Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
Mol Genet Genomic Med ; 12(1): e2313, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37942564

ABSTRACT

BACKGROUND: Noonan syndrome (NS) due to the RRAS2 gene, the pathogenic variant is an extremely rare RASopathies. Our objective was to identify the potential site of RRAS2, combined with the literature review, to find the correlation between clinical phenotype and genotype. De novo missense mutations affect different aspects of the RRAS2 function, leading to hyperactivation of the RAS-MAPK signaling cascade. METHODS: Conventional G-banding was used to analyze the chromosome karyotype of the patient. Copy number variation sequencing (CNV-seq) was used to detect the chromosomal gene microstructure of the patient and her parents. The exomes of the patient and her parents were sequenced using trio-based whole exome sequencing (trio-WES) technology. The candidate variant was verified by Sanger sequencing. The pathogenicity of the variant was predicted with a variety of bioinformatics tools. RESULTS: Chromosome analysis of the proband revealed 46, XX, and no abnormality was found by CNV-seq. After sequencing and bioinformatics filtering, the variant of RRAS2(c.67G>T; p. Gly23Cys) was found in the proband, while the mutation was absent in her parents. To the best of our knowledge, our patient was with the typical Noonan syndrome, such as short stature, facial dysmorphism, and developmental delay. Furthermore, our study is the first case of NS with embryonal rhabdomyosarcoma (ERMS) caused by the RRAS2 gene mutation reported in China. CONCLUSIONS: Our investigations suggested that the heterozygous missense of RRAS2 may be a potential causal variant in a rare cause of Noonan syndrome, expanding our understanding of the causally relevant mutations for this disorder.


Subject(s)
Monomeric GTP-Binding Proteins , Noonan Syndrome , Rhabdomyosarcoma, Embryonal , Humans , Female , Noonan Syndrome/pathology , Rhabdomyosarcoma, Embryonal/genetics , Rhabdomyosarcoma, Embryonal/complications , DNA Copy Number Variations , Mutation , Genotype , Membrane Proteins/genetics , Monomeric GTP-Binding Proteins/genetics
2.
J Neurol ; 271(3): 1331-1341, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37923938

ABSTRACT

The RASopathies are a group of genetic rare diseases caused by mutations affecting genes involved in the RAS/MAPK (RAS-mitogen activated protein kinase) pathway. Among them, PTPN11 pathogenic variants are responsible for approximately 50% of Noonan syndrome (NS) cases and, albeit to a lesser extent, of Leopard syndrome (LPRD1), which present a few overlapping clinical features, such as facial dysmorphism, developmental delay, cardiac defects, and skeletal deformities. Motor impairment and decreased muscle strength have been recently reported. The etiology of the muscle involvement in these disorders is still not clear but probably multifactorial, considering the role of the RAS/MAPK pathway in skeletal muscle development and Acetylcholine Receptors (AChR) clustering at the neuromuscular junction (NMJ). We report, herein, four unrelated children carrying three different heterozygous mutations in the PTPN11 gene. Intriguingly, their phenotypic features first led to a clinical suspicion of congenital myasthenic syndrome (CMS), due to exercise-induced fatigability with a variable degree of muscle weakness, and serum proteomic profiling compatible with a NMJ defect. Moreover, muscle fatigue improved after treatment with CMS-specific medication. Although the link between PTPN11 gene and neuromuscular transmission is unconfirmed, an increasing number of patients with RASopathies are affected by muscle weakness and fatigability. Hence, NS or LPDR1 should be considered in children with suspected CMS but negative genetic workup for known CMS genes or additional symptoms indicative of NS, such as facial dysmorphism or intellectual disability.


Subject(s)
Myasthenic Syndromes, Congenital , Noonan Syndrome , Child , Humans , Noonan Syndrome/genetics , Noonan Syndrome/diagnosis , Noonan Syndrome/pathology , Myasthenic Syndromes, Congenital/genetics , Proteomics , Mutation/genetics , Phenotype , Muscle Weakness , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
3.
J Appl Genet ; 65(2): 303-308, 2024 May.
Article in English | MEDLINE | ID: mdl-37987971

ABSTRACT

Noonan syndrome (NS; OMIM 163950) is an autosomal dominant RASopathy with variable clinical expression and genetic heterogeneity. Clinical manifestations include characteristic facial features, short stature, and cardiac anomalies. Variants in protein-tyrosine phosphatase, non-receptor-type 11 (PTPN11), encoding SHP-2, account for about half of NS patients, SOS1 in approximately 13%, RAF1 in 10%, and RIT1 each in 9%. Other genes have been reported to cause NS in less than 5% of cases including SHOC2, RASA2, LZTR1, SPRED2, SOS2, CBL, KRAS, NRAS, MRAS, PRAS, BRAF, PPP1CB, A2ML1, MAP2K1, and CDC42. Several additional genes associated with a Noonan syndrome-like phenotype have been identified. Clinical presentation and variants in patients with Noonan syndrome are this study's objectives. We performed Sanger sequencing of PTPN11 hotspot (exons 3, 8, and 13). We report molecular analysis of 61 patients with NS phenotype belonging to 58 families. We screened for hotspot variants (exons 3, 8, and 13) in PTPN11 gene by Sanger sequencing. Twenty-seven patients were carrying heterozygous pathogenic variants of PTPN11 gene with a similar frequency (41.4%) compared to the literature. Our findings expand the variant spectrum of Moroccan patients with NS phenotype in whom the analysis of hotspot variants showed a high frequency of exons 3 and 8. This screening test allowed us to establish a molecular diagnosis in almost half of the patients with a good benefit-cost ratio, with appropriate management and genetic counseling.


Subject(s)
Noonan Syndrome , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , alpha-Macroglobulins , Humans , Exons , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Noonan Syndrome/genetics , Noonan Syndrome/diagnosis , Noonan Syndrome/pathology , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , ras GTPase-Activating Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
4.
Sci Adv ; 9(28): eadf4766, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37450595

ABSTRACT

RIT1 is a RAS guanosine triphosphatase (GTPase) that regulates different aspects of signal transduction and is mutated in lung cancer, leukemia, and in the germline of individuals with Noonan syndrome. Pathogenic RIT1 proteins promote mitogen-activated protein kinase (MAPK) hyperactivation; however, this mechanism remains poorly understood. Here, we show that RAF kinases are direct effectors of membrane-bound mutant RIT1 necessary for MAPK activation. We identify critical residues in RIT1 that facilitate interaction with membrane lipids and show that these are necessary for association with RAF kinases and MAPK activation. Although mutant RIT1 binds to RAF kinases directly, it fails to activate MAPK signaling in the absence of classical RAS proteins. Consistent with aberrant RAF/MAPK activation as a driver of disease, we show that pathway inhibition alleviates cardiac hypertrophy in a mouse model of RIT1 mutant Noonan syndrome. These data shed light on the function of pathogenic RIT1 and identify avenues for therapeutic intervention.


Subject(s)
Lung Neoplasms , Noonan Syndrome , Animals , Mice , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Noonan Syndrome/pathology , Mitogen-Activated Protein Kinases/metabolism , Cardiomegaly/genetics , Signal Transduction
5.
Am J Med Genet A ; 191(8): 2074-2082, 2023 08.
Article in English | MEDLINE | ID: mdl-37194190

ABSTRACT

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.


Subject(s)
Costello Syndrome , Noonan Syndrome , Humans , Costello Syndrome/genetics , Costello Syndrome/pathology , Phenotype , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Failure to Thrive/genetics , Failure to Thrive/pathology , Germ-Line Mutation , Proto-Oncogene Proteins p21(ras)/genetics
6.
Eur J Med Genet ; 66(2): 104695, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36608738

ABSTRACT

Noonan syndrome is characterized by variable phenotypic expressivity with characteristic dysmorphic facial features, varying degrees of intellectual disability, developmental delay, short stature, and congenital heart defects in 50-80%. Other findings include a webbed neck, cryptorchidism, coagulation defects and eye abnormalities. Thus far, Noonan syndrome has mainly been attributed to heterozygous pathogenic variants in 10+ different genes, with the rare exception of cases due to biallelic pathogenic variants in LZTR1. Recently, homozygous loss-of-function variants in SPRED2 have been identified as a cause of a recessive Noonan syndrome-like phenotype. We present the phenotypes of two additional patients with homozygosity for a previously unreported loss-of-function variant in SPRED2, thereby adding relevant clinical information about the recently described Noonan syndrome-like SPRED2-related phenotype.


Subject(s)
Heart Defects, Congenital , Intellectual Disability , Noonan Syndrome , Humans , Male , Heterozygote , Homozygote , Intellectual Disability/genetics , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Phenotype , Repressor Proteins/genetics , Transcription Factors/genetics
7.
J Neuroimaging ; 33(2): 318-327, 2023 03.
Article in English | MEDLINE | ID: mdl-36480458

ABSTRACT

BACKGROUND AND PURPOSE: There are a few studies regarding intracranial findings in neonates with Noonan syndrome (NS); however, there are no quantitative analyses in a pediatric population. The aim of this study was to find characteristic intracranial abnormalities and to quantitatively analyze the posterior fossa and cranium base in children with NS. METHODS: A total of 30 patients (11 females and 19 males, mean age 13.1 ± 4.3 years) were retrospectively identified between July 2017 and June 2022. Twenty-one patients had MRI. Age at MRI examination, sex, genetic mutations, and clinical findings were noted. In patients with MRI, the presence of white matter lesions, basal ganglia lesions, corpus callosum abnormalities, sellar/parasellar lesions, and tonsillar ectopia was noted. For morphometric analysis, cerebellar diameter, vermis and clivus heights, cranial base, tentorial and infratentorial angles, and McRae's and Twining's lines were each measured twice by two radiologists individually. RESULTS: The most common lesions were focal white matter lesions, followed by abnormalities of the splenium of the corpus callosum. The cerebellar diameter, vermis and clivus heights, Twining's line, and infratentorial angle were significantly smaller; cranial base angle and tentorial angle were significantly larger in NS (p < .05). Interrater and intrarater agreements were the highest for cerebellar diameter and the lowest for tentorial angle measurements. CONCLUSION: Children with NS had characteristic callosal and tentorial findings and neuroimaging findings similar to other RASopathies. This study also shows that a small posterior fossa and flattening of the cranial base are present in children with NS, which may aid in diagnosis.


Subject(s)
Noonan Syndrome , Male , Infant, Newborn , Female , Humans , Child , Adolescent , Retrospective Studies , Noonan Syndrome/pathology , Skull Base , Neuroimaging , Cerebellum/pathology , Cranial Fossa, Posterior/pathology , Magnetic Resonance Imaging/methods
8.
Am J Med Genet C Semin Med Genet ; 190(4): 530-540, 2022 12.
Article in English | MEDLINE | ID: mdl-36533693

ABSTRACT

RASopathies are a set of clinical syndromes that have molecular and clinical overlap. Genetically, these syndromes are defined by germline pathogenic variants in RAS/MAPK pathway genes resulting in activation of this pathway. Clinically, their common molecular signature leads to comparable phenotypes, including cardiac anomalies, neurologic disorders and notably, elevated cancer risk. Cancer risk in individuals with RASopathies has been estimated from retrospective reviews and cohort studies. For example, in Costello syndrome, cancer incidence is significantly elevated over the general population, largely due to solid tumors. In some forms of Noonan syndrome, cancer risk is also elevated over the general population and is enriched for hematologic malignancies. Thus, cancer surveillance guidelines have been developed to monitor for the occurrence of such cancers in individuals with some RASopathies. These include abdominal ultrasound and urinalyses for individuals with Costello syndrome, while complete blood counts and splenic examination are recommended in Noonan syndrome. Improved cancer risk estimates and refinement of surveillance recommendations will improve the care of individuals with RASopathies.


Subject(s)
Costello Syndrome , Neoplasms , Noonan Syndrome , Humans , Noonan Syndrome/epidemiology , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Costello Syndrome/epidemiology , Costello Syndrome/genetics , Incidence , Retrospective Studies , ras Proteins/genetics , Neoplasms/epidemiology , Neoplasms/genetics
9.
Cells ; 11(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36231062

ABSTRACT

Noonan syndrome (NS) and related Noonan syndrome with multiple lentigines (NSML) contribute to the pathogenesis of human diseases in the RASopathy family. This family of genetic disorders constitute one of the largest groups of developmental disorders with variable penetrance and severity, associated with distinctive congenital disabilities, including facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was first clinically described decades ago, and several genes have since been identified, providing a molecular foundation to understand their physiopathology and identify targets for therapeutic strategies. These genes encode proteins that participate in, or regulate, RAS/MAPK signalling. The RAS pathway regulates cellular metabolism by controlling mitochondrial homeostasis, dynamics, and energy production; however, little is known about the role of mitochondrial metabolism in NS and NSML. This manuscript comprehensively reviews the most frequently mutated genes responsible for NS and NSML, covering their role in the current knowledge of cellular signalling pathways, and focuses on the pathophysiological outcomes on mitochondria and energy metabolism.


Subject(s)
LEOPARD Syndrome , Noonan Syndrome , Energy Metabolism/genetics , Germ-Line Mutation , Humans , LEOPARD Syndrome/genetics , Mitochondria/genetics , Mitochondria/pathology , Noonan Syndrome/genetics , Noonan Syndrome/pathology , ras Proteins/genetics
10.
Cornea ; 41(11): 1462-1464, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35867656

ABSTRACT

PURPOSE: Anterior segment abnormalities associated with Noonan syndrome are rare. We report our experience with 2 patients who developed keratopathy with significant visual sequelae. METHODS: case series. RESULTS: The first patient is a 9-year-old boy with genetically confirmed Noonan syndrome. At presentation, he was noted to have diffuse inferior epitheliopathy with vascularization and bilateral mild ptosis. Over 1 year, he developed focal areas of scarring with deterioration of vision and underwent superficial keratectomy in the left eye. However, over the following 2 years, he experienced recurrent corneal scarring and vascularization. The second patient is a 7-year-old boy with phenotypic Noonan syndrome. At presentation, he had an anterior subepithelial corneal scar inferiorly with epithelial defects in both eyes. He also had bilateral moderate ptosis and lagophthalmos. Despite lubrication, he developed recurrent bilateral corneal erosions with focal areas of scarring associated with vascularization and underwent superficial keratectomy for both eyes. Despite this, there was worsening corneal scarring and vascularization over time, eventually requiring deep anterior lamellar keratoplasty in the left eye. The host cornea showed a disturbed Bowman layer and an acellular mass of fibrous collagenous tissue between epithelium and stroma. CONCLUSIONS: Noonan syndrome may be associated with visually significant keratopathy, manifesting as focal corneal scarring with vascularization. These changes may due to an excessive fibrotic response in Noonan syndrome. Early recognition and treatment can help to delay the progression of keratopathy and need for surgical intervention.


Subject(s)
Corneal Diseases , Corneal Dystrophies, Hereditary , Corneal Edema , Noonan Syndrome , Child , Cicatrix/pathology , Cornea/pathology , Corneal Diseases/diagnosis , Corneal Diseases/etiology , Corneal Diseases/surgery , Corneal Dystrophies, Hereditary/surgery , Corneal Edema/pathology , Humans , Male , Noonan Syndrome/complications , Noonan Syndrome/diagnosis , Noonan Syndrome/pathology , Visual Acuity
11.
Am J Med Genet A ; 188(7): 2246-2250, 2022 07.
Article in English | MEDLINE | ID: mdl-35338599

ABSTRACT

Noonan syndrome-like disorder with loose anagen hair (NSLH) is a rare disease characterized by typical features of Noonan syndrome with additional findings of relative or absolute macrocephaly, loose anagen hair, and a higher incidence of intellectual disability. NSLH1 is caused by a heterozygous mutation in the SHOC2 gene on chromosome 10q25, and NLSH2 is caused by a heterozygous mutation in the Protein phosphatase one catalytic subunit beta (PPP1CB) gene on chromosome 2p23. Protein phosphatase1 (PP1), encoded by PPP1CB, forms a complex with SHOC2 and dephosphorylates RAFs, which results in activation of the signaling cascade and contribution to Noonan syndrome pathogenesis. Here, we report two genetically confirmed Japanese patients with NSLH2 having the same de novo mutation in PPP1CB presenting prominent-hyperteloric-appearing eyes and a tall forehead similar to individuals carrying a mutation in PPP1CB, c.146C > G; p.Pro49Arg, which is different from typical facial features of Noonan syndrome. They also showed short stature, absolute macrocephaly, and loose anagen hair like NSLH1: however, growth hormone deficiency often seen in NSLH1 caused by SHOC2 mutation was absent. Although a number of Noonan syndrome and NSLH1 patients have shown blunted or no response to GH therapy, linear growth was promoted by recombinant human growth hormone (rhGH) in one of our patients. Since another NSLH2 patient with good response to rhGH treatment was reported, rhGH therapy may be effective in patients with NSLH2.


Subject(s)
Abnormalities, Multiple , Human Growth Hormone , Loose Anagen Hair Syndrome , Megalencephaly , Noonan Syndrome , Abnormalities, Multiple/pathology , Hair/pathology , Human Growth Hormone/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Japan , Loose Anagen Hair Syndrome/diagnosis , Loose Anagen Hair Syndrome/genetics , Loose Anagen Hair Syndrome/pathology , Megalencephaly/pathology , Mutation , Noonan Syndrome/diagnosis , Noonan Syndrome/genetics , Noonan Syndrome/pathology
12.
Ann Pathol ; 42(3): 259-263, 2022 Apr.
Article in French | MEDLINE | ID: mdl-35058088

ABSTRACT

We report the case of a 10-year-old child with bilateral mandibular localization of a central giant cell granuloma occurring in the setting of Noonan syndrome. The histological appearance was classic with two intermigled components, one fibrous with non-atypical mononuclear cells, the other consisting of numerous osteoclast-like giant cells. This aspect is similar to that observed in the brown tumor as well as that of cherubism, which can also give multiple bone lesions. We will discuss the other lesions to consider in case of benign giant cell bone lesions affecting the jawbones, sometimes multiple and part of which falls within the scope of RASopathies.


Subject(s)
Cherubism , Granuloma, Giant Cell , Noonan Syndrome , Cherubism/genetics , Cherubism/pathology , Child , Giant Cells/pathology , Granuloma, Giant Cell/pathology , Humans , Jaw/pathology , Noonan Syndrome/complications , Noonan Syndrome/genetics , Noonan Syndrome/pathology
13.
Eur J Med Genet ; 64(9): 104284, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34242782

ABSTRACT

We describe a 13-years-old girl, previously diagnosed with PTPN11-associated Noonan Syndrome (NS), who presented to the pediatric emergency department for fever and drowsiness, which gradually worsened within 48 h. On admission, brain magnetic resonance imaging (MRI) scan showed diffuse, symmetric, multiple, poorly demarcated, confluent hyperintense lesions on MRI T2w-images, located in the Central Nervous System (CNS). In the absence of a better explanation and according to the current diagnostic criteria, a diagnosis of Acute Disseminated Encephalomyelitis (ADEM) was performed. The patient was first treated with intravenous methylprednisolone, then with intravenous immunoglobulin (IVIG). Owing to the poor clinical response, three sessions of therapeutic plasma exchange (TPE) were finally performed, with a progressive improvement. Follow-up MRI performed after three months from the onset revealed a considerable reduction in brain lesions, while cervical and dorsal ones were substantially unmodified. Neurological examination showed a full recovery of cognitive function and improved strength and tone of the upper limbs, while tetrahyporeflexia and proximal weakness of lower limbs were still appreciable. To date, this is the first described case of ADEM occurring in a patient with NS.


Subject(s)
Encephalomyelitis/etiology , Noonan Syndrome/complications , Adolescent , Brain/diagnostic imaging , Diagnosis, Differential , Encephalomyelitis/complications , Encephalomyelitis/pathology , Encephalomyelitis/therapy , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Methylprednisolone/therapeutic use , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Plasma Exchange
14.
Am J Med Genet A ; 185(12): 3623-3633, 2021 12.
Article in English | MEDLINE | ID: mdl-34184824

