Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Pediatr ; 181(10): 3691-3700, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35904599

RESUMO

Noonan syndrome (NS) is an autosomal dominant disorder characterized by clinical and genetic heterogeneity. It belongs to a wider group of pathologies, known as Rasopathies, due to the implication of genes encoding components of the Ras/MAPK signalling pathway. Recording the genetic alterations across populations helps assessing specific features to specific genes which is essential for better disease's recognition, prognosis and monitoring. Herein, we report the clinical and molecular data of a Greek cohort comprising of 86 NS or NS-like patients admitted at a single tertiary Centre in Athens, Greece. The analysis was performed using Sanger and next-generation sequencing, comprising 14 different genes. The mutational rates of the confirmed NS-associated genes in the Greek NS population are as follows: PTPN11 32.5%; RIT1 5.8%; SOS1 4.7%; BRAF 1.2%; CBL 1.2%; KRAS 1.2%; MAP2K1 1.2%; RAF1 1.2%; SHOC2 1.2%, corresponding to 50% of positivity in total NS population. The genotype-phenotype analysis showed statistically significant differences in craniofacial dysmorphisms (p = 0.005) and pulmonary valve stenosis (PS) (p < 0.001) frequencies between patients harbouring a pathogenic variant and patients without pathogenic variant in any of the tested genes. Patients with at least a pathogenic variant had 6.71 times greater odds to develop PS compared to pathogenic variant-negative patients (OR = 6.71, 95%; CI = (2.61, 17.27)). PTPN11 positive patients showed higher frequency of epicanthal folds (p = 0.004), ptosis (p = 0.001) and coarseness (p = 0.001) and lower frequency of neurological findings (p = 0.006), compared to patients carrying pathogenic variants in other genes. CONCLUSION: Craniofacial dysmorphism and PS prevail among pathogenic variant positive compared to pathogenic variant negative NS and NS-like patients while neurological defects are less common in PTPN11-affected NS patients compared to patients harbouring pathogenic variants in other genes. The significant prevalence of the Ras/MAPK pathogenic variants (17.4%), other than PTPN11, in Greek NS patients, highlights the necessity of a wider spectrum of molecular diagnosis. WHAT IS KNOWN: • Noonan syndrome (NS) has been associated with pathogenic variants in molecules-components of the Ras/MAPK pathway. • Clinical and genetic description of NS patients worldwide helps establishing personalized monitoring. WHAT IS NEW: • NS and NS-like mutational rate in Greece reaches 50% with pathogenic variants identified mostly in PTPN11 (32.5%), RIT1 (6%) and SOS1 (4.7%) genes. • The risk for pulmonary stenosis increases 6.71-fold in NS patients with a pathogenic variant compared to patients without genetic alterations.


Assuntos
Síndrome de Noonan , Grécia/epidemiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/epidemiologia , Síndrome de Noonan/genética , Fenótipo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética
2.
Am J Hum Genet ; 97(6): 922-32, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26637982

RESUMO

We describe an X-linked genetic syndrome associated with mutations in TAF1 and manifesting with global developmental delay, intellectual disability (ID), characteristic facial dysmorphology, generalized hypotonia, and variable neurologic features, all in male individuals. Simultaneous studies using diverse strategies led to the identification of nine families with overlapping clinical presentations and affected by de novo or maternally inherited single-nucleotide changes. Two additional families harboring large duplications involving TAF1 were also found to share phenotypic overlap with the probands harboring single-nucleotide changes, but they also demonstrated a severe neurodegeneration phenotype. Functional analysis with RNA-seq for one of the families suggested that the phenotype is associated with downregulation of a set of genes notably enriched with genes regulated by E-box proteins. In addition, knockdown and mutant studies of this gene in zebrafish have shown a quantifiable, albeit small, effect on a neuronal phenotype. Our results suggest that mutations in TAF1 play a critical role in the development of this X-linked ID syndrome.


Assuntos
Deficiências do Desenvolvimento/genética , Histona Acetiltransferases/genética , Deficiência Intelectual/genética , Doenças Neurodegenerativas/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Adolescente , Animais , Criança , Pré-Escolar , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Elementos E-Box , Fácies , Família , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Humanos , Lactente , Padrões de Herança , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Masculino , Mutação , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Linhagem , Fenótipo , Transdução de Sinais , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Adulto Jovem , Peixe-Zebra
3.
Pediatr Res ; 82(2): 253-260, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28422950

RESUMO

BackroundMicrocephaly can either be isolated or it may coexist with other neurological entities and/or multiple congenital anomalies, known as syndromic microcephaly. Although many syndromic cases can be classified based on the characteristic phenotype, some others remain uncertain and require further investigation. The present study describes the application of array-comparative genomic hybridization (array-CGH) as a diagnostic tool for the study of patients with clinically unknown syndromic microcephaly.MethodsFrom a cohort of 210 unrelated patients referred with syndromic microcephaly, we applied array-CGH analysis in 53 undiagnosed cases. In all the 53 cases except one, previous standard karyotype was negative. High-resolution 4 × 180K and 1 × 244K Agilent arrays were used in this study.ResultsIn 25 out of the 53 patients with microcephaly among other phenotypic anomalies, array-CGH revealed copy number variations (CNVs) ranging in size between 15 kb and 31.6 Mb. The identified CNVs were definitely causal for microcephaly in 11/53, probably causal in 7/53, and not causal for microcephaly in 7/53 patients. Genes potentially contributing to brain deficit were revealed in 16/53 patients.ConclusionsArray-CGH contributes to the elucidation of undefined syndromic microcephalic cases by permitting the discovery of novel microdeletions and/or microduplications. It also allows a more precise genotype-phenotype correlation by the accurate definition of the breakpoints in the deleted/duplicated regions.


Assuntos
Hibridização Genômica Comparativa/métodos , Microcefalia/genética , Criança , Estudos de Coortes , Feminino , Humanos , Cariotipagem , Masculino , Síndrome
4.
Mol Cell Probes ; 30(4): 254-260, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27417533

RESUMO

Dystrophinopathies are allelic X-linked myopathies caused by large deletions/duplications or small lesions along the DMD gene. An unexpected dynamic trinucleotide (GAA) expansion, ranging from ∼59 to 82 pure GAA repeats, within the DMD intron 62, was revealed to segregate through three family generations. From the pedigree, two female patients were referred for DMD investigation due to chronic myopathy and a muscle biopsy compatible with dystrophinopathy. As the size of the GAA repeat is limited to 11-33 within the general population our findings may provide a novel insight towards a Trinucleotide Repeat Expansion. Whether this TNR has an impact on the reported phenotype remains to be resolved.


Assuntos
Distrofina/genética , Repetições de Trinucleotídeos/genética , Adolescente , Adulto , Sequência de Bases , Criança , Metilação de DNA/genética , Distrofina/química , Feminino , Humanos , Masculino , Distrofia Muscular de Duchenne/genética , Linhagem
5.
Hum Mol Genet ; 20(10): 1925-36, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21349920

RESUMO

The recently described DNA replication-based mechanisms of fork stalling and template switching (FoSTeS) and microhomology-mediated break-induced replication (MMBIR) were previously shown to catalyze complex exonic, genic and genomic rearrangements. By analyzing a large number of isochromosomes of the long arm of chromosome X (i(Xq)), using whole-genome tiling path array comparative genomic hybridization (aCGH), ultra-high resolution targeted aCGH and sequencing, we provide evidence that the FoSTeS and MMBIR mechanisms can generate large-scale gross chromosomal rearrangements leading to the deletion and duplication of entire chromosome arms, thus suggesting an important role for DNA replication-based mechanisms in both the development of genomic disorders and cancer. Furthermore, we elucidate the mechanisms of dicentric i(Xq) (idic(Xq)) formation and show that most idic(Xq) chromosomes result from non-allelic homologous recombination between palindromic low copy repeats and highly homologous palindromic LINE elements. We also show that non-recurrent-breakpoint idic(Xq) chromosomes have microhomology-associated breakpoint junctions and are likely catalyzed by microhomology-mediated replication-dependent recombination mechanisms such as FoSTeS and MMBIR. Finally, we stress the role of the proximal Xp region as a chromosomal rearrangement hotspot.


Assuntos
Cromossomos Humanos X/genética , Replicação do DNA/genética , Isocromossomos/genética , Sequência de Bases , Quebra Cromossômica , Hibridização Genômica Comparativa , Humanos , Modelos Genéticos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Polimorfismo Genético , Recombinação Genética , Alinhamento de Sequência , Sequências de Repetição em Tandem/genética
6.
Pediatr Res ; 73(6): 772-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23481551

RESUMO

BACKGROUND: Congenital heart diseases (CHDs) are often associated with other congenital anomalies, dysmorphic features, and developmental delay, and only a few cases of chromosomal abnormalities are detected by conventional cytogenetic techniques. The microarray comparative genomic hybridization (CGH) analysis allows the identification of submicroscopic genomic rearrangements. METHODS: During the past 3 y, 55 of 330 patients referred for array CGH had CHD of unknown etiology plus at least one additional indication of abnormal chromosomal phenotype. High-resolution 1 × 244 K or 4 × 180 K Agilent arrays were used in this study (average resolution 7-13 kb). RESULTS: Copy-number variations were detected in 37 of 55 patients, and in 29 of 37 patients there were genes that have been associated with CHD. All 37 patients had at least one additional phenotypic abnormality: 30 of 37 had one or more other congenital anomalies, 23 of 37 had dysmorphic features, 16 of 37 had intellectual disability, 13 of 37 had abnormal magnetic resonance imaging, 10 of 37 had hypotonia, and 7 of 37 had seizures. In 9 of 55 patients, unexpected genomic rearrangements in relation to their phenotype were identified. CONCLUSION: In patients with CHD and at least one additional indication of abnormal chromosomal phenotype, array CGH analysis could detect possible submicroscopic chromosomal abnormalities and provide proper genetic counseling.


Assuntos
Hibridização Genômica Comparativa , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/genética , Humanos , Lactente , Recém-Nascido , Síndrome
7.
Birth Defects Res A Clin Mol Teratol ; 94(6): 494-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22511562

RESUMO

BACKGROUND: Pelizaeus-Merzbacher disease (PMD) is a recessive, X-linked leukoencephalopathy attributed to impaired myelination during central nervous system development, caused by defects in the proteolipid protein 1 (PLP1) gene. PMD presents clinical variability, ranging from the severe connatal form to the classic form. CASES: We report the clinical and molecular findings of two affected males, three carrier females, and an aborted male fetus with familial PMD. The two male probands presented with severe PMD phenotype and intellectual disability. High-resolution oligonucleotide-based array comparative genomic hybridization (aCGH) identified an Xq22.2 duplication of 320.6 kb (102641391-102961998, hg18), including the PLP1 gene and surrounding chromosomal region. Postmortem examination of the aborted fetus at 25 weeks' gestation showed focal subcortical white matter degeneration, focal gliosis, and cerebellar atrophy. CONCLUSIONS: Genotype-phenotype correlation is provided. In the connatal form of PMD, leukodystrophy and cerebellar atrophy can occur antenatally and be established at 25 weeks' gestation. The observation of degenerative brain lesions occurring before the onset of subcortical myelination suggests that the PLP1 gene has a more complex role in human brain development, exceeding its structural function in myelin formation.


Assuntos
Encéfalo/metabolismo , Duplicação Cromossômica/genética , Cromossomos Humanos X/genética , Proteína Proteolipídica de Mielina/genética , Doença de Pelizaeus-Merzbacher/genética , Adolescente , Adulto , Encéfalo/patologia , Criança , Feminino , Feto , Genes Recessivos , Estudos de Associação Genética , Genótipo , Humanos , Masculino , Linhagem , Fenótipo , Gravidez
8.
Birth Defects Res A Clin Mol Teratol ; 94(7): 561-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22730277

RESUMO

BACKGROUND Cat-Eye syndrome (CES) with teratoma has not been previously reported. We present the clinical and molecular findings of a 9-month-old girl with features of CES and also a palpable midline neck mass proved to be an extragonadal mature teratoma, additionally characterized by array comparative genomic hybridization (aCGH). RESULTS High resolution oligonucleotide-based aCGH confirmed that the supernumerary marker chromosome (SMC) derived from chromosome 22, as was indicated by molecular cytogenetic analysis with fluorescence in situ hybridization (FISH). Additionally, aCGH clarified the size, breakpoints, and gene content of the duplication (dup 22q11.1q11.21; size:1.6 Mb; breakpoints: 15,438,946-17,041,773; hg18). The teratoma tissue was also tested with aCGH, in which the CES duplication was not found, but the analysis revealed three aberrations: del Xp22.3 (108,864-2788,689; 2.7 Mb hg18), dup Yp11.2 (6688,491-7340,982; 0.65 Mb, hg18), and dup Yq11.2q11.23 (12,570,853-27,177,133; 14.61 Mb, hg18). These results indicated 46 XY (male) karyotype of the teratoma tissue, making this the second report of mature extragonadal teratoma in a female neonate, probably deriving from an included dizygotic twin of opposite sex (fetus in fetu). CONCLUSIONS Our findings extend the phenotypic spectrum of CES syndrome, a disorder with clinical variability, pointing out specific dosage-sensitive genes that might contribute to specific phenotypic features.


Assuntos
Transtornos Cromossômicos/genética , Neoplasias de Cabeça e Pescoço/genética , Teratoma/genética , Aneuploidia , Transtornos Cromossômicos/complicações , Cromossomos Humanos Par 22/genética , Hibridização Genômica Comparativa , Anormalidades do Olho , Feminino , Neoplasias de Cabeça e Pescoço/complicações , Humanos , Hibridização in Situ Fluorescente , Lactente , Cariotipagem , Fenótipo , Teratoma/complicações
9.
Eur J Pediatr ; 171(1): 51-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21590266

RESUMO

Noonan syndrome (NS) is a common multiple congenital anomaly entity, the diagnosis of which, on clinical grounds, is based on a comprehensive scoring system in order to select patients for molecular confirmation. Our aim was to evaluate the phenotypic characteristics in the light of PTPN11 mutations. The study revealed 80 patients who were referred with initial indication of NS or Noonan-like syndrome (NLS) and further assessed by a clinical geneticist; 60/80 index patients, mean age 5.9 ± 5.3 years, fulfilled the NS criteria. Molecular analysis of PTPN11 gene (exons and their flanking regions) of the total population revealed mutations in 17/80 patients, all belonging in the group of the patients screened with the scoring system. All mutations were heterozygous missense changes, mostly clustering in exon 3 (8/17), followed by exons 13 (3/17), 8 (2/17), 7 (2/17), 2 (1/17) and 4 (1/17). We conclude that (a) most of our clinically diagnosed NS cases were sporadic (b) PTPN11 analysis should be limited to those fulfilling the relevant NS criteria (c) Cardiovascular evaluation should comprise all NS patients, while pulmonary stenosis, short stature, and thorax deformities prevailed among those with PTPN11 mutations.


Assuntos
Mutação de Sentido Incorreto , Síndrome de Noonan/genética , Fenótipo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Marcadores Genéticos , Grécia , Heterozigoto , Humanos , Lactente , Masculino , Síndrome de Noonan/diagnóstico , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Adulto Jovem
10.
Am J Med Genet A ; 152A(8): 1925-32, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20575009

RESUMO

The 15q11-q13 PWS/AS critical region involves genes that are characterized by genomic imprinting. Multiple repeat elements within the region mediate rearrangements, including interstitial duplications, interstitial triplications, and supernumerary isodicentric marker chromosomes, as well as the deletions that cause Prader-Willi syndrome (PWS) and Angelman syndrome (AS). Recently, duplications of maternal origin concerning the same critical region have been implicated in autism spectrum disorders (ASD). We present a 6-month-old girl carrying a de novo duplication of maternal origin of the 15q11.2-q14 PWS/AS region (17.73 Mb in size) [46,XX,dup(15)(q11.2-q14)] detected with a high-resolution microarray-based comparative genomic hybridization (array-CGH). The patient is characterized by severe hypotonia, obesity, microstomia, long eyelashes, hirsutism, microretrognathia, short nose, severe psychomotor retardation, and multiple episodes of drug-resistant epileptic seizures, while her brain magnetic resonance imaging (MRI) documented partial corpus callosum dysplasia. In our patient the duplicated region is quite large extending beyond the Prader-Willi-Angelman critical region (PWACR), containing a number of genes that have been shown to be involved in ASD, exhibiting a severe phenotype, beyond the typical PWS/AS clinical manifestations. Reporting of similar well-characterized clinical cases with clearly delineated breakpoints of the duplicated region will clarify the contribution of specific genes to the phenotype.


Assuntos
Síndrome de Angelman/genética , Aberrações Cromossômicas , Cromossomos Humanos Par 15/genética , Hibridização Genômica Comparativa , Duplicação Gênica , Síndrome de Prader-Willi/genética , Síndrome de Angelman/patologia , Feminino , Impressão Genômica , Humanos , Hibridização in Situ Fluorescente , Lactente , Mães , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Síndrome de Prader-Willi/patologia
11.
Pediatr Res ; 67(5): 551-6, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20098342

RESUMO

The MECP2 gene mutations cause Rett syndrome (RTT) (OMIM: 312750), an X-linked dominant disorder primarily affecting girls. Until RTT was considered lethal in males, although now approximately 60 cases have been reported. Males with MECP2 mutations present with a broad spectrum of phenotypes ranging from neonatal encephalopathy to nonsyndromic mental retardation (MR). Four boys (aged, 3-11 y) were evaluated for MR. Patient 1 had autistic features. Patients 2 and 3 were brothers both presenting with psychomotor delay. Patient 4 showed dysmorphic features and behavioral problems reminiscent of FXS. All patients had a normal 46, XY karyotype and three were tested for FXS with negative results. MECP2 gene analysis of exons 3 and 4 was performed using methods based on the PCR, including Enzymatic Cleavage Mismatched Analysis (ECMA) and direct sequencing. Patient 1 presented somatic mosaicism for the classic RTT p.R106W mutation and patient 4 carried the p.T203M polymorphism. Analysis of the mothers in both cases revealed normal DNA sequences. Patients 2 and 3 had a novel deletion (c.1140del86) inherited from their unaffected mother. MECP2 gene mutations may be considered a rare cause of MR in males although great phenotypic variation hinders genotype-phenotype correlation.


Assuntos
Proteína 2 de Ligação a Metil-CpG/genética , Mosaicismo , Mutação Puntual , Síndrome de Rett/genética , Deleção de Sequência , Transtorno Autístico/genética , Criança , Transtornos do Comportamento Infantil/genética , Pré-Escolar , Análise Mutacional de DNA , Éxons , Predisposição Genética para Doença , Testes Genéticos , Humanos , Deficiência Intelectual/genética , Cariotipagem , Masculino , Linhagem , Fenótipo , Transtornos Psicomotores/genética , Estudos Retrospectivos , Síndrome de Rett/diagnóstico , Síndrome de Rett/psicologia , Índice de Gravidade de Doença
12.
In Vivo ; 24(3): 309-14, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20555004

RESUMO

BACKGROUND: Milroy's primary congenital lymphedema is a non-syndromic primary lymphedema caused mainly by autosomal dominant mutations in the FLT4 (VEGFR3) gene. Here, we report on a 6-month-old boy with congenital non-syndromic bilateral lymphedema at both feet and tibias, who underwent molecular investigation, consisted of PCR amplification and DHPLC analysis of exons 17-26 of the FLT4 gene. The clinical diagnosis of Milroy disease was confirmed by molecular analysis showing the c.3109G>C mutation in the FLT4 gene, inherited from the asymptomatic father. This is a known missense mutation, which substitutes an aspartic acid into a histidine on amino acid position 1037 of the resulting protein (p.D1037H), described in two other families with Milroy disease. A thorough genetic molecular investigation and clinical evaluation contributes to the provision of proper genetic counseling for parents of an affected child with Milroy disease. The herein described case, which is the third reported so far with c.3109G>C mutation, adds data on genotypic-phenotypic correlation of Milroy disease. The relative literature regarding the pathophysiology, molecular basis, clinical spectrum and treatment of Milroy disease is reviewed.


Assuntos
Linfedema , Mutação Puntual , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Pé/patologia , Genótipo , Humanos , Lactente , Linfedema/congênito , Linfedema/genética , Linfedema/patologia , Masculino , Fenótipo
13.
Horm Res ; 71(1): 45-51, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19039236

RESUMO

BACKGROUND: Sotos syndrome is an autosomal dominant disease characterized by tall stature, advanced bone age, typical morphological abnormalities of the face and developmental delay. It is caused by mutations in the NSD1 gene located on chromosome 5. NSD1 mutations are detected in the majority of the Sotos patients, and include intragenic NSD1 mutations and microdeletions in the 5q35 region. Cardiovascular and urogenital symptoms are more frequent in the microdeletion group. METHODS: Mutation analysis was performed in 4 patients with Sotos syndrome with typical phenotypic characteristics. RESULTS: In each of the 4 patients a NSD1 mutation was found (2 frame shifts, 1 nonsense and 1 missense mutation). Two of our patients presented dysplastic kidneys with cysts and psychosis, respectively. CONCLUSIONS: We describe 4 Greek patients with Sotos syndrome. Apart from the typical phenotypic characteristics, 2 of our patients presented rare clinical manifestations such as dysplastic kidneys and psychosis. The 3 detected mutations are novel.


Assuntos
Anormalidades Múltiplas/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Proteínas Nucleares/genética , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Fácies , Grécia , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Lactente , Fenótipo
14.
Acta Cardiol ; 64(1): 51-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19317298

RESUMO

OBJECTIVES: The objectives of this study were to investigate cell-free DNA daily concentration changes following an acute myocardial infarction (AMI) and to assess any correlations with complications during hospitalization. METHODS AND RESULTS: Serial cell-free DNA level determinations were performed by quantitative Real-Time PCR in 47 AMI patients once daily during hospitalization (235 samples) and once in 100 healthy subjects. Cell-free DNA concentrations are significantly higher in patients throughout hospitalization compared to healthy subject levels (2.644 (SE 0.0952) vs. 1.519 (SE 0.0566), P < 0.001). The median maximum cell-free DNA concentration was 3.5-fold higher (Mann Whitney P = 0.0035) in 20/47 patients with complicated post AMI course--group I--(1719.7, range 117.32-4996212.1 GenEq/ml plasma) compared with 27/47 patients without complications--group II--(492.9, range 56.43-4715.15 GenEq/ml plasma). Substantial differences exist between cell-free DNA concentrations measured on t(pre) (the day before the complication) and t(c) (the day the complication occurred) as well as t(post) (the day after the complication) in group I whereby cell-free DNA rises significantly in t(c) and remains elevated in t(post) (t(pre) vs. t(c), 2.445 vs. 2.965, P = 0.0171 and t(pre) vs. t(post) 2.445 vs. 2.913, P = 0.023). CONCLUSIONS: Cell-free DNA concentrations were elevated in AMI patients compared to healthy control subjects, rise significantly when complications occur and have a potential clinical value in monitoring patient progress during hospitalization.


Assuntos
DNA/sangue , Infarto do Miocárdio/sangue , Idoso , Análise de Variância , Biomarcadores/sangue , Estudos de Casos e Controles , Sistema Livre de Células , Feminino , Humanos , Pacientes Internados/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/fisiopatologia , Reação em Cadeia da Polimerase , Prognóstico , Fatores de Tempo
15.
Leuk Res ; 83: 106177, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31261022

RESUMO

Acute Lymphoblastic Leukemia (ALL) is a malignancy of the immature lymphoid cells mainly associated with numerical and structural chromosomal aberrations. The current standard for profiling the diverse genetic background comprises a combination of conventional karyotype and FISH analysis for the most common translocations, albeit with many limitations. Chromosomal Microarray Analysis (CMA) is a high throughput whole genome method that is gradually implemented in routine clinical practice, but not many studies have compared the two methods. Here we aim to investigate the added benefits of utilizing the high resolution 2 x 400 K G3 CGH + SNP CMA platform in routine diagnostics of pediatric ALL. From the 29 bone marrow samples that were analyzed, CMA identified clinically relevant findings in 83%, while detecting chromosomal aberrations in 75% of the patients with normal conventional karyotype. The most common finding was hyperdiploidy (20%), and the most common submicroscopic aberration involved CDKN2A/B genes. The smallest aberration detected was a 9 kb partial NF1 gene duplication. The prognosis of the patients when combining conventional cytogenetics and CMA was either changed or enhanced in 66% of the cases. A rare duplication possibly indicative of a cryptic ABL1-NUP214 fusion gene was found in one patient. We conclude that CMA, when combined with conventional cytogenetic analysis, can significantly enhance the genetic profiling of patients with pediatric ALL in a routine clinical setting.


Assuntos
Proteínas de Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos , Ploidias , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Proteínas de Neoplasias/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras B/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia
16.
Dev Med Child Neurol ; 50(12): 950-2, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19046188

RESUMO

We report the clinical and genetic evaluation of a 2-year-old Greek female with striking phenotypic similarities to the three previously published cases of Okamoto syndrome. The main features were characteristic facies, cleft palate, generalized hypotonia, severe developmental delay, congenital hydronephrosis, and congenital heart defects. Routine chromosome testing and whole-genome high-resolution comparative genetic hybridization analysis were negative for any gross numerical or structural chromosome aberrations and for microdeletions/duplications of more than 3 million base pairs respectively. Fluorescence in situ hybridization analysis for 22q11.2 deletion and DNA analysis of the protein tyrosine phosphatase, non-receptor type II gene were normal, thus excluding DiGeorge and Noonan syndromes. Our patient did not show most of the cardinal features of Schinzel-Giedion, otopalatodigital, and C-trigonocephaly syndromes. Moreover, in our patient some new malformations were identified: unilateral kidney hypoplasia and severe anal stenosis. The latter was considered as pertinent and is described here to establish a wider clinical spectrum of Okamoto syndrome. At the age of 3 years 6 months the child continues to show severe growth failure and significant global developmental delay. For the practising paediatrician it is prudent to bear Okamoto syndrome in mind, especially in children with learning disability and a pattern of dysmorphic features.


Assuntos
Anormalidades Múltiplas/genética , Fissura Palatina/genética , Anormalidades Craniofaciais/genética , Permeabilidade do Canal Arterial/genética , Fácies , Hidronefrose/genética , Deficiência Intelectual/genética , Hipotonia Muscular/genética , População Branca/genética , Anormalidades Múltiplas/diagnóstico , Pré-Escolar , Fissura Palatina/diagnóstico , Anormalidades Craniofaciais/diagnóstico , Análise Mutacional de DNA , Diagnóstico Diferencial , Permeabilidade do Canal Arterial/diagnóstico , Feminino , Seguimentos , Humanos , Hidronefrose/diagnóstico , Lactente , Deficiência Intelectual/diagnóstico , Rim/anormalidades , Hipotonia Muscular/diagnóstico , Fenótipo , Síndrome
17.
Eur J Pediatr ; 167(9): 1025-31, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18057963

RESUMO

The diagnosis of Noonan syndrome is essentially clinical, based upon the distinct phenotype and the involvement of the cardiovascular system. Tumor development is a rare manifestation of Noonan syndrome but can be explained by the molecular pathophysiology involved in the disorder. We present three Noonan patients who developed solid tumors. The first patient, a 4-year-old girl, developed granular cell tumors as did her mother in childhood. The second patient, a 1-year-old boy, had a low grade pilocytic astrocytoma, the clinical expression of which was persistent headache. MRI showed a pituitary mass in the posterior lobe. It was surgically removed. The third patient, a 7-year-old boy was found to have Sertoli tumors in his right cryptorchid testis. All three patients fulfilled the clinical criteria for Noonan syndrome. However, genetic testing was negative in patients 1 and 3. The diagnosis of Noonan syndrome was made based on distinct phenotypic findings in three patients who had different types of tumors.


Assuntos
Astrocitoma/complicações , Neoplasias Encefálicas/complicações , Tumor de Células Granulares/complicações , Síndrome de Noonan/complicações , Seminoma/complicações , Neoplasias Testiculares/complicações , Astrocitoma/genética , Astrocitoma/cirurgia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatologia , Criança , Pré-Escolar , Feminino , Tumor de Células Granulares/genética , Tumor de Células Granulares/patologia , Humanos , Lactente , Masculino , Síndrome de Noonan/genética , Síndrome de Noonan/fisiopatologia , Seminoma/fisiopatologia , Neoplasias Testiculares/fisiopatologia
18.
Hormones (Athens) ; 7(3): 217-29, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18694860

RESUMO

The end-organ resistance to androgens has been designated as androgen insensitivity syndrome (AIS), an X-linked disorder caused by mutations in the androgen receptor (AR) gene. It is generally accepted that defects in the AR gene prevent the normal development of both internal and external genital structures in 46,XY individuals, causing a variety of phenotypes ranging from male infertility to completely normal female external genitalia. Precise diagnosis requires clinical, hormonal and molecular investigation and is of great importance for appropriate gender assignment and management in general. The complexity of phenotypic presentation of AIS with genotype-phenotype variability of identical mutations complicates both the diagnostic procedure and genetic counseling of the affected families. More than 400 different AR gene mutations have thus far been reported but the receptor structure-function relationship and its phenotypic outcome is not yet fully understood. This review focuses on the clinical features and molecular pathophysiology of AIS and explores the relationship of the molecular defects in the AR gene to their clinical expression.


Assuntos
Síndrome de Resistência a Andrógenos/diagnóstico , Mutação , Receptores Androgênicos/genética , Síndrome de Resistência a Andrógenos/genética , Síndrome de Resistência a Andrógenos/metabolismo , Síndrome de Resistência a Andrógenos/terapia , Feminino , Aconselhamento Genético , Testes Genéticos , Genótipo , Hormônios/sangue , Humanos , Masculino , Fenótipo , Receptores Androgênicos/metabolismo , Índice de Gravidade de Doença
19.
PLoS One ; 13(10): e0205298, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30289920

RESUMO

The majority of apparently balanced translocation (ABT) carriers are phenotypically normal. However, several mechanisms were proposed to underlie phenotypes in affected ABT cases. In the current study, whole-genome mate-pair sequencing (WG-MPS) followed by Sanger sequencing was applied to further characterize de novo ABTs in three affected individuals. WG-MPS precisely mapped all ABT breakpoints and revealed three possible underlying molecular mechanisms. Firstly, in a t(X;1) carrier with hearing loss, a highly skewed X-inactivation pattern was observed and the der(X) breakpoint mapped ~87kb upstream an X-linked deafness gene namely POU3F4, thus suggesting an underlying long-range position effect mechanism. Secondly, cryptic complexity and a chromothripsis rearrangement was identified in a t(6;7;8;12) carrier with intellectual disability. Two translocations and a heterozygous deletion disrupted SOX5; a dominant nervous system development gene previously reported in similar patients. Finally, a direct gene disruption mechanism was proposed in a t(4;9) carrier with dysmorphic facial features and speech delay. In this case, the der(9) breakpoint directly disrupted NFIB, a gene involved in lung maturation and development of the pons with important functions in main speech processes. To conclude, in contrast to familial ABT cases with identical rearrangements and discordant phenotypes, where translocations are considered coincidental, translocations seem to be associated with phenotype presentation in affected de novo ABT cases. In addition, this study highlights the importance of investigating both coding and non-coding regions to decipher the underlying pathogenic mechanisms in these patients, and supports the potential introduction of low coverage WG-MPS in the clinical investigation of de novo ABTs.


Assuntos
Fácies , Perda Auditiva/genética , Deficiência Intelectual/genética , Transtornos do Desenvolvimento da Linguagem/genética , Translocação Genética , Sequência de Bases , Pontos de Quebra do Cromossomo , Feminino , Expressão Gênica , Perda Auditiva/diagnóstico , Perda Auditiva/fisiopatologia , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/fisiopatologia , Cariótipo , Transtornos do Desenvolvimento da Linguagem/diagnóstico , Transtornos do Desenvolvimento da Linguagem/fisiopatologia , Fatores de Transcrição NFI/deficiência , Fatores de Transcrição NFI/genética , Fatores do Domínio POU/deficiência , Fatores do Domínio POU/genética , Fenótipo , Fatores de Transcrição SOXD/deficiência , Fatores de Transcrição SOXD/genética , Sequenciamento Completo do Genoma
20.
Syst Biol Reprod Med ; 62(1): 69-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26636621

RESUMO

Preimplantation genetic diagnosis (PGD) to select histocompatible siblings to facilitate curative haematopoeitic stem-cell transplantation (HSCT) is now an acceptable option in the absence of an available human leukocyte antigen (HLA) compatible donor. We describe a case where the couple who requested HLA-PGD, were both carriers of two serious haematological diseases, beta-thalassaemia and sideroblastic anaemia. Their daughter, affected with sideroblastic anaemia, was programmed to have HSCT. A multiplex-fluorescent-touchdown-PCR protocol was optimized for the simultaneous amplification of: the two HBB-gene mutated regions (c.118C> T, c.25-26delAA), four short tandem repeats (STRs) in chr11p15.5 linked to the HBB gene, the SLC25A38 gene mutation (c.726C > T), two STRs in chr3p22.1 linked to the SLC25A38 gene, plus eleven informative STRs for HLA-haplotyping (chr6p22.1-21.3). This was followed by real-time nested PCR and high-resolution melting analysis (HRMA) for the detection of HBB and SLC25A38 gene mutations, as well as the analysis of all STRs on an automatic genetic analyzer (sequencer). The couple completed four clinical in vitro fertilization (IVF)/PGD cycles. At least one matched unaffected embryo was identified and transferred in each cycle. A twin pregnancy was established in the fourth PGD cycle and genotyping results at all loci were confirmed by prenatal diagnosis. Two healthy baby girls were delivered at week 38 of pregnancy. The need to exclude two familial disorders for HLA-PGD is rarely encountered. The methodological approach described here is fast, accurate, clinically-validated, and of relatively low cost.


Assuntos
Anemia Sideroblástica/diagnóstico , Anemia Sideroblástica/genética , Teste de Histocompatibilidade/métodos , Diagnóstico Pré-Implantação/métodos , Talassemia beta/diagnóstico , Talassemia beta/genética , Adulto , Anemia Sideroblástica/terapia , Feminino , Fertilização in vitro , Testes Genéticos , Genótipo , Transplante de Células-Tronco Hematopoéticas , Humanos , Recém-Nascido , Masculino , Proteínas de Transporte da Membrana Mitocondrial/biossíntese , Proteínas de Transporte da Membrana Mitocondrial/genética , Mutação/genética , Gravidez , Gravidez de Gêmeos , Talassemia beta/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA