Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Chemistry ; 25(59): 13491-13495, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31437315

ABSTRACT

Reaction of N-heterocyclic carbene (NHC)-stabilized PGeP-type germylene Ge{o-(PiPr2 )C6 H4 }2 ⋅Me IiPr (1) (Me IiPr=1,3-diisopropyl-4,5-dimethylimidazol-2-ylidene) with Ni(cod)2 gave pincer germylene complex Ni[Ge{o-(PiPr2 )C6 H4 }2 ](Me IiPr) (2), in which the Ge center of 2 is significantly pyramidalized. Theoretical calculation on 2 predicted the ambiphilicity of the germanium center, which was confirmed by reactivity studies. Thus, complex 2 reacted with both Lewis base Me IMe (Me IMe=1,3,4,5-tetramethylimidazol-2-ylidene) and Lewis acid BH3 ⋅SMe2 at the germanium center to afford the adducts Ni[Ge{o-(PiPr2 )C6 H4 }2 ⋅Me IMe](Me IiPr) (3) and Ni[Ge{o-(PiPr2 )C6 H4 }2 ⋅BH3 ](Me IiPr) (4), respectively. Furthermore, the former was slowly converted to dinuclear complex Ni2 [Ge{o-(PiPr2 )C6 H4 }2 ]2 (Me IMe)2 (5) at room temperature. Complex 5 can be regarded as a dimer of the Me IMe analog of 2 with a Ni-Ge-Ge-Ni linkage.

2.
Nat Commun ; 8(1): 767, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28974674

ABSTRACT

Although diabetes results in part from a deficiency of normal pancreatic beta cells, inducing human beta cells to regenerate is difficult. Reasoning that insulinomas hold the "genomic recipe" for beta cell expansion, we surveyed 38 human insulinomas to obtain insights into therapeutic pathways for beta cell regeneration. An integrative analysis of whole-exome and RNA-sequencing data was employed to extensively characterize the genomic and molecular landscape of insulinomas relative to normal beta cells. Here, we show at the pathway level that the majority of the insulinomas display mutations, copy number variants and/or dysregulation of epigenetic modifying genes, most prominently in the polycomb and trithorax families. Importantly, these processes are coupled to co-expression network modules associated with cell proliferation, revealing candidates for inducing beta cell regeneration. Validation of key computational predictions supports the concept that understanding the molecular complexity of insulinoma may be a valuable approach to diabetes drug discovery.Diabetes results in part from a deficiency of functional pancreatic beta cells. Here, the authors study the genomic and epigenetic landscapes of human insulinomas to gain insight into possible pathways for therapeutic beta cell regeneration, highlighting epigenetic genes and pathways.


Subject(s)
Cell Proliferation/genetics , Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/metabolism , Insulinoma/genetics , Pancreatic Neoplasms/genetics , Regeneration/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Diabetes Mellitus, Type 1/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Insulinoma/metabolism , Male , Middle Aged , Pancreatic Neoplasms/metabolism
3.
Cold Spring Harb Mol Case Stud ; 3(3): a001602, 2017 05.
Article in English | MEDLINE | ID: mdl-28487882

ABSTRACT

Cushing's disease (CD) is caused by pituitary corticotroph adenomas that secrete excess adrenocorticotropic hormone (ACTH). In these tumors, somatic mutations in the gene USP8 have been identified as recurrent and pathogenic and are the sole known molecular driver for CD. Although other somatic mutations were reported in these studies, their contribution to the pathogenesis of CD remains unexplored. No molecular drivers have been established for a large proportion of CD cases and tumor heterogeneity has not yet been investigated using genomics methods. Also, even in USP8-mutant tumors, a possibility may exist of additional contributing mutations, following a paradigm from other neoplasm types where multiple somatic alterations contribute to neoplastic transformation. The current study utilizes whole-exome discovery sequencing on the Illumina platform, followed by targeted amplicon-validation sequencing on the Pacific Biosciences platform, to interrogate the somatic mutation landscape in a corticotroph adenoma resected from a CD patient. In this USP8-mutated tumor, we identified an interesting somatic mutation in the gene RASD1, which is a component of the corticotropin-releasing hormone receptor signaling system. This finding may provide insight into a novel mechanism involving loss of feedback control to the corticotropin-releasing hormone receptor and subsequent deregulation of ACTH production in corticotroph tumors.


Subject(s)
ACTH-Secreting Pituitary Adenoma/genetics , ras Proteins/genetics , Adenoma/genetics , Adrenocorticotropic Hormone/genetics , Adult , Corticotrophs/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Female , Humans , Mutation , Pituitary ACTH Hypersecretion/genetics , Pituitary Neoplasms/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Sequence Analysis, DNA , Ubiquitin Thiolesterase/genetics
4.
Genome Med ; 8(1): 62, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27245685

ABSTRACT

BACKGROUND: Personalized therapy provides the best outcome of cancer care and its implementation in the clinic has been greatly facilitated by recent convergence of enormous progress in basic cancer research, rapid advancement of new tumor profiling technologies, and an expanding compendium of targeted cancer therapeutics. METHODS: We developed a personalized cancer therapy (PCT) program in a clinical setting, using an integrative genomics approach to fully characterize the complexity of each tumor. We carried out whole exome sequencing (WES) and single-nucleotide polymorphism (SNP) microarray genotyping on DNA from tumor and patient-matched normal specimens, as well as RNA sequencing (RNA-Seq) on available frozen specimens, to identify somatic (tumor-specific) mutations, copy number alterations (CNAs), gene expression changes, gene fusions, and also germline variants. To provide high sensitivity in known cancer mutation hotspots, Ion AmpliSeq Cancer Hotspot Panel v2 (CHPv2) was also employed. We integrated the resulting data with cancer knowledge bases and developed a specific workflow for each cancer type to improve interpretation of genomic data. RESULTS: We returned genomics findings to 46 patients and their physicians describing somatic alterations and predicting drug response, toxicity, and prognosis. Mean 17.3 cancer-relevant somatic mutations per patient were identified, 13.3-fold, 6.9-fold, and 4.7-fold more than could have been detected using CHPv2, Oncomine Cancer Panel (OCP), and FoundationOne, respectively. Our approach delineated the underlying genetic drivers at the pathway level and provided meaningful predictions of therapeutic efficacy and toxicity. Actionable alterations were found in 91 % of patients (mean 4.9 per patient, including somatic mutations, copy number alterations, gene expression alterations, and germline variants), a 7.5-fold, 2.0-fold, and 1.9-fold increase over what could have been uncovered by CHPv2, OCP, and FoundationOne, respectively. The findings altered the course of treatment in four cases. CONCLUSIONS: These results show that a comprehensive, integrative genomic approach as outlined above significantly enhanced genomics-based PCT strategies.


Subject(s)
Genetic Variation , Genomics/methods , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine/methods , Adolescent , Adult , Aged , Child , DNA Copy Number Variations , Exome , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Neoplasms/pathology , Polymorphism, Single Nucleotide , Prognosis , Young Adult
5.
Nat Commun ; 6: 6087, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25608663

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is a fatal bile duct cancer with dismal prognosis and limited therapeutic options. By performing RNA- and exome-sequencing analyses, we report a novel fusion event, FGFR2-PPHLN1 (16%), and damaging mutations in the ARAF oncogene (11%). Here we demonstrate that the chromosomal translocation t(10;12)(q26;q12) leading to FGFR2-PPHLN1 fusion possesses transforming and oncogenic activity, which is successfully inhibited by a selective FGFR2 inhibitor in vitro. Among the ARAF mutations, N217I and G322S lead to activation of the pathway and N217I shows oncogenic potential in vitro. Screening of a cohort of 107 iCCA patients reveals that FGFR2 fusions represent the most recurrent targetable alteration (45%, 17/107), while they are rarely present in other primary liver tumours (0/100 of hepatocellular carcinoma (HCC); 1/21 of mixed iCCA-HCC). Taken together, around 70% of iCCA patients harbour at least one actionable molecular alteration (FGFR2 fusions, IDH1/2, ARAF, KRAS, BRAF and FGF19) that is amenable for therapeutic targeting.


Subject(s)
Antigens, Neoplasm/genetics , Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation , Nuclear Proteins/genetics , Proto-Oncogene Proteins A-raf/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , 3T3 Cells , Aged , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Cohort Studies , Exome , Exons , Female , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Middle Aged , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Recombinant Fusion Proteins/chemistry , Sequence Analysis, RNA , Sequence Homology, Amino Acid , Translocation, Genetic
6.
BMC Med Genomics ; 7: 20, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24758382

ABSTRACT

BACKGROUND: Whole exome and genome sequencing (WES/WGS) is now routinely offered as a clinical test by a growing number of laboratories. As part of the test design process each laboratory must determine the performance characteristics of the platform, test and informatics pipeline. This report documents one such characterization of WES/WGS. METHODS: Whole exome and whole genome sequencing was performed on multiple technical replicates of five reference samples using the Illumina HiSeq 2000/2500. The sequencing data was processed with a GATK-based genome analysis pipeline to evaluate: intra-run, inter-run, inter-mode, inter-machine and inter-library consistency, concordance with orthogonal technologies (microarray, Sanger) and sensitivity and accuracy relative to known variant sets. RESULTS: Concordance to high-density microarrays consistently exceeds 97% (and typically exceeds 99%) and concordance between sequencing replicates also exceeds 97%, with no observable differences between different flow cells, runs, machines or modes. Sensitivity relative to high-density microarray variants exceeds 95%. In a detailed study of a 129 kb region, sensitivity was lower with some validated single-base insertions and deletions "not called". Different variants are "not called" in each replicate: of all variants identified in WES data from the NA12878 reference sample 74% of indels and 89% of SNVs were called in all seven replicates, in NA12878 WGS 52% of indels and 88% of SNVs were called in all six replicates. Key sources of non-uniformity are variance in depth of coverage, artifactual variants resulting from repetitive regions and larger structural variants. CONCLUSION: We report a comprehensive performance characterization of WES/WGS that will be relevant to offering laboratories, consumers of genome sequencing and others interested in the analytical validity of this technology.


Subject(s)
Exome/genetics , Genome, Human/genetics , Sequence Analysis, DNA/methods , Genotype , Humans , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide/genetics , Reference Standards , Reproducibility of Results
7.
Hepatology ; 58(3): 958-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23424026

ABSTRACT

UNLABELLED: Cholesteryl ester storage disease (CESD) and Wolman disease are autosomal recessive later-onset and severe infantile disorders, respectively, which result from the deficient activity of lysosomal acid lipase (LAL). LAL is encoded by LIPA (10q23.31) and the most common mutation associated with CESD is an exon 8 splice junction mutation (c.894G>A; E8SJM), which expresses only ∼3%-5% of normally spliced LAL. However, the frequency of c.894G>A is unknown in most populations. To estimate the prevalence of CESD in different populations, the frequencies of the c.894G>A mutation were determined in 10,000 LIPA alleles from healthy African-American, Asian, Caucasian, Hispanic, and Ashkenazi Jewish individuals from the greater New York metropolitan area and 6,578 LIPA alleles from African-American, Caucasian, and Hispanic subjects enrolled in the Dallas Heart Study. The combined c.894G>A allele frequencies from the two cohorts ranged from 0.0005 (Asian) to 0.0017 (Caucasian and Hispanic), which translated to carrier frequencies of 1 in 1,000 to ∼1 in 300, respectively. No African-American heterozygotes were detected. Additionally, by surveying the available literature, c.894G>A was estimated to account for 60% (95% confidence interval [CI]: 51%-69%) of reported mutations among multiethnic CESD patients. Using this estimate, the predicted prevalence of CESD in the Caucasian and Hispanic populations is ∼0.8 per 100,000 (∼1 in 130,000; 95% CI: ∼1 in 90,000 to 1 in 170,000). CONCLUSION: These data indicate that CESD may be underdiagnosed in the general Caucasian and Hispanic populations, which is important since clinical trials of enzyme replacement therapy for LAL deficiency are currently being developed. Moreover, future studies on CESD prevalence in African and Asian populations may require full-gene LIPA sequencing to determine heterozygote frequencies, since c.894G>A is not common in these racial groups.


Subject(s)
Cholesterol Ester Storage Disease/ethnology , Cholesterol Ester Storage Disease/genetics , Ethnicity/ethnology , Ethnicity/genetics , Mutation/genetics , Sterol Esterase/genetics , Adolescent , Adult , Black or African American/ethnology , Black or African American/genetics , Aged , Aged, 80 and over , Asian/ethnology , Asian/genetics , Exons/genetics , Heterozygote , Hispanic or Latino/ethnology , Hispanic or Latino/genetics , Humans , Jews/ethnology , Jews/genetics , Middle Aged , New York , Prevalence , Retrospective Studies , White People/ethnology , White People/genetics , Young Adult
8.
Anal Chem ; 82(24): 10186-93, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21086978

ABSTRACT

Tamoxifen (Tam), the antiestrogen used to treat estrogen receptor-positive breast cancer is a pro-drug that is converted to its major active metabolites, endoxifen and 4-hydroxy-tamoxifen (4-OH-Tam) by various biotransformation enzymes of which cytochrome P450-2D6 (CYP2D6) is key. The usual Tam dose is 20 mg daily; however, the plasma active metabolite concentrations vary due to common genetic variants encoding the biotransformation enzymes and environmental factors (e.g., concomitant drugs) that inhibit these enzymes. Effective treatment depends on adequate Tam conversion to its active isomers. To monitor metabolite plasma levels, a novel liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed to separate and quantitate Tam, N-desmethyl-tamoxifen (ND-Tam), and tamoxifen-N-oxide (Tam-N-oxide), and the E, Z, and Z' isomers of endoxifen and 4-OH-Tam. Known standards were used to identify each metabolite/isomer. Quantitation of these metabolites in plasma was linear from 0.6 to 2000 nM. Intra- and inter-assay reproducibilities were 0.2-8.4% and 0.6-6.3%, respectively. Accuracy determined by spike experiments with known standards was 86-103%. Endoxifen, 4-OH-Tam, and their isomers were stable in fresh frozen plasma for ≥6 months. This method provides the first sensitive, specific, accurate, and reproducible quantitation of Tam and its metabolite isomers for monitoring Tam-treated breast cancer patients.


Subject(s)
Tamoxifen/isolation & purification , Chromatography, Liquid , Humans , Isomerism , Selective Estrogen Receptor Modulators/isolation & purification , Tamoxifen/analysis , Tamoxifen/metabolism , Tandem Mass Spectrometry
9.
Am J Med Genet A ; 152A(2): 472-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20101700

ABSTRACT

Roberts syndrome (RBS) (OMIM #268300) is a rare autosomal recessive disorder characterized by tetraphocomelia (symmetrical limb reduction), craniofacial anomalies, growth retardation, mental retardation, cardiac and renal abnormalities. The syndrome is caused by mutations in the ESCO2 (establishment of cohesion 1 homolog 2) (Entrez 609353) gene, which is located at 8p21.1, and encodes a protein essential in establishing sister chromatid cohesion during S phase. SC phocomelia (SC) (OMIM #269000), has less severe symmetric limb reduction, flexion contractures of various joints, minor facial anomalies, growth retardation and occasionally, mental retardation. These two syndromes can be considered part of a spectrum, with RBS at the most severe range in which severely affected infants may be stillborn or die in the post-natal period, while individuals with SC phocomelia represent the milder end of the spectrum and typically survive to adulthood. In both presentations, karyotype investigations characteristically reveal premature centromere separation (PCS), otherwise known as heterochromatin repulsion or puffing. There is little literature about the follow-up of adults with the spectrum of RBS/SC phocomelia or their recommended management. We report on an adult presentation of RBS/SC phocomelia spectrum disorder with a history of major cardiac malformation in childhood, normal limbs on physical examination, mild facial anomalies, mild learning difficulties, and PCS. Molecular studies of ESCO2 have confirmed the diagnosis. A literature review, focussing on adult manifestations of this condition and a discussion of follow-up guidelines are presented.


Subject(s)
Ectromelia/genetics , Heart Defects, Congenital/genetics , Syndrome , Abnormalities, Multiple/genetics , Adult , Chromosome Banding , Craniofacial Abnormalities/genetics , DNA Mutational Analysis , Female , Growth Disorders/genetics , Heart Defects, Congenital/surgery , Homozygote , Humans , Karyotyping , Male , Polymerase Chain Reaction
10.
Per Med ; 7(6): 621-632, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21977050

ABSTRACT

The goal of personalized medicine is to treat each patient with the best drug: optimal therapeutic benefit with minimal side effects. The genomic revolution is rapidly identifying the genetic contribution to the diseased state as well as its contribution to drug efficacy and toxicity. The ability to perform genome-wide studies has led to an overwhelming number of candidate genes and/or their associated variants; however, understanding which are of therapeutic importance is becoming the greatest unmet need in the personalized medicine field. A related issue is the need to improve our methods of identifying and characterizing therapeutic drugs in the context of the complex genomic landscape of the intact body. Drosophila have proven to be a powerful tool for understanding the basic biological mechanisms of human development. This article will review Drosophila as a whole animal tool for gene and drug discovery. We will examine how Drosophila can be used to both sort through the myriad of hits coming from human genome-wide scans and to dramatically improve the early steps in pharmaceutical drug development.

12.
PLoS One ; 4(2): e4583, 2009.
Article in English | MEDLINE | ID: mdl-19240791

ABSTRACT

Deletions spanning chromosome 5q31.2 are among the most common recurring cytogenetic abnormalities detectable in myelodysplastic syndromes (MDS). Prior genomic studies have suggested that haploinsufficiency of multiple 5q31.2 genes may contribute to MDS pathogenesis. However, this hypothesis has never been formally tested. Therefore, we designed this study to systematically and comprehensively evaluate all 28 chromosome 5q31.2 genes and directly test whether haploinsufficiency of a single 5q31.2 gene may result from a heterozygous nucleotide mutation or microdeletion. We selected paired tumor (bone marrow) and germline (skin) DNA samples from 46 de novo MDS patients (37 without a cytogenetic 5q31.2 deletion) and performed total exonic gene resequencing (479 amplicons) and array comparative genomic hybridization (CGH). We found no somatic nucleotide changes in the 46 MDS samples, and no cytogenetically silent 5q31.2 deletions in 20/20 samples analyzed by array CGH. Twelve novel single nucleotide polymorphisms were discovered. The mRNA levels of 7 genes in the commonly deleted interval were reduced by 50% in CD34+ cells from del(5q) MDS samples, and no gene showed complete loss of expression. Taken together, these data show that small deletions and/or point mutations in individual 5q31.2 genes are not common events in MDS, and implicate haploinsufficiency of multiple genes as the relevant genetic consequence of this common deletion.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Genes, Neoplasm/genetics , Myelodysplastic Syndromes/genetics , Bone Marrow , Comparative Genomic Hybridization , Heterozygote , Humans , Mutation , Myelodysplastic Syndromes/etiology , Polymorphism, Single Nucleotide , Skin
13.
Cancer Res ; 68(14): 5524-8, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632602

ABSTRACT

Genetic lesions affecting a number of kinases and other elements within the epidermal growth factor receptor (EGFR) signaling pathway have been implicated in the pathogenesis of human non-small-cell lung cancer (NSCLC). We performed mutational profiling of a large cohort of lung adenocarcinomas to uncover other potential somatic mutations in genes of this pathway that could contribute to lung tumorigenesis. We have identified in 2 of 207 primary lung tumors a somatic activating mutation in exon 2 of MEK1 (i.e., mitogen-activated protein kinase kinase 1 or MAP2K1) that substitutes asparagine for lysine at amino acid 57 (K57N) in the nonkinase portion of the kinase. Neither of these two tumors harbored known mutations in other genes encoding components of the EGFR signaling pathway (i.e., EGFR, HER2, KRAS, PIK3CA, and BRAF). Expression of mutant, but not wild-type, MEK1 leads to constitutive activity of extracellular signal-regulated kinase (ERK)-1/2 in human 293T cells and to growth factor-independent proliferation of murine Ba/F3 cells. A selective MEK inhibitor, AZD6244, inhibits mutant-induced ERK activity in 293T cells and growth of mutant-bearing Ba/F3 cells. We also screened 85 NSCLC cell lines for MEK1 exon 2 mutations; one line (NCI-H1437) harbors a Q56P substitution, a known transformation-competent allele of MEK1 originally identified in rat fibroblasts, and is sensitive to treatment with AZD6244. MEK1 mutants have not previously been reported in lung cancer and may provide a target for effective therapy in a small subset of patients with lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , DNA Mutational Analysis , ErbB Receptors/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , MAP Kinase Kinase 1/metabolism , Mutation , Signal Transduction , Adenocarcinoma/metabolism , Amino Acid Sequence , Animals , Humans , Lung Neoplasms/metabolism , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid
14.
Blood ; 111(9): 4797-808, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18270328

ABSTRACT

Activating mutations in tyrosine kinase (TK) genes (eg, FLT3 and KIT) are found in more than 30% of patients with de novo acute myeloid leukemia (AML); many groups have speculated that mutations in other TK genes may be present in the remaining 70%. We performed high-throughput resequencing of the kinase domains of 26 TK genes (11 receptor TK; 15 cytoplasmic TK) expressed in most AML patients using genomic DNA from the bone marrow (tumor) and matched skin biopsy samples ("germline") from 94 patients with de novo AML; sequence variants were validated in an additional 94 AML tumor samples (14.3 million base pairs of sequence were obtained and analyzed). We identified known somatic mutations in FLT3, KIT, and JAK2 TK genes at the expected frequencies and found 4 novel somatic mutations, JAK1(V623A), JAK1(T478S), DDR1(A803V), and NTRK1(S677N), once each in 4 respective patients of 188 tested. We also identified novel germline sequence changes encoding amino acid substitutions (ie, nonsynonymous changes) in 14 TK genes, including TYK2, which had the largest number of nonsynonymous sequence variants (11 total detected). Additional studies will be required to define the roles that these somatic and germline TK gene variants play in AML pathogenesis.


Subject(s)
Germ-Line Mutation , Leukemia, Myeloid, Acute/genetics , Mutation , Protein-Tyrosine Kinases/genetics , DNA Mutational Analysis , Humans
15.
Blood ; 111(9): 4809-12, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18160671

ABSTRACT

Somatic mutations in JAK2 are frequently found in myeloproliferative diseases, and gain-of-function JAK3 alleles have been identified in M7 acute myeloid leukemia (AML), but a role for JAK1 in AML has not been described. We screened the entire coding region of JAK1 by total exonic resequencing of bone marrow DNA samples from 94 patients with de novo AML. We identified 2 novel somatic mutations in highly conserved residues of the JAK1 gene (T478S, V623A), in 2 separate patients and confirmed these by resequencing germ line DNA samples from the same patients. Overexpression of mutant JAK1 did not transform primary murine cells in standard assays, but compared with wild-type JAK1, JAK1(T478S), and JAK1(V623A) expression was associated with increased STAT1 activation in response to type I interferon and activation of multiple downstream signaling pathways. This is the first report to demonstrate somatic JAK1 mutations in AML and suggests that JAK1 mutations may function as disease-modifying mutations in AML pathogenesis.


Subject(s)
Janus Kinase 1/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Animals , DNA Mutational Analysis , Humans , Leukemia, Myeloid, Acute/etiology , Mice , Mutation, Missense , STAT1 Transcription Factor/metabolism , Transduction, Genetic
16.
Nat Genet ; 39(9): 1068-70, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17660820

ABSTRACT

Autosomal dominant retinal vasculopathy with cerebral leukodystrophy is a microvascular endotheliopathy with middle-age onset. In nine families, we identified heterozygous C-terminal frameshift mutations in TREX1, which encodes a 3'-5' exonuclease. These truncated proteins retain exonuclease activity but lose normal perinuclear localization. These data have implications for the maintenance of vascular integrity in the degenerative cerebral microangiopathies leading to stroke and dementias.


Subject(s)
Brain Diseases/genetics , Exodeoxyribonucleases/genetics , Mutation , Phosphoproteins/genetics , Retinal Diseases/genetics , Amino Acid Sequence , Brain Diseases/enzymology , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Genes, Dominant , Genetic Predisposition to Disease , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Diseases/enzymology , Sequence Homology, Amino Acid , Transfection
17.
Pediatr Res ; 62(2): 170-5, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17597650

ABSTRACT

Completely penetrant mutations in the surfactant protein B gene (SFTPB) and >75% reduction of SFTPB expression disrupt pulmonary surfactant function and cause neonatal respiratory distress syndrome. To inform studies of genetic regulation of SFTPB expression, we created a catalogue of SFTPB variants by comprehensive resequencing from an unselected, population-based cohort (n = 1,116). We found an excess of low-frequency variation [81 SNPs and five small insertion/deletions (in/dels)]. Despite its small genomic size (9.7 kb), SFTPB was characterized by weak linkage disequilibrium (LD) and high haplotype diversity. Using the HapMap Yoruban and European populations, we identified a recombination hot spot that spans SFTPB, was not detectable in our focused resequencing data, and accounts for weak LD. Using homology-based software tools, we discovered no definitively damaging exonic variants. We conclude that excess low-frequency variation, intragenic recombination and lack of common disruptive exonic variants favor complete resequencing as the optimal approach for genetic association studies to identify regulatory SFTPB variants that cause neonatal respiratory distress syndrome in genetically diverse populations.


Subject(s)
Gene Deletion , Genetic Testing , Mutagenesis, Insertional , Neonatal Screening/methods , Polymorphism, Single Nucleotide , Pulmonary Surfactant-Associated Protein B/genetics , Respiratory Distress Syndrome, Newborn/genetics , Black or African American/genetics , Asian People/genetics , Cohort Studies , DNA Mutational Analysis , Databases, Nucleic Acid , Evolution, Molecular , Exons , Genetic Predisposition to Disease , Haplotypes , Humans , Infant, Newborn , Introns , Linkage Disequilibrium , Population Surveillance , Recombination, Genetic , Respiratory Distress Syndrome, Newborn/ethnology , White People/genetics
18.
PLoS One ; 2(5): e426, 2007 May 09.
Article in English | MEDLINE | ID: mdl-17487277

ABSTRACT

BACKGROUND: Fifty percent of lung adenocarcinomas harbor somatic mutations in six genes that encode proteins in the EGFR signaling pathway, i.e., EGFR, HER2/ERBB2, HER4/ERBB4, PIK3CA, BRAF, and KRAS. We performed mutational profiling of a large cohort of lung adenocarcinomas to uncover other potential somatic mutations in genes of this signaling pathway that could contribute to lung tumorigenesis. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed genomic DNA from a total of 261 resected, clinically annotated non-small cell lung cancer (NSCLC) specimens. The coding sequences of 39 genes were screened for somatic mutations via high-throughput dideoxynucleotide sequencing of PCR-amplified gene products. Mutations were considered to be somatic only if they were found in an independent tumor-derived PCR product but not in matched normal tissue. Sequencing of 9MB of tumor sequence identified 239 putative genetic variants. We further examined 22 variants found in RAS family genes and 135 variants localized to exons encoding the kinase domain of respective proteins. We identified a total of 37 non-synonymous somatic mutations; 36 were found collectively in EGFR, KRAS, BRAF, and PIK3CA. One somatic mutation was a previously unreported mutation in the kinase domain (exon 16) of FGFR4 (Glu681Lys), identified in 1 of 158 tumors. The FGFR4 mutation is analogous to a reported tumor-specific somatic mutation in ERBB2 and is located in the same exon as a previously reported kinase domain mutation in FGFR4 (Pro712Thr) in a lung adenocarcinoma cell line. CONCLUSIONS/SIGNIFICANCE: This study is one of the first comprehensive mutational analyses of major genes in a specific signaling pathway in a sizeable cohort of lung adenocarcinomas. Our results suggest the majority of gain-of-function mutations within kinase genes in the EGFR signaling pathway have already been identified. Our findings also implicate FGFR4 in the pathogenesis of a subset of lung adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation , Receptor, Fibroblast Growth Factor, Type 4/genetics , Signal Transduction/genetics , Amino Acid Sequence , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Sequence Homology, Amino Acid
19.
Blood ; 110(5): 1648-55, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17494858

ABSTRACT

Severe congenital neutropenia (SCN) is an inborn disorder of granulopoiesis. Like most other bone marrow failure syndromes, it is associated with a marked propensity to transform into a myelodysplastic syndrome (MDS) or acute leukemia, with a cumulative rate of transformation to MDS/leukemia that exceeds 20%. The genetic (and/or epigenetic) changes that contribute to malignant transformation in SCN are largely unknown. In this study, we performed mutational profiling of 14 genes previously implicated in leukemogenesis using 14 MDS/leukemia samples from patients with SCN. We used high-throughput exon-based resequencing of whole-genome-amplified genomic DNA with a semiautomated method to detect mutations. The sensitivity and specificity of the sequencing pipeline was validated by determining the frequency of mutations in these 14 genes using 188 de novo AML samples. As expected, mutations of tyrosine kinase genes (FLT3, KIT, and JAK2) were common in de novo AML, with a cumulative frequency of 30%. In contrast, no mutations in these genes were detected in the SCN samples; instead, mutations of CSF3R, encoding the G-CSF receptor, were common. These data support the hypothesis that mutations of CSF3R may provide the "activated tyrosine kinase signal" that is thought to be important for leukemogenesis.


Subject(s)
Genetic Diseases, Inborn/genetics , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Neutropenia/genetics , Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Adult , DNA Mutational Analysis , Enzyme Activation/genetics , Epigenesis, Genetic , Genetic Diseases, Inborn/complications , Genome, Human/genetics , Humans , Leukemia, Myeloid, Acute/etiology , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/genetics , Neutropenia/complications , Neutropenia/congenital
20.
Genome Res ; 17(5): 659-66, 2007 May.
Article in English | MEDLINE | ID: mdl-17416743

ABSTRACT

Small insertions and deletions (indels) and single nucleotide polymorphisms (SNPs) are common genetic variants that are thought to be associated with a wide variety of human diseases. Owing to the genome's size and complexity, manually characterizing each one of these variations in an individual is not practical. While significant progress has been made in automated single-base mutation discovery from the sequences of diploid PCR products, automated and reliable detection of indels continues to pose difficult challenges. In this paper, we present PolyScan, an algorithm and software implementation designed to provide de novo heterozygous indel detection and improved SNP identification in the context of high-throughput medical resequencing. Tests on a human diploid PCR-based sequence data set, consisting of 90,270 traces from 13 genes, indicate that PolyScan identified approximately 90% of the 151 consensus indel sites and approximately 84% of the 1546 heterozygous indels previously identified by manual inspection. Tests on tumor-derived data show that PolyScan better identifies high-quality, low-level mutations as compared with other mutation detection software. Moreover, SNP identification improves when reprocessing the results of other programs. These results suggest that PolyScan may play a useful role in the post human genome project research era.


Subject(s)
Algorithms , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Sequence Deletion , Software , Base Sequence , Humans , Molecular Sequence Data , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...