ABSTRACT

RASopathies are a group of disorders caused by pathogenic variants in the genes encoding Ras/mitogen-activated protein kinase pathway and share overlapping clinical and molecular features. This study is aimed to describe the clinical and molecular features of 38 patients with RASopathies. Sanger or targeted next-generation sequencing of related genes and multiplex ligation-dependent-probe amplification analysis for NF1 were performed. The pathogenic variant detection rate was 94.4%. While PTPN11 was responsible for 50% of 18 patients with Noonan syndrome (NS), SOS1, LZTR1, RIT1, and RAF1 were responsible for the remaining 27.8%, 11.1%, 5.5%, and 5.5%, respectively. Three variants in LZTR1 were novel, of which two were identified in the compound heterozygous state in a patient with intellectual disability and hypertrophic cardiomyopathy, whereas the third variant was found in the heterozygous state in a patient with pulmonary stenosis and normal intelligence. We described pyloric stenosis, knee dislocation, and cleft palate in patients with SOS1, RIT1, and RAF1 variants, respectively, that was not previously reported. We detected a PTPN11 variant in three patients from same family with NS with multiple lentigines. BRAF and MAP2K2 variants were found in eight patients with Cardiofaciocutaneous syndrome. Two variants in HRAS were detected in two Costello syndrome patients, one with a mild and the other with a severe phenotype. While large NF1 deletions were identified in four Neurofibromatosis-NS patients with intellectual disability, intelligence was normal in one patient with missense variant. In conclusion, this study provided three novel variants in LZTR1 and expanded the clinical phenotype of rare RASopathies.


Subject(s)
Neurofibromatoses/genetics , Neurofibromin 1/genetics , Noonan Syndrome/genetics , Proto-Oncogene Proteins c-raf/genetics , Transcription Factors/genetics , ras Proteins/genetics , Adolescent , Adult , Child , Child, Preschool , Cleft Palate/genetics , Cleft Palate/physiopathology , Costello Syndrome/genetics , Costello Syndrome/physiopathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/physiopathology , Facies , Failure to Thrive/genetics , Failure to Thrive/physiopathology , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Humans , Infant , Knee Dislocation/genetics , Knee Dislocation/physiopathology , Male , Mutation , Neurofibromatoses/epidemiology , Neurofibromatoses/pathology , Noonan Syndrome/epidemiology , Noonan Syndrome/pathology , Phenotype , Pyloric Stenosis/genetics , Pyloric Stenosis/physiopathology , Young Adult
15.
Am J Med Genet A ; 185(10): 3099-3103, 2021 10.
Article in English | MEDLINE | ID: mdl-34080768

ABSTRACT

Noonan syndrome (NS) is a Mendelian phenotype, member of a group of disorders sharing neurocardiofaciocutaneous involvement, known as RASopathies, caused by germline variants in genes coding for components of the RAS/MAPK signaling pathway. Recently, a novel gene of the RAS family (MRAS) was reported to be associated with NS in five children, all of them presenting, among the cardinal features of NS, the same cardiac finding, hypertrophic cardiomyopathy (HCM). We report on a 2-month-old infant boy also presenting this cardiac anomaly that evolved to a fatal outcome after a surgical myectomy. In addition, a thick walled left ventricle apical aneurysm, rarely described in NS, was also disclosed. Next-generation sequencing revealed a missense, previously reported variant in MRAS (p.Thr68Ile). This report reinforces the high frequency of HCM among individuals harboring MRAS variants, contrasting to the 20% overall prevalence of this cardiac anomaly in NS. Thus, these preliminary data suggest that variants in MRAS per se are high risk factors for the development of an early, severe HCM, mostly of them with left ventricle outflow tract obstruction, with poor prognosis. Because of the severity of the cardiac involvement, other clinical findings could not be addressed in detail. Therefore, long-term follow-up of these individuals and further descriptions are required to fully understand the complete phenotypic spectrum of NS associated with MRAS germline variants, including if these individuals present an increased risk for cancer.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Heart Defects, Congenital/genetics , Noonan Syndrome/genetics , ras Proteins/genetics , Adolescent , Cardiomyopathy, Hypertrophic/diagnosis , Cardiomyopathy, Hypertrophic/pathology , Child , Child, Preschool , Female , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/pathology , Heterozygote , Humans , Infant , MAP Kinase Signaling System/genetics , Male , Mutation/genetics , Noonan Syndrome/diagnosis , Noonan Syndrome/pathology
16.
Am J Med Genet A ; 185(10): 3048-3052, 2021 10.
Article in English | MEDLINE | ID: mdl-34032360

ABSTRACT

Noonan syndrome (NS) is one of the common RASopathies. While the clinical phenotype in NS is variable, it is typically characterized by distinctive craniofacial features, cardiac defects, reduced growth, bleeding disorders, learning issues, and an increased risk of cancer. Several different genes cause NS, all of which are involved in the Ras/mitogen-activated protein kinase (Ras/MAPK) pathway. Juvenile xanthogranuloma (JXG) is an uncommon, proliferative, self-limited cutaneous disorder that affects young individuals and may be overlooked or misdiagnosed due to its transient nature. A RASopathy that is known to be associated with JXG is neurofibromatosis type 1 (NF1). JXG in NF1 has also been reported in association with a juvenile myelomonocytic leukemia (JMML). As RASopathies, both NS and NF1 have an increased incidence of JMML. We report a 10-month-old female with NS who has a PTPN11 pathogenic variant resulting in a heterozygous SHP2 p.Y62D missense mutation. She was found to have numerous, small, yellow-pink smooth papules that were histopathologically confirmed to be JXG. In understanding the common underlying pathogenetic dysregulation of the Ras/MAPK pathway in both NS and NF1, this report suggests a possible molecular association for why NS individuals may be predisposed to JXG.


Subject(s)
Genetic Predisposition to Disease , Leukemia, Myelomonocytic, Juvenile/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Xanthogranuloma, Juvenile/genetics , Female , Humans , Infant , Leukemia, Myelomonocytic, Juvenile/complications , Leukemia, Myelomonocytic, Juvenile/pathology , Mutation, Missense/genetics , Neurofibromin 1/genetics , Noonan Syndrome/complications , Noonan Syndrome/pathology , Phenotype , Xanthogranuloma, Juvenile/complications , Xanthogranuloma, Juvenile/pathology , ras Proteins/genetics
17.
Am J Med Genet A ; 185(6): 1883-1887, 2021 06.
Article in English | MEDLINE | ID: mdl-33779033

ABSTRACT

Noonan syndrome (NS) is an autosomal dominant condition with variable expressivity most commonly due to a germline pathogenic variant in PTPN11, which encodes the protein tyrosine phosphatase SHP-2. Gain-of-function variants in PTPN11 are known to promote oncogenic behavior in affected tissues. We report the clinical description of a young adult male presenting with relapsing ganglioneuromas, dysmorphic features, cardiac abnormalities, and multiple lentigines, strongly suspicious for NS. Solid tumor testing identified the recurrent pathogenic c.922G>A (p.Asn308Asp) in PTPN11. Proband and parental blood sampling testing confirmed c.922G>A as a de novo germline alteration. Comprehensive literature review of solid tumors specifically associated to PTPN11, indicates that this is the first documentation of ganglioneuroma and its clinical recurrence after resection in conjunction with a genetically confirmed NS diagnosis. The findings in our patient further extend the list of neuroblastic and neural crest-derived neoplasms associated with this condition.


Subject(s)
Ganglioneuroma/genetics , Heart Defects, Congenital/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Ganglioneuroma/pathology , Genetic Predisposition to Disease , Heart Defects, Congenital/pathology , Humans , Male , Mutation, Missense/genetics , Neoplasm Recurrence, Local/genetics , Noonan Syndrome/pathology , Phenotype , Young Adult
18.
Am J Med Genet A ; 185(6): 1897-1902, 2021 06.
Article in English | MEDLINE | ID: mdl-33750022

ABSTRACT

RASopathies are a group of syndromes with partially overlapping clinical features caused by germline mutations of the RAS/MAPK signaling pathway genes. The most common disorder is Noonan syndrome (NS; MIM 163950). We report the first prenatal case of NS with SOS2 (NM_006939.4) mutation in a euploid fetus with a severe increase in nuchal translucency (NT > 12 mm). Trio-based custom next-generation sequencing detected a de novo heterozygous missense mutation in the SOS2 gene: c.800 T > A (p.Met267Lys). Owing to the marked variable expressivity of NS and the scarcity of SOS2 mutation-related NS cases reported in the literature, it is difficult to provide appropriate genetic counseling. Several issues such as the best management technique and optimal NT cutoff have been discussed. In addition, in general, the fine balance between the advantages of an early prenatal diagnosis and the challenge of determining if the detected gene variant is pathogenic and, primarily, the stress of the counselees when providing a genetic counseling with limited information on the prenatal phenotype have been discussed. A prenatal path comprising examinations and multidisciplinary counseling is essential to support couples in a shared decision-making process.


Subject(s)
Early Diagnosis , Genetic Predisposition to Disease , Noonan Syndrome/diagnosis , Son of Sevenless Proteins/genetics , Female , Fetus/diagnostic imaging , Fetus/pathology , Genetic Counseling , Humans , Male , Mutation, Missense , Noonan Syndrome/diagnostic imaging , Noonan Syndrome/genetics , Noonan Syndrome/pathology , Pedigree , Prenatal Diagnosis
19.
Eur J Med Genet ; 64(4): 104187, 2021 04.
Article in English | MEDLINE | ID: mdl-33676063

ABSTRACT

RASopathies are a group of genetic conditions caused by germline variants in genes encoding signal transducers and modulators of the RAS-MAPK cascade. These disorders are multisystem diseases with considerable clinical overlap, even though distinct hallmarks are recognizable for each specific syndrome. Here we report on the presence of enlarged spinal nerve roots resembling neurofibromas, a typical neuroradiological finding of neurofibromatosis type 1, in two patients with a molecularly confirmed diagnosis of Noonan syndrome and cardio-facio-cutaneous syndrome, respectively. This evidence add enlarged spinal nerve roots as features shared among RASopathies. Future studies aiming to a better understanding of the molecular mechanisms leading to neurogenic tumor development in these patients are necessary to define their biological nature, evolution, prognosis and possible treatments.


Subject(s)
Ectodermal Dysplasia/pathology , Failure to Thrive/pathology , Heart Defects, Congenital/pathology , Noonan Syndrome/pathology , Spinal Nerve Roots/pathology , Child , Ectodermal Dysplasia/diagnostic imaging , Ectodermal Dysplasia/genetics , Facies , Failure to Thrive/diagnostic imaging , Failure to Thrive/genetics , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/genetics , Humans , Male , Noonan Syndrome/diagnostic imaging , Noonan Syndrome/genetics , Spinal Nerve Roots/diagnostic imaging , ras Proteins/genetics
20.
Am J Med Genet A ; 185(4): 1151-1158, 2021 04.
Article in English | MEDLINE | ID: mdl-33554457

ABSTRACT

Computer-assisted pattern recognition platforms, such as Face2Gene® (F2G), can facilitate the diagnosis of children with rare genetic syndromes by comparing a patient's features to known genetic diagnoses. Our work designed, implemented, and evaluated an innovative model of care in clinical genetics in a heterogeneous and multicultural patient population that utilized this facial phenotyping software at the point-of-care. We assessed the performance of F2G by comparing the suggested diagnoses to the patient's confirmed molecular diagnosis. Providers' overall experiences with the technology and trainees' educational experiences were assessed with questionnaires. We achieved an overall diagnostic yield of 57%. This increased to 82% when cases diagnosed with syndromes not recognized by F2G were removed. The mean rank of a confirmed diagnosis in the top 10 was 2.3 (CI 1.5-3.2) and the mean gestalt score 37.6%. The most commonly suggested diagnoses were Noonan syndrome, mucopolysaccharidosis, and 22q11.2 deletion syndrome. Our qualitative assessment revealed that clinicians and trainees saw value using the tool in practice. Overall, this work helped to implement an innovative patient care delivery model in clinical genetics that utilizes a facial phenotyping tool at the point-of-care. Our data suggest that F2G has utility in the genetics clinic as a clinical decision support tool in diverse populations, with a majority of patients having their eventual diagnosis listed in the top 10 suggested syndromes based on a photograph alone. It shows promise for further integration into clinical care and medical education, and we advocate for its continued use, adoption and refinement along with transparent and accountable industrial partnerships.


Subject(s)
Face/physiopathology , Facial Recognition , Genetic Counseling , Image Processing, Computer-Assisted/methods , Child , Child, Preschool , DiGeorge Syndrome/diagnosis , DiGeorge Syndrome/diagnostic imaging , DiGeorge Syndrome/pathology , Female , Humans , Machine Learning , Male , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/diagnostic imaging , Mucopolysaccharidoses/pathology , Noonan Syndrome/diagnosis , Noonan Syndrome/diagnostic imaging , Noonan Syndrome/pathology , Pattern Recognition, Automated/methods , Phenotype , Point-of-Care Systems , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...