Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Hum Genet ; 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38448605

RESUMEN

Rare heterozygous variants in exons 33-34 of the SRCAP gene are associated with Floating-Harbor syndrome and have a dominant-negative mechanism of action. At variance, heterozygous null alleles falling in other parts of the same gene cause developmental delay, hypotonia, musculoskeletal defects, and behavioral abnormalities (DEHMBA) syndrome. We report an 18-year-old man with DEHMBA syndrome and obstructive sleep apnea, who underwent exome sequencing (ES) and whole transcriptome sequencing (WTS) on peripheral blood. Trio analysis prioritized the de novo heterozygous c.5658+5 G > A variant. WTS promptly demostrated four different abnormal transcripts affecting >40% of the reads, three of which leading to a frameshift. This study demonstrated the efficacy of a combined ES-WTS approach in solving undiagnosed cases. We also speculated that sleep respiratory disorder may be an underdiagnosed complication of DEHMBA syndrome.

2.
J Hum Genet ; 69(1): 53-58, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37697026

RESUMEN

Heterozygous deleterious variants in SKI cause Shprintzen-Goldberg Syndrome, which is mainly characterized by craniofacial features, neurodevelopmental disorder and thoracic aorta dilatations/aneurysms. The encoded protein is a member of the transforming growth factor beta signaling. Paucity of reported studies exploring the SGS molecular pathogenesis hampers disease recognition and clinical interpretation of private variants. Here, the unpublished c.349G>A, p.[Gly117Ser] and the recurrent c.539C>T, p.[Thr180Met] SKI variants were studied combining in silico and in vitro approach. 3D comparative modeling and calculation of the interaction energy predicted that both variants alter the SKI tertiary protein structure and its interactions. Computational data were functionally corroborated by the demonstration of an increase of MAPK phosphorylation levels and alteration of cell cycle in cells expressing the mutant SKI. Our findings confirmed the effects of SKI variants on MAPK and opened the path to study the role of perturbations of the cell cycle in SGS.


Asunto(s)
Síndrome de Marfan , Simulación de Dinámica Molecular , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ciclo Celular/genética , Factor de Crecimiento Transformador beta
3.
Front Endocrinol (Lausanne) ; 14: 1205977, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37600704

RESUMEN

Introduction: Hypophosphatasia (HPP) is a rare genetic disease caused by inactivating variants of the ALPL gene. Few data are available on the clinical presentation in Italy and/or on Italian HPP surveys. Methods: There were 30 suspected HPP patients recruited from different Italian tertiary cares. Biological samples and related clinical, biochemical, and anamnestic data were collected and the ALPL gene sequenced. Search for large genomic deletions at the ALPL locus (1p36) was done. Phylogenetic conservation and modeling were applied to infer the effect of the variants on the protein structure. Results: There were 21 ALPL variants and one large genomic deletion found in 20 out of 30 patients. Unexpectedly, NGS-driven differential diagnosis allowed uncovering three hidden additional HPP cases, for a total of 33 HPP subjects. Eight out of 24 coding variants were novel and classified as "pathogenic", "likely pathogenic", and "variants of uncertain significance". Bioinformatic analysis confirmed that all the variants strongly destabilize the homodimer structure. There were 10 cases with low ALP and high VitB6 that resulted negative to genetic testing, whereas two positive cases have an unexpected normal ALP value. No association was evident with other biochemical/clinical parameters. Discussion: We present the survey of HPP Italian patients with the highest ALPL mutation rate so far reported and confirm the complexity of a prompt recognition of the syndrome, mostly for HPP in adults. Low ALP and high VitB6 values are mandatory for the genetic screening, this latter remaining the gold standard not only to confirm the clinical diagnosis but also to make differential diagnosis, to identify carriers, to avoid likely dangerous therapy in unrecognized cases.


Asunto(s)
Hipofosfatasia , Adulto , Humanos , Hipofosfatasia/diagnóstico , Hipofosfatasia/epidemiología , Hipofosfatasia/genética , Filogenia , Biología Computacional , Diagnóstico Diferencial , Italia/epidemiología , Enfermedades Raras
4.
Hum Genet ; 142(6): 785-808, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37079061

RESUMEN

Deleterious variants in collagen genes are the most common cause of hereditary connective tissue disorders (HCTD). Adaptations of the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) criteria are still lacking. A multidisciplinary team was set up for developing specifications of the ACMG/AMP criteria for COL1A1, COL1A2, COL2A1, COL3A1, COL5A1, COL5A2, COL11A1, COL11A2 and COL12A1, associated with various forms of HCTD featuring joint hypermobility, which is becoming one of the most common reasons of referral for molecular testing in this field. Such specifications were validated against 209 variants, and resulted effective for classifying as pathogenic and likely pathogenic null alleles without downgrading of the PVS1 level of strength and recurrent Glycine substitutions. Adaptations of selected criteria reduced uncertainties on private Glycine substitutions, intronic variants predicted to affect the splicing, and null alleles with a downgraded PVS1 level of strength. Segregation and multigene panel sequencing data mitigated uncertainties on non-Glycine substitutions by the attribution of one or more benignity criteria. These specifications may improve the clinical utility of molecular testing in HCTD by reducing the number of variants with neutral/conflicting interpretations. Close interactions between laboratory and clinicians are crucial to estimate the a priori utility of molecular test and to improve medical reports.


Asunto(s)
Variación Genética , Inestabilidad de la Articulación , Humanos , Estados Unidos , Pruebas Genéticas/métodos , Inestabilidad de la Articulación/diagnóstico , Inestabilidad de la Articulación/genética , Análisis de Secuencia de ADN/métodos
5.
Biochim Biophys Acta Mol Cell Res ; 1870(5): 119474, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37030452

RESUMEN

In the central nervous system, thrombin-mediated activation of protease-activated receptors (PARs) results in neuroinflammation and increased vascular permeability. These events have been linked to cancer and neurodegeneration. Endothelial cells (ECs) isolated from sporadic cerebral cavernous malformation (CCM) specimens showed dysregulation of genes involved in "thrombin-mediated PAR-1 activation" signaling. CCM is a vascular disease involving brain capillaries. In CCM, ECs show defective cell junctions. Oxidative stress and neuroinflammation play a key role in disease onset and progression. In order to confirm the possible role of thrombin pathway in sporadic CCM pathogenesis, we evaluated PARs expression in CCM-ECs. We found that sporadic CCM-ECs overexpress PAR1, PAR3 and PAR4, together with other coagulation factor encoding genes. Moreover, we investigated about expression of the three familial CCM genes (KRIT1, CCM2 and PDCD10) in human cerebral microvascular ECs, following thrombin exposure, as well as protein level. Thrombin exposure affects EC viability and results in dysregulation of CCM gene expression and, then, in decreased protein level. Our results confirm amplification of PAR pathway in CCM suggesting, for the first time, the possible role of PAR1-mediated thrombin signaling in sporadic CCM. Thrombin-mediated PARs over activation results in increased blood-brain barrier permeability due to loss of cell junction integrity and, in this context, also the three familial CCM genes may be involved.


Asunto(s)
Hemangioma Cavernoso del Sistema Nervioso Central , Humanos , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Células Endoteliales/metabolismo , Enfermedades Neuroinflamatorias , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Trombina/farmacología , Proteínas Proto-Oncogénicas/genética
6.
Pediatr Res ; 93(5): 1208-1215, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35953512

RESUMEN

BACKGROUND: Loss-of-function variants in MID1 are the most common cause of Opitz G/BBB syndrome (OS). The interpretation of intronic variants affecting the splicing is a rising issue in OS. METHODS: Exon sequencing of a 2-year-old boy with OS showed that he was a carrier of the de novo c.1286-10G>T variant in MID1. In silico predictions and minigene assays explored the effect of the variant on splicing. The minigene approach was also applied to two previously identified MID1 c.864+1G>T and c.1285+1G>T variants. RESULTS: Minigene assay demonstrated that the c.1286-10G>T variant generated the inclusion of eight nucleotides that predicted generation of a frameshift. The c.864+1G>T and c.1285+1G>T variants resulted in an in-frame deletion predicted to generate a shorter MID1 protein. In hemizygous males, this allowed reclassification of all the identified variants from "of unknown significance" to "likely pathogenic." CONCLUSIONS: Minigene assay supports functional effects from MID1 intronic variants. This paves the way to the introduction of similar second-tier investigations in the molecular diagnostics workflow of OS. IMPACT: Causative intronic variants in MID1 are rarely investigated in Opitz syndrome. MID1 is not expressed in blood and mRNA studies are hardly accessible in routine diagnostics. Minigene assay is an alternative for assessing the effect of intronic variants on splicing. This is the first study characterizing the molecular consequences of three MID1 variants for diagnostic purposes and demonstrating the efficacy of minigene assays in supporting their clinical interpretation. Review of the criteria according to the American College of Medical Genetics reassessed all variants as likely pathogenic.


Asunto(s)
Fisura del Paladar , Hipertelorismo , Masculino , Humanos , Preescolar , Mutación , Fisura del Paladar/genética , Hipertelorismo/genética , Ubiquitina-Proteína Ligasas/genética
7.
Genes (Basel) ; 13(6)2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35741725

RESUMEN

Cerebral cavernous malformations (CCM) are capillary malformations affecting the central nervous system and commonly present with headaches, epilepsy and stroke. Treatment of CCM is symptomatic, and its prevention is limited. CCM are often sporadic but sometimes may be multifocal and/or affect multiple family members. Heterozygous pathogenic variants in PDCD10 cause the rarest and apparently most severe genetic variant of familial CCM. We carried out an RNA-Seq and a Q-PCR validation analysis in Pdcd10-silenced and wild-type mouse endothelial cells in order to better elucidate CCM molecular pathogenesis. Ninety-four differentially expressed genes presented an FDR-corrected p-value < 0.05. A functionally clustered dendrogram showed that differentially expressed genes cluster in cell proliferation, oxidative stress, vascular processes and immune response gene-ontology functions. Among differentially expressed genes, the major cluster fell in signaling related to inflammation and pathogen recognition, including HIF1α and Nos2 signaling and immune regulation. Validation analysis performed on wild-type, Pdcd10-null and Pdcd10-null reconstituted cell lines was consistent with RNA-Seq data. This work confirmed previous mouse transcriptomic data in endothelial cells, which are recognized as a critical tissue for CCM formation and expands the potential molecular signatures of PDCD10-related familial CCM to alterations in inflammation and pathogen recognition pathways.


Asunto(s)
Células Endoteliales , Inflamación , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Hemangioma Cavernoso del Sistema Nervioso Central , Hipoxia/metabolismo , Inflamación/genética , Inflamación/metabolismo , Ratones
8.
Int J Mol Sci ; 24(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36614110

RESUMEN

Achalasia is an esophageal smooth muscle motility disorder with unknown pathogenesis. Taking into account our previous results on the downexpression of miR-200c-3p in tissues of patients with achalasia correlated with an increased expression of PRKG1, SULF1, and SYDE1 genes, our aim was to explore the unknown biological interaction between these genes and human miR-200c-3p and if this relation could unravel their functional role in the etiology of achalasia. To search for putative miR-200c-3p binding sites in the 3'-UTR of PRKG1, SULF1 and SYDE1, a bioinformatics tool was used. To test whether PRKG1, SULF1, and SYDE1 are targeted by miR-200c-3p, a dual-luciferase reporter assay and quantitative PCR on HEK293 and fibroblast cell lines were performed. To explore the biological correlation between PRKG1 and miR-200c-3p, an immunoblot analysis was carried out. The overexpression of miR-200c-3p reduced the luciferase activity in cells transfected with a luciferase reporter containing a fragment of the 3'-UTR regions of PRKG1, SULF1, and SYDE1 which included the miR-200c-3p seed sequence. The deletion of the miR-200c-3p seed sequence from the 3'-UTR fragments abrogated this reduction. A negative correlation between miR-200c-3p and PRKG1, SULF1, and SYDE1 expression levels was observed. Finally, a reduction of the endogenous level of PRKG1 in cells overexpressing miR-200c-3p was detected. Our study provides, for the first time, functional evidence about the PRKG1 gene as a direct target and SULF1 and SYDE1 as potential indirect substrates of miR-200c-3p and suggests the involvement of NO/cGMP/PKG signaling in the pathogenesis of achalasia.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Acalasia del Esófago , MicroARNs , Humanos , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Acalasia del Esófago/genética , Células HEK293 , MicroARNs/genética , MicroARNs/metabolismo
9.
Genet Med ; 24(2): 439-453, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906501

RESUMEN

PURPOSE: This study aimed to describe a multisystemic disorder featuring cardiovascular, facial, musculoskeletal, and cutaneous anomalies caused by heterozygous loss-of-function variants in TAB2. METHODS: Affected individuals were analyzed by next-generation technologies and genomic array. The presumed loss-of-function effect of identified variants was assessed by luciferase assay in cells transiently expressing TAB2 deleterious alleles. In available patients' fibroblasts, variant pathogenicity was further explored by immunoblot and osteoblast differentiation assays. The transcriptomic profile of fibroblasts was investigated by RNA sequencing. RESULTS: A total of 11 individuals from 8 families were heterozygotes for a novel TAB2 variant. In total, 7 variants were predicted to be null alleles and 1 was a missense change. An additional subject was heterozygous for a 52 kb microdeletion involving TAB2 exons 1 to 3. Luciferase assay indicated a decreased transcriptional activation mediated by NF-κB signaling for all point variants. Immunoblot analysis showed a reduction of TAK1 phosphorylation while osteoblast differentiation was impaired. Transcriptomic analysis identified deregulation of multiple pleiotropic pathways, such as TGFß-, Ras-MAPK-, and Wnt-signaling networks. CONCLUSION: Our data defined a novel disorder associated with loss-of-function or, more rarely, hypomorphic alleles in a restricted linker region of TAB2. The pleiotropic manifestations in this disorder partly recapitulate the 6q25.1 (TAB2) microdeletion syndrome and deserve the definition of cardio-facial-cutaneous-articular syndrome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , FN-kappa B , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Exones/genética , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación , Transducción de Señal
10.
Clin Genet ; 99(6): 829-835, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33604894

RESUMEN

Cerebral cavernous malformation (CCM) is a vascular malformation of the central nervous system which may occur sporadically or segregate within families due to heterozygous variants in KRIT1/CCM1, MGC4607/CCM2 or PDCD10/CCM3. Intronic variants are not uncommon in familial CCM, but their clinical interpretation is often hampered by insufficient data supporting in silico predictions. Here, the mRNA analysis for two intronic unpublished variants (KRIT1 c.1147-7 T > G and PDCD10 c.395 + 2 T > G) and three previously published variants in KRIT1 but without data supporting their effects was carried out. This study demonstrated that all variants can induce a frameshift with the lack of residues located in the C-terminal regions and involved in protein-protein complex formation, which is essential for vascular homeostasis. These results support the introduction of mRNA analysis in the diagnostic pathway of familial CCM and expand the knowledge of abnormal splicing patterning in this disorder.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteína KRIT1/genética , Proteínas de la Membrana/genética , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Humanos , Empalme del ARN/genética , ARN Mensajero/genética
11.
Am J Med Genet A ; 185(3): 955-965, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33369135

RESUMEN

ATP6V0A2-related cutis laxa, also known as autosomal recessive cutis laxa type 2A (ARCL2A), is a subtype of hereditary cutis laxa originally characterized by skin, skeletal, and neurological involvement, and a combined defect of N-glycosylation and O-glycosylation. The associated clinical spectrum subsequently expanded to a less severe phenotype dominated by cutaneous involvement. At the moment, ARCL2A was described in a few case reports and series only. An Italian adult woman ARCL2A with a phenotype restricted to skin and the two novel c.3G>C and c.1101dup ATP6V0A2 variants has been reported. A systematic literature review allowed us to identify 69 additional individuals from 64 families. Available data were scrutinized in order to describe the clinical and molecular variability of ARCL2A. About 78.3% of known variants were predicted null alleles, while 11 were missense and 2 affected noncanonical splice sites. Age at ascertainment appeared as the unique phenotypic discriminator with earlier age more commonly associated with facial dysmorphism (p .02), high/cleft palate (p .005), intellectual disability/global developmental delay (p .013), and seizures (p .024). No specific genotype-phenotype correlations were identified. This work confirmed the existence of an attenuated phenotype associated with ATP6V0A2 biallelic variants and offers an updated critique to the clinical and molecular variability of ARCL2A.


Asunto(s)
Cutis Laxo/genética , ATPasas de Translocación de Protón/genética , Adulto , Factores de Edad , Alelos , Secuencia de Bases , Codón sin Sentido , Cutis Laxo/diagnóstico , Exones/genética , Femenino , Mutación del Sistema de Lectura , Genes Recesivos , Estudios de Asociación Genética , Heterogeneidad Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Esperanza de Vida , Mutación con Pérdida de Función , Mutación Missense , Linaje , Fenotipo , ATPasas de Translocación de Protón/deficiencia , Sitios de Empalme de ARN/genética , Piel/patología
12.
Genes (Basel) ; 11(12)2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33348901

RESUMEN

Stickler syndrome (SS) is a hereditary connective tissue disorder affecting bones, eyes, and hearing. Type 2 SS and the SS variant otospondylomegaepiphyseal dysplasia (OSMED) are caused by deleterious variants in COL11A1 and COL11A2, respectively. In both genes, available database information indicates a high rate of potentially deleterious intronic variants, but published evidence of their biological effect is usually insufficient for a definite clinical interpretation. We report four previously unpublished intronic variants in COL11A1 (c.2241 + 5G>T, c.2809 - 2A>G, c.3168 + 5G>C) and COL11A2 (c.4392 + 1G>A) identified in type 2 SS/OSMED individuals. The pathogenic effect of these variants was first predicted in silico and then investigated by an exon-trapping assay. We demonstrated that all variants can induce exon in-frame deletions, which lead to the synthesis of shorter collagen XI α1 or 2 chains. Lacking residues are located in the α-triple helical region, which has a crucial role in regulating collagen fibrillogenesis. In conclusion, this study suggests that these alternative COL11A1 and COL11A2 transcripts might result in aberrant triple helix collagen. Our approach may help to improve the diagnostic molecular pathway of COL11-related disorders.


Asunto(s)
Colágeno Tipo XI/deficiencia , Enfermedades del Tejido Conjuntivo/genética , Enanismo/genética , Intrones/genética , Osteocondrodisplasias/genética , Mutación Puntual , Desprendimiento del Vítreo/genética , Adulto , Secuencia de Bases , Colágeno Tipo XI/química , Colágeno Tipo XI/genética , Enfermedades del Tejido Conjuntivo/diagnóstico , Discapacidades del Desarrollo/genética , Diagnóstico Diferencial , Enanismo/diagnóstico , Síndrome de Ehlers-Danlos/diagnóstico , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Osteocondrodisplasias/diagnóstico , Isoformas de Proteínas/genética , Estructura Secundaria de Proteína , Empalme del ARN , ARN Mensajero/genética , Desprendimiento del Vítreo/diagnóstico
13.
Int J Mol Sci ; 21(14)2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32698527

RESUMEN

Transforming growth factor ß (TGF-ß) superfamily signaling pathways are ubiquitous and essential for several cellular and physiological processes. The overexpression of TGF-ß results in excessive fibrosis in multiple human disorders. Among them, stiff skin syndrome (SSS) is an ultrarare and untreatable condition characterized by the progressive thickening and hardening of the dermis, and acquired joint limitations. SSS is distinct in a widespread form, caused by recurrent germline variants of FBN1 encoding a key molecule of the TGF-ß signaling, and a segmental form with unknown molecular basis. Here, we report a 12-year-old female with segmental SSS, affecting the right upper limb with acquired thickening of the dermis evident at the magnetic resonance imaging, and progressive limitation of the elbow and shoulder. To better explore the molecular and cellular mechanisms that drive segmental SSS, several functional studies on patient's fibroblasts were employed. We hypothesized an impairment of TGF-ß signaling and, consequently, a dysregulation of the associated downstream signaling. Lesional fibroblast studies showed a higher phosphorylation level of extracellular signal-regulated kinase 1/2 (ERK1/2), increased levels of nuclear factor-kB (NFkB), and a nuclear accumulation of phosphorylated Smad2 via Western blot and microscopy analyses. Quantitative PCR expression analysis of genes encoding key extracellular matrix proteins revealed increased levels of COL1A1, COL3A1, AGT, LTBP and ITGB1, while zymography assay reported a reduced metalloproteinase 2 enzymatic activity. In vitro exposure of patient's fibroblasts to losartan led to the partial restoration of normal transforming growth factor ß (TGF-ß) marker protein levels. Taken together, these data demonstrate that in our patient, segmental SSS is characterized by the overactivation of multiple TGF-ß signaling pathways, which likely results in altered extracellular matrix composition and fibroblast homeostasis. Our results for the first time reported that aberrant TGF-ß signaling may drive the pathogenesis of segmental SSS and might open the way to novel therapeutic approaches.


Asunto(s)
Contractura/patología , Transducción de Señal , Enfermedades Cutáneas Genéticas/patología , Piel/patología , Factor de Crecimiento Transformador beta/metabolismo , Adolescente , Contractura/diagnóstico por imagen , Contractura/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Imagen por Resonancia Magnética , Fosforilación , Piel/diagnóstico por imagen , Piel/metabolismo , Enfermedades Cutáneas Genéticas/diagnóstico por imagen , Enfermedades Cutáneas Genéticas/metabolismo
14.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165742, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32105826

RESUMEN

Transforming growth factor beta-activated kinase 1 (TAK1) is a highly conserved kinase protein encoded by MAP3K7, and activated by multiple extracellular stimuli, growth factors and cytokines. Heterozygous variants in MAP3K7 cause the cardiospondylocarpofacial syndrome (CSCFS) which is characterized by short stature, dysmorphic facial features, cardiac septal defects with valve dysplasia, and skeletal anomalies. CSCFS has been described in seven patients to date and its molecular pathogenesis is only partially understood. Here, the functional effects of the MAP3K7 c.737-7A > G variant, previously identified in a girl with CSCFS and additional soft connective tissue features, were explored. This splice variant generates an in-frame insertion of 2 amino acid residues in the kinase domain of TAK1. Computational analysis revealed that this in-frame insertion alters protein dynamics in the kinase activation loop responsible for TAK1 autophosphorylation after binding with its interactor TAB1. Co-immunoprecipitation studies demonstrate that the ectopic expression of TAK1-mutated protein impairs its ability to physically bind TAB1. In patient's fibroblasts, MAP3K7 c.737-7A > G variant results in reduced TAK1 autophosphorylation and dysregulation of the downstream TAK1-dependent signaling pathway. TAK1 loss-of-function is associated with an impaired TGFß-mediated α-SMA cytoskeleton assembly and cell migration, and defective autophagy process. These findings contribute to our understanding of the molecular pathogenesis of CSCFS and might offer the rationale for the design of novel therapeutic targets.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Autofagia/genética , Pérdida Auditiva Bilateral/genética , Quinasas Quinasa Quinasa PAM/genética , Insuficiencia de la Válvula Mitral/genética , Osteosclerosis/genética , Anomalías Múltiples/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Niño , Citoesqueleto/genética , Femenino , Fibroblastos/metabolismo , Pérdida Auditiva Bilateral/fisiopatología , Humanos , Mutación con Pérdida de Función/genética , Insuficiencia de la Válvula Mitral/fisiopatología , Mutación/genética , Osteosclerosis/fisiopatología , Fosforilación/genética , Polimorfismo de Nucleótido Simple/genética , Unión Proteica/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
15.
Hum Mutat ; 40(10): 1886-1898, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31250519

RESUMEN

Transforming growth factor ß-activated kinase 1 (TAK1) mediates multiple biological processes through the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and the mitogen-activated protein kinase (MAPK) signaling pathways. TAK1 activation is tightly regulated by its binding partners (TABs). In particular, binding with TAB2 is crucial for cardiovascular development and extracellular matrix (ECM) homeostasis. In our previous work, we reported a novel multisystem disorder associated with the heterozygous TAB2 c.1398dup variant. Here, we dissect the functional effects of this variant in order to understand its molecular pathogenesis. We demonstrate that TAB2 c.1398dup considerably undergoes to nonsense-mediated messenger RNA decay and encodes a truncated protein that loses its ability to bind TAK1. We also show an alteration of the TAK1 autophosphorylation status and of selected downstream signaling pathways in patients' fibroblasts. Immunofluorescence analyses and ECM-related polymerase chain reaction-array panels highlight that patient fibroblasts display ECM disorganization and altered expression of selected ECM components and collagen-related pathways. In conclusion, we deeply dissect the molecular pathogenesis of the TAB2 c.1398dup variant and show that the resulting phenotype is well explained by TAB2 loss-of-function. Our data also offer initial insights on the ECM homeostasis impairment as a molecular mechanism probably underlying a multisystem disorder linked to TAB2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Matriz Extracelular/metabolismo , Variación Genética , Haploinsuficiencia , Homeostasis , Proteínas Adaptadoras Transductoras de Señales/química , Secuencia de Aminoácidos , Línea Celular , Proliferación Celular , Análisis Mutacional de ADN , Fibroblastos/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Mutación , Degradación de ARNm Mediada por Codón sin Sentido , Fosforilación , Unión Proteica , Transducción de Señal
16.
Hum Mutat ; 40(11): e24-e36, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31254430

RESUMEN

Familial cerebral cavernous malformation (FCCM) is an autosomal dominant vascular disorder caused by heterozygous deleterious variants in KRIT1, CCM2 or PDCD10. In a previous study, we presented the clinical and molecular findings in 140 FCCM individuals. In the present work, we report supporting information on (a) applied diagnostic workflow; (b) clinical significance of molecular findings according to the American College of Medical Genetics and Genomics/Association for Molecular Pathology recommendations; (c) standardization of molecular and clinical data according to the Human Phenotype Ontology; (d) preliminary genotype-phenotype correlations on a subgroup of patients by considering sex, age at diagnosis, neurological symptoms, and number and anatomical site(s) of vascular anomalies; (e) datasets submitted to the Leiden Open Variation Database. An overview of the changes of our diagnostic approach before and after the transition to next-generation sequencing is also reported. This work presents the full procedure that we apply for molecular testing, data interpretation and storing in public databases in FCCM.


Asunto(s)
Interpretación Estadística de Datos , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Hemangioma Cavernoso del Sistema Nervioso Central/diagnóstico , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Flujo de Trabajo , Alelos , Biología Computacional/métodos , Bases de Datos Genéticas , Femenino , Estudios de Asociación Genética/métodos , Genotipo , Humanos , Masculino , Técnicas de Diagnóstico Molecular , Fenotipo
17.
Eur J Hum Genet ; 27(7): 1113-1120, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30787447

RESUMEN

Hartsfield syndrome (HS) is an ultrarare developmental disorder mainly featuring holoprosencephaly and ectrodactyly. It is caused by heterozygous or biallelic variants in FGFR1. Recently, a dominant-negative effect was suggested for FGFR1 variants associated with HS. Here, exome sequencing analysis in a 12-year-old boy with HS disclosed a novel de novo heterozygous variant c.1934C>T in FGFR1 predicted to cause the p.(Ala645Val) amino-acid substitution. In order to evaluate whether the variant, changing a highly conserved residue of the kinase domain, affects FGFR1 function, biochemical studies were employed. We measured the FGFR1 receptor activity in FGF2-treated cell lines exogenously expressing wild-type or Ala645Val FGFR1 by monitoring the activation status of FGF2/FGFR1 downstream pathways. Our analysis highlighted that RAS/ERK1/2 signaling was significantly perturbed in cells expressing mutated FGFR1, in comparison with control cells. We also provided preliminary evidence showing a modulation of the autophagic process in cells expressing mutated FGFR1. This study expands the FGFR1 mutational spectrum associated with HS, provides functional evidence further supporting a dominant-negative effect of this category of FGFR1 variants and offers initial insights on dysregulation of autophagy in HS.


Asunto(s)
Labio Leporino , Fisura del Paladar , Dedos/anomalías , Deformidades Congénitas de la Mano , Holoprosencefalia , Discapacidad Intelectual , Sistema de Señalización de MAP Quinasas , Mutación Missense , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Sustitución de Aminoácidos , Labio Leporino/genética , Labio Leporino/metabolismo , Labio Leporino/patología , Fisura del Paladar/genética , Fisura del Paladar/metabolismo , Fisura del Paladar/patología , Femenino , Dedos/patología , Genes Dominantes , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/metabolismo , Deformidades Congénitas de la Mano/patología , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Holoprosencefalia/patología , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Hum Mol Genet ; 28(13): 2133-2142, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30806661

RESUMEN

Hereditary multiple osteochondromas (HMO) is a rare autosomal dominant skeletal disorder, caused by heterozygous variants in either EXT1 or EXT2, which encode proteins involved in the biogenesis of heparan sulphate. Pathogenesis and genotype-phenotype correlations remain poorly understood. We studied 114 HMO families (158 affected individuals) with causative EXT1 or EXT2 variants identified by Sanger sequencing, or multiplex ligation-dependent probe amplification and qPCR. Eighty-seven disease-causative variants (55 novel and 32 known) were identified including frameshift (42%), nonsense (32%), missense (11%), splicing (10%) variants and genomic rearrangements (5%). Informative clinical features were available for 42 EXT1 and 27 EXT2 subjects. Osteochondromas were more frequent in EXT1 as compared to EXT2 patients. Anatomical distribution of lesions showed significant differences based on causative gene. Microscopy analysis for selected EXT1 and EXT2 variants verified that EXT1 and EXT2 mutants failed to co-localize each other and loss Golgi localization by surrounding the nucleus and/or assuming a diffuse intracellular distribution. In a cell viability study, cells expressing EXT1 and EXT2 mutants proliferated more slowly than cells expressing wild-type proteins. This confirms the physiological relevance of EXT1 and EXT2 Golgi co-localization and the key role of these proteins in the cell cycle. Taken together, our data expand genotype-phenotype correlations, offer further insights in the pathogenesis of HMO and open the path to future therapies.


Asunto(s)
Exostosis Múltiple Hereditaria/genética , N-Acetilglucosaminiltransferasas/genética , Proliferación Celular , Supervivencia Celular , Femenino , Estudios de Asociación Genética , Aparato de Golgi/enzimología , Células HEK293 , Humanos , Masculino , Mutación , N-Acetilglucosaminiltransferasas/análisis
19.
Hum Mutat ; 39(12): 1885-1900, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30161288

RESUMEN

Cerebral cavernous malformation (CCM) is a capillary malformation arising in the central nervous system. CCM may occur sporadically or cluster in families with autosomal dominant transmission, incomplete penetrance, and variable expressivity. Three genes are associated with CCM KRIT1, CCM2, and PDCD10. This work is a retrospective single-center molecular study on samples from multiple Italian clinical providers. From a pool of 317 CCM index patients, we found germline variants in either of the three genes in 80 (25.2%) probands, for a total of 55 different variants. In available families, extended molecular analysis found segregation in 60 additional subjects, for a total of 140 mutated individuals. From the 55 variants, 39 occurred in KRIT1 (20 novel), 8 in CCM2 (4 novel), and 8 in PDCD10 (4 novel). Effects of the three novel KRIT1 missense variants were characterized in silico. We also investigated a novel PDCD10 deletion spanning exon 4-10, on patient's fibroblasts, which showed significant reduction of interactions between KRIT1 and CCM2 encoded proteins and impaired autophagy process. This is the largest study in Italian CCM patients and expands the known mutational spectrum of KRIT1, CCM2, and PDCD10. Our approach highlights the relevance of seeking supporting information to pathogenicity of new variants for the improvement of management of CCM.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas Portadoras/genética , Neoplasias del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Proteína KRIT1/genética , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Eliminación de Secuencia , Adulto , Anciano , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Proteínas Portadoras/metabolismo , Células Cultivadas , Neoplasias del Sistema Nervioso Central/metabolismo , Niño , Preescolar , Simulación por Computador , Exones , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Humanos , Italia , Proteína KRIT1/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Mutación Missense , Linaje , Proteínas Proto-Oncogénicas/metabolismo , Estudios Retrospectivos , Adulto Joven
20.
Radiother Oncol ; 77(1): 25-31, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15919126

RESUMEN

BACKGROUND AND PURPOSE: The goal of this study was to compare the inter-observer variability between CT and MRI for the delineation of pharyngo-laryngeal SCC, parotid glands and spinal cord. PATIENTS AND METHODS: Twenty pharyngo-laryngeal tumors were delineated by five observers on CT and MRI, using consistent delineation guidelines. Spinal cords and parotid glands were also delineated on CT and MRI by three observers. Mean GTVs and coefficients of variation were calculated for each observer and compared using ANOVA and its derived Pearson intra-class coefficient (R). For GTVs, a mismatch analysis (ratio between intersection and union volumes) was also performed. RESULTS: Regarding oropharyngeal GTVs (n=10), no significant difference was observed between observers either with CT (33.9, 31.1, 32, 34 and 34.7 ml, five observers, P=0.47) or with MRI (30.5, 29.4, 30.1 and 31.5 ml, four observers, P=0.59). CVs (13.6 vs 12.9%), (0.98 vs 0.99) and mismatches (0.43 vs 0.42) between CT and MRI did not significantly differ. Regarding laryngeal-hypopharyngeal GTVs (n=10), no significant difference was observed between observers either on CT (18.1, 20.7, 20.9, 19.3 and 21.9 ml, five observers, P=0.29) or on MRI (19.3, 21.5, 20, 22.1 and 21.8 ml, five observers, P=0.16). CVs (20.2 vs 13.8%), (0.94 vs 0.94) and mismatches (0.31 vs 0.41) were comparable. Regarding OARs, a small but significant difference in mean parotid volume was observed between observers (P<0.001) and between modalities (P<0.001) (CT: 34.8, 29.4, and 26.8 ml; MRI: 30.6, 27.9 and 20.4 ml). Similar results were obtained for mean spinal cord volumes (CT: 10.7, 10.6, and 9.5 ml; MRI: 8.7, 8.5 and 8.2 ml; P=0.05).


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Neoplasias Laríngeas/diagnóstico por imagen , Imagen por Resonancia Magnética , Glándula Parótida/diagnóstico por imagen , Neoplasias Faríngeas/diagnóstico por imagen , Médula Espinal/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Carcinoma de Células Escamosas/radioterapia , Fraccionamiento de la Dosis de Radiación , Humanos , Neoplasias Laríngeas/radioterapia , Variaciones Dependientes del Observador , Neoplasias Faríngeas/radioterapia , Traumatismos por Radiación/prevención & control , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...