Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.658
Filtrar
1.
J Pregnancy ; 2024: 1758662, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38961858

RESUMEN

Congenital Zika syndrome (CZS) is a major concern in India and highlights the multifaceted challenges posed by the Zika virus (ZIKV). The alarming increase in CZS cases in India, a condition that has serious effects on both public health and newborns, has raised concerns. This review highlights the importance of raising concern and awareness and taking preventive measures by studying the epidemiology, clinical symptoms, and potential long-term consequences of CZS. The review also contributes to worldwide research and information sharing to improve the understanding and prevention of CZS. As India deals with the changing nature of CZS, this thorough review is an important tool for policymakers, health workers, and researchers to understand what is happening now, plan for what to do in the future, and work together as a team, using medical knowledge, community involvement, and study projects to protect newborns' health and reduce the public health impact of these syndromes.


Asunto(s)
Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika , Humanos , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/congénito , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/complicaciones , India/epidemiología , Embarazo , Recién Nacido , Complicaciones Infecciosas del Embarazo/epidemiología , Complicaciones Infecciosas del Embarazo/prevención & control , Femenino , Virus Zika , Microcefalia/epidemiología , Microcefalia/virología , Microcefalia/etiología
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 , Humanos , Microcefalia/genética , Microcefalia/patología , Discapacidad Intelectual/genética , Animales , Masculino , Femenino , Ratones , Discapacidades del Desarrollo/genética , Células HEK293 , Pez Cebra/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Preescolar , Chlorocebus aethiops , Células COS , Niño , Neuronas/metabolismo , Neuronas/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(4): 649-654, 2024 Apr 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-39019795

RESUMEN

O'Donnell-Luria-Rodan (ODLURO) syndrome is an autosomal dominant genetic disorder caused by mutations in the KMT2E (lysine methyltransferase 2E) gene. The Third Xiangya Hospital of Central South University admitted a 12-year and 9-month-old male patient who presented with growth retardation, intellectual disability, and distinctive facial features. Peripheral blood was collected from the patient, and DNA was extracted for genetic testing. Chromosome karyotyping showed 46XY. Whole-exome sequencing and low-coverage massively parallel copy number variation sequencing (CNV-seq) revealed a 506 kb heterozygous deletion in the 7q22.3 region, which includes 6 genes, including KMT2E. The patient was diagnosed with ODLURO syndrome. Both the patient's parents and younger brother had normal clinical phenotypes and genetic test results, indicating that this deletion was a de novo mutation. The clinical and genetic characteristics of this case can help increase clinicians' awareness of ODLURO syndrome.


Asunto(s)
Discapacidad Intelectual , Humanos , Masculino , Discapacidad Intelectual/genética , Niño , N-Metiltransferasa de Histona-Lisina/genética , Mutación , Trastornos del Crecimiento/genética , Anomalías Múltiples/genética , Cromosomas Humanos Par 7/genética , Cariotipificación , Fenotipo , Variaciones en el Número de Copia de ADN , Secuenciación del Exoma , Heterocigoto , Contractura , Microcefalia , Facies
4.
Mol Biol Rep ; 51(1): 783, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926176

RESUMEN

BACKGROUND: Autosomal recessive primary microcephaly (MCPH) is a rare neurodevelopmental and genetically heterogeneous disorder, characterized by small cranium size (> - 3 SD below mean) and often results in varying degree of intellectual disability. Thirty genes have been identified for the etiology of this disorder due to its clinical and genetic heterogeneity. METHODS AND RESULTS: Here, we report two consanguineous Pakistani families affected with MCPH exhibiting mutation in WDR62 gene. The investigation approach involved Next Generation Sequencing (NGS) gene panel sequencing coupled with linkage analysis followed by validation of identified variants through automated Sanger sequencing and Barcode-Tagged (BT) sequencing. The molecular genetic analysis revealed one novel splice site variant (NM_001083961.2(WDR62):c.1372-1del) in Family A and one known exonic variant NM_001083961.2(WDR62):c.3936dup (p.Val1313Argfs*18) in Family B. Magnetic Resonance Imaging (MRI) scans were also employed to gain insights into the structural architecture of affected individuals. Neurological assessments showed the reduced gyral and sulcal patterns along with normal corpus callosum in affected individuals harboring novel variant. In silico assessments of the identified variants were conducted using different tools to confirm the pathogenicity of these variants. Through In silico analyses, both variants were identified as disease causing and protein modeling of exonic variant indicates subtle conformational alterations in prophesied protein structure. CONCLUSION: This study identifies a novel variant (c.1372-1del) and a recurrent pathogenic variant c.3936dup (p.Val1313Argfs*18) in the WDR62 gene among the Pakistani population, expanding the mutation spectrum for MCPH. These findings emphasize the importance of genetic counseling and awareness to reduce consanguinity and address the burden of this disorder.


Asunto(s)
Consanguinidad , Microcefalia , Mutación , Proteínas del Tejido Nervioso , Linaje , Humanos , Microcefalia/genética , Femenino , Masculino , Pakistán , Mutación/genética , Proteínas del Tejido Nervioso/genética , Neuroimagen/métodos , Niño , Imagen por Resonancia Magnética/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Preescolar , Adolescente , Proteínas de Ciclo Celular
5.
J Child Neurol ; 39(5-6): 209-217, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38847106

RESUMEN

Background: Genetic microcephaly is linked to an increased risk of developmental disabilities, epilepsy, and motor impairment. The aim of this study is to describe the spectrum of identifiable genetic etiologies, clinical characteristics, and radiologic features of genetic microcephaly in patients referred to a tertiary center in Saudi Arabia. Method: This is a retrospective chart review study of all patients with identifiable genetic microcephaly presenting to a tertiary center in Saudi Arabia. The patients' demographics, clinical, laboratory, radiologic, and molecular findings were collected. Results: Of the total 128 cases referred, 52 cases (40%) had identifiable genetic causes. Monogenic disorders were found in 48 cases (92%), whereas chromosomal disorders were found in only 4 cases (8%). Developmental disability was observed in 40 cases (84%), whereas only 8 cases (16%) had borderline IQ or mild developmental delay. Epilepsy was seen in 29 cases (56%), and motor impairment was seen in 26 cases (50%). Brain magnetic resonance imaging (MRI) revealed abnormalities in 26 (50%) of the cohort. Hereditary neurometabolic disorders were seen in 7 (15%) of the 48 cases with monogenic disorders. The most common gene defect was ASPM, which is responsible for primary microcephaly type 5 and was seen in 10 cases (19%). A novel PLK1 gene pathogenic mutation was seen in 3 cases (6%). Conclusion: Single gene defect is common in this Saudi population, with the ASPM gene being the most common. Hereditary neurometabolic disorders are a common cause of genetic microcephaly. Furthermore, we propose the PKL1 gene mutation as a possible novel cause of genetic microcephaly.


Asunto(s)
Microcefalia , Humanos , Microcefalia/genética , Arabia Saudita , Masculino , Femenino , Estudios Retrospectivos , Preescolar , Niño , Lactante , Adolescente , Proteínas del Tejido Nervioso/genética , Imagen por Resonancia Magnética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Mutación/genética , Encéfalo/diagnóstico por imagen , Encéfalo/patología
6.
Dialogues Clin Neurosci ; 26(1): 24-27, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38829782

RESUMEN

INTRODUCTION: Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder with a multifaceted etiology. This case report explores the ischemic cryptogenic vascular dissection as a potential underlying cause of ASD. METHODS: A 9-year-old child presented with symptoms of ASD, including social interaction difficulties, repetitive behaviors, and cognitive challenges. Despite conventional ASD treatments, significant improvement was only observed after addressing an underlying ischemic cryptogenic vascular dissection identified through DCE-CT. RESULTS: Following a reconstructive treatment approach to the vascular dissection, the patient showed marked improvement in cognitive functions, social abilities, and a reduction in ASD-related symptoms whether during the perioperative period or during approximately 5-month follow-up. CONCLUSION: This case suggests that ischemic cryptogenic vascular dissection may contribute to the symptoms of ASD. Identifying and treating underlying vascular anomalies may offer a new avenue for mitigating ASD symptoms, emphasizing the need for comprehensive diagnostic estimations in ASD management.


Asunto(s)
Trastorno del Espectro Autista , Humanos , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/complicaciones , Niño , Masculino , Microcefalia/complicaciones , Microcefalia/diagnóstico
7.
Neuron ; 112(13): 2157-2176.e12, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38697111

RESUMEN

Mutations in human nonsense-mediated mRNA decay (NMD) factors are enriched in neurodevelopmental disorders. We show that deletion of key NMD factor Upf2 in mouse embryonic neural progenitor cells causes perinatal microcephaly but deletion in immature neurons does not, indicating NMD's critical roles in progenitors. Upf2 knockout (KO) prolongs the cell cycle of radial glia progenitor cells, promotes their transition into intermediate progenitors, and leads to reduced upper-layer neurons. CRISPRi screening identified Trp53 knockdown rescuing Upf2KO progenitors without globally reversing NMD inhibition, implying marginal contributions of most NMD targets to the cell cycle defect. Integrated functional genomics shows that NMD degrades selective TRP53 downstream targets, including Cdkn1a, which, without NMD suppression, slow the cell cycle. Trp53KO restores the progenitor cell pool and rescues the microcephaly of Upf2KO mice. Therefore, one physiological role of NMD in the developing brain is to degrade selective TRP53 targets to control progenitor cell cycle and brain size.


Asunto(s)
Encéfalo , Ratones Noqueados , Células-Madre Neurales , Degradación de ARNm Mediada por Codón sin Sentido , Proteína p53 Supresora de Tumor , Animales , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Ratones , Encéfalo/metabolismo , Células-Madre Neurales/metabolismo , Degradación de ARNm Mediada por Codón sin Sentido/genética , Epistasis Genética , Microcefalia/genética , Ciclo Celular/fisiología , Ciclo Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
8.
Birth Defects Res ; 116(5): e2346, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38761025

RESUMEN

AIM: Nijmegen breakage syndrome (NBS) is an autosomal recessive DNA repair disorder which is characterized by immunodeficiency and increased risk of lymphoproliferative malignancy. CASE: We observed an increase in the rate of chromosomal rearrangements in the cultured cells following an incidental radiograph for craniosynostosis in a newborn who was followed up due to microcephaly. We identified a homozygous deletion of c.657_661delACAAA/p.Lys219fs (rs587776650) in the NBN gene through whole exome sequencing. CONCLUSION: It is crucial to thoroughly examine the clinical features of newborns with microcephaly and consider chromosomal instability syndromes just like Nijmegen breakage syndrome. Not overlooking radiosensitivity, which is a characteristic feature of this syndrome, is a vital condition to the patient's survival time.


Asunto(s)
Microcefalia , Síndrome de Nijmegen , Tolerancia a Radiación , Humanos , Síndrome de Nijmegen/genética , Recién Nacido , Tolerancia a Radiación/genética , Microcefalia/genética , Proteínas de Ciclo Celular/genética , Proteínas Nucleares/genética , Masculino , Secuenciación del Exoma/métodos , Femenino , Homocigoto
9.
Neurogenetics ; 25(3): 179-191, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38795246

RESUMEN

Primary microcephaly is a rare neurogenic and genetically heterogeneous disorder characterized by significant brain size reduction that results in numerous neurodevelopmental disorders (NDD) problems, including mild to severe intellectual disability (ID), global developmental delay (GDD), seizures and other congenital malformations. This disorder can arise from a mutation in genes involved in various biological pathways, including those within the brain. We characterized a recessive neurological disorder observed in nine young adults from five independent consanguineous Pakistani families. The disorder is characterized by microcephaly, ID, developmental delay (DD), early-onset epilepsy, recurrent infection, hearing loss, growth retardation, skeletal and limb defects. Through exome sequencing, we identified novel homozygous variants in five genes that were previously associated with brain diseases, namely CENPJ (NM_018451.5: c.1856A > G; p.Lys619Arg), STIL (NM_001048166.1: c.1235C > A; p.(Pro412Gln), CDK5RAP2 (NM_018249.6 c.3935 T > G; p.Leu1312Trp), RBBP8 (NM_203291.2 c.1843C > T; p.Gln615*) and CEP135 (NM_025009.5 c.1469A > G; p.Glu490Gly). These variants were validated by Sanger sequencing across all family members, and in silico structural analysis. Protein 3D homology modeling of wild-type and mutated proteins revealed substantial changes in the structure, suggesting a potential impact on function. Importantly, all identified genes play crucial roles in maintaining genomic integrity during cell division, with CENPJ, STIL, CDK5RAP2, and CEP135 being involved in centrosomal function. Collectively, our findings underscore the link between erroneous cell division, particularly centrosomal function, primary microcephaly and ID.


Asunto(s)
Proteínas de Ciclo Celular , Discapacidad Intelectual , Microcefalia , Linaje , Humanos , Microcefalia/genética , Discapacidad Intelectual/genética , Masculino , Femenino , Proteínas de Ciclo Celular/genética , Adulto , Proteínas Cromosómicas no Histona/genética , Proteínas del Tejido Nervioso/genética , División Celular/genética , Mutación , Péptidos y Proteínas de Señalización Intracelular/genética , Genómica , Adulto Joven , Consanguinidad , Secuenciación del Exoma , Homocigoto , Discapacidades del Desarrollo/genética , Adolescente , Pakistán , Proteínas Asociadas a Microtúbulos
10.
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
11.
Psychiatr Genet ; 34(3): 74-80, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690959

RESUMEN

BACKGROUND: Biallelic loss-of-function variants in SMPD4 cause a rare and severe neurodevelopmental disorder. These variants have been identified in a group of children with neurodevelopmental disorders with microcephaly, arthrogryposis, and structural brain anomalies. SMPD4 encodes a sphingomyelinase that hydrolyzes sphingomyelin into ceramide at neutral pH and can thereby affect membrane lipid homeostasis. SMPD4 localizes to the membranes of the endoplasmic reticulum and nuclear envelope and interacts with nuclear pore complexes. MATERIALS AND METHODS: For the efficient prenatal diagnosis of rare and undiagnosed diseases, the parallel detection of copy number variants (CNVs) and single nucleotide variants using whole-exome analysis is required. A physical examination of the parents was performed. Karyotype and whole-exome analysis were performed for the fetus and the parents. RESULTS: A fetus with microcephaly and arthrogryposis; biallelic null variants (c.387-1G>A; Chr2[GRCh38]: g.130142742_130202459del) were detected by whole-exome sequencing (WES). We have reported for the first time the biallelic loss-of-function mutations in SMPD4 in patients born to unrelated parents in China. CONCLUSION: WES could replace chromosomal microarray analysis and copy number variation sequencing as a more cost-effective genetic test for detecting CNVs and diagnosing highly heterogeneous conditions.


Asunto(s)
Variaciones en el Número de Copia de ADN , Secuenciación del Exoma , Microcefalia , Polimorfismo de Nucleótido Simple , Diagnóstico Prenatal , Esfingomielina Fosfodiesterasa , Humanos , Variaciones en el Número de Copia de ADN/genética , Secuenciación del Exoma/métodos , Femenino , Diagnóstico Prenatal/métodos , Esfingomielina Fosfodiesterasa/genética , Polimorfismo de Nucleótido Simple/genética , Embarazo , Microcefalia/genética , Heterocigoto , Artrogriposis/genética , Artrogriposis/diagnóstico , Masculino , Exoma/genética , Mutación/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/diagnóstico
12.
Seizure ; 118: 148-155, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38704883

RESUMEN

PURPOSE: This study aimed to identify continuous epileptiform discharges (CEDs) on electroencephalograms (EEG) and to determine their clinical significance in children with congenital Zika syndrome (CZS). METHODS: This prospective cohort study included 75 children diagnosed with CZS born from March 2015 and followed up until September 2018 (age up to 36 months). EEG was performed to detect CEDs up to 24 months old. Data on obstetric, demographic, and clinical signs; cranial computed tomography (CT); ophthalmology examination; anti-seizure medication; growth; and motor development were collected. Fisher's exact test was used to verify the associations between categorical variables, and the T- test was used to compare the mean z-scores of anthropometric measurements between the groups with and without CED. RESULTS: CEDs were identified in 41 (54.67 %) children. The mean age of CEDs identification was 12.24 ± 6.86 months. Bilateral CEDs were shown in 62.89 % of EEGs. CEDs were associated with severe congenital microcephaly, defined by z-score >3 standard deviation of head circumference (HC) below the mean for sex and age (p = 0.025), and worse outcomes, including first seizure before 6 months (p = 0.004), drug-resistant epilepsy (p < 0.001), chorioretinal scarring or mottling (p = 0.002), and severe CT findings (p = 0.002). The CED group had lower mean z-scores of HC up to 24 months of age. CONCLUSION: This is the first description of the prevalence and significance of CEDs that also remains during wakefulness in patients with CZS. New investigations may suggest that it is more appropriate to classify the EEG not as a CED, but as a periodic pattern. Anyway, CEDs may be a marker of neurological severity in children with CSZ.


Asunto(s)
Electroencefalografía , Infección por el Virus Zika , Humanos , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/fisiopatología , Infección por el Virus Zika/congénito , Femenino , Masculino , Lactante , Estudios Prospectivos , Preescolar , Microcefalia/fisiopatología , Microcefalia/diagnóstico por imagen , Epilepsia/fisiopatología , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/fisiopatología
13.
Am J Med Genet A ; 194(8): e63581, 2024 Aug.
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
15.
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
16.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...