Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Hum Genomics ; 18(1): 103, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39285482

RESUMO

BACKGROUND: Deletion or duplication in the DMD gene is one of the most common causes of Duchenne and Becker muscular dystrophy (DMD/BMD). However, the pathogenicity of complex rearrangements involving DMD, especially segmental duplications with unknown breakpoints, is not well understood. This study aimed to evaluate the structure, pattern, and potential impact of rearrangements involving DMD duplication. METHODS: Two families with DMD segmental duplications exhibiting phenotypical differences were recruited. Optical genome mapping (OGM) was used to explore the cryptic pattern of the rearrangements. Breakpoints were validated using long-range polymerase chain reaction combined with next-generation sequencing and Sanger sequencing. RESULTS: A multi-copy duplication involving exons 64-79 of DMD was identified in Family A without obvious clinical symptoms. Family B exhibited typical DMD neuromuscular manifestations and presented a duplication involving exons 10-13 of DMD. The rearrangement in Family A involved complex in-cis tandem repeats shown by OGM but retained a complete copy (reading frame) of DMD inferred from breakpoint validation. A reversed insertion with a segmental repeat was identified in Family B by OGM, which was predicted to disrupt the normal structure and reading frame of DMD after confirming the breakpoints. CONCLUSIONS: Validating breakpoint and rearrangement pattern is crucial for the functional annotation and pathogenic classification of genomic structural variations. OGM provides valuable insights into etiological analysis of DMD/BMD and enhances our understanding for cryptic effects of complex rearrangements.


Assuntos
Distrofina , Éxons , Distrofia Muscular de Duchenne , Linhagem , Fenótipo , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Distrofina/genética , Masculino , Éxons/genética , Feminino , Mapeamento Cromossômico , Rearranjo Gênico/genética , Criança , Duplicações Segmentares Genômicas/genética , Sequenciamento de Nucleotídeos em Larga Escala , Duplicação Gênica/genética , Adolescente
2.
J Thromb Haemost ; 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39260745

RESUMO

BACKGROUND: Targeted long-read sequencing (LRS) is expected to comprehensively analyse diverse complex variants in haemophilia A (HA) and B (HB), caused by the F8 and F9 genes, respectively. However, its clinical applicability still requires extensive validation. OBJECTIVES: To evaluate the clinical applicability of targeted LRS-based analysis, compared with routine PCR-based methods. METHODS: Gene variants of retrieved subjects were retrospectively and prospectively analysed. Whole-genome sequencing (WGS) was performed to further analyse undiagnosed cases. Breakpoints of novel genomic rearrangements were mapped and validated using long-distance-PCR and long-range-PCR combined with sequencing. RESULTS: Totally, 122 subjects were retrieved. In retrospective analysis of the 90 HA cases, HA-LRS assay showed consistent results in 84 cases compared with routine methods, and characterized six large deletions with their exact breakpoints confirmed by further validation in six cases (routine methods only presented failure in amplifying the involved exons). In prospective analysis of the 21 HA subjects, 20 variants of F8 were identified in 20 cases. For the remaining HA patient, no duplication/deletion or SNV/InDel was found, but a potential recombination involving exons 14 and 21 of F8 was observed by LRS. WGS analysis and further verification defined a 30,478bp tandem repeat involving exons 14-21 of F8. Among the 11 HB patients, HB-LRS analysis detected 11 SNVs/InDels in F9, consistent with routine methods. CONCLUSIONS: Targeted LRS-based analysis is efficient and comprehensive to identify SNVs/InDels and genomic rearrangements of haemophilia genes, especially we first expanding the panel including F9. However, further investigation for complex gross rearrangement is still essential.

3.
BMC Med Genomics ; 17(1): 135, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773466

RESUMO

BACKGROUND: Thoracic aortic aneurysm/dissection (TAAD) and patent ductus arteriosus (PDA) are serious autosomal-dominant diseases affecting the cardiovascular system. They are mainly caused by variants in the MYH11 gene, which encodes the heavy chain of myosin 11. The aim of this study was to evaluate the genotype-phenotype correlation of MYH11 from a distinctive perspective based on a pair of monozygotic twins. METHODS: The detailed phenotypic characteristics of the monozygotic twins from the early fetal stage to the infancy stage were traced and compared with each other and with those of previously documented cases. Whole-exome and Sanger sequencing techniques were used to identify and validate the candidate variants, facilitating the analysis of the genotype-phenotype correlation of MYH11. RESULTS: The monozygotic twins were premature and presented with PDA, pulmonary hypoplasia, and pulmonary hypertension. The proband developed heart and brain abnormalities during the fetal stage and died at 18 days after birth, whereas his sibling was discharged after being cured and developed normally post follow-up. A novel variant c.766 A > G p. (Ile256Val) in MYH11 (NM_002474.2) was identified in the monozygotic twins and classified as a likely pathogenic variant according to the American College of Medical Genetics/Association for Molecular Pathology guidelines. Reviewing the reported cases (n = 102) showed that the penetrance of MYH11 was 82.35%, and the most common feature was TAAD (41.18%), followed by PDA (22.55%), compound TAAD and PDA (9.80%), and other vascular abnormalities (8.82%). The constituent ratios of null variants among the cases with TAAD (8.60%), PDA (43.8%), or compound TAAD and PDA (28.6%) were significantly different (P = 0.01). Further pairwise comparison of the ratios among these groups showed that there were significant differences between the TAAD and PDA groups (P = 0.006). CONCLUSION: This study expands the mutational spectrum of MYH11 and provides new insights into the genotype-phenotype correlation of MYH11 based on the monozygotic twins with variable clinical features and outcomes, indicating that cryptic modifiers and complex mechanisms beside the genetic variants may be involved in the condition.


Assuntos
Estudos de Associação Genética , Cadeias Pesadas de Miosina , Gêmeos Monozigóticos , Humanos , Gêmeos Monozigóticos/genética , Cadeias Pesadas de Miosina/genética , Masculino , Recém-Nascido , Fenótipo , Miosinas Cardíacas/genética , Aneurisma da Aorta Torácica/genética , Permeabilidade do Canal Arterial/genética , Feminino , Mutação , Dissecção Aórtica/genética
4.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(2): 234-238, 2024 Feb 10.
Artigo em Chinês | MEDLINE | ID: mdl-38311566

RESUMO

OBJECTIVE: To explore the clinical features and genetic etiology for a child with developmental delay, impaired growth, facial dysmorphism, and axonal neuropathy (DIGFAN). METHODS: A child who was admitted to the Second Affiliated Hospital of Guangxi Medical University on March 22, 2021 was selected the study subject. Clinical data of the child was collected. Following extraction of genomic DNA, the child and his parents were subjected to whole exome sequencing (WES), and candidate variant was verified by Sanger sequencing and bioinformatic analysis. RESULTS: The child, a 10-year-and-9-month-old boy, had manifested with short stature, intellectual disability, delayed speech, motor and language development, and facial dysmorphism. WES and Sanger sequencing revealed that he has harbored a novel de novo c.800T>C (p.Leu267Pro) variant of the MORC2 gene. The Leucine at position 267, which is highly conserved among various species, is located in the S5 domain of ribosome protein in the ATPase binding region of MORC2. And the Leu267Pro may affect the function of MORC2 by altering the spatial conformation and activity of ATPase. Based on the guidelines from the American College of Medical Genetics and Genomics, the c.800T>C variant was classified as likely pathogenic (PS2+PM2_Supporting+PP2+PP3). CONCLUSION: The MORC2: c.800T>C (p.Leu267Pro) variant probably underlay the pathogenesis of DIGFAN syndrome in this child.


Assuntos
Adenosina Trifosfatases , Nanismo , Criança , Humanos , Masculino , China , Biologia Computacional , Nanismo/genética , Genômica , Mutação , Síndrome , Fatores de Transcrição
5.
Front Genet ; 14: 1246712, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38174045

RESUMO

Vascular Ehlers-Danlos syndrome (vEDS), the most severe type of Ehlers-Danlos syndrome, is caused by an autosomal-dominant defect in the COL3A1 gene. In this report, we describe the clinical history, specific phenotype, and genetic diagnosis of a man who died of vEDS. The precise diagnosis of this case using whole-exome sequencing provided solid evidence for the cause of death, demonstrating the practical value of genetic counseling and analysis. Early diagnosis for the proband's son, who was also affected by vEDS, revealed initial complications of vEDS in early childhood, which have rarely been reported. We also reviewed the literature on COL3A1 missense mutations and related phenotypes. We identified an association between digestion tract events and non-glycine missense variants, which disproves a previous hypothesis regarding the genotype-phenotype correlation of vEDS. Our results demonstrate the necessity of offering comprehensive genetic testing for every patient suspected of having vEDS.

6.
Front Genet ; 13: 1056127, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36506332

RESUMO

Cornelia de Lange syndrome (CdLS) is an autosomal dominant or X-linked genetic disease with significant genetic heterogeneity. Variants of the NIPBL gene are responsible for CdLS in 60% of patients. Herein, we report the case of a patient with CdLS showing distinctive facial features, microcephaly, developmental delay, and growth retardation. Whole exome sequencing was performed for the patient, and a novel de novo heterozygous synonymous variant was identified in the deep region of exon 40 in the NIPBL gene (NM_133433.4: c. 6819G > T, p. Gly2273 = ). The clinical significance of the variant was uncertain according to the ACMG/AMP guidelines; however, based on in silico analysis, it was predicted to alter mRNA splicing. To validate the prediction, a reverse transcriptase-polymerase chain reaction was conducted. The variant activated a cryptic splice donor, generating a short transcript of NIPBL. A loss of 137 bp at the 3' end of NIPBL exon 40 was detected, which potentially altered the open reading frame by inserting multiple premature termination codons. Quantitative real-time PCR analysis showed that the ratio of the transcription level of the full-length transcript to that of the altered short transcript in the patient was 5:1, instead of 1:1. These findings may explain the relatively mild phenotype of the patient, regardless of the loss of function of the truncated protein due to a frameshift in the mRNA. To the best of our knowledge, this study is the first to report a synonymous variant in the deep exon regions of the NIPBL gene responsible for CdLS. The identified variant expands the mutational spectrum of the NIPBL gene. Furthermore, synonymous variations may be pathogenic, which should not be ignored in the clinical and genetic diagnosis of the disease.

7.
Mol Genet Genomic Med ; 10(4): e1901, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35235708

RESUMO

OBJECTIVE: Craniosynostosis is the result of the early fusion of cranial sutures. Syndromic craniosynostosis includes but not limited by Crouzon syndrome and Pfeiffer syndrome. Considerable phenotypic overlap exists among these syndromes and mutations in FGFR2 may cause different syndromes. This study aims to investigate the explanation of the phenotypic variability via clinical and genetic evaluation for eight patients in a large pedigree. METHODS: For each patient, comprehensive physical examination, cranial plain CT scan with three-dimensional CT reconstruction (3D-CT), and eye examinations were conducted. Whole exome sequencing was applied for genetic diagnosis of the proband. Variants were analyzed and interpreted following the ACMG/AMP guidelines. Sanger sequencing was performed to reveal genotypes of all the family members. RESULTS: A pathogenic variant in the FGFR2 gene, c.833G > T (p.C278F), was identified and proved to be co-segregate with the disease. Some symptoms of head, hearing, vision, mouth, teeth expressed differently by affected individuals. Nonetheless, all the eight patients manifested core symptoms of Crouzon syndrome without abnormality in the limbs, which could exclude diagnosis of Pfeiffer syndrome. CONCLUSION: We have established clinical and genetic diagnosis of Crouzon syndrome for eight patients in a five-generation Chinese family. Variability of clinical features among these familial patients was slighter than that in previously reported sporadic cases.


Assuntos
Acrocefalossindactilia , Disostose Craniofacial , Craniossinostoses , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Acrocefalossindactilia/genética , Variação Biológica da População , Disostose Craniofacial/genética , Craniossinostoses/genética , Humanos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Síndrome
8.
Front Cell Dev Biol ; 9: 661747, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33937263

RESUMO

PURPOSE: ROR2, a member of the ROR family, is essential for skeletal development as a receptor of Wnt5a. The present study aims to investigate the mutational spectrum of ROR2 in children with short stature and to identify the underlying molecular mechanisms. METHODS: We retrospectively analyzed clinical phenotype and whole-exome sequencing (WES) data of 426 patients with short stature through mutation screening of ROR2. We subsequently examined the changes in protein expression and subcellular location in ROR2 caused by the mutations. The mRNA expression of downstream signaling molecules of the Wnt5a-ROR2 pathway was also examined. RESULTS: We identified 12 mutations in ROR2 in 21 patients, including 10 missense, one nonsense, and one frameshift. Among all missense variants, four recurrent missense variants [c.1675G > A(p.Gly559Ser), c.2212C > T(p.Arg738Cys), c.1930G > A(p.Asp644Asn), c.2117G > A(p.Arg706Gln)] were analyzed by experiments in vitro. The c.1675G > A mutation significantly altered the expression and the cellular localization of the ROR2 protein. The c.1675G > A mutation also caused a significantly decreased expression of c-Jun. In contrast, other missense variants did not confer any disruptive effect on the biological functions of ROR2. CONCLUSION: We expanded the mutational spectrum of ROR2 in patients with short stature. Functional experiments potentially revealed a novel molecular mechanism that the c.1675G > A mutation in ROR2 might affect the expression of downstream Wnt5a-ROR2 pathway gene by disturbing the subcellular localization and expression of the protein.

9.
J Clin Lab Anal ; 34(2): e23046, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31556165

RESUMO

OBJECTIVE: To develop a technique for non-invasive prenatal diagnosis of spinal muscular atrophy and validate its performance. STUDY DESIGN: Pregnant women with 1 copy of SMN1 and male fetuses were enrolled. Seventeen women were included in test set A, and 10 of them were selected into test set B randomly and blinded. The two sets were tested independently by two different researchers blinded to fetal genotypes. Fetal DNA fractions were calculated based on the relative proportion of mapped chromosome Y sequencing reads. An algorithm was developed to decide fetal SMN1 copy numbers. RESULTS: The concordance rate with the results of MLPA testing of amniocyte DNA was 94.12% in test set A and 90% in set B. For all tests with a classifiable result, the percent of agreement with the results of MLPA testing of amniocyte DNA was up to 100% (25/25). CONCLUSION: We have developed a direct, rapid, and low-cost technique, which has a potential to be utilized for first-trimester non-invasive prenatal diagnosis and screening for spinal muscular atrophy with considerable reliability and feasibility.


Assuntos
Atrofia Muscular Espinal/diagnóstico , Teste Pré-Natal não Invasivo/métodos , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Ácidos Nucleicos Livres/sangue , Feminino , Dosagem de Genes , Haplótipos , Humanos , Masculino , Testes para Triagem do Soro Materno/métodos , Atrofia Muscular Espinal/genética , Reação em Cadeia da Polimerase/métodos , Gravidez , Segundo Trimestre da Gravidez , Proteína 1 de Sobrevivência do Neurônio Motor/sangue
10.
Mol Genet Genomic Med ; 7(9): e901, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31364285

RESUMO

BACKGROUND: Primary carnitine deficiency (PCD) is an autosomal recessive disorder of carnitine transportation caused by mutations in the SLC22A5 that lead to low serum carnitine levels and decreased intracellular carnitine accumulation. Characteristic clinical findings are hypoketotic hypoglycemia and skeletal and cardiac myopathy. OBJECTIVE: To genetically diagnose 24 unrelated Chinese patients with PCD, including 18 infants and six adults. METHODS: The entire coding region and the intron-exon boundaries of SLC22A5 were amplified by polymerase chain reaction (PCR). In silico analyses and reverse transcription-polymerase chain reaction (RT-PCR) were used to predict variants' impact on protein structure and function. RESULTS: Disease-causing variants in the SLC22A5 were identified in all 24 subjects, and c.288delG, c.495C>A, c.774_775insTCG, c.824+1G>A, and c.1418G>T were novel. The novel variant c.824+1G>A caused a truncated protein p.Phe276Tyrfs*8. CONCLUSIONS: We identified 13 variants in the SLC22A5 in 24 PCD patients, and five of these variants are novel mutations. c.824+1G>A was confirmed to alter mRNA splicing by reverse transcription PCR. Furthermore, our findings broaden the mutation spectrum of SLC22A5 and the understanding of the diverse and variable effects of PCD variants.


Assuntos
Povo Asiático/genética , Cardiomiopatias/genética , Carnitina/deficiência , Hiperamonemia/genética , Mutação INDEL , Doenças Musculares/genética , Membro 5 da Família 22 de Carreadores de Soluto/genética , Adulto , Carnitina/genética , China , Feminino , Humanos , Lactente , Masculino
11.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(4): 540-543, 2018 Aug 10.
Artigo em Chinês | MEDLINE | ID: mdl-30098252

RESUMO

OBJECTIVE: To detect mutations of the XPC (XPC complex subunit, DNA damage recognition and repair factor) gene in a family affected with xeroderma pigmentosum group C (XP-C). METHODS: The patient was subjected to next-generation sequencing and Sanger sequencing. Suspected mutations were validated by Sanger sequencing. Effect of splicing mutation was confirmed by reverse transcription-PCR (RT-PCR). RESULTS: Compound heterozygous mutations of c.2098G to T and c.2034-7_2040del were found in the XPC gene in the proband. Among these, c.2098G to T (p.G700X) is a nonsense mutation resulting in a truncated XPC protein. C.2034-7_2040del involves the -1 position, which may alter the splice donor site of the intron 11 of XPC and result in a truncated XPC protein with loss of amino acids from 940 to 679 positions. The two mutations were not detected among 100 unrelated healthy controls. CONCLUSION: Mutations of c.2098 G to T and c.2034-7_2040del of the XPC gene may lead to abnormal XPC expression and reduction or elimination of normal XPC functions, which may underlie the disease in this family.


Assuntos
Proteínas de Ligação a DNA/genética , Xeroderma Pigmentoso/genética , Códon sem Sentido , Análise Mutacional de DNA , Reparo do DNA , Humanos , Sítios de Splice de RNA
12.
Sci Rep ; 7: 44446, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28322228

RESUMO

Copy number variation (CNV) is of great significance in human evolution and disorders. Through tracing the parent-of-origin of de novo pathogenic CNVs, we are expected to investigate the relative contributions of germline genomic stability on reproductive health. In our study, short tandem repeat (STR) and single nucleotide polymorphism (SNP) were used to determine the parent-of-origin of 87 de novo pathogenic CNVs found in unrelated patients with intellectual disability (ID), developmental delay (DD) and multiple congenital anomalies (MCA). The results shown that there was a significant difference on the distribution of the parent-of-origin for different CNVs types (Chi-square test, p = 4.914 × 10-3). An apparently paternal bias existed in deletion CNVs and a maternal bias in duplication CNVs, indicating that the relative contribution of paternal germline variations is greater than that of maternal to the origin of deletions, and vice versa to the origin of duplications. By analyzing the sequences flanking the breakpoints, we also confirmed that non-allelic homologous recombination (NAHR) served as the major mechanism for the formation of recurrent CNVs whereas non-SDs-based mechanisms played a part in generating rare non-recurrent CNVs and might relate to the paternal germline bias in deletion CNVs.


Assuntos
Anormalidades Múltiplas/genética , Variações do Número de Cópias de DNA , Deficiências do Desenvolvimento/genética , Padrões de Herança , Deficiência Intelectual/genética , Polimorfismo de Nucleotídeo Único , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/fisiopatologia , Viés , Criança , Aberrações Cromossômicas , Bases de Dados Genéticas , Deficiências do Desenvolvimento/diagnóstico , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Mutação em Linhagem Germinativa , Recombinação Homóloga , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/fisiopatologia , Masculino , Repetições de Microssatélites
13.
Fam Cancer ; 16(3): 417-422, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28185117

RESUMO

Peutz-Jeghers syndrome (PJS) is a rare autosomal dominant disorder characterized by mucocutaneous hyperpigmentation, gastrointestinal (GI) hamartmatous polyps, and an increased risk of various malignancies. Pathogenic variants in the LKB1 tumor suppressor gene (also known as STK11) are the major cause of PJS. In this study, compound heterozygous variants of LKB1, c.890G > A/ c.1062C > G and del(exon1)/ c.1062C > G, were identified in two sporadic Chinese PJS cases respectively. Although all these three variants had been related to the autosomal dominant PJS in previous studies, all evidences collected in this study including de novo data, segregation data, population data, in-silico data, and functional data indicated that del(exon1) and c.890G > A are pathogenic in these two PJS families rather than c.1062C > G. This finding would contribute to genetic counseling for individuals carrying the variant c.1062C > G with or without PJS phenotypes. Moreover, this finding reminds genetic counselors that it is necessary to reevaluate the pathogenicity of reported variants in a known Mendelian disorder in order to avoid a misleading decision.


Assuntos
Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Adolescente , Adulto , Povo Asiático/genética , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença/genética , Heterozigoto , Humanos , Masculino , Linhagem , Adulto Jovem
14.
J Hum Genet ; 62(4): 513-516, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28077840

RESUMO

GATA zinc finger domain-containing 2B (GATAD2B) is a subunit of the methyl-CpG-binding protein-1 complex (MECP1), which deacetylates methylated nucleosomes and regresses transcriptional activity. Recently, GATAD2B has been elucidated as a candidate gene in patients with intellectual disability (ID). In this study, we identified two novel heterozygous frameshift mutations of GATAD2B in two unrelated ID cases through next-generation sequencing (NGS). Both of the mutations c.80_81insGATGT and c.552_555delGAAA cause truncated proteins that might be detrimental to neurodevelopment. We performed western blotting and observed a reduction in the target protein compared with normal controls. This is the first report of GATAD2B in Chinese ID patients. Our findings will broaden the spectrum of GATAD2B mutations and facilitate genetic diagnosis and counseling.


Assuntos
Fatores de Transcrição GATA/genética , Estudos de Associação Genética , Deficiência Intelectual/genética , Criança , Pré-Escolar , Feminino , Mutação da Fase de Leitura , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Deficiência Intelectual/patologia , Masculino , Proteínas Repressoras
15.
J Genet Couns ; 26(1): 72-78, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27422779

RESUMO

Spinal muscular atrophy is an autosomal recessive neuromuscular disease mainly caused by homozygous deletion of SMN1. The 2-copy SMN1 allele may present in the families of SMA patients with homozygous deletion of SMN1, one of whose parents has two SMN1 copies. In such families, individuals having two SMN1 copies still have a chance to be "2 + 0" carriers. In this study, the risks for the parents, fetuses and other siblings having two SMN1 copies to be "2 + 0" carriers were estimated based on Chinese meta-analysis data and turned out to be rather striking. Our findings would help to optimize genetic counseling regarding spinal muscular atrophy.


Assuntos
Dosagem de Genes , Predisposição Genética para Doença , Heterozigoto , Atrofia Muscular Espinal/metabolismo , Mutação , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Alelos , Feminino , Humanos , Masculino , Atrofia Muscular Espinal/genética
16.
Clin Chim Acta ; 464: 24-29, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27983999

RESUMO

Galloway-Mowat syndrome (GMS) is a very rare autosomal-recessive disorder characterized by nephrotic syndrome associated with microcephaly, and various central nervous system abnormalities, mostly cerebral hypoplasia or cerebellar atrophy, intellectual disability and neural-migration defects. WDR73 is the only gene known to cause GMS, and has never been implicated in other disease. Here we present a Chinese consanguineous family with infantile onset intellectual disability and cerebellar hypoplasia but no microcephaly. Whole exome sequencing identified a WDR73 p.W371G missense mutation. The mutation is confirmed to be segregated in this family by Sanger sequencing according to a recessive inheritance pattern. It is predicted to be deleterious by multiple algorithms and affect highly conserved site. Structural modeling revealed conformational differences between the wild type protein and the p.W371G protein. Real-time PCR and Western blotting revealed altered mRNA and protein levels in mutated samples. Our study indicates the novel WDR73 p.W371G missense mutation causes infantile onset intellectual disability and cerebellar hypoplasia in recessive mode of inheritance. Our findings imply that microcephaly is a variable phenotype in WDR73-related disease, suggest WDR73 to be a candidate gene of severe intellectual disability and cerebellar hypoplasia, and expand the molecular spectrum of WDR73-related disease.


Assuntos
Cerebelo/anormalidades , Consanguinidade , Deficiência Intelectual/genética , Mutação de Sentido Incorreto , Malformações do Sistema Nervoso/genética , Linhagem , Proteínas/genética , Adulto , Idade de Início , Sequência de Aminoácidos , Animais , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/genética , Feminino , Humanos , Lactente , Deficiência Intelectual/complicações , Deficiência Intelectual/etiologia , Masculino , Modelos Moleculares , Malformações do Sistema Nervoso/complicações , Malformações do Sistema Nervoso/etiologia , Conformação Proteica , Proteínas/química
17.
BMC Med Genet ; 17(1): 77, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27821076

RESUMO

BACKGROUND: Peutz-Jeghers syndrome (PJS) is a rare autosomal dominant inherited disorder characterized by gastrointestinal (GI) hamartomatous polyps, mucocutaneous hyperpigmentation, and an increased risk of cancer. Mutations in the serine-threonine kinase 11 gene (SKT11) are the major cause of PJS. CASE PRESENTATION: Blood samples were collected from six PJS families including eight patients. Mutation screening of STK11 gene was performed in these six families by Sanger sequencing and multiplex ligation-dependent probe amplification (MLPA) assay. Three novel mutations (c.721G > C, c.645_726del82, and del(exon2-5)) and three recurrent mutations (c.752G > A, c.545 T > C and del(exon1)) in STK11 were detected in six Chinese PJS families. Genotype-phenotype correlations suggested that truncating mutations trend to result in severe complications. CONCLUSION: These findings broaden the mutation spectrum of the STK11 gene and would help clinicians and genetic counselors provide better clinical surveillance for PJS patients, especially for ones carrying truncating mutation.


Assuntos
Mutação , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Análise de Sequência de DNA/métodos , Quinases Proteína-Quinases Ativadas por AMP , Adolescente , Adulto , Povo Asiático/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Linhagem , Adulto Jovem
18.
Am J Med Genet A ; 170(6): 1613-21, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27028180

RESUMO

Kabuki syndrome (KS) is a rare condition with multiple congenital anomalies and mental retardation. Exonic deletions, disrupting the lysine (K)-specific demethylase 6A (KDM6A) gene have been demonstrated as rare cause of KS. Here, we report a de novo 227-kb deletion in chromosome Xp11.3 of a 7-year-old Chinese girl with KS. Besides the symptoms of KS, the patient also presented with skin allergic manifestations, which were considered to be a new, rare feature of the phenotypic spectrum. The deletion includes the upstream region and exons 1-2 of KDM6A and potentially causes haploinsuffiency of the gene. We also discuss the mutation spectrum of KDM6A and clinical variability of patients with KDM6A deletion through a literature review. © 2016 Wiley Periodicals, Inc.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Éxons , Face/anormalidades , Doenças Hematológicas/diagnóstico , Doenças Hematológicas/genética , Histona Desmetilases/genética , Proteínas Nucleares/genética , Deleção de Sequência , Doenças Vestibulares/diagnóstico , Doenças Vestibulares/genética , China , Análise Mutacional de DNA , Fácies , Feminino , Genes Ligados ao Cromossomo X , Estudos de Associação Genética , Estudo de Associação Genômica Ampla , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Polimorfismo de Nucleotídeo Único , Inativação do Cromossomo X
19.
Clin Chim Acta ; 453: 75-9, 2016 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-26656312

RESUMO

Glutaric aciduria type I (GA-I) is a rare autosomal recessive metabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH), leading to an abnormal metabolism of lysine, hydroxylysine and tryptophan. It results in accumulations of glutaric acid, 3-hydroxyglutaric acid and glutaconic acid. Clinical features include the sudden onset of encephalopathy, hypotonia and macrocephaly usually before age 18months. Here we report five cases of GA-I confirmed with mutation analysis. GCDH gene mutations were identified in all five probands with GA-I. Three of them had compound heterozygous mutations and two had homozygous mutations. Mutations of two alleles (c.334G>T and IVS11-11A>G) were novel and both of them were confirmed to be splice site mutations by reverse transcription PCR.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/genética , Glutaril-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Sequência de Bases , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/metabolismo , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/química , Glutaril-CoA Desidrogenase/genética , Glutaril-CoA Desidrogenase/metabolismo , Heterozigoto , Humanos , Masculino , Modelos Moleculares , Mutação , Conformação Proteica
20.
Gene ; 569(2): 313-7, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25936346

RESUMO

Familial hypercholesterolemia (FH MIM# 143890) is one of the most common autosomal inherited diseases. FH is characterized by elevated plasma levels of total cholesterol and low-density lipoprotein-cholesterol. Mutation in the LDLR gene, which encodes the LDL receptor protein, is responsible for most of the morbidity of FH. The incidence of heterozygous FH is about 1/500, whereas the incidence of homozygous FH is only 1/1,000,000 in Caucasian population. In this study, we report a homozygous LDLR mutation (c.298G>A) in a familial hypercholesterolemia patient, who exhibited intracranial cholesterol deposition, which is a rare addition to the common FH phenotypes. The proband's consanguineous parents have the same heterozygous mutation with elevated concentrations of LDL-C but no xanthoma.


Assuntos
Hiperlipoproteinemia Tipo II/genética , Receptores de LDL/genética , Sequência de Aminoácidos , Animais , Povo Asiático/genética , Encéfalo/patologia , Consanguinidade , Análise Mutacional de DNA , Feminino , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/patologia , Lipoproteínas/sangue , Masculino , Dados de Sequência Molecular , Linhagem , Receptores de LDL/química , Alinhamento de Sequência , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA