Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 345
Filtrar
Más filtros

Intervalo de año de publicación
1.
Am J Med Genet A ; 191(5): 1293-1300, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36810866

RESUMEN

Our study of 61 children with NAA10-related neurodevelopmental syndrome, an X-linked disorder due to NAA10 gene variants, demonstrated a high prevalence of growth failure, with weight and height percentiles often in the failure-to-thrive diagnostic range; however, dramatic weight fluctuations and phenotypic variability is evidenced in the growth parameters of this population. Although never previously explored in depth, the gastrointestinal pathology associated with NAA10-related neurodevelopmental syndrome includes feeding difficulties in infancy, dysphagia, GERD/silent reflux, vomiting, constipation, diarrhea, bowel incontinence, and presence of eosinophils on esophageal endoscopy, in order from most to least prevalent. Additionally, the gastrointestinal symptom profile for children with this syndrome has been expanded to include eosinophilic esophagitis, cyclic vomiting syndrome, Mallory Weiss tears, abdominal migraine, esophageal dilation, and subglottic stenosis. Although the exact cause of poor growth in NAA10-related neurodevelopmental syndrome probands is unclear and the degree of contribution to this problem by GI symptomatology remains uncertain, an analysis including nine G-tube or GJ-tube fed probands demonstrates that G/GJ-tubes are overall efficacious with respect to improvements in weight gain and caregiving. The choice to insert a gastrostomy or gastrojejunal tube to aid with weight gain is often a challenging decision to make for parents, who may alternatively choose to rely on oral feeding, caloric supplementation, calorie tracking, and feeding therapy. In this case, if NAA10-related neurodevelopmental syndrome children are not tracking above the failure to thrive (FTT) range past 1 year of age despite such efforts, the treating physicians should be consulted regarding possibly undergoing G-tube placement to avoid prolonged growth failure. If G-tubes are not immediately inducing weight gain after insertion, recommendations could include altering formula, increasing caloric input, or exchanging a G-tube for a GJ-tube by means of a minimally invasive procedure.


Asunto(s)
Nutrición Enteral , Reflujo Gastroesofágico , Niño , Humanos , Nutrición Enteral/métodos , Gastrostomía/métodos , Reflujo Gastroesofágico/cirugía , Síndrome , Insuficiencia de Crecimiento/genética , Aumento de Peso , Variación Biológica Poblacional , Acetiltransferasa A N-Terminal/genética , Acetiltransferasa E N-Terminal
2.
Am J Med Genet A ; 191(8): 2074-2082, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37194190

RESUMEN

Costello syndrome is a clinically recognizable, severe neurodevelopmental disorder caused by heterozygous activating variants in HRAS. The vast majority of affected patients share recurring variants affecting HRAS codons 12 and 13 and a relatively uniform phenotype. Here, we report the unique and attenuated phenotype of six individuals of an extended family affected by the HRAS variant c.176C>T p.(Ala59Gly), which, to our knowledge, has never been reported as a germline variant in patients so far. HRAS Alanine 59 has been previously functionally investigated as an oncogenic hotspot and the p.Ala59Gly substitution was shown to impair intrinsic GTP hydrolysis. All six individuals we report share a phenotype of ectodermal anomalies and mild features suggestive of a RASopathy, reminiscent of patients with Noonan syndrome-like disorder with loose anagen hair. All six are of normal intelligence, none have a history of failure to thrive or malignancy, and they have no known cardiac or neurologic pathologies. Our report adds to the previous reports of patients with rare variants affecting amino acids located in the SWITCH II/G3 region of HRAS and suggests a consistent, attenuated phenotype distinct from classical Costello syndrome. We propose the definition of a new distinct HRAS-related RASopathy for patients carrying HRAS variants affecting codons 58, 59, 60.


Asunto(s)
Síndrome de Costello , Síndrome de Noonan , Humanos , Síndrome de Costello/genética , Síndrome de Costello/patología , Fenotipo , Síndrome de Noonan/genética , Síndrome de Noonan/patología , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/patología , Mutación de Línea Germinal , Proteínas Proto-Oncogénicas p21(ras)/genética
3.
Am J Med Genet A ; 191(11): 2723-2727, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37668308

RESUMEN

Missense variants in the RNF13 gene have been previously known to cause congenital microcephaly, epileptic encephalopathy, blindness, and failure to thrive through a gain-of-function disease mechanism. Here, we identify a nonsense variant, expected to result in protein truncation, in a similarly affected patient. We show that this nonsense variant, residing in the terminal exon, is likely to escape nonsense-mediated decay while removing a critical region for protein function, thus resulting in a gain-of-function effect. We review the literature and disease databases and identify several other affected individuals with overlapping phenotypes carrying distinct truncating variants in the terminal exon upstream of the putative critical region. Furthermore, we analyze truncating variants from the general population, namely, the Genome Aggregation Database (gnomAD), and provide additional evidence supporting our hypothesis, and ruling out haploinsufficiency as an alternative disease mechanism. In summary, our case report, literature review, and analysis of disease and population databases strongly support the hypothesis that heterozygous gain-of-function variants in a critical region of RNF13 cause congenital microcephaly, epileptic encephalopathy, blindness, and failure to thrive.


Asunto(s)
Microcefalia , Espasmos Infantiles , Humanos , Microcefalia/genética , Insuficiencia de Crecimiento/genética , Mutación con Ganancia de Función , Espasmos Infantiles/genética , Ceguera , Ubiquitina-Proteína Ligasas/genética
4.
Cardiovasc Drugs Ther ; 37(6): 1193-1204, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35156148

RESUMEN

The RAS/mitogen-activated protein kinase (MAPK) pathway controls a plethora of developmental and post-developmental processes. It is now clear that mutations in the RAS-MAPK pathway cause developmental diseases collectively referred to as the RASopathies. The RASopathies include Noonan syndrome, Noonan syndrome with multiple lentigines, cardiofaciocutaneous syndrome, neurofibromatosis type 1, and Costello syndrome. RASopathy patients exhibit a wide spectrum of congenital heart defects (CHD), such as valvular abnormalities and hypertrophic cardiomyopathy (HCM). Since the cardiovascular defects are the most serious and recurrent cause of mortality in RASopathy patients, it is critical to understand the pathological signaling mechanisms that drive the disease. Therapies for the treatment of HCM and other RASopathy-associated comorbidities have yet to be fully realized. Recent developments have shown promise for the use of repurposed antineoplastic drugs that target the RAS-MAPK pathway for the treatment of RASopathy-associated HCM. However, given the impact of the RAS-MAPK pathway in post-developmental physiology, establishing safety and evaluating risk when treating children will be paramount. As such insight provided by preclinical and clinical information will be critical. This review will highlight the cardiovascular manifestations caused by the RASopathies and will discuss the emerging therapies for treatment.


Asunto(s)
Síndrome de Costello , Displasia Ectodérmica , Cardiopatías Congénitas , Síndrome de Noonan , Niño , Humanos , Cardiopatías Congénitas/tratamiento farmacológico , Cardiopatías Congénitas/genética , Síndrome de Noonan/tratamiento farmacológico , Síndrome de Noonan/genética , Síndrome de Costello/genética , Insuficiencia de Crecimiento/tratamiento farmacológico , Insuficiencia de Crecimiento/genética , Displasia Ectodérmica/genética
5.
J Med Genet ; 59(12): 1241-1246, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35906012

RESUMEN

BACKGROUND: Two imprinting control centres, H19/IGF2:IG-differentialy methylated region (DMR) and KCNQ1OT1:TSS-DMR, reside on chromosome 11p15.5. Paternal deletions involving the KCNQ1OT1:TSS-DMR result in variable phenotypes, namely, normal phenotype, Silver-Russel syndrome (SRS) and fetal demise. However, expression analyses for CDKN1C in these patients are very limited. CASES: Patient 1 (adult woman) and patient 2 (boy in early childhood) showed prenatal and postnatal growth failure and clinical suspicion of SRS. MOLECULAR ANALYSES: Both patients showed hypermethylation of the KCNQ1OT1:TSS-DMR caused by the paternal heterozygous de novo deletions involving the KCNQ1OT1:TSS-DMR, but not including CDKN1C enhancers. The deletion sizes were 5 kb and 12 kb for patients 1 and 2, respectively. CDKN1C gene expressions in immortalised leucocytes of both patients were increased compared with those of controls. CONCLUSION: Paternal deletions involving the KCNQ1OT1:TSS-DMR, but not including CDKN1C enhancers, disrupt KCNQ1OT1 expression, strongly activate CDKN1C expression and consequently cause severe growth failure.


Asunto(s)
ARN Largo no Codificante , Síndrome de Silver-Russell , Embarazo , Femenino , Humanos , Preescolar , Impresión Genómica/genética , Herencia Paterna , Síndrome de Silver-Russell/genética , Metilación de ADN/genética , Fenotipo , Insuficiencia de Crecimiento/genética , ARN Largo no Codificante/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética
6.
Hum Mutat ; 43(12): 1852-1855, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36054331

RESUMEN

RASopathies are disorders caused by germline mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. These syndromes share features of developmental delay, facial dysmorphisms, and defects in various organs, as well as cancer predisposition. Somatic mutations of the same pathway are one of the primary causes of cancer. It is thought that germline cancer-causing mutations would be embryonic lethal, as a more severe phenotype was shown in Drosophila and zebrafish embryos with cancer MAP2K1 mutations than in those with RASopathy mutations. Here we report the case of a patient with RASopathy caused by a cancer-associated MAP2K1 p.Phe53Leu mutation. The postzygotic mosaic nature of this mutation could explain the patient's survival.


Asunto(s)
Displasia Ectodérmica , Cardiopatías Congénitas , Síndromes de Inmunodeficiencia , Neoplasias , Animales , Humanos , Pez Cebra/genética , Insuficiencia de Crecimiento/genética , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Facies , Cardiopatías Congénitas/genética , Mutación , MAP Quinasa Quinasa 1/genética
7.
Am J Med Genet C Semin Med Genet ; 190(4): 414-424, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36428239

RESUMEN

RASopathies comprise a group of clinically overlapping developmental disorders caused by genetic variations affecting components or modulators of the RAS-MAPK signaling cascade, which lead to dysregulation of signal flow through this pathway. Noonan syndrome and the less frequent, clinically related disorders, Costello syndrome, cardiofaciocutaneous syndrome, Noonan syndrome with multiple lentigines, and Noonan syndrome-like disorder with loose anagen hair are part of the RASopathy spectrum and share a recognizable pattern of multisystem involvement. This review describes the "Noonan syndrome-like" phenotype as a common phenotypic signature of generalized developmental RAS pathway dysregulation. Distinctive features of the different entities are revisited against the background of the understanding of underlying genetic alterations and genotype correlations, which has evolved rapidly during the past 20 years, thereby leading to suggestions regarding the nosology of RASopathies.


Asunto(s)
Síndrome de Costello , Cardiopatías Congénitas , Síndrome de Noonan , Humanos , Síndrome de Noonan/genética , Cardiopatías Congénitas/genética , Síndrome de Costello/genética , Insuficiencia de Crecimiento/genética , Mutación
8.
Am J Med Genet C Semin Med Genet ; 190(4): 440-451, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36408797

RESUMEN

The cardiovascular phenotype associated with RASopathies has expanded far beyond the original descriptions of pulmonary valve stenosis by Dr Jaqueline Noonan in 1968 and hypertrophic cardiomyopathy by Hirsch et al. in 1975. Because of the common underlying RAS/MAPK pathway dysregulation, RASopathy syndromes usually present with a typical spectrum of overlapping cardiovascular anomalies, although less common cardiac defects can occur. The identification of the causative genetic variants has enabled the recognition of specific correlations between genotype and cardiac phenotype. Characterization and understanding of genotype-phenotype associations is not only important for counseling a family of an infant with a new diagnosis of a RASopathy condition but is also critical for their clinical prognosis with respect to cardiac disease, neurodevelopment and other organ system involvement over the lifetime of the patient. This review will focus on the cardiac manifestations of the most common RASopathy syndromes, the relationship between cardiac defects and causal genetic variation, the contribution of cardiovascular abnormalities to morbidity and mortality and the most relevant follow-up issues for patients affected by RAS/MAPK pathway diseases, with respect to cardiac clinical outcomes and management, in children and in the adult population.


Asunto(s)
Displasia Ectodérmica , Cardiopatías Congénitas , Síndrome de Noonan , Humanos , Síndrome de Noonan/genética , Síndrome de Noonan/diagnóstico , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/diagnóstico , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/diagnóstico , Proteínas ras/genética , Displasia Ectodérmica/genética , Mutación
9.
Am J Hum Genet ; 104(1): 179-185, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30595371

RESUMEN

Accumulation of unfolded proteins in the endoplasmic reticulum (ER) initiates a stress response mechanism to clear out the unfolded proteins by either facilitating their re-folding or inducing their degradation. When this fails, an apoptotic cascade is initiated so that the affected cell is eliminated. IRE1α is a critical sensor of the unfolded-protein response, essential for initiating the apoptotic signaling. Here, we report an infantile neurodegenerative disorder associated with enhanced activation of IRE1α and increased apoptosis. Three unrelated affected individuals with congenital microcephaly, infantile epileptic encephalopathy, and profound developmental delay were found to carry heterozygous variants (c.932T>C [p.Leu311Ser] or c.935T>C [p.Leu312Pro]) in RNF13, which codes for an IRE1α-interacting protein. Structural modeling predicted that the variants, located on the surface of the protein, would not alter overall protein folding. Accordingly, the abundance of RNF13 and IRE1α was not altered in affected individuals' cells. However, both IRE1α-mediated stress signaling and stress-induced apoptosis were increased in affected individuals' cells. These results indicate that the RNF13 variants confer gain of function to the encoded protein and thereby lead to altered signaling of the ER stress response associated with severe neurodegeneration in infancy.


Asunto(s)
Ceguera/congénito , Ceguera/genética , Insuficiencia de Crecimiento/genética , Mutación con Ganancia de Función , Heterocigoto , Microcefalia/genética , Espasmos Infantiles/genética , Ubiquitina-Proteína Ligasas/genética , Secuencia de Aminoácidos , Apoptosis , Niño , Preescolar , Discapacidades del Desarrollo/genética , Estrés del Retículo Endoplásmico , Humanos , Lactante , Masculino , Modelos Moleculares , Espasmos Infantiles/congénito , Ubiquitina-Proteína Ligasas/química , Respuesta de Proteína Desplegada
10.
Clin Genet ; 101(4): 403-410, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34988996

RESUMEN

Here, we delineate the phenotype of two siblings with a bi-allelic frameshift variant in MMP15 gene with congenital cardiac defects, cholestasis, and dysmorphism. Genome sequencing analysis revealed a recently reported homozygous frameshift variant (c.1058delC, p.Pro353Glnfs*102) in MMP15 gene that co-segregates with the phenotype in the family in a recessive mode of inheritance. Relative quantification of MMP15 mRNA showed evidence of degradation of the mutated transcript, presumably by nonsense mediated decay. Likewise, MMP15: p.Gly231Arg, a concurrently reported homozygous missense variant in another patient exhibiting a similar phenotype, was predicted to disrupt zinc ion binding to the MMP-15 enzyme catalytic domain, which is essential for substrate proteolysis, by structural modeling. Previous animal models and cellular findings suggested that MMP15 plays a crucial role in the formation of endocardial cushions. These findings confirm that MMP15 is an important gene in human development, particularly cardiac, and that its loss of function is likely to cause a severe disorder phenotype.


Asunto(s)
Colestasis , Cardiopatías Congénitas , Ictericia , Metaloproteinasa 15 de la Matriz/genética , Animales , Insuficiencia de Crecimiento/genética , Cardiopatías Congénitas/genética , Homocigoto , Humanos , Fenotipo
11.
Clin Genet ; 102(1): 56-60, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35246835

RESUMEN

Genetic defect in the nuclear encoded subunits of cytochrome c oxidase are very rare. To date, most deleterious variants affect the mitochondrially encoded subunits of complex IV and the nuclear genes encoded for assembly factors. A biallelic pathogenic variant in the mitochondrial complex IV subunit COX5A was previously reported in a couple of sibs with failure to thrive, lactic acidosis and pulmonary hypertension and a lethal phenotype. Here, we describe a second family with a 11-year-old girl presenting with failure to thrive, lactic acidosis, hypoglycemia and short stature. Clinical exome revealed the homozygous missense variant c.266 T > G in COX5A, which produces a drop of the corresponding protein and a reduction of the COX activity. Compared to the previous observation, this girl showed an attenuated metabolic derangement without involvement of the cardiovascular system and neurodevelopment. Our observation confirms that COX5A recessive variants may cause mitochondrial disease and expands the associated phenotype to less severe presentations.


Asunto(s)
Acidosis Láctica , Enanismo , Hipoglucemia , Acidosis Láctica/genética , Acidosis Láctica/patología , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Insuficiencia de Crecimiento/genética , Homocigoto , Humanos , Hipoglucemia/genética , Fenotipo
12.
Am J Med Genet A ; 188(2): 414-421, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34854525

RESUMEN

Cardio-facio-cutaneous syndrome (CFCS) is a rare disorder characterized by distinctive craniofacial appearance, cardiac, neurologic, cutaneous, and musculoskeletal abnormalities. It is due to heterozygous mutations in BRAF, MAP2K1, MAP2K2, and KRAS genes, belonging to the RAS/MAPK pathway. The role of RAS signaling in bone homeostasis is highly recognized, but data on bone mineral density (BMD) in CFCS are lacking. In the present study we evaluated bone parameters, serum and urinary bone metabolites in 14 individuals with a molecularly confirmed diagnosis of CFCS. Bone assessment was performed through dual X-ray absorptiometry (DXA); height-adjusted results were compared to age- and sex-matched controls. Blood and urinary bone metabolites were also analyzed and compared to the reference range. Despite vitamin D supplementation and almost normal bone metabolism biomarkers, CFCS patients showed significantly decreased absolute values of DXA-assessed subtotal and lumbar BMD (p ≤ 0.05), compared to controls. BMD z-scores and t-scores (respectively collected for children and adults) were below the reference range in CFCS, while normal in healthy controls. These findings confirmed a reduction in BMD in CFCS and highlighted the importance of monitoring bone health in these affected individuals.


Asunto(s)
Displasia Ectodérmica , Insuficiencia de Crecimiento , Absorciometría de Fotón , Adulto , Densidad Ósea/genética , Niño , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/genética , Cardiopatías Congénitas , Homeostasis , Humanos
13.
Am J Med Genet A ; 188(10): 3106-3109, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35938194

RESUMEN

EEF1A2 is a gene whose protein product, eukaryotic translation elongation factor 1 alpha 2 (eEF1A2), plays an important role in neurodevelopment. Reports of individuals with pathogenic variants in EEF1A2 are rare, with less than 40 individuals reported world-wide, however a common feature is the association of the variant with developmental and epileptic encephalopathy. Thus far, there have been limited reports of other organ systems or body functions affected by variants in this gene. Here, we present a case of a child with EEF1A2-related disorder who presented at 3 months of age with hypotonia, microcephaly, failure to thrive, and respiratory insufficiency with central apneas requiring respiratory support. Our case highlights the notion that the respiratory system may be highly implicated in EEF1A2-related disorder, allowing for better phenotypic characterization of the disorder.


Asunto(s)
Insuficiencia de Crecimiento , Factor 1 de Elongación Peptídica , Apnea , Niño , Insuficiencia de Crecimiento/diagnóstico , Insuficiencia de Crecimiento/genética , Humanos , Lactante , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Sistema Respiratorio/metabolismo
14.
Am J Med Genet A ; 188(9): 2732-2737, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35801299

RESUMEN

Cardio-facio-cutaneous (CFC) syndrome (OMIM #:115150, 615278, 615279, 615280) is a rare genetic condition caused by variants in the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Up to 75% of cases are caused by mutations in the BRAF gene, whereas KRAS gene mutation has only been reported in <2% of cases. CFC syndrome is characterized by cardiac abnormalities, distinctive craniofacial dysmorphism, and various cutaneous abnormalities. Musculoskeletal and orthopedic manifestations are also prevalent in patients with CFC syndrome, among which the most common are skeletal deformities and joint laxities. Dysplastic bone disorders, on the other hand, have not been reported in CFC syndrome before. We report on a case of symmetrical polyostotic fibrous dysplasia (FD) in a patient with CFC syndrome with the KRAS(NM_004985.5):c.57G>C; p.Leu19Phe variant. The FDs were incidentally picked up, and patient was conservatively managed and remained asymptomatic on follow-up. The same variant was reported previously in a patient with Oculoectodermal Syndrome (OES), who developed polyostotic non-ossifying fibroma (NOF). This case explores FD as a possible new clinical feature of CFC syndrome, and when linked to the historical case of OES, explores whether the KRAS(NM_004985.5):c.57G>C; p.Leu19Phe mutation may potentially contribute to the development of dysplastic bone lesions in patients with this particular mutation.


Asunto(s)
Displasia Ectodérmica , Cardiopatías Congénitas , Quiste Dermoide , Displasia Ectodérmica/complicaciones , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/patología , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Humanos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)
15.
Am J Med Genet A ; 188(12): 3505-3509, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35943247

RESUMEN

Cardio-facio-cutaneous syndrome (CFC) (OMIM 115150) is a congenital disease caused by constitutive activation of the Raf/MEK/ERK signaling cascade. Unlike aspects of morphological anomalies, metabolic functions related to the disease have garnered little attention. We present severe neuroglycopenic symptoms due to nonketotic hypoglycemia in two children with CFC (Case 1, a 4-year-old male with c.389A > G heterozygous variant in MAP2K1; Case 2, a 3-year-old male with c.770A > G heterozygous variant in BRAF). Case 1 exhibited a nonketotic hypoglycemic coma and clustered left-hemispheric convulsions despite receiving infusion therapy, leading to severe sequelae with choreoathetosis. Brain magnetic resonance imaging of Case 1 showed T2-elongation with restricted diffusion on the bilateral basal ganglia and thalamus, with the dominance of the right putamen. Case 2 presented a prolonged generalized seizure as an initial clinical symptom but fully recovered. The presence of growth hormone and cortisol deficiency was ruled out in both cases. Blood spots acylcarnitine profiles excluded the co-occurrence of mitochondrial HMG-CoA synthase deficiency and HMG-CoA lyase deficiency. These cases demonstrate the potential vulnerability to nonketotic hypoglycemia, especially during lipid shortages. As children with CFC frequently have difficulties feeding, we suggest great attention should be paid to the potential risk of severe nonketotic hypoglycemia.


Asunto(s)
Displasia Ectodérmica , Cardiopatías Congénitas , Hipoglucemia , Enfermedades del Sistema Nervioso , Niño , Masculino , Humanos , Preescolar , Proteínas Proto-Oncogénicas B-raf , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/patología , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Displasia Ectodérmica/patología , Facies , Cardiopatías Congénitas/diagnóstico , Hipoglucemia/complicaciones , Hipoglucemia/genética
16.
Neuroradiology ; 64(1): 185-195, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34309696

RESUMEN

PURPOSE: To evaluate the brain volumetric changes caused by BRAF gene mutation in non-epileptic CFC patients and the influence of the age of epilepsy onset on brain development in 2 cohorts of epileptic CFC patients. METHODS: We enrolled CFC patients carrying BRAF gene mutations without epilepsy (4 patients) and with epilepsy (16 patients). CFC epileptic patients were divided into two cohorts based on the age of seizure onset: early-age onset (7 children) and late-age onset (9 adolescents). All three cohorts of patients underwent 3D FSPGR T1-weighted imaging to assess supratentorial and infratentorial brain volumes. Moreover, for each compartment, gray matter (GM), white matter (WM), and cerebrospinal fluid (CSF) volumes were measured. All measurements were compared with those of age-matched controls without neuroimaging abnormalities. RESULTS: All CFC patients showed supratentorial and infratentorial WM reduction and supratentorial ventricular enlargement (p < 0.01). However, patients with early age of epilepsy onset, compared with the other two cohorts of CFC patients, showed both GM and a more pronounced WM volume reduction (p < 0.01). CONCLUSION: In non-epileptic CFC children, we demonstrated WM volumetric reduction suggesting a direct effect of BRAF gene mutation on brain development. Nevertheless, in CFC epileptic patients, the age of epilepsy onset may contribute to brain atrophy. Brain atrophy in CFC patients, in part due to the natural history of the disease, may be worsened by epilepsy when it begins in the early ages because of interference with brain growth at that critical age of development.


Asunto(s)
Encéfalo/patología , Epilepsia , Proteínas Proto-Oncogénicas B-raf , Adolescente , Encéfalo/diagnóstico por imagen , Estudios de Casos y Controles , Niño , Displasia Ectodérmica/genética , Epilepsia/diagnóstico por imagen , Epilepsia/genética , Facies , Insuficiencia de Crecimiento/genética , Cardiopatías Congénitas/genética , Humanos , Imagen por Resonancia Magnética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética
17.
J Med Genet ; 58(3): 213-216, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32332102

RESUMEN

Newly synthesised glycoproteins enter the rough endoplasmic reticulum through a translocation pore. The translocon associated protein (TRAP) complex is located close to the pore. In a patient with a homozygous start codon variant in TRAPγ (SSR3), absence of TRAPγ causes disruption of the TRAP complex, impairs protein translocation into the endoplasmic reticulum and affects transport, for example, into the brush-border membrane. Furthermore, we observed an unbalanced non-occupancy of N-glycosylation sites. The major clinical features are intrauterine growth retardation, facial dysmorphism, congenital diarrhoea, failure to thrive, pulmonary disease and severe psychomotor disability.


Asunto(s)
Retículo Endoplásmico Rugoso/genética , Retardo del Crecimiento Fetal/genética , Glicoproteínas/genética , Fosfatasa Ácida Tartratorresistente/genética , Niño , Preescolar , Diarrea/genética , Diarrea/patología , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/patología , Femenino , Retardo del Crecimiento Fetal/patología , Glicoproteínas/biosíntesis , Glicosilación , Humanos , Lactante , Recién Nacido , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Masculino , Trastornos Psicomotores/genética , Trastornos Psicomotores/patología , Fosfatasa Ácida Tartratorresistente/deficiencia
18.
Dev Dyn ; 250(8): 1074-1095, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33522658

RESUMEN

BACKGROUND: Cardio-facio-cutaneous (CFC) syndrome is a human multiple congenital anomaly syndrome that is caused by activating heterozygous mutations in either BRAF, MEK1, or MEK2, three protein kinases of the Ras/mitogen-activated protein kinase (MAPK) pathway. CFC belongs to a group of syndromes known as RASopathies. Skeletal muscle hypotonia is a ubiquitous phenotype of RASopathies, especially in CFC syndrome. To better understand the underlying mechanisms for the skeletal myopathy in CFC, a mouse model with an activating BrafL597V allele was utilized. RESULTS: The activating BrafL597V allele resulted in phenotypic alterations in skeletal muscle characterized by a reduction in fiber size which leads to a reduction in muscle size which are functionally weaker. MAPK pathway activation caused inhibition of myofiber differentiation during embryonic myogenesis and global transcriptional dysregulation of developmental pathways. Inhibition in differentiation can be rescued by MEK inhibition. CONCLUSIONS: A skeletal myopathy was identified in the CFC BrafL597V mouse validating the use of models to study the effect of Ras/MAPK dysregulation on skeletal myogenesis. RASopathies present a novel opportunity to identify new paradigms of myogenesis and further our understanding of Ras in development. Rescue of the phenotype by inhibitors may help advance the development of therapeutic options for RASopathy patients.


Asunto(s)
Displasia Ectodérmica/genética , Insuficiencia de Crecimiento/genética , Cardiopatías Congénitas/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Proteínas Proto-Oncogénicas B-raf/genética , Alelos , Animales , Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patología , Facies , Insuficiencia de Crecimiento/metabolismo , Insuficiencia de Crecimiento/patología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Fenotipo , Proteínas Proto-Oncogénicas B-raf/metabolismo
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(10): 1129-1134, 2022 Oct 10.
Artículo en Zh | MEDLINE | ID: mdl-36184098

RESUMEN

OBJECTIVE: To explore the genotype-phenotype correlation of a patient with cardio-facio-cutaneous syndrome (CFCS) due to variant of the MAP2K1 gene. METHODS: DNA was extracted from peripheral blood samples of the infant and his parents and subjected to whole exome sequencing. Candidate variant was verified by Sanger sequencing. RESULTS: The patient had typical CFCS facies and developmental delay, and was found to harbor a de novo heterozygous c.389A>G (p.Tyr130Cys) missense variant in exon 3 of the MAP2K1 gene. Based on the American college of Medical Genetics and Genomics guidelines, this variant was classified as likely pathogenic. CONCLUSION: This patient has differed from previously reported cases by having no cardiac anomaly or seizures but typical facial features and skin abnormalities accompanied by growth retardation, intellectual impairment, and urinary malformation. It has therefore enriched the phenotypic spectrum of CFCS due to variants of the MAP2K1 gene.


Asunto(s)
Displasia Ectodérmica , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/genética , Cardiopatías Congénitas , Humanos , MAP Quinasa Quinasa 1/genética , Mutación
20.
J Hum Genet ; 66(4): 389-399, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33040082

RESUMEN

Noonan syndrome (NS) and cardio-facio-cutaneous (CFC) syndrome are the most common subtypes of RASopathy. As an effector of Ras, BRAF is one of the molecules responsible for RASopathy. We investigated the phenotypic and genotypic features of 26 patients with BRAF-associated RASopathy. The clinical diagnoses were CFC (n = 21, 80.8%), NS (n = 3, 11.5%), NS/CFC (n = 1, 3.8%), and undefined syndromic intellectual disability (ID) (n = 1, 3.8%). The mostly shared phenotypes were ID (90.5%), cutaneous manifestations (84.6%), congenital heart defects (76.9%), short stature (76.9%), and dysmorphic features such as short neck (65.4%) and low-set ears (65.4%). Importantly, moderate to severe ID (57.1%) and epilepsy (26.9%) were noted. Eighteen different missense mutations were found, including a novel mutation, p.Phe498Tyr. p.Gln257Arg (n = 9, 34.6%) was the most common mutation, and the mutations were clustered in the cysteine-rich domain or protein kinase domain. A review of previously reported cases along with our findings revealed the existence of multiple sub-phenotypes of RASopathy within a single genotype, indicating that BRAF-associated RASopathy is not variant-specific. Our study further delineated the diverse and expanded clinical phenotypes of BRAF-associated RASopathy with their molecular genetic characteristics.


Asunto(s)
Displasia Ectodérmica/patología , Insuficiencia de Crecimiento/patología , Cardiopatías Congénitas/patología , Mutación , Síndrome de Noonan/patología , Proteínas Proto-Oncogénicas B-raf/genética , Adolescente , Niño , Preescolar , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/genética , Femenino , Cardiopatías Congénitas/genética , Humanos , Lactante , Masculino , Síndrome de Noonan/genética , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA