Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Cell ; 140(1): 74-87, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-20074521

RESUMO

We report that eight heterozygous missense mutations in TUBB3, encoding the neuron-specific beta-tubulin isotype III, result in a spectrum of human nervous system disorders that we now call the TUBB3 syndromes. Each mutation causes the ocular motility disorder CFEOM3, whereas some also result in intellectual and behavioral impairments, facial paralysis, and/or later-onset axonal sensorimotor polyneuropathy. Neuroimaging reveals a spectrum of abnormalities including hypoplasia of oculomotor nerves and dysgenesis of the corpus callosum, anterior commissure, and corticospinal tracts. A knock-in disease mouse model reveals axon guidance defects without evidence of cortical cell migration abnormalities. We show that the disease-associated mutations can impair tubulin heterodimer formation in vitro, although folded mutant heterodimers can still polymerize into microtubules. Modeling each mutation in yeast tubulin demonstrates that all alter dynamic instability whereas a subset disrupts the interaction of microtubules with kinesin motors. These findings demonstrate that normal TUBB3 is required for axon guidance and maintenance in mammals.


Assuntos
Tubulina (Proteína)/metabolismo , Sequência de Aminoácidos , Animais , Axônios/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Sobrevivência Celular , Criança , Deficiências do Desenvolvimento , Feminino , Humanos , Cinesinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Transporte Proteico , Tubulina (Proteína)/química , Tubulina (Proteína)/genética
2.
PLoS Genet ; 15(4): e1008088, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31034465

RESUMO

PIK3C2A is a class II member of the phosphoinositide 3-kinase (PI3K) family that catalyzes the phosphorylation of phosphatidylinositol (PI) into PI(3)P and the phosphorylation of PI(4)P into PI(3,4)P2. At the cellular level, PIK3C2A is critical for the formation of cilia and for receptor mediated endocytosis, among other biological functions. We identified homozygous loss-of-function mutations in PIK3C2A in children from three independent consanguineous families with short stature, coarse facial features, cataracts with secondary glaucoma, multiple skeletal abnormalities, neurological manifestations, among other findings. Cellular studies of patient-derived fibroblasts found that they lacked PIK3C2A protein, had impaired cilia formation and function, and demonstrated reduced proliferative capacity. Collectively, the genetic and molecular data implicate mutations in PIK3C2A in a new Mendelian disorder of PI metabolism, thereby shedding light on the critical role of a class II PI3K in growth, vision, skeletal formation and neurological development. In particular, the considerable phenotypic overlap, yet distinct features, between this syndrome and Lowe's syndrome, which is caused by mutations in the PI-5-phosphatase OCRL, highlight the key role of PI metabolizing enzymes in specific developmental processes and demonstrate the unique non-redundant functions of each enzyme. This discovery expands what is known about disorders of PI metabolism and helps unravel the role of PIK3C2A and class II PI3Ks in health and disease.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Catarata/genética , Transtornos da Motilidade Ciliar/genética , Nanismo/genética , Mutação , Fosfatidilinositol 3-Quinases/genética , Adolescente , Adulto , Criança , Consanguinidade , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Linhagem , Fenótipo , Adulto Jovem
3.
Hum Mol Genet ; 27(11): 1913-1926, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29566152

RESUMO

Primary ovarian insufficiency (POI) is characterized by amenorrhea and loss or dysfunction of ovarian follicles prior to the age of 40. POI has been associated with autosomal recessive mutations in genes involving hormonal signaling and folliculogenesis, however, the genetic etiology of POI most often remains unknown. Here we report MRPS22 homozygous missense variants c.404G>A (p.R135Q) and c.605G>A (p.R202H) identified in four females from two independent consanguineous families as a novel genetic cause of POI in adolescents. Both missense mutations identified in MRPS22 are rare, occurred in highly evolutionarily conserved residues, and are predicted to be deleterious to protein function. In contrast to prior reports of mutations in MRPS22 associated with severe mitochondrial disease, the POI phenotype is far less severe. Consistent with this genotype-phenotype correlation, mitochondrial defects in oxidative phosphorylation or rRNA levels were not detected in fibroblasts derived from the POI patients, suggesting a non-bioenergetic or tissue-specific mitochondrial defect. Furthermore, we demonstrate in a Drosophila model that mRpS22 deficiency specifically in somatic cells of the ovary had no effect on fertility, whereas flies with mRpS22 deficiency specifically in germ cells were infertile and agametic, demonstrating a cell autonomous requirement for mRpS22 in germ cell development. These findings collectively identify that MRPS22, a component of the small mitochondrial ribosome subunit, is critical for ovarian development and may therefore provide insight into the pathophysiology and treatment of ovarian dysfunction.


Assuntos
Proteínas de Drosophila/genética , Fertilidade/genética , Proteínas Mitocondriais/genética , Insuficiência Ovariana Primária/genética , Proteínas Ribossômicas/genética , Adolescente , Adulto , Amenorreia/genética , Amenorreia/patologia , Animais , Modelos Animais de Doenças , Drosophila/genética , Feminino , Fertilidade/fisiologia , Homozigoto , Humanos , Menopausa Precoce/genética , Mutação de Sentido Incorreto/genética , Folículo Ovariano/patologia , Insuficiência Ovariana Primária/patologia , Adulto Jovem
4.
Am J Hum Genet ; 99(5): 1172-1180, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27773429

RESUMO

Glycine is a major neurotransmitter that activates inhibitory glycine receptors and is a co-agonist for excitatory glutamatergic N-methyl-D-aspartate (NMDA) receptors. Two transporters, GLYT1 and GLYT2, regulate extracellular glycine concentrations within the CNS. Dysregulation of the extracellular glycine has been associated with hyperekplexia and nonketotic hyperglycinemia. Here, we report four individuals from two families who presented at birth with facial dysmorphism, encephalopathy, arthrogryposis, hypotonia progressing to hypertonicity with startle-like clonus, and respiratory failure. Only one individual survived the respiratory failure and was weaned off ventilation but has significant global developmental delay. Mildly elevated cerebrospinal fluid (CSF) glycine and normal serum glycine were observed in two individuals. In both families, we identified truncating mutations in SLC6A9, encoding GLYT1. We demonstrate that pharmacologic or genetic abolishment of GlyT1 activity in mice leads to mildly elevated glycine in the CSF but not in blood. Additionally, previously reported slc6a9-null mice and zebrafish mutants also display phenotypes consistent with the affected individuals we examined. Our data suggest that truncating SLC6A9 mutations lead to a distinct human neurological syndrome hallmarked by mildly elevated CSF glycine and normal serum glycine.


Assuntos
Artrogripose/genética , Proteínas da Membrana Plasmática de Transporte de Glicina/genética , Glicina/líquido cefalorraquidiano , Hiperglicinemia não Cetótica/genética , Animais , Artrogripose/diagnóstico , Pré-Escolar , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Glicina/sangue , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Knockout , Linhagem
5.
J Med Genet ; 55(11): 779-784, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29875123

RESUMO

BACKGROUND: Intestinal integrity is essential for proper nutrient absorption and tissue homeostasis, with damage leading to enteric protein loss, that is, protein-losing enteropathy (PLE). Recently, homozygous nonsense variants in the plasmalemma vesicle-associated protein gene (PLVAP) were reported in two patients with severe congenital PLE. PLVAP is the building block of endothelial cell (EC) fenestral diaphragms; its importance in barrier function is supported by mouse models of Plvap deficiency. OBJECTIVE: To genetically diagnose two first-degree cousins once removed, who presented with PLE at ages 22 and 2.5 years. METHODS: Family-based whole exome sequencing was performed based on an autosomal recessive inheritance model. In silico analyses were used to predict variant impact on protein structure and function. RESULTS: We identified a rare homozygous variant (NM_031310.2:c.101T>C;p.Leu34Pro) in PLVAP, which co-segregated with the disease. Leu34 is predicted to be located in a highly conserved, hydrophobic, α-helical region within the protein's transmembrane domain, suggesting Leu34Pro is likely to disrupt protein function and/or structure. Electron microscopy and PLVAP immunohistochemistry demonstrated apparently normal diaphragm morphology, predicted to be functionally affected. CONCLUSIONS: Biallelic missense variants in PLVAP can cause an attenuated form of the PLE and hypertriglyceridaemia syndrome. Our findings support the role of PLVAP in the pathophysiology of PLE, expand the phenotypic and mutation spectrums and underscore PLVAP's importance in EC barrier function in the gut.


Assuntos
Proteínas de Transporte/genética , Estudos de Associação Genética , Homozigoto , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Fenótipo , Enteropatias Perdedoras de Proteínas/diagnóstico , Enteropatias Perdedoras de Proteínas/genética , Adulto , Substituição de Aminoácidos , Biomarcadores , Biópsia , Proteínas de Transporte/química , Biologia Computacional/métodos , Consanguinidade , Feminino , Humanos , Recém-Nascido , Masculino , Proteínas de Membrana/química , Modelos Moleculares , Linhagem , Conformação Proteica , Enteropatias Perdedoras de Proteínas/metabolismo , Relação Estrutura-Atividade , Adulto Jovem
6.
Gastroenterology ; 152(8): 1876-1880.e1, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28242209

RESUMO

Hereditary mixed polyposis syndrome is a rare colon cancer predisposition syndrome caused by a duplication of a noncoding sequence near the gremlin 1, DAN family BMP antagonist gene (GREM1) originally described in Ashkenazi Jews. Few families with GREM1 duplications have been described, so there are many questions about detection and management. We report 4 extended families with the duplication near GREM1 previously found in Ashkenazi Jews; 3 families were identified at cancer genetic clinics in Israel and 1 family was identified in a cohort of patients with familial colorectal cancer. Their clinical features include extracolonic tumors, onset of polyps in adolescence, and rapid progression of some polyps to advanced adenomas. One family met diagnostic criteria for Lynch syndrome. Expansion of the hereditary mixed polyposis syndrome phenotype can inform surveillance strategies for carriers of GREM1 duplications.


Assuntos
Polipose Adenomatosa do Colo/diagnóstico , Polipose Adenomatosa do Colo/genética , Biomarcadores Tumorais/genética , Colo/patologia , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Neoplasias Colorretais Hereditárias sem Polipose/genética , Detecção Precoce de Câncer/métodos , Duplicação Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Polipose Adenomatosa do Colo/etnologia , Polipose Adenomatosa do Colo/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Colonoscopia , Neoplasias Colorretais Hereditárias sem Polipose/etnologia , Neoplasias Colorretais Hereditárias sem Polipose/patologia , Análise Mutacional de DNA , Progressão da Doença , Feminino , Predisposição Genética para Doença , Hereditariedade , Humanos , Israel , Judeus/genética , Masculino , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular , Mutação , Linhagem , Fenótipo , Fatores de Tempo , Adulto Jovem
7.
Blood Cells Mol Dis ; 68: 115-116, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-27864021

RESUMO

Heterozygote GBA (glucosylceramidase beta) mutations increase the risk of Parkinson's disease (PD). Data based on the measured frequencies of GBA mutated alleles in the healthy population suggest that severe GBA mutations are associated with even higher risk for PD. These data, however, are prone to methodological biases resulting from the rarity of severe mutations and from ethnic-dependent differences in allele frequencies. To overcome these biases, we traced 13 Gaucher disease (GD) patients who were compound heterozygotes for one mild (N370S) and one severe GBA mutation and who reported a parent with PD. We determined the GBA mutation status of all parents and examined them whenever possible. While 50% of the parents carried a mild GBA mutation, we hypothesized that PD cases would be more likely to carry a severe mutation. We found that 10/13 PD parents had a severe mutation and only 3/10 carried a mild mutation (binomial test P<0.05). Using an unbiased methodology, we show that carriers of severe GBA mutations are at higher risk for PD relative to carriers of the mild mutations.


Assuntos
Glucosilceramidase/genética , Mutação , Doença de Parkinson/genética , Idoso , Alelos , Feminino , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Mutação Puntual
8.
Pediatr Res ; 83(4): 825-828, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29320483

RESUMO

BackgroundThe objective of our study was to examine the risk for submicroscopic chromosomal aberrations among fetuses with apparently isolated solitary kidney.MethodsData acquisition was performed retrospectively by searching Israeli Ministry of Health-computerized database. All cases having chromosomal microarray analysis (CMA), referred because of an indication of isolated unilateral kidney agenesis between January 2013 and September 2016, were included. Rate of clinically significant CMA findings in these pregnancies was compared to pregnancies with normal ultrasound, based on a systematic review encompassing 9,792 cases and local data of 5,541 pregnancies undergoing CMA because of maternal request.ResultsOf the 81 pregnancies with isolated solitary kidney, 2 (2.47%) loss-of-copy number variants compatible with well-described deletion syndromes were reported (16p11.2-16p12.2 and 22q11.21 microdeletion syndromes). In addition, one variant of unknown significance was demonstrated. The relative risk for pathogenic CMA findings among pregnancies with isolated unilateral renal agenesis was not significantly different compared with the control population.ConclusionCMA analysis in pregnancies with unilateral renal agenesis might still be useful, to the same degree as it can be in the general population.


Assuntos
Aberrações Cromossômicas/embriologia , Rim/anormalidades , Rim/embriologia , Análise de Sequência com Séries de Oligonucleotídeos , Rim Único/diagnóstico por imagem , Rim Único/embriologia , Adulto , Deleção Cromossômica , Cromossomos Humanos Par 16 , Cromossomos Humanos Par 22/genética , Feminino , Estudos de Associação Genética , Humanos , Masculino , Idade Materna , Gravidez , Estudos Retrospectivos , Risco , Rim Único/genética , Ultrassonografia Pré-Natal
9.
Hum Mutat ; 38(1): 7-15, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27667302

RESUMO

Werner syndrome (WS) is a rare autosomal recessive disorder characterized by a constellation of adult onset phenotypes consistent with an acceleration of intrinsic biological aging. It is caused by pathogenic variants in the WRN gene, which encodes a multifunctional nuclear protein with exonuclease and helicase activities. WRN protein is thought to be involved in optimization of various aspects of DNA metabolism, including DNA repair, recombination, replication, and transcription. In this update, we summarize a total of 83 different WRN mutations, including eight previously unpublished mutations identified by the International Registry of Werner Syndrome (Seattle, WA) and the Japanese Werner Consortium (Chiba, Japan), as well as 75 mutations already reported in the literature. The Seattle International Registry recruits patients from all over the world to investigate genetic causes of a wide variety of progeroid syndromes in order to contribute to the knowledge of basic mechanisms of human aging. Given the unusually high prevalence of WS patients and heterozygous carriers in Japan, the major goal of the Japanese Consortium is to develop effective therapies and to establish management guidelines for WS patients in Japan and elsewhere. This review will also discuss potential translational approaches to this disorder, including those currently under investigation.


Assuntos
Mutação , Helicase da Síndrome de Werner/genética , Síndrome de Werner/genética , Fatores Etários , Animais , Bases de Dados Genéticas , Modelos Animais de Doenças , Éxons , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Geografia , Humanos , Japão , Camundongos , Fenótipo , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Sistema de Registros , Pesquisa Translacional Biomédica , Navegador , Síndrome de Werner/diagnóstico , Síndrome de Werner/epidemiologia
10.
Hum Mol Genet ; 24(13): 3742-51, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25859010

RESUMO

Retinitis pigmentosa (RP), the most common form of inherited retinal degeneration, is clinically and genetically heterogeneous and can appear as syndromic or non-syndromic. Mucopolysaccharidosis type IIIC (MPS IIIC) is a lethal disorder, caused by mutations in the heparan-alpha-glucosaminide N-acetyltransferase (HGSNAT) gene and characterized by progressive neurological deterioration, with retinal degeneration as a prominent feature. We identified HGSNAT mutations in six patients with non-syndromic RP. Whole exome sequencing (WES) in an Ashkenazi Jewish Israeli RP patient revealed a novel homozygous HGSNAT variant, c.370A>T, which leads to partial skipping of exon 3. Screening of 66 Ashkenazi RP index cases revealed an additional family with two siblings homozygous for c.370A>T. WES in three Dutch siblings with RP revealed a complex HGSNAT variant, c.[398G>C; 1843G>A] on one allele, and c.1843G>A on the other allele. HGSNAT activity levels in blood leukocytes of patients were reduced compared with healthy controls, but usually higher than those in MPS IIIC patients. All patients were diagnosed with non-syndromic RP and did not exhibit neurological deterioration, or any phenotypic features consistent with MPS IIIC. Furthermore, four of the patients were over 60 years old, exceeding by far the life expectancy of MPS IIIC patients. HGSNAT is highly expressed in the mouse retina, and we hypothesize that the retina requires higher HGSNAT activity to maintain proper function, compared with other tissues associated with MPS IIIC, such as the brain. This report broadens the spectrum of phenotypes associated with HGSNAT mutations and highlights the critical function of HGSNAT in the human retina.


Assuntos
Acetiltransferases/genética , Mucopolissacaridose III/enzimologia , Mutação Puntual , Retinose Pigmentar/enzimologia , Adulto , Idoso , Animais , Doenças Assintomáticas , Sequência de Bases , Éxons , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mucopolissacaridose III/genética , Linhagem , Retina/enzimologia , Retinose Pigmentar/genética
11.
Am J Hematol ; 92(11): 1170-1176, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28762527

RESUMO

Eliglustat, an oral substrate reduction therapy, is a first-line treatment for adults with Gaucher disease type 1 (GD1) who are poor, intermediate, or extensive CYP2D6 metabolizers (>90% of patients). In the primary analysis of the Phase 3 ENGAGE trial (NCT00891202), eliglustat treatment for 9 months resulted in significant reductions in spleen and liver volumes and increases in hemoglobin concentration and platelet count compared with placebo. We report 18-month outcomes of patients who entered the trial extension period, in which all patients received eliglustat. Of 40 trial patients, 39 entered the extension period, and 38 completed 18 months. Absolute values and percent change over time were determined for spleen and liver volume, hemoglobin concentration, platelet count, bone mineral density, bone marrow burden, and Gaucher disease biomarkers. For patients randomized to eliglustat in the double-blind period, continuing treatment with eliglustat for 9 more months resulted in incremental improvement of all disease parameters. For patients randomized to placebo in the double-blind period, eliglustat treatment during the 9-month, open-label period resulted in significant decrease of spleen and liver volumes and significant increase of hemoglobin and platelets, with a similar rate of change to patients who had received eliglustat in the double-blind period. Eliglustat treatment was also associated with improvement in bone marrow burden score, bone mineral density, and established biomarkers of Gaucher disease, including reduction of the bioactive lipid, glucosylsphingosine. These findings underscore the efficacy of eliglustat in treatment-naïve patients. Eliglustat was well-tolerated, and there were no new safety concerns with longer-term exposure.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Terapia de Reposição de Enzimas , Doença de Gaucher/tratamento farmacológico , Pirrolidinas/uso terapêutico , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/efeitos adversos , Seguimentos , Doença de Gaucher/diagnóstico , Doença de Gaucher/enzimologia , Glucosilceramidase/antagonistas & inibidores , Humanos , Fígado/patologia , Tamanho do Órgão , Pirrolidinas/administração & dosagem , Pirrolidinas/efeitos adversos , Baço/patologia , Resultado do Tratamento
12.
BMC Genomics ; 17 Suppl 2: 444, 2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27357693

RESUMO

BACKGROUND: Next generation sequencing (NGS) provides a key technology for deciphering the genetic underpinnings of human diseases. Typical NGS analyses of a patient depict tens of thousands non-reference coding variants, but only one or very few are expected to be significant for the relevant disorder. In a filtering stage, one employs family segregation, rarity in the population, predicted protein impact and evolutionary conservation as a means for shortening the variation list. However, narrowing down further towards culprit disease genes usually entails laborious seeking of gene-phenotype relationships, consulting numerous separate databases. Thus, a major challenge is to transition from the few hundred shortlisted genes to the most viable disease-causing candidates. RESULTS: We describe a novel tool, VarElect ( http://ve.genecards.org ), a comprehensive phenotype-dependent variant/gene prioritizer, based on the widely-used GeneCards, which helps rapidly identify causal mutations with extensive evidence. The GeneCards suite offers an effective and speedy alternative, whereby >120 gene-centric automatically-mined data sources are jointly available for the task. VarElect cashes on this wealth of information, as well as on GeneCards' powerful free-text Boolean search and scoring capabilities, proficiently matching variant-containing genes to submitted disease/symptom keywords. The tool also leverages the rich disease and pathway information of MalaCards, the human disease database, and PathCards, the unified pathway (SuperPaths) database, both within the GeneCards Suite. The VarElect algorithm infers direct as well as indirect links between genes and phenotypes, the latter benefitting from GeneCards' diverse gene-to-gene data links in GenesLikeMe. Finally, our tool offers an extensive gene-phenotype evidence portrayal ("MiniCards") and hyperlinks to the parent databases. CONCLUSIONS: We demonstrate that VarElect compares favorably with several often-used NGS phenotyping tools, thus providing a robust facility for ranking genes, pointing out their likelihood to be related to a patient's disease. VarElect's capacity to automatically process numerous NGS cases, either in stand-alone format or in VCF-analyzer mode (TGex and VarAnnot), is indispensable for emerging clinical projects that involve thousands of whole exome/genome NGS analyses.


Assuntos
Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Algoritmos , Mineração de Dados , Bases de Dados Genéticas , Genoma Humano , Humanos , Fenótipo
13.
Am J Gastroenterol ; 111(2): 275-84, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26729549

RESUMO

OBJECTIVES: Hereditary biallelic mismatch repair deficiency (BMMRD) is caused by biallelic mutations in the mismatch repair (MMR) genes and manifests features of neurofibromatosis type 1, gastrointestinal (GI) polyposis, and GI, brain, and hematological cancers. This is the first study to characterize the GI phenotype in BMMRD using both retrospective and prospective surveillance data. METHODS: The International BMMRD Consortium was created to collect information on BMMRD families referred from around the world. All patients had germline biallelic MMR mutations or lack of MMR protein staining in normal and tumor tissue. GI screening data were obtained through medical records with annual updates. RESULTS: Thirty-five individuals from seven countries were identified with BMMRD. GI data were available on 24 of 33 individuals (73%) of screening age, totaling 53 person-years. The youngest age of colonic adenomas was 7, and small bowel adenoma was 11. Eight patients had 19 colorectal adenocarcinomas (CRC; median age 16.7 years, range 8-25), and 11 of 18 (61%) CRC were distal to the splenic flexure. Eleven patients had 15 colorectal surgeries (median 14 years, range 9-25). Four patients had five small bowel adenocarcinomas (SBC; median 18 years, range 11-33). Two CRC and two SBC were detected during surveillance within 6-11 months and 9-16 months, respectively, of last consecutive endoscopy. No patient undergoing surveillance died of a GI malignancy. Familial clustering of GI cancer was observed. CONCLUSIONS: The prevalence and penetrance of GI neoplasia in children with BMMRD is high, with rapid development of carcinoma. Colorectal and small bowel surveillance should commence at ages 3-5 and 8 years, respectively.


Assuntos
Adenocarcinoma/cirurgia , Adenoma/cirurgia , Neoplasias Encefálicas/fisiopatologia , Neoplasias Colorretais/cirurgia , Intestino Delgado/cirurgia , Síndromes Neoplásicas Hereditárias/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/etiologia , Adenocarcinoma/genética , Adenoma/etiologia , Adenoma/genética , Adenosina Trifosfatases/genética , Adolescente , Adulto , Alelos , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/etiologia , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Neoplasias Colorretais/complicações , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/fisiopatologia , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Feminino , Mutação em Linhagem Germinativa , Glioma/etiologia , Humanos , Neoplasias Intestinais/etiologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/cirurgia , Neoplasias Renais/etiologia , Leucemia/etiologia , Linfoma/etiologia , Masculino , Melanoma/etiologia , Endonuclease PMS2 de Reparo de Erro de Pareamento , Proteína 1 Homóloga a MutL , Síndromes Neoplásicas Hereditárias/complicações , Síndromes Neoplásicas Hereditárias/genética , Proteínas Nucleares/genética , Fenótipo , Estudos Prospectivos , Estudos Retrospectivos , Tumor de Wilms/etiologia , Adulto Jovem
14.
Blood Cells Mol Dis ; 60: 65-72, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26051481

RESUMO

Bone crises in type 1 Gaucher disease are reported in long bones and occasionally in weight bearing bones and other bones, but rarely in small bones of the hands and feet. We retrospectively examined the incidence of bone pain in patients followed at the Rabin Medical Center, Israel, before and following the initiation of enzyme replacement therapy (ERT) and evaluated them for bone crises. Of 100 type I Gaucher disease patients, 30 (30%) experienced one or more bone crises. Small bone crises represented 31.5% of all bone crises and were always preceded by crises in other bones. While the incidence of long bone crises reduced after the initiation of ERT, small bone crises increased. Almost 60% of patients with bone crises were of the N370S/84GG genotype suggesting a greater susceptibility of N370S/84GG patients to severe bone complications. These patients also underwent the greatest number of splenectomies (70.6% of splenectomised patients). Splenectomised patients showed a trend towards increased long and small bone crises after surgery. Active investigation of acute pain in the hands and feet in patients in our cohort has revealed a high incidence of small bone crises. Physicians should consider imaging studies to investigate unexplained pain in these areas.


Assuntos
Osso e Ossos/patologia , Ossos do Pé/patologia , Doença de Gaucher/complicações , Ossos da Mão/patologia , Ossos da Perna/patologia , Dor/etiologia , Adolescente , Adulto , Criança , Terapia de Reposição de Enzimas/efeitos adversos , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Esplenectomia , Adulto Jovem
15.
Am J Med Genet A ; 170(9): 2338-48, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27271787

RESUMO

One of the goals of evaluating a patient in the genetics clinic is to find the diagnosis that would explain his or her clinical presentation. Sometimes the patient's diagnosis remains undefined or does not explain all of the clinical findings. As clinicians are often guided by a "single disorder" paradigm, diagnosing multiple genetic conditions in the same patient requires a heightened sense of awareness. Over the last few years, we evaluated several patients (n = 14) who were found to have more than one genetic diagnosis. In this paper, we will describe their natural history and diagnoses, and draw on the lessons learned from this phenomenon, which we expect to grow in this era of next-generation diagnostic technologies. To our knowledge, this is by far the largest series of patients with double diagnoses. Based on our findings, we strongly recommend that physicians question every diagnosis to determine whether it indeed explains all of the patients' symptoms, and consider whether they should continue the diagnostic evaluation to look for a more accurate and complete set of diagnoses. © 2016 Wiley Periodicals, Inc.


Assuntos
Estudos de Associação Genética , Doenças Genéticas Inatas/diagnóstico , Doenças Genéticas Inatas/genética , Adolescente , Adulto , Aneuploidia , Criança , Pré-Escolar , Deleção Cromossômica , Duplicação Cromossômica , Tomada de Decisão Clínica , Feminino , Doenças Genéticas Inatas/terapia , Testes Genéticos , Variação Genética , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Adulto Jovem
16.
Pediatr Blood Cancer ; 63(3): 418-27, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26544533

RESUMO

BACKGROUND: Heterozygous germline mutations in any of the mismatch repair (MMR) genes, MLH1, MSH2, MSH6, and PMS2, cause Lynch syndrome (LS), an autosomal dominant cancer predisposition syndrome conferring a high risk of colorectal, endometrial, and other cancers in adulthood. Offspring of couples where both spouses have LS have a 1:4 risk of inheriting biallelic MMR gene mutations. These cause constitutional MMR deficiency (CMMRD) syndrome, a severe recessively inherited cancer syndrome with a broad tumor spectrum including mainly hematological malignancies, brain tumors, and colon cancer in childhood and adolescence. Many CMMRD children also present with café au lait spots and axillary freckling mimicking neurofibromatosis type 1. PROCEDURE: We describe our experience in seven CMMRD families demonstrating the role and importance of founder mutations and consanguinity on its prevalence. Clinical presentations included brain tumors, colon cancer, lymphoma, and small bowel cancer. RESULTS: In children from two nonconsanguineous Ashkenazi Jewish (AJ) families, the common Ashkenazi founder mutations were detected; these were homozygous in one family and compound heterozygous in the other. In four consanguineous families of various ancestries, different homozygous mutations were identified. In a nonconsanguineous Caucasus/AJ family, lack of PMS2 was demonstrated in tumor and normal tissues; however, mutations were not identified. CONCLUSIONS: CMMRD is rare, but, especially in areas where founder mutations for LS and consanguinity are common, pediatricians should be aware of it since they are the first to encounter these children. Early diagnosis will enable tailored cancer surveillance in the entire family and a discussion regarding prenatal genetic diagnosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina Trifosfatases/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Consanguinidade , Reparo de Erro de Pareamento de DNA/genética , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Efeito Fundador , Proteína 2 Homóloga a MutS/genética , Proteínas Nucleares/genética , Adolescente , Manchas Café com Leite/genética , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Israel , Linfoma/genética , Masculino , Endonuclease PMS2 de Reparo de Erro de Pareamento , Proteína 1 Homóloga a MutL , Mutação , Linhagem , Adulto Jovem
17.
Breast J ; 22(6): 662-666, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27533489

RESUMO

Data on genetic anticipation in breast cancer are sparse. We sought to evaluate age at diagnosis of breast cancer in daughters with a BRCA mutation and their mothers. A review of all carriers of the BRCA mutation diagnosed with breast cancer at the Genetics Institute of a tertiary medical center in 2000-2013 yielded 80 women who could be paired with a mother with breast cancer who was either a carrier of the BRCA mutation or an obligate carrier according to pedigree analysis. Age at diagnosis, type of mutation (BRCA1, BRCA2), year of birth, and ethnicity were recorded. Paired t-test was used to analyze differences in age at cancer diagnosis between groups and subgroups. Mean age at diagnosis of breast cancer was 50.74 years (range 22-88) in the mothers and 43.85 years (range 24-75) in the daughters. The difference was statistically significant (p < 0.001). These findings were consistent regardless of type of BRCA mutation, ethnicity, or mother's year of birth. However, on separate analysis of pairs in which the mother was diagnosed before the age of 50 years, there was no significant difference in mean age at diagnosis between mothers and daughters (~42 years for both). Daughters who carry a BRCA mutation are diagnosed with breast cancer at an earlier age than their carrier mothers, with the exception of pairs in which the mother was diagnosed before the age of 50 years. Future breast-screening guidelines may need to target specific subpopulations of BRCA mutation carriers.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias da Mama/genética , Mutação , Adulto , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Feminino , Predisposição Genética para Doença , Heterozigoto , Humanos , Israel , Judeus/genética , Pessoa de Meia-Idade , Adulto Jovem
20.
Blood Cells Mol Dis ; 55(4): 415-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26460268

RESUMO

BACKGROUND: Lifelong intravenous (IV) enzyme replacement therapy (ERT) every other week for Gaucher disease is appreciated as decreasing quality of life in a palpable way. OBJECTIVE: To review the Israeli experience with the home therapy option for IV velaglucerase alfa (Shire, Lexington MA USA) infusions every-other-week in the clinical trial context, in the early access program (EAP) during a shortage with the standard commercial ERT, and currently with the commercially available drug (VPRIV, Shire). RESULTS: Among 24 patients participating in trials, 1654 infusions were at home; in the EAP and commercial setting, 12,191 infusions were performed at home for a total of 154 patients with 98.4% compliance. There were no incidents of serious adverse events. CONCLUSION: This is the first review of experience of 174 patients and 13,845 intravenous infusions of velaglucerase alfa for Gaucher in the home setting, underscoring its safety.


Assuntos
Terapia de Reposição de Enzimas , Doença de Gaucher/tratamento farmacológico , Glucosilceramidase/administração & dosagem , Terapia por Infusões no Domicílio , Esquema de Medicação , Terapia de Reposição de Enzimas/efeitos adversos , Terapia de Reposição de Enzimas/métodos , Terapia por Infusões no Domicílio/efeitos adversos , Humanos , Adesão à Medicação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA