation, myelination, neurite formation, axon specification, outgrowth, dendrite, and synapse formation. We report eleven individuals from seven families harboring predicted pathogenic biallelic, de novo, and heterozygous variants in the NAV3 gene, which encodes the microtubule positive tip protein neuron navigator 3 (NAV3). All affected individuals have intellectual disability (ID), microcephaly, skeletal deformities, ocular anomalies, and behavioral issues. In mouse brain, Nav3 is expressed throughout the nervous system, with more prominent signatures in postmitotic, excitatory, inhibiting, and sensory neurons. When overexpressed in HEK293T and COS7 cells, pathogenic variants impaired NAV3 ability to stabilize microtubules. Further, knocking-down nav3 in zebrafish led to severe morphological defects, microcephaly, impaired neuronal growth, and behavioral impairment, which were rescued with co-injection of WT NAV3 mRNA and not by transcripts encoding the pathogenic variants. Our findings establish the role of NAV3 in neurodevelopmental disorders, and reveal its involvement in neuronal morphogenesis, and neuromuscular responses.


Asunto(s)
Discapacidades del Desarrollo , Discapacidad Intelectual , Microcefalia , Animales , Niño , Preescolar , Femenino , Humanos , Masculino , Ratones , Chlorocebus aethiops , Células COS , Discapacidades del Desarrollo/genética , Células HEK293 , Discapacidad Intelectual/genética , Microcefalia/genética , Microcefalia/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Pez Cebra/genética
3.
Genes (Basel) ; 15(7)2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39062689

RESUMEN

Structural variation is a source of genetic variation that, in some cases, may trigger pathogenicity. Here, we describe two cases, a mother and son, with the same partial inverted duplication of the long arm of chromosome 8 [invdup(8)(q24.21q24.21)] of 17.18 Mb, showing different clinical manifestations: microcephaly, dorsal hypertrichosis, seizures and neuropsychomotor development delay in the child, and a cleft lip/palate, down-slanted palpebral fissures and learning disabilities in the mother. The deleterious outcome, in general, is reflected by the gain or loss of genetic material. However, discrepancies among the clinical manifestations raise some concerns about the genomic configuration within the chromosome and other genetic modifiers. With that in mind, we also performed a literature review of research published in the last 20 years about the duplication of the same, or close, chromosome region, seeking the elucidation of at least some relevant clinical features.


Asunto(s)
Cromosomas Humanos Par 8 , Humanos , Femenino , Masculino , Cromosomas Humanos Par 8/genética , Adulto , Inversión Cromosómica/genética , Microcefalia/genética , Microcefalia/patología , Duplicación Cromosómica/genética , Niño , Preescolar
4.
Bioessays ; 46(8): e2300245, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38778437

RESUMEN

Entosis, a form of cell cannibalism, is a newly discovered pathogenic mechanism leading to the development of small brains, termed microcephaly, in which P53 activation was found to play a major role. Microcephaly with entosis, found in Pals1 mutant mice, displays P53 activation that promotes entosis and apoptotic cell death. This previously unappreciated pathogenic mechanism represents a novel cellular dynamic in dividing cortical progenitors which is responsible for cell loss. To date, various recent models of microcephaly have bolstered the importance of P53 activation in cell death leading to microcephaly. P53 activation caused by mitotic delay or DNA damage manifests apoptotic cell death which can be suppressed by P53 removal in these animal models. Such genetic studies attest P53 activation as quality control meant to eliminate genomically unfit cells with minimal involvement in the actual function of microcephaly associated genes. In this review, we summarize the known role of P53 activation in a variety of microcephaly models and introduce a novel mechanism wherein entotic cell cannibalism in neural progenitors is triggered by P53 activation.


Asunto(s)
Apoptosis , Entosis , Microcefalia , Proteína p53 Supresora de Tumor , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Humanos , Ratones , Modelos Animales de Enfermedad
5.
Am J Med Genet A ; 194(9): e63658, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38712921

RESUMEN

We present a case study of a patient exhibiting acquired microcephaly along with global developmental delay and drug-resistant epilepsy. Brain magnetic resonance imaging revealed distinctive features, including a Z-shaped morphology of the brainstem, volumetric reduction of white matter, diffuse thinning of the corpus callosum, and partial fusion of the cerebellar hemispheres at their most cranial portion. Whole-exome sequencing uncovered a pathogenic variant in the ARF3 gene c.200A>T, p.(Asp67Val). The neurodevelopmental disorder associated with the ARF3 gene is exceptionally rare, with only two previously documented cases in the literature. This disorder is characterized by global developmental delay and brain malformations, particularly affecting the white matter, cerebellum, and brainstem. It can also manifest as acquired microcephaly and epilepsy. These phenotypic characteristics align with Golgipathies, underscoring the significance of considering this group of conditions in relevant clinical contexts. In cases where a Z-shaped morphology of the brainstem is observed, ARF3-associated disorder should be included in the list of differential diagnoses.


Asunto(s)
Factores de Ribosilacion-ADP , Trastornos del Neurodesarrollo , Femenino , Humanos , Factores de Ribosilacion-ADP/genética , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Secuenciación del Exoma , Predisposición Genética a la Enfermedad , Imagen por Resonancia Magnética , Microcefalia/genética , Microcefalia/patología , Microcefalia/diagnóstico , Mutación/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/diagnóstico por imagen , Fenotipo , Sustancia Blanca/patología , Sustancia Blanca/diagnóstico por imagen , Preescolar
6.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731817

RESUMEN

MCPH1 has been identified as the causal gene for primary microcephaly type 1, a neurodevelopmental disorder characterized by reduced brain size and delayed growth. As a multifunction protein, MCPH1 has been reported to repress the expression of TERT and interact with transcriptional regulator E2F1. However, it remains unclear whether MCPH1 regulates brain development through its transcriptional regulation function. This study showed that the knockout of Mcph1 in mice leads to delayed growth as early as the embryo stage E11.5. Transcriptome analysis (RNA-seq) revealed that the deletion of Mcph1 resulted in changes in the expression levels of a limited number of genes. Although the expression of some of E2F1 targets, such as Satb2 and Cdkn1c, was affected, the differentially expressed genes (DEGs) were not significantly enriched as E2F1 target genes. Further investigations showed that primary and immortalized Mcph1 knockout mouse embryonic fibroblasts (MEFs) exhibited cell cycle arrest and cellular senescence phenotype. Interestingly, the upregulation of p19ARF was detected in Mcph1 knockout MEFs, and silencing p19Arf restored the cell cycle and growth arrest to wild-type levels. Our findings suggested it is unlikely that MCPH1 regulates neurodevelopment through E2F1-mediated transcriptional regulation, and p19ARF-dependent cell cycle arrest and cellular senescence may contribute to the developmental abnormalities observed in primary microcephaly.


Asunto(s)
Puntos de Control del Ciclo Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Microcefalia , Animales , Ratones , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Fibroblastos/metabolismo , Ratones Noqueados , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/patología
7.
J Med Genet ; 61(8): 759-768, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-38670634

RESUMEN

BACKGROUND: Pontocerebellar hypoplasia (PCH) may present with supratentorial phenotypes and is often accompanied by microcephaly. Damaging mutations in the X-linked gene CASK produce self-limiting microcephaly with PCH in females but are often lethal in males. CASK deficiency leads to early degeneration of cerebellar granule cells but its role in other regions of the brain remains uncertain. METHOD: We generated a conditional Cask knockout mice and deleted Cask ubiquitously after birth at different times. We examined the clinical features in several subjects with damaging mutations clustered in the central part of the CASK protein. We have performed phylogenetic analysis and RT-PCR to assess the splicing pattern within the same protein region and performed in silico structural analysis to examine the effect of splicing on the CASK's structure. RESULT: We demonstrate that deletion of murine Cask after adulthood does not affect survival but leads to cerebellar degeneration and ataxia over time. Intriguingly, damaging hemizygous CASK mutations in boys who display microcephaly and cerebral dysfunction but without PCH are known. These mutations are present in two vertebrate-specific CASK exons. These exons are subject to alternative splicing both in forebrain and hindbrain. Inclusion of these exons differentially affects the molecular structure and hence possibly the function/s of the CASK C-terminus. CONCLUSION: Loss of CASK function disproportionately affects the cerebellum. Clinical data, however, suggest that CASK may have additional vertebrate-specific function/s that play a role in the mammalian forebrain. Thus, CASK has an ancient function shared between invertebrates and vertebrates as well as novel vertebrate-specific function/s.


Asunto(s)
Guanilato-Quinasas , Ratones Noqueados , Animales , Guanilato-Quinasas/genética , Guanilato-Quinasas/química , Ratones , Masculino , Humanos , Femenino , Microcefalia/genética , Microcefalia/patología , Mutación , Exones/genética , Empalme Alternativo/genética , Filogenia , Cerebelo/metabolismo , Cerebelo/anomalías , Cerebelo/patología
8.
Genet Med ; 26(7): 101143, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38641995

RESUMEN

PURPOSE: Neurodevelopmental disorders exhibit clinical and genetic heterogeneity, ergo manifest dysfunction in components of diverse cellular pathways; the precise pathomechanism for the majority remains elusive. METHODS: We studied 5 affected individuals from 3 unrelated families manifesting global developmental delay, postnatal microcephaly, and hypotonia. We used exome sequencing and prioritized variants that were subsequently characterized using immunofluorescence, immunoblotting, pulldown assays, and RNA sequencing. RESULTS: We identified biallelic variants in ZFTRAF1, encoding a protein of yet unknown function. Four affected individuals from 2 unrelated families segregated 2 homozygous frameshift variants in ZFTRAF1, whereas, in the third family, an intronic splice site variant was detected. We investigated ZFTRAF1 at the cellular level and signified it as a nucleocytoplasmic protein in different human cell lines. ZFTRAF1 was completely absent in the fibroblasts of 2 affected individuals. We also identified 110 interacting proteins enriched in mRNA processing and autophagy-related pathways. Based on profiling of autophagy markers, patient-derived fibroblasts show irregularities in the protein degradation process. CONCLUSION: Thus, our findings suggest that biallelic variants of ZFTRAF1 cause a severe neurodevelopmental disorder.


Asunto(s)
Mutación con Pérdida de Función , Microcefalia , Hipotonía Muscular , Trastornos del Neurodesarrollo , Linaje , Humanos , Microcefalia/genética , Microcefalia/patología , Hipotonía Muscular/genética , Hipotonía Muscular/patología , Masculino , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Femenino , Preescolar , Mutación con Pérdida de Función/genética , Alelos , Niño , Lactante , Secuenciación del Exoma , Fibroblastos/metabolismo , Fibroblastos/patología , Autofagia/genética
9.
Am J Med Genet A ; 194(8): e63581, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38600862

RESUMEN

Alu elements are short, interspersed elements located throughout the genome, playing a role in human diversity, and occasionally causing genetic diseases. Here, we report a novel Alu insertion causing Mowat-Wilson syndrome, a rare neurodevelopmental disorder, in an 8-year-old boy displaying the typical clinical features for Mowat-Wilson syndrome. The variant was not initially detected in genome sequencing data, but through deep phenotyping, which pointed to only one plausible candidate gene, manual inspection of genome sequencing alignment data enabled us to identify a de novo heterozygous Alu insertion in exon 8 of the ZEB2 gene. Nanopore long-read sequencing confirmed the Alu insertion, leading to the formation of a premature stop codon and likely haploinsufficiency of ZEB2. This underscores the importance of deep phenotyping and mobile element insertion analysis in uncovering genetic causes of monogenic disorders as these elements might be overlooked in standard next-generation sequencing protocols.


Asunto(s)
Elementos Alu , Facies , Enfermedad de Hirschsprung , Discapacidad Intelectual , Microcefalia , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Humanos , Elementos Alu/genética , Microcefalia/genética , Microcefalia/patología , Masculino , Niño , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Fenotipo , Mutagénesis Insercional/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Exones/genética
10.
Eur J Hum Genet ; 32(8): 920-927, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38605125

RESUMEN

Copy number variants (CNVs) represent the genetic cause of about 15-20% of neurodevelopmental disorders (NDDs). We identified a ~67 kb de novo intragenic deletion on chromosome 2q22.3 in a female individual showing a developmental encephalopathy characterised by epilepsy, severe intellectual disability, speech delay, microcephaly, and thin corpus callosum with facial dysmorphisms. The microdeletion involved exons 5-6 of GTDC1, encoding a putative glycosyltransferase, whose expression is particularly enriched in the nervous system. In a previous study, a balanced de novo translocation encompassing GTDC1 was reported in a male child with global developmental delay and delayed speech and language development. Based on these premises, we explored the transcriptomic profile of our proband to evaluate the functional consequences of the novel GTDC1 de novo intragenic deletion in relation to the observed neurodevelopmental phenotype. RNA-seq on the proband's lymphoblastoid cell line (LCL) showed expression changes of glycine/serine and cytokine/chemokine signalling pathways, which are related to neurodevelopment and epileptogenesis. Subsequent analysis by ELISA (enzyme-linked immunosorbent assay) and HPLC (high-performance liquid chromatography) revealed increased levels of glycine in the proband's LCL and serum compared to matched controls. Given that an increased level of glycine has been observed in the plasma samples of individuals with Rett syndrome, a condition sharing epilepsy, microcephaly, and intellectual disability with our proband, we proposed that the GTDC1 downregulation is implicated in neurodevelopmental impairment by altering glycine metabolism. Furthermore, our findings expanded the phenotypic spectrum of the novel GTDC1-related condition, including microcephaly and epilepsy among relevant clinical features.


Asunto(s)
Glicina , Trastornos del Neurodesarrollo , Humanos , Femenino , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Glicina/metabolismo , Glicina/genética , Cromosomas Humanos Par 2/genética , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/patología , Niño , Microcefalia/genética , Microcefalia/patología , Microcefalia/metabolismo , Deleción Cromosómica , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Discapacidad Intelectual/metabolismo , Preescolar
11.
EMBO Rep ; 25(5): 2418-2440, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38605277

RESUMEN

Microcephaly is a common feature in inherited bone marrow failure syndromes, prompting investigations into shared pathways between neurogenesis and hematopoiesis. To understand this association, we studied the role of the microcephaly gene Mcph1 in hematological development. Our research revealed that Mcph1-knockout mice exhibited congenital macrocytic anemia due to impaired terminal erythroid differentiation during fetal development. Anemia's cause is a failure to complete cell division, evident from tetraploid erythroid progenitors with DNA content exceeding 4n. Gene expression profiling demonstrated activation of the p53 pathway in Mcph1-deficient erythroid precursors, leading to overexpression of Cdkn1a/p21, a major mediator of p53-dependent cell cycle arrest. Surprisingly, fetal brain analysis revealed hypertrophied binucleated neuroprogenitors overexpressing p21 in Mcph1-knockout mice, indicating a shared pathophysiological mechanism underlying both erythroid and neurological defects. However, inactivating p53 in Mcph1-/- mice failed to reverse anemia and microcephaly, suggesting that p53 activation in Mcph1-deficient cells resulted from their proliferation defect rather than causing it. These findings shed new light on Mcph1's function in fetal hematopoietic development, emphasizing the impact of disrupted cell division on neurogenesis and erythropoiesis - a common limiting pathway.


Asunto(s)
Proteínas de Ciclo Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Eritropoyesis , Ratones Noqueados , Microcefalia , Proteína p53 Supresora de Tumor , Animales , Ratones , Anemia Macrocítica/genética , Anemia Macrocítica/patología , Anemia Macrocítica/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Microcefalia/genética , Microcefalia/patología , Mutación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
12.
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
13.
Eur J Med Genet ; 69: 104942, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677542

RESUMEN

Telangiectasia-ectodermal dysplasia-brachydactyly-cardiac anomaly (TEBC) syndrome is a rare autosomal dominant condition, recently linked to the protein kinase D1 (PRKD1) gene. The phenotype of TEBC remains incomplete at this point. Our aim is to improve the characterization of the clinical and molecular aspects of the TEBC syndrome. We report on the 8th patient carrying a heterozygous de novo variation of PRKD1 c.2134G > A, p. (Val712Met) identified by trio exome sequencing. The proband presents with partial atrioventricular septal defect, brachydactyly, ectodermal dysplasia, telangiectasia that developed in childhood, intellectual disability with microcephaly, multicystic renal dysplasia and moderate hormonal resistance. In view of this 8th description and review of the literature, it appears that neurodevelopmental disorders and microcephaly are frequently associated with PRKD1 missense variants, adding to the four main clinical signs described initially in the TEBC syndrome. Further descriptions are required to confirm the observed endocrine and kidney abnormalities. This should contribute to a more comprehensive understanding of the phenotypic spectrum and may help establish genotype-phenotype correlations. In the context of genotype-first strategy, accurate patient descriptions are fundamental. Characterization of specific syndromic associations is essential for variant interpretation support and patient follow-up, even in very rare diseases, such as the TEBC syndrome.


Asunto(s)
Displasia Ectodérmica , Cardiopatías Congénitas , Humanos , Displasia Ectodérmica/genética , Displasia Ectodérmica/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Fenotipo , Braquidactilia/genética , Braquidactilia/patología , Masculino , Telangiectasia/genética , Telangiectasia/patología , Femenino , Mutación Missense , Síndrome , Microcefalia/genética , Microcefalia/patología , Niño , Proteína Quinasa C
14.
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
15.
Mol Genet Genomic Med ; 12(4): e2426, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38562046

RESUMEN

BACKGROUND: Mandibulofacial dysostosis with microcephaly (MFDM, OMIM# 610536) is a rare monogenic disease that is caused by a mutation in the elongation factor Tu GTP binding domain containing 2 gene (EFTUD2, OMIM* 603892). It is characterized by mandibulofacial dysplasia, microcephaly, malformed ears, cleft palate, growth and intellectual disability. MFDM can be easily misdiagnosed due to its phenotypic overlap with other craniofacial dysostosis syndromes. The clinical presentation of MFDM is highly variable among patients. METHODS: A patient with craniofacial anomalies was enrolled and evaluated by a multidisciplinary team. To make a definitive diagnosis, whole-exome sequencing was performed, followed by validation by Sanger sequencing. RESULTS: The patient presented with extensive facial bone dysostosis, upward slanting palpebral fissures, outer and middle ear malformation, a previously unreported orbit anomaly, and spina bifida occulta. A novel, pathogenic insertion mutation (c.215_216insT: p.Tyr73Valfs*4) in EFTUD2 was identified as the likely cause of the disease. CONCLUSIONS: We diagnosed this atypical case of MFDM by the detection of a novel pathogenetic mutation in EFTUD2. We also observed previously unreported features. These findings enrich both the genotypic and phenotypic spectrum of MFDM.


Asunto(s)
Discapacidad Intelectual , Disostosis Mandibulofacial , Microcefalia , Humanos , Microcefalia/patología , Disostosis Mandibulofacial/genética , Disostosis Mandibulofacial/patología , Fenotipo , Mutación , Discapacidad Intelectual/genética , Factores de Elongación de Péptidos/genética , Factores de Elongación de Péptidos/metabolismo , Ribonucleoproteína Nuclear Pequeña U5/genética , Ribonucleoproteína Nuclear Pequeña U5/metabolismo
16.
Sci Rep ; 14(1): 9497, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664418

RESUMEN

Raine syndrome (RNS) is a rare autosomal recessive osteosclerotic dysplasia. RNS is caused by loss-of-function disease-causative variants of the FAM20C gene that encodes a kinase that phosphorylates most of the secreted proteins found in the body fluids and extracellular matrix. The most common RNS clinical features are generalized osteosclerosis, facial dysmorphism, intracerebral calcifications and respiratory defects. In non-lethal RNS forms, oral traits include a well-studied hypoplastic amelogenesis imperfecta (AI) and a much less characterized gingival phenotype. We used immunomorphological, biochemical, and siRNA approaches to analyze gingival tissues and primary cultures of gingival fibroblasts of two unrelated, previously reported RNS patients. We showed that fibrosis, pathological gingival calcifications and increased expression of various profibrotic and pro-osteogenic proteins such as POSTN, SPARC and VIM were common findings. Proteomic analysis of differentially expressed proteins demonstrated that proteins involved in extracellular matrix (ECM) regulation and related to the TGFß/SMAD signaling pathway were increased. Functional analyses confirmed the upregulation of TGFß/SMAD signaling and subsequently uncovered the involvement of two closely related transcription cofactors important in fibrogenesis, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). Knocking down of FAM20C confirmed the TGFß-YAP/TAZ interplay indicating that a profibrotic loop enabled gingival fibrosis in RNS patients. In summary, our in vivo and in vitro data provide a detailed description of the RNS gingival phenotype. They show that gingival fibrosis and calcifications are associated with, and most likely caused by excessed ECM production and disorganization. They furthermore uncover the contribution of increased TGFß-YAP/TAZ signaling in the pathogenesis of the gingival fibrosis.


Asunto(s)
Anomalías Múltiples , Proteínas Adaptadoras Transductoras de Señales , Fisura del Paladar , Hipoplasia del Esmalte Dental , Exoftalmia , Fibroblastos , Fibrosis , Encía , Osteosclerosis , Proteómica , Transducción de Señal , Factores de Transcripción , Factor de Crecimiento Transformador beta , Proteínas Señalizadoras YAP , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Encía/metabolismo , Encía/patología , Proteómica/métodos , Fibrosis/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Osteosclerosis/metabolismo , Osteosclerosis/genética , Osteosclerosis/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Hipoplasia del Esmalte Dental/metabolismo , Hipoplasia del Esmalte Dental/genética , Hipoplasia del Esmalte Dental/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Microcefalia/metabolismo , Microcefalia/genética , Microcefalia/patología , Femenino , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Masculino , Transactivadores/metabolismo , Transactivadores/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasa de la Caseína I/metabolismo , Quinasa de la Caseína I/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Amelogénesis Imperfecta/metabolismo , Amelogénesis Imperfecta/genética , Amelogénesis Imperfecta/patología , Células Cultivadas
17.
Am J Med Genet A ; 194(7): e63598, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38501757

RESUMEN

Primary microcephaly is characterized by a head circumference prenatally or at birth that falls below three standard deviations from age-, ethnic-, and sex-specific norms. Genetic defects are one of the underlying causes of primary microcephaly. Since 2014, five variants of the SASS6 gene have been identified as the cause of MCPH 14 in three reported families. In this study, we present the genetic findings of members of a nonconsanguineous Chinese couple with a history of microcephaly and fetal growth restriction (FGR) during their first pregnancy. Utilizing trio whole-exome sequencing, we identified compound heterozygous variants involving a frameshift NM_194292.3:c.450_453del p.(Lys150AsnfsTer7) variant and a splice region NM_194292.3:c.1674+3A>G variant within the SASS6 gene in the affected fetus. Moreover, reverse transcriptase-polymerase chain reaction from RNA of the mother's peripheral blood leukocytes revealed that the c.1674+3A>G variant led to the skipping of exon 14 and an inframe deletion. To the best of our knowledge, the association between FGR and SASS6-related microcephaly has not been reported, and our findings confirm the pivotal role of SASS6 in microcephaly pathogenesis and reveal an expanded view of the phenotype and mutation spectrum associated with this gene.


Asunto(s)
Alelos , Secuenciación del Exoma , Retardo del Crecimiento Fetal , Microcefalia , Humanos , Microcefalia/genética , Microcefalia/patología , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/patología , Femenino , Masculino , Embarazo , Linaje , Mutación/genética , Adulto , Predisposición Genética a la Enfermedad , Fenotipo
18.
Am J Med Genet A ; 194(7): e63599, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38517182

RESUMEN

Pathogenic variants in TRIO, encoding the guanine nucleotide exchange factor, are associated with two distinct neurodevelopmental delay phenotypes: gain-of-function missense mutations within the spectrin repeats are causative for a severe developmental delay with macrocephaly (MIM: 618825), whereas loss-of-function missense variants in the GEF1 domain and truncating variants throughout the gene lead to a milder developmental delay and microcephaly (MIM: 617061). In three affected family members with mild intellectual disability/NDD and microcephaly, we detected a novel heterozygous TRIO variant at the last coding base of exon 31 (NM_007118.4:c.4716G>A). RNA analysis from patient-derived lymphoblastoid cells confirmed aberrant splicing resulting in the skipping of exon 31 (r.4615_4716del), leading to an in-frame deletion in the first Pleckstrin homology subdomain of the GEF1 domain: p.(Thr1539_Lys1572del). To test for a distinct gestalt, facial characteristics of the family members and 41 previously published TRIO cases were systematically evaluated via GestaltMatcher. Computational analysis of the facial gestalt suggests a distinguishable facial TRIO-phenotype not outlined in the existing literature.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido , Linaje , Fenotipo , Sitios de Empalme de ARN , Humanos , Factores de Intercambio de Guanina Nucleótido/genética , Masculino , Femenino , Sitios de Empalme de ARN/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Microcefalia/genética , Microcefalia/patología , Niño , Preescolar , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Exones/genética , Empalme del ARN/genética , Facies , Proteínas Serina-Treonina Quinasas
19.
Am J Med Genet A ; 194(8): e63593, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38549403

RESUMEN

Biallelic pathogenic variants in ZNF335 are one of the genetic causes of microcephaly, reported only in the past decade. It regulates neural progenitor proliferation and neurogenesis by interacting with a H3K4 methyltransferase complex. Biallelic pathogenic ZNF335 variants predispose to neuronal cell death and aberrant differentiation, thus causing secondary microcephaly. These neurodevelopmental anomalies lead to imaging findings in the cortex, posterior fossa, and basal ganglia. We report an individual of Nepalese ancestry with a novel homozygous ZNF335 variant (c.3591 + 2dup) (p.?) (NM_022095.3) which on further RNA analysis confirmed a splice site variant in intron 23. The patient presented with primary microcephaly with atrophic cerebral hemispheres, oversimplification of gyri, basal ganglia, and corpus callosal atrophy. Literature review on the topic revealed a spectrum of brain abnormalities, which can present either with a primary or secondary microcephaly depending upon the underlying genetic variant.


Asunto(s)
Alelos , Proteínas de Unión al ADN , Microcefalia , Factores de Transcripción , Humanos , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Encéfalo/anomalías , Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Homocigoto , Imagen por Resonancia Magnética , Microcefalia/genética , Microcefalia/patología , Mutación/genética , Factores de Transcripción/genética
20.
Genet Med ; 26(6): 101104, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38411040

RESUMEN

PURPOSE: The functionality of many cellular proteins depends on cofactors; yet, they have only been implicated in a minority of Mendelian diseases. Here, we describe the first 2 inherited disorders of the cytosolic iron-sulfur protein assembly system. METHODS: Genetic testing via genome sequencing was applied to identify the underlying disease cause in 3 patients with microcephaly, congenital brain malformations, progressive developmental and neurologic impairments, recurrent infections, and a fatal outcome. Studies in patient-derived skin fibroblasts and zebrafish models were performed to investigate the biochemical and cellular consequences. RESULTS: Metabolic analysis showed elevated uracil and thymine levels in body fluids but no pathogenic variants in DPYD, encoding dihydropyrimidine dehydrogenase. Genome sequencing identified compound heterozygosity in 2 patients for missense variants in CIAO1, encoding cytosolic iron-sulfur assembly component 1, and homozygosity for an in-frame 3-nucleotide deletion in MMS19, encoding the MMS19 homolog, cytosolic iron-sulfur assembly component, in the third patient. Profound alterations in the proteome, metabolome, and lipidome were observed in patient-derived fibroblasts. We confirmed the detrimental effect of deficiencies in CIAO1 and MMS19 in zebrafish models. CONCLUSION: A general failure of cytosolic and nuclear iron-sulfur protein maturation caused pleiotropic effects. The critical function of the cytosolic iron-sulfur protein assembly machinery for antiviral host defense may well explain the recurrent severe infections occurring in our patients.


Asunto(s)
Proteínas Hierro-Azufre , Pez Cebra , Animales , Humanos , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Masculino , Femenino , Fenotipo , Fibroblastos/metabolismo , Fibroblastos/patología , Citosol/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Microcefalia/genética , Microcefalia/patología , Lactante , Metalochaperonas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA