Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Prenat Diagn ; 44(2): 237-246, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37632214

RESUMO

OBJECTIVE: Recurrent deletions involving 17q12 are associated with a variety of clinical phenotypes, including congenital abnormalities of the kidney and urinary tract (CAKUT), maturity onset diabetes of the young, type 5, and neurodevelopmental disorders. Structural and/or functional renal disease is the most common phenotypic feature, although the prenatal renal phenotypes and the postnatal correlates have not been well characterized. METHOD: We reviewed pre- and postnatal medical records of 26 cases with prenatally or postnatally identified 17q12/HNF1B microdeletions (by chromosomal microarray or targeted gene sequencing), obtained through a multicenter collaboration. We specifically evaluated 17 of these cases (65%) with reported prenatal renal ultrasound findings. RESULTS: Heterogeneous prenatal renal phenotypes were noted, most commonly renal cysts (41%, n = 7/17) and echogenic kidneys (41%), although nonspecific dysplasia, enlarged kidneys, hydronephrosis, pelvic kidney with hydroureter, and lower urinary tract obstruction were also reported. Postnatally, most individuals developed renal cysts (73%, 11/15 live births), and there were no cases of end-stage renal disease during childhood or the follow-up period. CONCLUSION: Our findings demonstrate that copy number variant analysis to assess for 17q12 microdeletion should be considered for a variety of prenatally detected renal anomalies. It is important to distinguish 17q12 microdeletion from other etiologies of CAKUT as the prognosis for renal function and presence of associated findings are distinct and may influence pregnancy and postnatal management.


Assuntos
Doenças Renais Císticas , Nefropatias , Anormalidades Urogenitais , Refluxo Vesicoureteral , Gravidez , Feminino , Humanos , Deleção Cromossômica , Rim/diagnóstico por imagem , Rim/anormalidades , Nefropatias/congênito , Fenótipo , Doenças Renais Císticas/diagnóstico por imagem , Doenças Renais Císticas/genética , Fator 1-beta Nuclear de Hepatócito/genética , Estudos Multicêntricos como Assunto
2.
Res Sq ; 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37790327

RESUMO

Activation-induced cytidine deaminase (AID) is a B cell-specific base editor required during class switch recombination and somatic hypermutation for B cell maturation and antibody diversification. However, it has also been implicated as a factor in the etiology of several B cell malignancies. Evaluating the AID-induced mutation load in patients at-risk for certain types of blood cancers is critical in assessing disease severity and treatment options. Here, we have developed a digital PCR (dPCR) assay that allows us to track the mutational landscape resulting from AID modification or DNA double-strand break (DSB) formation and repair at sites known to be prone to DSBs. Implementation of this new assay showed that increased AID levels in immature B cells increases genome instability at loci linked to translocation formation. This included the CRLF2 locus that is often involved in chromosomal translocations associated with a subtype of acute lymphoblastic leukemia (ALL) that disproportionately affects Latin Americans (LAs). To support this LA-specific identification of AID mutation signatures, we characterized DNA from immature B cells isolated from the bone marrow of ALL patients. Our ability to detect and quantify these mutation signatures will potentiate future risk identification, early detection of cancers, and reduction of associated cancer health disparities.

3.
Cancer Genet ; 248-249: 39-48, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33065430

RESUMO

Trisomy 3 has been previously reported in association with T-cell lymphomas and less commonly in different types of non-Hodgkin B-cell lymphomas. Trisomy 3 has also been reported in two cases of pediatric post-transplant lymphoproliferative disorder (PTLD). We present comprehensive clinicopathologic review of two pediatric patients with cardiac and liver/intestinal allografts that developed polymorphic PTLD characterized by trisomy 3. Both patients had Epstein-Barr virus (EBV) viremia and EBV was positive in tissue by EBER in situ hybridization. Using karyotype analysis and fluorescence in situ hybridization, we identified trisomy 3 in both patients. Both patients responded to treatment and are now free of the PTLD. Trisomy 3, an uncommon cytogenetic finding in pediatric polymorphic PTLD, may be a recurrent cytogenetic aberration if confirmed in a larger study of pediatric PTLDs. Further clinical follow up might help stratify significance of trisomy 3 as a prognostic factor.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 3/genética , Infecções por Vírus Epstein-Barr/epidemiologia , Transplante de Coração/efeitos adversos , Transplante de Fígado/efeitos adversos , Transtornos Linfoproliferativos/etiologia , Trissomia/patologia , Adolescente , Cardiomiopatia Dilatada/cirurgia , Criança , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/virologia , Feminino , Herpesvirus Humano 4/isolamento & purificação , Humanos , Enteropatias/cirurgia , Intestinos/transplante , Cariotipagem , Hepatopatias/cirurgia , Transtornos Linfoproliferativos/complicações , Transtornos Linfoproliferativos/virologia , Recidiva , Trissomia/genética
4.
J Mol Med (Berl) ; 97(12): 1711-1722, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31834445

RESUMO

The phenotypic spectrum of congenital heart defects (CHDs) is contributed by both genetic and environmental factors. Their interactions are profoundly heterogeneous but may operate on common pathways as in the case of hypoxia signaling during postnatal heart development in the context of CHDs. Tetralogy of Fallot (TOF) is the most common cyanotic (hypoxemic) CHD. However, how the hypoxic environment contributes to TOF pathogenesis after birth is poorly understood. We performed Genome-wide transcriptome analysis on right ventricle outflow tract (RVOT) specimens from cyanotic and noncyanotic TOF. Co-expression network analysis identified gene modules specifically associated with clinical diagnosis and hypoxemia status in the TOF hearts. In particular, hypoxia-dependent induction of myocyte proliferation is associated with E2F1-mediated cell cycle regulation and repression of the WNT11-RB1 axis. Genes enriched in epithelial mesenchymal transition (EMT), fibrosis, and sarcomere were also repressed in cyanotic TOF patients. Importantly, transcription factor analysis of the hypoxia-regulated modules suggested CREB1 as a putative regulator of hypoxia/WNT11-RB1 circuit. The study provides a high-resolution landscape of transcriptome programming associated with TOF phenotypes and unveiled hypoxia-induced regulatory circuit in cyanotic TOF. Hypoxia-induced cardiomyocyte proliferation involves negative modulation of CREB1 activity upstream of the WNT11-RB1 axis. KEY MESSAGES: Genetic and environmental factors contribute to congenital heart defects (CHDs). How hypoxia contributes to Tetralogy of Fallot (TOF) pathogenesis after birth is unclear. Systems biology-based analysis revealed distinct molecular signature in CHDs. Gene expression modules specifically associated with cyanotic TOF were uncovered. Key regulatory circuits induced by hypoxia in TOF pathogenesis after birth were unveiled.


Assuntos
Ventrículos do Coração/metabolismo , Hipóxia/metabolismo , Tetralogia de Fallot/metabolismo , Transcriptoma/genética , Criança , Pré-Escolar , Estudos de Coortes , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fator de Transcrição E2F1/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Genoma , Ventrículos do Coração/patologia , Humanos , Lactente , Masculino , Transdução de Sinais/genética , Tetralogia de Fallot/genética , Transcriptoma/fisiologia , Proteínas Wnt/metabolismo
5.
Cancer Genet ; 228-229: 197-217, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30377088

RESUMO

Multiple studies have demonstrated the utility of chromosomal microarray (CMA) testing to identify clinically significant copy number alterations (CNAs) and copy-neutral loss-of-heterozygosity (CN-LOH) in myeloid malignancies. However, guidelines for integrating CMA as a standard practice for diagnostic evaluation, assessment of prognosis and predicting treatment response are still lacking. CMA has not been recommended for clinical work-up of myeloid malignancies by the WHO 2016 or the NCCN 2017 guidelines but is a suggested test by the European LeukaemiaNet 2013 for the diagnosis of primary myelodysplastic syndrome (MDS). The Cancer Genomics Consortium (CGC) Working Group for Myeloid Neoplasms systematically reviewed peer-reviewed literature to determine the power of CMA in (1) improving diagnostic yield, (2) refining risk stratification, and (3) providing additional genomic information to guide therapy. In this manuscript, we summarize the evidence base for the clinical utility of array testing in the workup of MDS, myelodysplastic/myeloproliferative neoplasms (MDS/MPN) and myeloproliferative neoplasms (MPN). This review provides a list of recurrent CNAs and CN-LOH noted in this disease spectrum and describes the clinical significance of the aberrations and how they complement gene mutation findings by sequencing. Furthermore, for new or suspected diagnosis of MDS or MPN, we present suggestions for integrating genomic testing methods (CMA and mutation testing by next generation sequencing) into the current standard-of-care clinical laboratory testing (karyotype, FISH, morphology, and flow).


Assuntos
Variações do Número de Cópias de DNA , Perda de Heterozigosidade , Síndromes Mielodisplásicas/genética , Transtornos Mieloproliferativos/genética , Humanos
6.
Cancer Genet ; 228-229: 218-235, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30344013

RESUMO

Structural genomic abnormalities, including balanced chromosomal rearrangements, copy number gains and losses and copy-neutral loss-of-heterozygosity (CN-LOH) represent an important category of diagnostic, prognostic and therapeutic markers in acute myeloid leukemia (AML). Genome-wide evaluation for copy number abnormalities (CNAs) is at present performed by karyotype analysis which has low resolution and is unobtainable in a subset of cases. Furthermore, examination for possible CN-LOH in leukemia cells is at present not routinely performed in the clinical setting. Chromosomal microarray (CMA) analysis is a widely available assay for CNAs and CN-LOH in diagnostic laboratories, but there are currently no guidelines how to best incorporate this technology into clinical testing algorithms for neoplastic diseases including AML. The Cancer Genomics Consortium Working Group for Myeloid Neoplasms performed an extensive review of peer-reviewed publications focused on CMA analysis in AML. Here we summarize evidence regarding clinical utility of CMA analysis in AML extracted from published data, and provide recommendations for optimal utilization of CMA testing in the diagnostic workup. In addition, we provide a list of CNAs and CN-LOH regions which have documented clinical significance in diagnosis, prognosis and treatment decisions in AML.


Assuntos
Variações do Número de Cópias de DNA , Medicina Baseada em Evidências , Leucemia Mieloide Aguda/genética , Perda de Heterozigosidade , Humanos
7.
Am J Med Genet A ; 176(12): 2829-2834, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30244526

RESUMO

Nonimmune hydrops fetalis (NIHF) is a rare disorder with a high perinatal mortality of at least 50%. One cause of NIHF is generalized lymphatic dysplasia (GLD), a rare form of primary lymphedema of the extremities and systemic involvement including chylothoraces and pericardial effusions. An autosomal recessive form of GLD has been described, caused by variants in the PIEZO1 gene. It has been reported clinically to cause NIHF and childhood onset of facial and limb lymphedema, most of which were diagnosed postnatally. We present a case of a woman with recurrent pregnancies affected by NIHF because of novel compound heterozygous variants in the PIEZO1 gene diagnosed prenatally using exome sequencing (ES). Two variants in PIEZO1 (c.3206G>A and c.6208A>C) were identified that were inherited from the father and mother, and are predicted to cause a nonsense and missense change, respectively, in the PIEZO1 subunits. Ultrasound demonstrated severe bilateral pleural effusions, whole body edema and polyhydramnios. Histopathology revealed an increased number of lymphatic channels, many of which showed failure of luminal canalization. Sanger sequencing confirmed the same variants in a prior fetal demise. We provide phenotypic correlation with ultrasound and autopsy finding, review PIEZO1 variants as a cause of GLD and discuss the uses of prenatal ES to date.


Assuntos
Exoma , Variação Genética , Hidropisia Fetal/diagnóstico , Hidropisia Fetal/genética , Canais Iônicos/genética , Adulto , Autopsia , Biópsia , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Recém-Nascido , Gravidez , Ultrassonografia Pré-Natal , Sequenciamento do Exoma
8.
Int J Surg Pathol ; 24(8): 709-714, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27363433

RESUMO

Triple hit lymphomas are a subset of so-called double hit non-Hodgkin lymphomas exhibiting simultaneous gene translocations/disruption of MYC, BCL2, and BCL6; however, their overlapping morphologic features and complex genetic rearrangements can render classification and prognostication vexing. Clinically triple hit lymphomas are thought to demonstrate aggressive behavior, similar to or worse than that of double hit lymphomas. Only rare reports of long term survivors exist and raise the possibility that unidentified morphologic, immunologic, or cytogenetic differences may impart a less adverse prognosis than current literature and opinion may suggest. Here we report 3 such cases with less aggressive behavior. Cases such as these may prove useful in comparing outcomes, and underlying mechanisms of tumor progression, in aggressive non-Hodgkin lymphomas.


Assuntos
Linfoma não Hodgkin/genética , Linfoma não Hodgkin/patologia , Feminino , Genes myc , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-6/genética , Translocação Genética
9.
Hum Mutat ; 37(2): 148-54, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26507355

RESUMO

Mandibulofacial dysostosis with microcephaly (MFDM) is a multiple malformation syndrome comprising microcephaly, craniofacial anomalies, hearing loss, dysmorphic features, and, in some cases, esophageal atresia. Haploinsufficiency of a spliceosomal GTPase, U5-116 kDa/EFTUD2, is responsible. Here, we review the molecular basis of MFDM in the 69 individuals described to date, and report mutations in 38 new individuals, bringing the total number of reported individuals to 107 individuals from 94 kindreds. Pathogenic EFTUD2 variants comprise 76 distinct mutations and seven microdeletions. Among point mutations, missense substitutions are infrequent (14 out of 76; 18%) relative to stop-gain (29 out of 76; 38%), and splicing (33 out of 76; 43%) mutations. Where known, mutation origin was de novo in 48 out of 64 individuals (75%), dominantly inherited in 12 out of 64 (19%), and due to proven germline mosaicism in four out of 64 (6%). Highly penetrant clinical features include, microcephaly, first and second arch craniofacial malformations, and hearing loss; esophageal atresia is present in an estimated ∼27%. Microcephaly is virtually universal in childhood, with some adults exhibiting late "catch-up" growth and normocephaly at maturity. Occasionally reported anomalies, include vestibular and ossicular malformations, reduced mouth opening, atrophy of cerebral white matter, structural brain malformations, and epibulbar dermoid. All reported EFTUD2 mutations can be found in the EFTUD2 mutation database (http://databases.lovd.nl/shared/genes/EFTUD2).


Assuntos
Anormalidades Múltiplas/genética , Perda Auditiva/genética , Deficiência Intelectual/genética , Disostose Mandibulofacial/genética , Microcefalia/genética , Mutação , Fatores de Alongamento de Peptídeos/genética , Ribonucleoproteína Nuclear Pequena U5/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/patologia , Motivos de Aminoácidos , Bases de Dados Genéticas , Expressão Gênica , Haploinsuficiência , Perda Auditiva/diagnóstico , Perda Auditiva/patologia , Humanos , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/patologia , Disostose Mandibulofacial/diagnóstico , Disostose Mandibulofacial/patologia , Microcefalia/diagnóstico , Microcefalia/patologia , Modelos Moleculares , Dados de Sequência Molecular , Penetrância , Fenótipo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Splicing de RNA , Spliceossomos/genética
10.
Hum Mol Genet ; 24(8): 2375-89, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25574029

RESUMO

Cardiac left ventricular outflow tract (LVOT) defects represent a common but heterogeneous subset of congenital heart disease for which gene identification has been difficult. We describe a 46,XY,t(1;5)(p36.11;q31.2)dn translocation carrier with pervasive developmental delay who also exhibited LVOT defects, including bicuspid aortic valve (BAV), coarctation of the aorta (CoA) and patent ductus arteriosus (PDA). The 1p breakpoint disrupts the 5' UTR of AHDC1, which encodes AT-hook DNA-binding motif containing-1 protein, and AHDC1-truncating mutations have recently been described in a syndrome that includes developmental delay, but not congenital heart disease [Xia, F., Bainbridge, M.N., Tan, T.Y., Wangler, M.F., Scheuerle, A.E., Zackai, E.H., Harr, M.H., Sutton, V.R., Nalam, R.L., Zhu, W. et al. (2014) De Novo truncating mutations in AHDC1 in individuals with syndromic expressive language delay, hypotonia, and sleep apnea. Am. J. Hum. Genet., 94, 784-789]. On the other hand, the 5q translocation breakpoint disrupts the 3' UTR of MATR3, which encodes the nuclear matrix protein Matrin 3, and mouse Matr3 is strongly expressed in neural crest, developing heart and great vessels, whereas Ahdc1 is not. To further establish MATR3 3' UTR disruption as the cause of the proband's LVOT defects, we prepared a mouse Matr3(Gt-ex13) gene trap allele that disrupted the 3' portion of the gene. Matr3(Gt-ex13) homozygotes are early embryo lethal, but Matr3(Gt-ex13) heterozygotes exhibit incompletely penetrant BAV, CoA and PDA phenotypes similar to those in the human proband, as well as ventricular septal defect (VSD) and double-outlet right ventricle (DORV). Both the human MATR3 translocation breakpoint and the mouse Matr3(Gt-ex13) gene trap insertion disturb the polyadenylation of MATR3 transcripts and alter Matrin 3 protein expression, quantitatively or qualitatively. Thus, subtle perturbations in Matrin 3 expression appear to cause similar LVOT defects in human and mouse.


Assuntos
Coartação Aórtica/genética , Valva Aórtica/anormalidades , Permeabilidade do Canal Arterial/genética , Doenças das Valvas Cardíacas/genética , Proteínas Associadas à Matriz Nuclear/genética , Proteínas de Ligação a RNA/genética , Adolescente , Animais , Coartação Aórtica/metabolismo , Valva Aórtica/metabolismo , Doença da Válvula Aórtica Bicúspide , Pré-Escolar , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Permeabilidade do Canal Arterial/metabolismo , Feminino , Inativação Gênica , Doenças das Valvas Cardíacas/metabolismo , Ventrículos do Coração/anormalidades , Ventrículos do Coração/metabolismo , Humanos , Recém-Nascido , Masculino , Camundongos , Mutagênese Insercional , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Translocação Genética
11.
Am J Med Genet A ; 164A(12): 3076-82, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25256560

RESUMO

Deletions of chromosome 17q12 [OMIM 614527] encompass a wide range of phenotypes, including renal cysts, diabetes mellitus, pancreatic structural abnormalities, genital tract anomalies, developmental delay, learning difficulties, and more recently, autism spectrum disorder and schizophrenia. To date, gastrointestinal malformations have not been fully characterized in this syndrome. In this case report, we describe a four-year-old girl with a 17q12 microdeletion who was born with duodenal atresia, bilateral renal cysts, left kidney dysplasia, a midline cystic structure at the conus medullaris, and dysmorphic features. Both the patient and her affected father were found to have a deletion of 17q12, which encompasses the HNF1B (hepatocyte nuclear factor beta). It is hypothesized that HNF1B may play a role in intestinal differentiation and development. Our clinical report further expands the pre-and post-natal presentation of this rare microdeletion syndrome.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Aberrações Cromossômicas , Cromossomos Humanos Par 17/genética , Obstrução Duodenal/genética , Fator 1-beta Nuclear de Hepatócito/deficiência , Fenótipo , Feminino , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Atresia Intestinal , Análise em Microsséries , Síndrome
12.
BMC Med Genet ; 15: 49, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24886118

RESUMO

BACKGROUND: Wiedemann-Steiner Syndrome (WSS) is characterized by short stature, a variety of dysmorphic facial and skeletal features, characteristic hypertrichosis cubiti (excessive hair on the elbows), mild-to-moderate developmental delay and intellectual disability. [MIM#: 605130]. Here we report two unrelated children for whom clinical exome sequencing of parent-proband trios was performed at UCLA, resulting in a molecular diagnosis of WSS and atypical clinical presentation. CASE PRESENTATION: For patient 1, clinical features at 9 years of age included developmental delay, craniofacial abnormalities, and multiple minor anomalies. Patient 2 presented at 1 year of age with developmental delay, microphthalmia, partial 3-4 left hand syndactyly, and craniofacial abnormalities. A de novo missense c.4342T>C variant and a de novo splice site c.4086+G>A variant were identified in the KMT2A gene in patients 1 and 2, respectively. CONCLUSIONS: Based on the clinical and molecular findings, both patients appear to have novel presentations of WSS. As the hallmark hypertrichosis cubiti was not initially appreciated in either case, this syndrome was not suspected during the clinical evaluation. This report expands the phenotypic spectrum of the clinical phenotypes and KMT2A variants associated with WSS.


Assuntos
Anormalidades Múltiplas/genética , Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Proteína de Leucina Linfoide-Mieloide/genética , Anormalidades Múltiplas/diagnóstico , Criança , Biologia Computacional/métodos , Deficiências do Desenvolvimento/diagnóstico , Exoma , Fácies , Feminino , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Histona-Lisina N-Metiltransferase , Humanos , Lactente , Deficiência Intelectual/diagnóstico , Masculino , Fenótipo , Síndrome
13.
Am J Med Genet A ; 161A(7): 1773-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23696469

RESUMO

Deletions of the long arm of chromosome 18 have been previously reported in many patients. Most cases involve the more distal regions of the long arm (18q21.1->qter). However, proximal interstitial deletions involving 18q11.2 are extremely rare. Here we report on a 14-month-old female with a 4.7 Mb (19,667,062-24,401,876 hg19) de novo interstitial deletion within chromosomal band 18q11.2, which includes GATA6 and 24 other RefSeq genes. The clinical features of our patient include complex congenital heart defects, a double outlet right ventricle, a subaortic ventricular septal defect, D-malposed great arteries, an atrial septal defect, a dysplastic aortic valve and patent ductus arteriosus. In addition, she had renal anomalies-a duplicated collecting system on the left and mild right hydronephrosis. These heart and renal defects are not reported in other patients with 18q proximal interstitial deletions. Heterozygous point mutations in GATA6, encoding for a zinc finger transcription factor, have been shown to cause congenital heart defects. Given the well-established biological role of GATA6 in cardiac development, a deletion of GATA6 is very likely responsible for our patient's complex congenital heart defects. This is the smallest and most proximal 18q11.2 deletion involving GATA6 that is associated with complex congenital heart disease and renal anomalies.


Assuntos
Transtornos Cromossômicos/genética , Cardiopatias Congênitas/genética , Rim/anormalidades , Deleção Cromossômica , Transtornos Cromossômicos/etiologia , Cromossomos Humanos Par 18/genética , Feminino , Fator de Transcrição GATA6/genética , Humanos , Lactente , Recém-Nascido , Gravidez
14.
Am J Med Genet A ; 161A(5): 1167-72, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23532965

RESUMO

Pierre Robin sequence (PRS) is a malformation pattern characterized by the core triad of retrognathia, glossoptosis, and cleft palate that causes difficulty in glossopharyngeal-laryngeal-vagal functions. The etiology of PRS remains largely unknown; previous reports have suggested that it is caused by intrauterine constriction or external conditions such as oligohydramnios, breech position, or abnormal uterine anatomy. Genetic causes include occurrence as a manifestation of many single gene conditions and chromosomal rearrangements. Positional effect on some loci or genes, including SOX9 has also been posited as a cause. Here, we report on an 18-month-old girl born with isolated PRS. Clinical chromosome microarray analysis (CMA) revealed a maternally inherited ~623 kb microdeletion that is -725 kb upstream of 5' SOX9 at chromosome locus 17q24.3. Her mother had cleft palate. This region, although devoid of any genes, is known to have a position effect on SOX9 due to elimination of highly conserved non-coding cis-regulatory elements. This report supports the evidence that deregulation of an intact SOX9 coding region is a cause of or associated with isolated PRS, and provides further evidence that CMA in the clinical setting is a powerful tool in detecting microdeletions in gene "desert" regions that have pathogenic position effect on specific genes.


Assuntos
Fibromatose Gengival/genética , Hipertricose/genética , Síndrome de Pierre Robin/genética , Fatores de Transcrição SOX9/genética , Deleção Cromossômica , Cromossomos Humanos Par 17/genética , Feminino , Humanos , Lactente , Análise de Sequência com Séries de Oligonucleotídeos
15.
Pediatr Blood Cancer ; 56(3): 470-3, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21225931

RESUMO

Duplication 22q11.2 syndrome is the result of a microduplication of the same chromosomal region that is deleted in DiGeorge and Velocardiofacial syndromes. We describe a patient with dysmorphic features who was diagnosed with pre-B acute lymphoblastic leukemia, and developed cystinuria and pancreatitis during treatment. Duplication 22q11.2 has not been previously described in association with hematologic abnormalities. Chromosomal microarray technology was used to diagnose duplication 22q11.2 syndrome. In this era of advanced genomics, this technology has become an important method for helping to determine the molecular basis of diseases, best treatments and ultimately patient outcomes.


Assuntos
Cistinúria/etiologia , Duplicação Gênica , Pancreatite/etiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/etiologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Criança , Cromossomos Humanos Par 22/genética , Cistinúria/tratamento farmacológico , Síndrome de DiGeorge/complicações , Síndrome de DiGeorge/genética , Face/anormalidades , Perfilação da Expressão Gênica , Humanos , Hibridização in Situ Fluorescente , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Pancreatite/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Resultado do Tratamento
16.
Am J Med Genet A ; 152A(10): 2459-67, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20830797

RESUMO

The screening of individuals with mild dysmorphic features and mental retardation using whole genome scanning technologies has resulted in the delineation of several previously unrecognized microdeletion syndromes. Microdeletion of 3q29 has been recently described as one such new syndrome. The clinical phenotype is variable despite an almost identical submicroscopic deletion size in most cases. We report on two individuals that further expand the clinical presentation of this rare disorder and compare the findings with earlier reports to refine the 3q29 microdeletion syndrome phenotype. The propositi are a 10-year-old female and a 15-year-old male, who have in common intellectual disabilities, a history of autism and psychiatric symptoms ranging from bipolar disorder presenting with increasing suicidal ideation to aggressive behavior and general anxiety. Other shared physical findings include asymmetric face, high-nasal bridge, crowded/dysplastic teeth, and tapered fingers. Oligonucleotide array-based chromosomal microarray analysis (CMA) using a genome-wide SNP array identified a de novo subtelomeric microdeletion of chromosome region 3q29 ranging from 1.6 to 2.1 Mb. The region of overlap encompasses 20 RefSeq genes, including FBX045, DLG1, and PAK2. These genes are related to neuronal postsynaptic membrane function and PTEN signaling, suggesting a role for synaptic connectivity dysfunction in the etiology of autism in these children. The novel clinical presentation of our patients expands the clinical spectrum of the 3q29 microdeletion syndrome and provides additional insights into the pathophysiology of autism and psychiatric disorders.


Assuntos
Transtorno Autístico/genética , Transtorno Bipolar/genética , Cromossomos Humanos Par 3 , Deficiência Intelectual/genética , Transtornos Psicóticos/genética , Deleção de Sequência , Adolescente , Adulto , Criança , Mapeamento Cromossômico , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , PTEN Fosfo-Hidrolase/genética , Idade Paterna , Fenótipo , Sinapses/fisiologia , Síndrome
18.
Pediatr Hematol Oncol ; 26(6): 426-31, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19657992

RESUMO

The authors report a male infant born at 35 weeks gestational age with an atypical presentation of homozygous alpha-thalassemia. The live-born infant displayed abnormalities of the upper limbs and genitalia, which are vascular-type disruptive defects associated with this disease. Cardiomegaly and placentomegaly were the only evidence of fetal hydrops. Postnatal karyotype revealed mosaicism for trisomy 7, yet another rare finding in a live-born. The authors discuss their institutional experience with each of these rare conditions and the potential contribution of each to the overall unusual clinical presentation in this patient. This is the first report of these simultaneous diagnoses.


Assuntos
Anormalidades Múltiplas/genética , Cromossomos Humanos Par 7/genética , Mosaicismo , Trissomia/genética , Talassemia alfa/genética , Adulto , Evolução Fatal , Feminino , Homozigoto , Humanos , Hidropisia Fetal/genética , Lactente , Masculino , Gravidez , Ultrassonografia Pré-Natal
19.
Cancer Genet Cytogenet ; 190(1): 33-9, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19264231

RESUMO

In a retrospective analysis of a large group of cases (n=291) with B-cell CLL diagnosis, the various characteristics of IGH aberrations as identified by fluorescence in situ hybridization (FISH) probes were studied. Conventional cytogenetic and FISH studies with the standard panel (13q14, 11q13, 17p13, 12 centromere), and with IGH break-apart probes were done for each case. Abnormal karyotypes were detected with conventional cytogenetics in 29% of cases, and FISH detected abnormalities in 70%. Deletion of 13q14 was the most frequent anomaly, followed by trisomy 12, deletion of 11q, and deletion of 17p. Among the IGH abnormalities detected, translocations with unknown partners (split signals) occurred in only a small group of patients (15%). Instead, deletion of 5'IGH, corresponding to the variable IGH segment (IGH(V)) was the most recurrent aberration, observed in 82% (the second most common finding among our patients). This deletion was associated with good prognostic markers: 13q14 deletion, normal karyotype, and CD38 and ZAP-70 negative expression. Although not exclusive to CLL, the deletion occurred in a high frequency, in contrast to its rarity in other B-cell lymphoproliferative disorders. Longitudinal studies are warranted, to determine when in the disease progression this abnormality is acquired, as a potential early marker, and its impact on the natural history of CLL.


Assuntos
Região 5'-Flanqueadora/genética , Deleção de Genes , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia Linfocítica Crônica de Células B/genética , ADP-Ribosil Ciclase 1/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Aberrações Cromossômicas , Cromossomos Humanos Par 12 , Cromossomos Humanos Y , Feminino , Regulação Leucêmica da Expressão Gênica , Frequência do Gene , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Proteína-Tirosina Quinase ZAP-70/genética
20.
Prenat Diagn ; 26(10): 966-72, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16906598

RESUMO

Apert syndrome was diagnosed in a newborn with typical facial and digital features whose only detected prenatal abnormality had been agenesis of the corpus callosum. This prompted a review of the central nervous system findings in all cases of Apert syndrome treated at the Craniofacial Center Boston Children's Hospital between 1978 and 2004. Two of 30 patients with Apert syndrome had prenatal identification of mild dilatation of the lateral cerebral ventricles and complete agenesis of the corpus callosum (ACC) documented with both ultrasound and MRI. Both had the common S252W mutation of FGFR2. Though cranial and orbital malformations typical of Apert were eventually seen in these fetuses in the third-trimester, even in retrospect, these were not detectable at mid second-trimester, ultrasound screening for congenital malformations. Hand malformations also went undetected in the second-trimester despite extensive imaging by experienced radiologists. We conclude that prenatal ultrasonographic identification of mild ventriculomegaly or ACC should stimulate a careful search for features of Apert syndrome and prompt follow-up imaging to look for bony abnormalities that have later onset. Prenatal molecular testing for Apert mutations should be considered in cases of mild ventriculomegaly and ACC.


Assuntos
Acrocefalossindactilia/diagnóstico , Diagnóstico Pré-Natal , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Acrocefalossindactilia/genética , Adolescente , Feminino , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Mutação , Gravidez , Resultado da Gravidez , Tomografia Computadorizada por Raios X , Ultrassonografia Pré-Natal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA