Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Clin Genet ; 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38747114

RESUMO

Type IV collagen is an integral component of basement membranes. Mutations in COL4A1, one of the key genes encoding Type IV collagen, can result in a variety of diseases. It is clear that a significant proportion of mutations that affect splicing can cause disease directly or contribute to the susceptibility or severity of disease. Here, we analyzed exonic mutations and intronic mutations described in the COL4A1 gene using bioinformatics programs and identified candidate mutations that may alter the normal splicing pattern through a minigene system. We identified seven variants that induce splicing alterations by disrupting normal splice sites, creating new ones, or altering splice regulatory elements. These mutations are predicted to impact protein function. Our results help in the correct molecular characterization of variants in COL4A1 and may help develop more personalized treatment options.

2.
Hum Mutat ; 42(9): 1153-1164, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34157794

RESUMO

Primary distal renal tubular acidosis (dRTA) is a rare tubular disease associated with variants in SLC4A1, ATP6V0A4, ATP6V1B1, FOXⅠ1, or WDR72 genes. Currently, there is growing evidence that all types of exonic variants can alter splicing regulatory elements, affecting the precursor messenger RNA (pre-mRNA) splicing process. This study was to determine the consequences of variants associated with dRTA on pre-mRNA splicing combined with predictive bioinformatics tools and minigene assay. As a result, among the 15 candidate variants, 7 variants distributed in SLC4A1 (c.1765C>T, p.Arg589Cys), ATP6V1B1 (c.368G>T, p.Gly123Val; c.370C>T, p.Arg124Trp; c.484G>T, p.Glu162* and c.1102G>A, p.Glu368Lys) and ATP6V0A4 genes (c.322C>T, p.Gln108* and c.1572G>A, p.Pro524Pro) were identified to result in complete or incomplete exon skipping by either disruption of exonic splicing enhancers (ESEs) and generation of exonic splicing silencers, or interference with the recognition of the classic splicing site, or both. To our knowledge, this is the first study on pre-mRNA splicing of exonic variants in the dRTA-related genes. These results highlight the importance of assessing the effects of exonic variants at the mRNA level and suggest that minigene analysis is an effective tool for evaluating the effects of splicing on variants in vitro.


Assuntos
Acidose Tubular Renal , ATPases Vacuolares Próton-Translocadoras , Acidose Tubular Renal/genética , Proteína 1 de Troca de Ânion do Eritrócito/genética , Éxons/genética , Fatores de Transcrição Forkhead/genética , Humanos , Proteínas/genética , Splicing de RNA/genética , ATPases Vacuolares Próton-Translocadoras/genética
3.
Int J Mol Sci ; 21(24)2020 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-33322357

RESUMO

BACKGROUND: Cutaneous malignant melanoma (CMM) is one of the most common skin cancers worldwide. CMM pathogenesis involves genetic and environmental factors. Recent studies have led to the identification of new genes involved in CMM susceptibility: beyond CDKN2A and CDK4, BAP1, POT1, and MITF were recently identified as potential high-risk melanoma susceptibility genes. OBJECTIVE: This study is aimed to evaluate the genetic predisposition to CMM in patients from central Italy. METHODS: From 1998 to 2017, genetic testing was performed in 888 cases with multiple primary melanoma and/or familial melanoma. Genetic analyses included the sequencing CDKN2A, CDK4, BAP1, POT1, and MITF in 202 cases, and of only CDKN2A and CDK4 codon 24 in 686 patients. By the evaluation of the personal and familial history, patients were divided in two clinical categories: "low significance" and "high significance" cases. RESULTS: 128 patients (72% belonging to the "high significance" category, 28% belonging to the "low significance" category) were found to carry a DNA change defined as pathogenic, likely pathogenic, variant of unknown significance (VUS)-favoring pathogenic or VUS. CONCLUSIONS: It is important to verify the genetic predisposition in CMM patients for an early diagnosis of further melanomas and/or other tumors associated with the characterized genotype.


Assuntos
Predisposição Genética para Doença/genética , Melanoma/genética , Melanoma/metabolismo , Adulto , Idoso , Quinase 4 Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Humanos , Itália , Masculino , Fator de Transcrição Associado à Microftalmia/genética , Pessoa de Meia-Idade , Estudos Retrospectivos , Complexo Shelterina , Proteínas de Ligação a Telômeros/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética
4.
Dermatol Online J ; 26(8)2020 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-32941720

RESUMO

Pancreatic cancer-melanoma syndrome (PCMS) is an inherited condition in which mutation carriers have an increased risk of malignant melanoma and/or pancreatic cancer. About 30% of PCMS cases carry mutations in CDKN2A. This gene encodes several protein isoforms, one of which, known as p16, regulates the cell-cycle by interacting with CDK4/CDK6 kinases and with several non-CDK proteins. Herein, we report on a novel CDKN2A germline in-frame deletion (c.52_57delACGGCC) found in an Italian family with PCMS. By segregation analysis, the c.52_57delACGGCC was proven to segregate in kindred with cutaneous melanoma (CM), in kindred with CM and pancreatic cancer, and in a single case presenting only with pancreatic cancer. In the literature, duplication mapping in the same genic region has been already reported at the germline level in several unrelated CM cases as a variant of unknown clinical significance. A computational approach for studying the effect of mutational changes over p16 protein structure showed that both the deletion and the duplication of the c.52_57 nucleotides result in protein misfolding and loss of interactors' binding. In conclusion, the present results argue that the quantitative alteration of nucleotides c.52_57 has a pathogenic role in p16 function and that the c.52_57delACGGCC is associated with PCMS.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Mutação em Linhagem Germinativa , Melanoma/genética , Síndromes Neoplásicas Hereditárias/genética , Neoplasias Pancreáticas/genética , Inibidor p16 de Quinase Dependente de Ciclina/ultraestrutura , Feminino , Deleção de Genes , Humanos , Masculino , Melanoma/etiologia , Pessoa de Meia-Idade , Síndromes Neoplásicas Hereditárias/etiologia , Neoplasias Pancreáticas/etiologia , Linhagem , Estrutura Quaternária de Proteína
5.
BMC Med Genet ; 19(1): 173, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30236073

RESUMO

BACKGROUND: Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by hamartomas in any organ systems. Mutations in the TSC1 or TSC2 gene lead to the dysfunction of hamartin or tuberin proteins, which cause tuberous sclerosis complex. CASE PRESENTATION: We describe the clinical characteristics of patients from a Chinese family with tuberous sclerosis complex and analyze the functional consequences of their causal genetic mutations. A novel heterozygous mutation (c.3610G > A) at the last nucleotide of exon 29 in TSC2 was identified. On the protein level, this variant was presumed to be a missense mutation (p.Gly1204Arg). However, the splicing assay revealed that this mutation also leads to the whole TSC2 exon 29 skipping, besides the wild-type transcript. The mutated transcript results in an in-frame deletion of 71 amino acids (p.Gly1133_Thr1203del) and its ratio with the normal splice product is of about 44:56. CONCLUSIONS: The novel c.3610G > A TSC2 mutation was identified in association with tuberous sclerosis complex. And it was proven to code both for a missense-carrying transcript (56%), and for an isoform lacking exon 29 (44%).


Assuntos
Sequência de Bases , Mutação de Sentido Incorreto , Splicing de RNA , RNA Mensageiro/genética , Deleção de Sequência , Proteína 2 do Complexo Esclerose Tuberosa/genética , Esclerose Tuberosa/genética , Povo Asiático , Éxons , Feminino , Genes Dominantes , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Pessoa de Meia-Idade , Linhagem , Esclerose Tuberosa/diagnóstico , Esclerose Tuberosa/etnologia , Esclerose Tuberosa/patologia , Proteína 2 do Complexo Esclerose Tuberosa/deficiência
6.
BMC Nephrol ; 19(1): 181, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30005619

RESUMO

BACKGROUND: Sixty mutations of claudin 16 coding gene have been reported in familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC) patients. Recent investigations revealed that a highly conserved glycine-leucine-tryptophan (115G-L-W117) motif in the first extracellular segment (ESC1) of claudin 16 might be essential for stabilization of the appropriately folded ECS1 structure and conservation of normal claudin 16 function. However, neither missense nor nonsense mutation has ever been described in this motif. Our study aimed at identifying mutations in a Chinese patient with FHHNC and exploring the association between genotype and phenotype. CASE PRESENTATION: A 33-year-old female presented with 4 years history of recurrent acute pyelonephritis without other notable past medical history. Her healthy parents, who aged 56 and 53 respectively, were second cousins, and her only sibling died from renal failure without definite cause at age 25. Renal ultrasound imaging demonstrated atrophic kidneys and bilateral nephrocalcinosis. The laboratory workup revealed impaired renal function (Stage CKD IV), hypocalcemia and mild hypomagnesemia, accompanied with marked renal loss of magnesium and hypercalciuria. During the follow-up, treatment with calcitriol and calcium but not with magnesium was difficult to achieve normal serum calcium levels, whereas her serum magnesium concentration fluctuated within normal ranges. In the end, the patient unavoidably reached ESRD at 36 years old. The clinical features and family history suggested the diagnosis of FHHNC. To make a definite diagnosis, we use whole-exome sequencing to identify the disease-causing mutations and Sanger sequencing to confirm the mutation co-segregation in the family. As a result, a novel homozygous mutation (c.346C > G, p.Leu116Val) in 115G-L-W117 motif of claudin 16 was identified. Her parents, grandmother and one of her cousins carried heterozygous p.Leu116Val, whereas 200 unrelated controls did not carry this mutation. CONCLUSIONS: We described a delayed diagnosis patient with FHHNC in the Chinese population and identified a novel missense mutation in the highly conserved 115G-L-W117 motif of claudin 16 for the first time. According to the reported data and the information deduced from 3D modeling, we speculate that this mutation probably reserve partial residual function which might be related to the slight phenotype of the patient.


Assuntos
Povo Asiático/genética , Claudinas/genética , Códon sem Sentido/genética , Hipercalciúria/genética , Deficiência de Magnésio/genética , Nefrocalcinose/genética , Adulto , Claudinas/química , Diagnóstico Tardio , Feminino , Humanos , Hipercalciúria/complicações , Hipercalciúria/diagnóstico , Leucina/genética , Deficiência de Magnésio/complicações , Deficiência de Magnésio/diagnóstico , Nefrocalcinose/complicações , Nefrocalcinose/diagnóstico , Linhagem , Estrutura Secundária de Proteína
7.
Cell Physiol Biochem ; 44(4): 1559-1577, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29197877

RESUMO

BACKGROUND/AIMS: Truncating LMNA gene mutations occur in many inherited cardiomyopathy cases, but the molecular mechanisms involved in the disease they cause have not yet been systematically investigated. Here, we studied a novel frameshift LMNA variant (p.D243Gfs*4) identified in three members of an Italian family co-segregating with a severe form of cardiomyopathy with conduction defects. METHODS: HEK293 cells and HL-1 cardiomyocytes were transiently transfected with either Lamin A or D243Gfs*4 tagged with GFP (or mCherry). D243Gfs*4 expression, cellular localization and its effects on diverse cellular mechanisms were evaluated with western blotting, laser-scanning confocal microscopy and video-imaging analysis in single cells. RESULTS: When expressed in HEK293 cells, GFP- (or mCherry)-tagged LMNA D243Gfs*4 colocalized with calnexin within the ER. ER mislocalization of LMNA D243Gfs*4 did not significantly induce ER stress response, abnormal Ca2+ handling and apoptosis when compared with HEK293 cells expressing another truncated mutant of LMNA (R321X) which similarly accumulates within the ER. Of note, HEK293-LMNA D243Gfs*4 cells showed a significant reduction of connexin 43 (CX43) expression level, which was completely rescued by activation of the WNT/ß-catenin signaling pathway. When expressed in HL-1 cardiomyocytes, D243Gfs*4 significantly impaired the spontaneous Ca2+ oscillations recorded in these cells as result of propagation of the depolarizing waves through the gap junctions between non-transfected cells surrounding a cell harboring the mutation. Furthermore, mCh-D243Gfs*4 HL-1 cardiomyocytes showed reduced CX43-dependent Lucifer Yellow (LY) loading and propagation. Of note, activation of ß-catenin rescued both LY loading and LMNA D243Gfs*4 -HL-1 cells spontaneous activity propagation. CONCLUSION: Overall, the present results clearly indicate the involvement of the aberrant CX43 expression/activity as a pathogenic mechanism for the conduction defects associated to this LMNA truncating alteration.


Assuntos
Doença do Sistema de Condução Cardíaco/genética , Cardiomiopatias/genética , Lamina Tipo A/genética , Apoptose , Sequência de Bases , Cálcio/metabolismo , Calnexina/metabolismo , Doença do Sistema de Condução Cardíaco/complicações , Doença do Sistema de Condução Cardíaco/patologia , Cardiomiopatias/complicações , Cardiomiopatias/patologia , Linhagem Celular , Conexina 43 , Retículo Endoplasmático/metabolismo , Feminino , Junções Comunicantes/metabolismo , Células HEK293 , Humanos , Lamina Tipo A/metabolismo , Repetições de Microssatélites/genética , Microscopia Confocal , Pessoa de Meia-Idade , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Linhagem , Polimorfismo de Nucleotídeo Único , Imagem com Lapso de Tempo , Via de Sinalização Wnt
9.
Birth Defects Res A Clin Mol Teratol ; 106(1): 61-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26663529

RESUMO

BACKGROUND: Campomelic dysplasia and acampomelic campomelic dysplasia (ACD) are allelic disorders due to heterozygous mutations in or around SOX9. Translocations and deletions involving the SOX9 5' regulatory region are rare causes of these disorders, as well as Pierre Robin sequence (PRS) and 46,XY gonadal dysgenesis. Genotype-phenotype correlations are not straightforward due to the complex epigenetic regulation of SOX9 expression during development. METHODS: We report a three-generation pedigree with a novel ∼1 Mb deletion upstream of SOX9 and including KCNJ2 and KCNJ16, and ascertained for dominant transmission of PRS. RESULTS: Further characterization of the family identified subtle appendicular anomalies and a variable constellation of axial skeletal features evocative of ACD in several members. Affected males showed learning disability. CONCLUSION: The identified deletion was smaller than all other chromosome rearrangements associated with ACD. Comparison with other reported translocations and deletions involving this region allowed further refining of genotype-phenotype correlations and an update of the smallest regions of overlap associated with the different phenotypes. Intrafamilial variability in this pedigree suggests a phenotypic continuity between ACD and PRS in patients carrying mutations in the SOX9 5' regulatory region.


Assuntos
Displasia Campomélica/genética , Deficiência Intelectual/genética , Síndrome de Pierre Robin/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Fatores de Transcrição SOX9/genética , Adulto , Sequência de Bases , Displasia Campomélica/diagnóstico , Displasia Campomélica/patologia , Feminino , Expressão Gênica , Genes Dominantes , Estudos de Associação Genética , Variação Genética , Humanos , Lactente , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/patologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Síndrome de Pierre Robin/diagnóstico , Síndrome de Pierre Robin/patologia , Canais de Potássio Corretores do Fluxo de Internalização/deficiência , Deleção de Sequência
10.
Am J Nephrol ; 42(1): 78-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26340091

RESUMO

BACKGROUND: Twenty-six HOGA1 mutations have been reported in primary hyperoxaluria (PH) type 3 (PH3) patients with c.700 + 5G>T accounting for about 50% of the total alleles. However, PH3 has never been described in Asians. METHODS: A Chinese child with early-onset nephrolithiasis was suspected of having PH. We searched for AGXT, GRHPR and HOGA1 gene mutations in this patient and his parents. All coding regions, including intron-exon boundaries, were analyzed using PCR followed by direct sequence analysis. RESULTS: Two heterozygous mutations not previously described in the literature about HOGA1 were identified (compound heterozygous). One mutation was a successive 2 bp substitution at the last nucleotide of exon 6 and at the first nucleotide of intron 6, respectively (c.834_834 + 1GG>TT), while the other one was a guanine to adenine substitution of the last nucleotide of exon 6 (c.834G>A). Direct sequencing analysis failed to find these mutations in 100 unrelated healthy subjects and the functional role on splicing of both variants found in this study was confirmed by a minigene assay based on the pSPL3 exon trapping vector. In addition, we found a SNP in this family (c.715G>A, p.V239I). There were no mutations detected in AGXT and GRHPR. CONCLUSION: Two novel HOGA1 mutations were identified in association with PH3. This is the first description and investigation on mutant gene analysis of PH3 in an Asian.


Assuntos
Hiperoxalúria Primária/genética , Nefrolitíase/genética , Oxo-Ácido-Liases/genética , Cálculos Urinários/etiologia , Pré-Escolar , China , Análise Mutacional de DNA , Humanos , Hiperoxalúria Primária/complicações , Masculino , Mutação , Radiografia , Cálculos Urinários/diagnóstico por imagem
11.
Am J Hum Genet ; 87(2): 265-73, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20691405

RESUMO

Ectodermal dysplasias form a large disease family with more than 200 members. The combination of hair and tooth abnormalities, alopecia, and cutaneous syndactyly is characteristic of ectodermal dysplasia-syndactyly syndrome (EDSS). We used a homozygosity mapping approach to map the EDSS locus to 1q23 in a consanguineous Algerian family. By candidate gene analysis, we identified a homozygous mutation in the PVRL4 gene that not only evoked an amino acid change but also led to exon skipping. In an Italian family with two siblings affected by EDSS, we further detected a missense and a frameshift mutation. PVRL4 encodes for nectin-4, a cell adhesion molecule mainly implicated in the formation of cadherin-based adherens junctions. We demonstrated high nectin-4 expression in hair follicle structures, as well as in the separating digits of murine embryos, the tissues mainly affected by the EDSS phenotype. In patient keratinocytes, mutated nectin-4 lost its capability to bind nectin-1. Additionally, in discrete structures of the hair follicle, we found alterations of the membrane localization of nectin-afadin and cadherin-catenin complexes, which are essential for adherens junction formation, and we found reorganization of actin cytoskeleton. Together with cleft lip and/or palate ectodermal dysplasia (CLPED1, or Zlotogora-Ogur syndrome) due to an impaired function of nectin-1, EDSS is the second known "nectinopathy" caused by mutations in a nectin adhesion molecule.


Assuntos
Moléculas de Adesão Celular/genética , Displasia Ectodérmica/complicações , Displasia Ectodérmica/genética , Mutação/genética , Sindactilia/complicações , Sindactilia/genética , Anormalidades Múltiplas/genética , Adulto , Sequência de Aminoácidos , Animais , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/metabolismo , Criança , Extremidades/embriologia , Família , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Cabelo/patologia , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Pele/patologia , Síndrome
13.
Genes (Basel) ; 14(2)2023 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-36833228

RESUMO

Asperger syndrome (AS) is a pervasive developmental disorder characterized by general impairment in socialization, stereotypical behavior, defective adaptation to the social context usually without intellectual disability, and some high functioning areas related to memory and mathematics. Clinical criteria are not well defined and the etiology is heterogeneous and mostly unknown. Like in typical autism spectrum disorders (ASD), the genetic background plays a crucial role in AS, and often an almost mendelian segregation can be observed in some families. We performed a whole exome sequencing (WES) in three relatives of a family with vertical transmission of AS-ASD to identify variants in candidate genes segregating with the phenotype. Variant p.(Cys834Ser) in the RADX gene was the only one segregating among all the affected family members. This gene encodes a single-strand DNA binding factor, which mediates the recruitment of genome maintenance proteins to sites of replication stress. Replication stress and genome instability have been reported recently in neural progenitor cells derived from ASD patients, leading to a disruption of long neural genes involved in cell-cell adhesion and migration. We propose RADX as a new gene that when mutated could represent a predisposing factor to AS-ASD.


Assuntos
Síndrome de Asperger , Transtorno do Espectro Autista , Deficiência Intelectual , Humanos , Transtorno do Espectro Autista/genética , Proteínas/genética , Deficiência Intelectual/genética , Fenótipo
14.
Mol Genet Genomic Med ; 11(4): e2128, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36597580

RESUMO

BACKGROUND: Gitelman syndrome (GS) is a type of salt-losing tubular disease, most of which is caused by SLC12A3 gene variants, and missense variants account for the majority. Recently, the phenomenon of exon skipping, in which variants disrupt normal pre-mRNA splicing, has been related to a variety of diseases. Therefore, we hypothesize that a certain proportion of SLC12A3 variants can result in disease via interfering with the normal splicing process. METHODS: We analyzed 342 previously presumed SLC12A3 missense variants using bioinformatics programs and identified candidate variants that may alter the splicing of pre-mRNA through minigene assays. RESULTS: Our study revealed that, among ten candidate variants, six variants (c.602G>A, c.602G>T, c.1667C>T, c.1925G>A, c.2548G>C, and c.2549G>C) led to complete or incomplete exon skipping by affecting exonic splicing regulatory elements and/or disturbing canonical splice sites. CONCLUSION: It is worth mentioning that this is the largest study on pre-mRNA splicing of SLC12A3 exonic variants. In addition, our study emphasizes the importance of detecting splicing function at the mRNA level in GS and indicates that minigene analysis is a valuable tool for splicing functional assays of variants in vitro.


Assuntos
Síndrome de Gitelman , Membro 3 da Família 12 de Carreador de Soluto , Humanos , Éxons , Síndrome de Gitelman/genética , Mutação de Sentido Incorreto , Precursores de RNA/genética , Splicing de RNA , Membro 3 da Família 12 de Carreador de Soluto/genética
15.
Eur J Hum Genet ; 31(4): 474-478, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36529819

RESUMO

Constitutional heterozygous mutations in CHEK2 gene have been associated with hereditary cancer risk. To date, only a few homozygous CHEK2 mutations have been reported in families with cancer susceptibility. Here, we report two unrelated individuals with a personal and familial cancer history in whom biallelic CHEK2 alterations were identified. The first case resulted homozygous for the CHEK2 c.793-1 G > A (p.Asp265Thrfs*10) variant, and the second one was found to be compound heterozygous for the c.1100delC (p.Thr367Metfs*15) and the c.1312 G > T (p.Asp438Tyr) variants. Multiple cytogenetic anomalies were demonstrated on peripheral lymphocytes of both patients. A literature revision showed that a single other CHEK2 homozygous variant was previously associated to a constitutional randomly occurring multi-translocation karyotype from peripheral blood in humans. We hypothesize that, at least some biallelic CHEK2 mutations might be associated with a novel disorder, further expanding the group of chromosome instability syndromes. Additional studies on larger cohorts are needed to confirm if chromosomal instability could represent a marker for CHEK2 constitutionally mutated recessive genotypes, and to investigate the cancer risk and the occurrence of other anomalies typically observed in chromosome instability syndromes.


Assuntos
Neoplasias da Mama , Proteínas Serina-Treonina Quinases , Humanos , Feminino , Proteínas Serina-Treonina Quinases/genética , Predisposição Genética para Doença , Quinase do Ponto de Checagem 2/genética , Mutação , Genótipo , Instabilidade Cromossômica
16.
Front Neurosci ; 17: 1304080, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38249578

RESUMO

Introduction: Folliculin, encoded by FLCN gene, plays a role in the mTORC1 autophagy cascade and its alterations are responsible for the Birt-Hogg-Dubé (BHD) syndrome, characterized by follicle hamartomas, kidney tumors and pneumothorax. Patient and results: We report a 74-years-old woman diagnosed with dementia and carrying a FLCN alteration in absence of any sign of BHD. She also carried an alteration of MAT1A gene, which is also implicated in the regulation of mTORC1. Discussion: The MAT1A variant could have prevented the development of a FLCN-related oncological phenotype. Conversely, our patient presented with dementia that, to date, has yet to be documented in BHD. Folliculin belongs to the DENN family proteins, which includes C9orf72 whose alteration has been associated to neurodegeneration. The folliculin perturbation could affect the C9orf72 activity and our patient could represent the first human model of a relationship between FLCN and C9orf72 across the path of autophagy.

17.
Genes (Basel) ; 13(7)2022 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-35886051

RESUMO

Choroideremia is an X-linked recessive condition presenting in males, with progressive degeneration of retinal and choroidal tissues leading to progressive visual loss. Its pathological mechanism is due to alterations in the CHM gene that encodes for REP1, a protein required for prenylation of Rab by the Rab geranylgeranyl transferase (RGGT). Even though female carriers are predicted to be not affected by the disease, a wide phenotypic spectrum ranging from mild to severe cases has been reported in women. The reason why Choroideremia manifests in female carriers remains elusive. While X chromosome inactivation (XCI) skewing has been proposed as a leading putative mechanism, emerging evidence has shown that CHM could variably escape from XCI. We described a family with an initial clinical suspicion of Retinitis Pigmentosa in which a novel CHM pathogenic splicing variant was found by exome sequencing. The variant, initially found in the 63-year-old female presenting with impaired visual acuity and severe retinal degeneration, segregated in the 31-year-old daughter and the 37-year-old son, both presenting with fundus anomalies. mRNA studies revealed a shorter in-frame CHM isoform lacking exon 10. Molecular modeling of the ternary REP1/Rab/RGGT protein complex predicted significant impairing of REP1/Rab binding without alteration of REP1/RGGT interaction. We suggest that, in our female cases, the biallelic expression of CHM may have led to the production of both the mutant and wild type REP1. The mutant isoform, sequestrating RGGT, could reduce its available amount for Rab prenylation, thus exerting a dominant-negative effect. If confirmed with further studies and in large cohorts of female carriers, the here proposed molecular mechanism could help to explain the complexity of manifestation of Choroideremia in females.


Assuntos
Coroideremia , Degeneração Retiniana , Retinose Pigmentar , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Coroideremia/diagnóstico , Coroideremia/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Retina/patologia , Degeneração Retiniana/genética , Retinose Pigmentar/metabolismo
18.
Genes (Basel) ; 13(12)2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36553533

RESUMO

As a consequence of the implementation of NGS technologies, the diagnostic yield of neurodevelopmental disorders has dramatically increased during the past two decades. Among neurodevelopmental genes, transcription-related genes and chromatin remodeling genes are the most represented category of disease-causing genes. Indeed, the term "chromatinopathies" is now widely used to describe epigenetic disorders caused by mutations in these genes. We hereby describe a twenty-seven-year-old female patient diagnosed with moderate intellectual disability comorbid with other neuropsychiatric and behavioral issues carrying a de novo heterozygous stop variant in the KDM5C gene (NM_004187.5: c. 3847G>T, p.Glu1283*), encoding a histone demethylase that specifically acts on the H3K4 lysines. The gene is located on the X chromosome and has been associated with Claes-Jensen-type intellectual disability, an X-linked syndromic disorder. We discuss our case in relation to previously reported affected females harboring pathogenic mutations in the KDM5C gene with the objective of delineating genotype-phenotype correlations and further defining a common recognizable phenotype. We also highlight the importance of reverse phenotyping in relation to whole-exome sequencing results.


Assuntos
Deficiência Intelectual , Transtornos do Neurodesenvolvimento , Feminino , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/diagnóstico , Histona Desmetilases/genética , Mutação , Transtornos do Neurodesenvolvimento/diagnóstico , Transtornos do Neurodesenvolvimento/genética , Estudos de Associação Genética
19.
Genes (Basel) ; 13(12)2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36553625

RESUMO

In 2018, a new clinical subtype, caused by biallelic variants in the AEBP1 gene, encoding the ACLP protein, was added to the current nosological classification of the Ehlers-Danlos Syndromes (EDS). This new phenotype, provisionally termed EDS classical-like type 2 (clEDS2), has not yet been fully characterized, as only nine cases have been reported to date. Here we describe a patient, homozygous for a novel AEBP1 pathogenic variant (NM_001129.5 c.2123_2124delTG (p.Val708AlafsTer5)), whose phenotype is reminiscent of classical EDS but also includes previously unreported multiple congenital malformations. Furthermore, we briefly summarize the current principal clinical manifestations of clEDS2 and the molecular evidence surrounding the role of AEBP1 in the context of extracellular matrix homeostasis and connective tissue development. Although a different coexisting etiology for the multiple congenital malformations of our patient cannot be formally excluded, the emerging role of ACLP in TGF-ß and WNT pathways may explain their occurrence and the phenotypical variability of clEDS2.


Assuntos
Síndrome de Ehlers-Danlos , Humanos , Mutação , Síndrome de Ehlers-Danlos/patologia , Matriz Extracelular/genética , Fenótipo , Homozigoto , Carboxipeptidases/genética , Proteínas Repressoras/genética
20.
Neurogenetics ; 12(3): 233-40, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21365283

RESUMO

We report the detailed clinical presentation and molecular features of a spinal neurofibromatosis familial case where a 40-year-old woman, presenting with multiple bilateral spinal neurofibromas and no other clinical feature of neurofibromatosis type 1 (NF1), inherited a paternal large multiexonic deletion (c.5944-?_7126+?del) which resulted in NF1 gene haploinsufficiency at the RNA level. In the clinically unaffected 73-year-old father, spinal cord MRI disclosed bilateral and symmetrical hypertrophy of spinal lumbosacral roots. Our study widens the phenotypic and mutational spectrum of NF1 and illustrates the difficulties of counseling patients with border-line or atypical presentation of this disorder.


Assuntos
Deleção de Genes , Genes da Neurofibromatose 1 , Neurofibromatoses/genética , Nervos Espinhais/patologia , Adulto , Idoso , Análise Mutacional de DNA , Éxons , Família , Feminino , Humanos , Masculino , Neurofibromatoses/diagnóstico , Neurofibromatoses/diagnóstico por imagem , Neurofibromatoses/patologia , Linhagem , Radiografia , Nervos Espinhais/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa