Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Genet Med ; 21(7): 1486-1496, 2019 07.
Article in English | MEDLINE | ID: mdl-30504929

ABSTRACT

PURPOSE: To enhance classification of variants of uncertain significance (VUS) in the DNA mismatch repair (MMR) genes in the cancer predisposition Lynch syndrome, we developed the cell-free in vitro MMR activity (CIMRA) assay. Here, we calibrate and validate the assay, enabling its integration with in silico and clinical data. METHODS: Two sets of previously classified MLH1 and MSH2 variants were selected from a curated MMR gene database, and their biochemical activity determined by the CIMRA assay. The assay was calibrated by regression analysis followed by symmetric cross-validation and Bayesian integration with in silico predictions of pathogenicity. CIMRA assay reproducibility was assessed in four laboratories. RESULTS: Concordance between the training runs met our prespecified validation criterion. The CIMRA assay alone correctly classified 65% of variants, with only 3% discordant classification. Bayesian integration with in silico predictions of pathogenicity increased the proportion of correctly classified variants to 87%, without changing the discordance rate. Interlaboratory results were highly reproducible. CONCLUSION: The CIMRA assay accurately predicts pathogenic and benign MMR gene variants. Quantitative combination of assay results with in silico analysis correctly classified the majority of variants. Using this calibration, CIMRA assay results can be integrated into the diagnostic algorithm for MMR gene variants.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , Genetic Techniques , 3T3 Cells , Animals , Bayes Theorem , Calibration , Computer Simulation , Humans , In Vitro Techniques , Mice , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Reproducibility of Results , Sensitivity and Specificity
2.
Cell Physiol Biochem ; 49(1): 144-159, 2018.
Article in English | MEDLINE | ID: mdl-30184526

ABSTRACT

BACKGROUND/AIMS: Classical Hodgkin lymphoma (cHL) is among the most frequent lymphoma subtypes. The tumor cells originate from crippled germinal center (GC)-B cells that escaped from apoptosis. MicroRNAs (miRNAs) play important roles in B-cell maturation and aberrant expression of miRNAs contributes to the pathogenesis of cHL. Our aim was to identify oncogenic miRNAs relevant for growth of cHL using a high-throughput screening approach. METHODS: A lentiviral pool of 63 miRNA inhibition constructs was used to identify miRNAs essential to cell growth in three cHL cell lines in duplicate. As a negative control we also infected cHL cell lines with a lentiviral barcoded empty vector pool consisting of 222 constructs. The abundance of individual constructs was followed over time by a next generation sequencing approach. The effect on growth was confirmed using individual GFP competition assays and on apoptosis using Annexin-V staining. Our previously published Argonaute 2 (Ago2) immunoprecipitation (IP) data were used to identify target genes relevant for cell growth / apoptosis. Luciferase assays and western blotting were performed to confirm targeting by miRNAs. RESULTS: Four miRNA inhibition constructs, i.e. miR-449a-5p, miR-625-5p, let-7f-2-3p and miR-21-5p, showed a significant decrease in abundance in at least 4 of 6 infections. In contrast, none of the empty vector constructs showed a significant decrease in abundance in 3 or more of the 6 infections. The most abundantly expressed miRNA, i.e. miR-21-5p, showed significantly higher expression levels in cHL compared to GC-B cells. GFP competition assays confirmed the negative effect of miR-21-5p inhibition on HL cell growth. Annexin-V staining of cells infected with miR-21-5p inhibitor indicated a significant increase in apoptosis at day 7 and 9 after viral infection, consistent with the decrease in growth. Four miR-21-5p cell growth- and apoptosis-associated targets were AGO2-IP enriched in cHL cell lines and showed a significant decrease in expression in cHL cell lines in comparison to normal GC-B cells. For the two most abundantly expressed, i.e. BTG2 and PELI1, we confirmed targeting by miR-21-5p using luciferase assays and for PELI1 we also confirmed this at the protein level by western blotting. CONCLUSION: Using a miRNA loss-of-function high-throughput screen we identified four miRNAs with oncogenic effects in cHL and validated the results for the in cHL abundantly expressed miR-21-5p. MiR-21-5p is upregulated in cHL compared to GC-B cells and protects cHL cells from apoptosis possibly via targeting BTG2 and PELI1.


Subject(s)
MicroRNAs/metabolism , 3' Untranslated Regions , Antagomirs/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation/genetics , HEK293 Cells , High-Throughput Nucleotide Sequencing , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogenes/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Gastroenterology ; 155(1): 118-129.e6, 2018 07.
Article in English | MEDLINE | ID: mdl-29601828

ABSTRACT

BACKGROUND & AIMS: Hirschsprung disease (HSCR) is an inherited congenital disorder characterized by absence of enteric ganglia in the distal part of the gut. Variants in ret proto-oncogene (RET) have been associated with up to 50% of familial and 35% of sporadic cases. We searched for variants that affect disease risk in a large, multigenerational family with history of HSCR in a linkage region previously associated with the disease (4q31.3-q32.3) and exome wide. METHODS: We performed exome sequencing analyses of a family in the Netherlands with 5 members diagnosed with HSCR and 2 members diagnosed with functional constipation. We initially focused on variants in genes located in 4q31.3-q32.3; however, we also performed an exome-wide analysis in which known HSCR or HSCR-associated gene variants predicted to be deleterious were prioritized for further analysis. Candidate genes were expressed in HEK293, COS-7, and Neuro-2a cells and analyzed by luciferase and immunoblot assays. Morpholinos were designed to target exons of candidate genes and injected into 1-cell stage zebrafish embryos. Embryos were allowed to develop and stained for enteric neurons. RESULTS: Within the linkage region, we identified 1 putative splice variant in the lipopolysaccharide responsive beige-like anchor protein gene (LRBA). Functional assays could not confirm its predicted effect on messenger RNA splicing or on expression of the mab-21 like 2 gene (MAB21L2), which is embedded in LRBA. Zebrafish that developed following injection of the lrba morpholino had a shortened body axis and subtle gut morphological defects, but no significant reduction in number of enteric neurons compared with controls. Outside the linkage region, members of 1 branch of the family carried a previously unidentified RET variant or an in-frame deletion in the glial cell line derived neurotrophic factor gene (GDNF), which encodes a ligand of RET. This deletion was located 6 base pairs before the last codon. We also found variants in the Indian hedgehog gene (IHH) and its mediator, the transcription factor GLI family zinc finger 3 (GLI3). When expressed in cells, the RET-P399L variant disrupted protein glycosylation and had altered phosphorylation following activation by GDNF. The deletion in GDNF prevented secretion of its gene product, reducing RET activation, and the IHH-Q51K variant reduced expression of the transcription factor GLI1. Injection of morpholinos that target ihh reduced the number of enteric neurons to 13% ± 1.4% of control zebrafish. CONCLUSIONS: In a study of a large family with history of HSCR, we identified variants in LRBA, RET, the gene encoding the RET ligand (GDNF), IHH, and a gene encoding a mediator of IHH signaling (GLI3). These variants altered functions of the gene products when expressed in cells and knockout of ihh reduced the number of enteric neurons in the zebrafish gut.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Hedgehog Proteins/genetics , Hirschsprung Disease/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-ret/genetics , Zinc Finger Protein Gli3/genetics , Animals , COS Cells , Chlorocebus aethiops , Family , Female , Genetic Predisposition to Disease , Genetic Variation , HEK293 Cells , Humans , Male , Morpholinos , Netherlands , Pedigree , Protein Isoforms , Proto-Oncogene Mas , Sequence Analysis, DNA , Signal Transduction , Zebrafish
4.
Neoplasia ; 18(6): 339-46, 2016 06.
Article in English | MEDLINE | ID: mdl-27292023

ABSTRACT

SET domain-containing 2 (SETD2) is responsible for the trimethylation of histone H3 lysine36 (H3K36me3) and is one of the genes most frequently mutated in clear cell renal cell carcinoma (ccRCC). It is located at 3p21, one copy of which is lost in the majority of ccRCC tumors, suggesting that SETD2 might function as a tumor suppressor gene. However, the manner in which loss of SETD2 contributes to ccRCC development has not been studied in renal primary tubular epithelial cells (PTECs). Therefore, we studied the consequences of SETD2 knockdown through lentiviral shRNA in human PTECs. Consistent with its known function, SETD2 knockdown (SETD-KD) led to loss of H3K36me3 in PTECs. In contrast to SETD2 wild-type PTECs, which have a limited proliferation capacity; the SETD2-KD PTECs continued to proliferate. The expression profiles of SETD2-KD PTECs showed a large overlap with the expression profile of early-passage, proliferating PTECs, whereas nonproliferating PTECs showed a significantly different expression profile. Gene set enrichment analysis revealed a significant enrichment of E2F targets in SETD2-KD and proliferating PTECs as compared with nonproliferating PTECs and in proliferating PTEC compared with SETD2-KD. The SETD2-KD PTECs maintained low expression of CDKN2A and high expression of E2F1, whereas their levels changed with continuing passages in untreated PTECs. In contrast to the nonproliferating PTECs, SETD2-KD PTECs showed no ß-galactosidase staining, confirming the protection against senescence. Our results indicate that SETD2 inactivation enables PTECs to bypass the senescence barrier, facilitating a malignant transformation toward ccRCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p18/biosynthesis , E2F1 Transcription Factor/biosynthesis , Histone-Lysine N-Methyltransferase/genetics , Kidney Neoplasms/pathology , Carcinoma, Renal Cell/genetics , Cell Line , Cell Proliferation , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Epithelial Cells/metabolism , Genes, Tumor Suppressor , HEK293 Cells , Histones/metabolism , Humans , Kidney Neoplasms/genetics , Kidney Tubules/cytology , Methylation , RNA Interference , RNA, Small Interfering/genetics , beta-Galactosidase/metabolism
5.
Hum Mutat ; 35(12): 1514-23, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231886

ABSTRACT

Microsatellite instability (MSI) in tumors results in an accumulation of mutations in (target) genes. Previous studies suggest that the profile of target genes differs according to tumor type. This paper describes the first genome-wide search for target genes for mismatch repair-deficient endometrial cancers. Genes expressed in normal endometrium containing coding repeats were analyzed for mutations in tumors. We identified 44 possible genes of which seven are highly mutated (>15%). Some candidates were also found mutated in colorectal and gastric tumors. The most frequently mutated gene, NRIP1 encoding nuclear receptor-interacting protein 1, was silenced in an endometrial tumor cell line and expression microarray experiments were performed. Silencing of NRIP1 was associated with differences in the expression of several genes in the estrogen-receptor network. Furthermore, an enrichment of genes related to cell cycle (regulation) and replication was observed. We present a new profile of target genes, some of them tissue specific, whereas others seem to play a more general role in MSI tumors. The high-mutation frequency combined with the expression data suggest, for the first time, an involvement of NRIP1 in endometrial cancer development.


Subject(s)
Endometrial Neoplasms/genetics , Microsatellite Repeats/genetics , Receptors, Estrogen/metabolism , Adaptor Proteins, Signal Transducing/genetics , Base Sequence , Cell Line, Tumor , DNA Primers , Endometrial Neoplasms/metabolism , Female , Gene Knockdown Techniques , Humans , Mutation , Nuclear Proteins/genetics , Nuclear Receptor Interacting Protein 1 , Real-Time Polymerase Chain Reaction
6.
Gene ; 534(1): 40-3, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24161884

ABSTRACT

Cardiovascular manifestations in patients with Marfan syndrome (MFS) are related to aortic and valvular abnormalities. However, dilatation of the left ventricle (LV) can occur, even in the absence of aortic surgery or valvular abnormalities. We evaluated genetic characteristics of patients with MFS with LV dilatation. One hundred eighty-two patients fulfilling the MFS criteria, without valvular abnormalities or previous aortic surgery, with a complete FBN1 analysis, were studied. FBN1 mutations were identified in over 81% of patients. Twenty-nine patients (16%) demonstrated LV dilatation (LV end diastolic diameter corrected for age and body surface area >112%). FBN1-positive patients carrying a non-missense mutation more often had LV dilatation than missense mutation carriers (14/74 versus 5/75; p<0.05). Finally, FBN1-negative MFS patients significantly more often demonstrated LV dilatation than FBN1-positive patients (10/33 versus 19/149; p<0.05). It is concluded that LV dilatation in MFS patients is more often seen in patients with a non-missense mutation and in those patients without an FBN1 mutation. Therefore physicians should be aware of the possibility of LV dilatation in these patients even in the absence of valvular pathology.


Subject(s)
Genotype , Heart Ventricles/pathology , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Adult , Female , Fibrillin-1 , Fibrillins , Humans , Male , Microfilament Proteins/genetics , Mutation, Missense , Phenotype
7.
J Clin Endocrinol Metab ; 96(6): E991-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21470995

ABSTRACT

CONTEXT: Medullary and papillary thyroid carcinoma (MTC and PTC) are two types of thyroid cancer that can originate from activating mutations or rearrangements in the RET gene. Therapeutic options are limited in recurrent disease, but because RET is a tyrosine kinase (TK) receptor involved in cellular growth and proliferation, treatment with a TK inhibitor might be promising. Several TK inhibitors have been tested in clinical trials, but it is unknown which inhibitor is most effective and whether there is any specificity for particular RET mutations. OBJECTIVE: We aimed to compare the effect of four TK inhibitors (axitinib, sunitinib, vandetanib, and XL184) on cell proliferation, RET expression and autophosphorylation, and ERK activation in cell lines expressing a MEN2A (MTC-TT), a MEN2B (MZ-CRC-1) mutation, and a RET/PTC (TPC-1) rearrangement. DESIGN: The three cell lines were cultured and treated with the four TK inhibitors. Effects on cell proliferation and RET and ERK expression and activation were determined. RESULTS: XL184 and vandetanib most effectively inhibited cell proliferation, RET autophosphorylation in combination with a reduction of RET expression, and ERK phosphorylation in MTC-TT and MZ-CRC-1, respectively. TPC-1 cells showed a decrease in RET autophosphorylation after treatment with XL184, but no effect was observed on ERK activation. CONCLUSION: There is indeed specificity for different RET mutations, with XL184 being the most potent inhibitor in MEN2A and PTC and vandetanib the most effective in MEN2B in vitro. No TK inhibitor was superior for all the cell lines tested, indicating that mutation-specific therapies could be beneficial in treating MTC and PTC.


Subject(s)
Cell Proliferation/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thyroid Gland/drug effects , Anilides/pharmacology , Axitinib , Blotting, Western , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , Indoles/pharmacology , Piperidines/pharmacology , Proto-Oncogene Proteins c-ret/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sunitinib , Thyroid Gland/cytology , Thyroid Gland/metabolism , Tumor Cells, Cultured
8.
Gastroenterology ; 140(2): 572-582.e2, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20977903

ABSTRACT

BACKGROUND & AIMS: Two noncoding variations in RET-the T allele of the single nucleotide polymorphism (SNP) rs2435357 (Enh1:C>T) and the A allele of the SNP rs2506004 (Enh2:C>A)-are associated with Hirschsprung's disease. These SNPs are in strong linkage disequilibrium and located in an enhancer element in intron 1 of the RET gene. The T allele of the Enh1 variant results in reduced expression of RET, compared with the C allele, because the T allele disrupts binding to the transcription factor SOX10. We studied whether the A allele of Enh2 (Enh2-A) also affects RET gene expression. METHODS: We evaluated the function of Enh1 and Enh2 using luciferase reporter assays with constructs that contained each allele, separately or in combination. We performed in silico analysis to identify transcription activators or repressors that bind to Enh2-C. RESULTS: The Enh1-T and the Enh2-A alleles reduced expression of the luciferase reporter gene. In silico analysis identified the sequence of Enh2-C and its surrounding sequence (ACGTG) as a potential binding site for the NXF-ARNT2 and SIM2-ARNT2 transcription factor heterodimers. The affinity of NXF-ARNT2 for Enh2-C was confirmed by electrophoresis mobility shift and supershift assays. Transfection of neuroblastoma cell lines with NXF-ARNT2 or SIM2-ARNT2 increased and decreased expression of RET, respectively. CONCLUSIONS: More than one SNP on an associated haplotype can influence gene expression and ultimately disease phenotype. Binding of the transcription factors NXF, ARNT2, and SIM2 to RET depend on the RET polymorphism of Enh2 and affect RET expression and the development of Hirschsprung's disease.


Subject(s)
Gene Expression , Genetic Variation , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Cells, Cultured , Exons , Haplotypes , Hirschsprung Disease/metabolism , Humans , Introns , Linkage Disequilibrium , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neural Stem Cells/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-ret/metabolism
9.
PLoS One ; 5(12): e15144, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21187955

ABSTRACT

Hirschsprung disease (HSCR) is a congenital malformation characterized by the absence of enteric neurons in the distal part of the colon. Several genes have been implicated in the development of this disease that together account for 20% of all cases, implying that other genes are involved. Since HSCR is frequently associated with other congenital malformations, the functional characterization of the proteins encoded by the genes involved in these syndromes can provide insights into the protein-network involved in HSCR development. Recently, we found that KBP, encoded by the gene involved in a HSCR- associated syndrome called Goldberg-Shprintzen syndrome, interacts with SCG10, a stathmin-like protein. To determine if SCG10 is involved in the etiology of HSCR, we determined SCG10 expression levels during development and screened 85 HSCR patients for SCG10 mutations. We showed that SCG10 expression increases during development but no germline mutation was found in any of these patients. In conclusion, this study shows that SCG10 is not directly implicated in HSCR development. However, an indirect involvement of SCG10 cannot be ruled out as this can be due to a secondary effect caused by its direct interactors.


Subject(s)
Hirschsprung Disease/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Animals , Calcium-Binding Proteins , DNA Mutational Analysis , Disease Models, Animal , Gene Expression Regulation, Developmental , Germ-Line Mutation , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Neural Crest/cytology , Protein Interaction Mapping , Stathmin , Stem Cells/cytology , Time Factors
10.
Hum Mol Genet ; 19(18): 3642-51, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20621975

ABSTRACT

Goldberg-Shprintzen syndrome (GOSHS) is a rare clinical disorder characterized by central and enteric nervous system defects. This syndrome is caused by inactivating mutations in the Kinesin Binding Protein (KBP) gene, which encodes a protein of which the precise function is largely unclear. We show that KBP expression is up-regulated during neuronal development in mouse cortical neurons. Moreover, KBP-depleted PC12 cells were defective in nerve growth factor-induced differentiation and neurite outgrowth, suggesting that KBP is required for cell differentiation and neurite development. To identify KBP interacting proteins, we performed a yeast two-hybrid screen and found that KBP binds almost exclusively to microtubule associated or related proteins, specifically SCG10 and several kinesins. We confirmed these results by validating KBP interaction with one of these proteins: SCG10, a microtubule destabilizing protein. Zebrafish studies further demonstrated an epistatic interaction between KBP and SCG10 in vivo. To investigate the possibility of direct interaction between KBP and microtubules, we undertook co-localization and in vitro binding assays, but found no evidence of direct binding. Thus, our data indicate that KBP is involved in neuronal differentiation and that the central and enteric nervous system defects seen in GOSHS are likely caused by microtubule-related defects.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microtubules/metabolism , Neurogenesis , Serpins/metabolism , Stathmin/metabolism , Zebrafish Proteins/metabolism , Animals , Calcium-Binding Proteins , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Disease Models, Animal , HeLa Cells , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Microtubule Proteins , NIH 3T3 Cells , Neurons/cytology , Neurons/metabolism , PC12 Cells , Protein Binding , Rats , Serpins/genetics , Stathmin/genetics , Zebrafish Proteins/genetics
11.
Cancer Res ; 70(11): 4287-91, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20501857

ABSTRACT

Sporadic clear cell renal cell carcinoma (cRCC) is genetically characterized by the recurrent loss of the short arm of chromosome 3, with a hotspot for copy number loss in the 3p21 region. We applied a method called "gene identification by nonsense-mediated mRNA decay inhibition" to a panel of 10 cRCC cell lines with 3p21 copy number loss to identify biallelic inactivated genes located at 3p21. This revealed inactivation of the histone methyltransferase gene SETD2, located on 3p21.31, as a common event in cRCC cells. SETD2 is nonredundantly responsible for trimethylation of the histone mark H3K36. Consistent with this function, we observed loss or a decrease of H3K36me3 in 7 out of the 10 cRCC cell lines. Identification of missense mutations in 2 out of 10 primary cRCC tumor samples added support to the involvement of loss of SETD2 function in the development of cRCC tumors.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Renal Cell/genetics , Histone-Lysine N-Methyltransferase/genetics , Kidney Neoplasms/genetics , Adenocarcinoma, Clear Cell/enzymology , Carcinoma, Renal Cell/enzymology , Cell Line, Tumor , Chromosomes, Human, Pair 3 , Comparative Genomic Hybridization , DNA Methylation , DNA Mutational Analysis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Kidney Neoplasms/enzymology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
J Am Coll Cardiol ; 49(25): 2430-9, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17599607

ABSTRACT

OBJECTIVES: The goal of this study was to identify the underlying gene defect in a family with inherited myocardial fibrosis. BACKGROUND: A large family with an autosomal dominantly inherited form of myocardial fibrosis with a highly malignant clinical outcome has been investigated. Because myocardial fibrosis preceded the clinical and echocardiographic signs, we consider the disease to be a hereditary form of cardiac fibrosis. METHODS: Twenty-five family members were clinically evaluated, and 5 unaffected and 8 affected family members were included in a genome-wide linkage study. RESULTS: The highest logarithm of the odds (LOD) score (LOD = 2.6) was found in the region of the lamin AC (LMNA) gene. The LMNA mutation analysis, both by denaturing gradient gel electrophoresis and sequencing, failed to show a mutation. Subsequent Southern blotting, complementary deoxyribonucleic acid sequencing, and multiplex ligation-dependent probe amplification analysis, however, revealed a deletion of the start codon-containing exon and an adjacent noncoding exon. In vitro studies demonstrated that the deletion results in the formation of nuclear aggregates of lamin, suggesting that the mutant allele is being transcribed. CONCLUSIONS: This novel LMNA deletion causes a distinct, highly malignant cardiomyopathy with early-onset primary cardiac fibrosis likely due to an effect of the shortened mutant protein, which secondarily leads to arrhythmias and end-stage cardiac failure.


Subject(s)
Endomyocardial Fibrosis/epidemiology , Endomyocardial Fibrosis/genetics , Gene Deletion , Genetic Predisposition to Disease , Lamin Type A/genetics , Mutation , Adult , Age Distribution , Biopsy, Needle , Blotting, Southern , Electrocardiography , Endomyocardial Fibrosis/pathology , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Incidence , Male , Middle Aged , Pedigree , Prognosis , Risk Assessment , Severity of Illness Index , Sex Distribution , Survival Rate
13.
Hum Genet ; 119(1-2): 206-11, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16408224

ABSTRACT

Biallelic germline mutations of MUTYH-a gene encoding a base excision repair protein-are associated with an increased susceptibility of colorectal cancer. Whether monoallelic MUTYH mutations also increase cancer risk is not yet clear, although there is some evidence suggesting a slight increase of risk. As the MUTYH protein interacts with the mismatch repair (MMR) system, we hypothesised that the combination of a monoallelic MUTYH mutation with an MMR gene mutation increases cancer risk. We therefore investigated the prevalence of monoallelic MUTYH mutations in carriers of a germline MMR mutation: 40 carriers of a truncating mutation (group I) and 36 of a missense mutation (group II). These patients had been diagnosed with either colorectal or endometrial cancer. We compared their MUTYH mutation frequencies with those observed in a group of 134 Dutch colorectal and endometrial cancer patients without an MMR gene mutation (0.7%) and those reported for Caucasian controls (1.5%). In group I one monoallelic MUTYH mutation was found (2.5%). In group II five monoallelic germline MUTYH mutations were found (14%), four of them in MSH6 missense mutation carriers (20%). Of all patients with an MMR gene mutation, only those with a missense mutation showed a significantly higher frequency of (monoallelic) MUTYH mutations than the Dutch cancer patients without MMR gene mutations (P = 0.002) and the published controls (P = 0.001). These results warrant further study to test the hypothesis of mutations in MMR genes (in particular MSH6) and MUTYH acting together to increase cancer risk.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Glycosylases/genetics , DNA Mismatch Repair , Mutation , Adaptor Proteins, Signal Transducing , Carrier Proteins/analysis , Carrier Proteins/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA Mutational Analysis , DNA Repair/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Germ-Line Mutation , Humans , Immunohistochemistry , Microsatellite Instability , MutL Protein Homolog 1 , MutS Homolog 2 Protein/analysis , MutS Homolog 2 Protein/genetics , Mutation, Missense , Nuclear Proteins/analysis , Nuclear Proteins/genetics
14.
Am J Hum Genet ; 77(1): 120-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15883926

ABSTRACT

We identified, by homozygosity mapping, a novel locus on 10q21.3-q22.1 for Goldberg-Shprintzen syndrome (GOSHS) in a consanguineous Moroccan family. Phenotypic features of GOSHS in this inbred family included microcephaly and mental retardation, which are both central nervous system defects, as well as Hirschsprung disease, an enteric nervous system defect. Furthermore, since bilateral generalized polymicogyria was diagnosed in all patients in this family, this feature might also be considered a key feature of the syndrome. We demonstrate that homozygous nonsense mutations in KIAA1279 at 10q22.1, encoding a protein with two tetratrico peptide repeats, underlie this syndromic form of Hirschsprung disease and generalized polymicrogyria, establishing the importance of KIAA1279 in both enteric and central nervous system development.


Subject(s)
Codon, Nonsense , Enteric Nervous System/abnormalities , Nervous System Malformations/genetics , Abnormalities, Multiple , Base Sequence , Chromosomes, Human, Pair 10 , Consanguinity , Female , Hirschsprung Disease/genetics , Humans , Intellectual Disability/genetics , Male , Nerve Tissue Proteins , Pedigree , Syndrome
15.
Am J Hum Genet ; 76(5): 850-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15759212

ABSTRACT

Patients with sporadic Hirschsprung disease (HSCR) show increased allele sharing at markers in the 5' region of the RET locus, indicating the presence of a common ancestral RET mutation. In a previous study, we found a haplotype of six SNPs that was transmitted to 55.6% of our patients, whereas it was present in only 16.2% of the controls we used. Among the patients with that haplotype, 90.8% had it on both chromosomes, which led to a much higher risk of developing HSCR than when the haplotype occurred heterozygously. To more precisely define the HSCR-associated region and to identify candidate disease-associated variant(s), we sequenced the shared common haplotype region from 10 kb upstream of the RET gene through intron 1 and exon 2 (in total, 33 kb) in a patient homozygous for the common risk haplotype and in a control individual homozygous for the most common nonrisk haplotype. A comparison of these sequences revealed 86 sequence differences. Of these 86 variations, 8 proved to be in regions highly conserved among different vertebrates and within putative transcription factor binding sites. We therefore considered these as candidate disease-associated variants. Subsequent genotyping of these eight variants revealed a strong disease association for six of the eight markers. These six markers also showed the largest distortions in allele transmission. Interspecies comparison showed that only one of the six variations was located in a region also conserved in a nonmammalian species, making it the most likely candidate HSCR-associated variant.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Hirschsprung Disease/genetics , Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Consensus Sequence , Conserved Sequence , Gene Frequency , Genetic Markers , Haplotypes , Humans , Molecular Sequence Data , Mutation , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-ret , Risk
16.
Cancer Res ; 65(5): 1729-37, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15753368

ABSTRACT

The RET proto-oncogene encodes a receptor tyrosine kinase whose dysfunction plays a crucial role in the development of several neural crest disorders. Distinct activating RET mutations cause multiple endocrine neoplasia type 2A (MEN2A), type 2B (MEN2B), and familial medullary thyroid carcinoma (FMTC). Despite clear correlations between the mutations found in these cancer syndromes and their phenotypes, the molecular mechanisms connecting the mutated receptor to the different disease phenotypes are far from completely understood. Luciferase reporter assays in combination with immunoprecipitations, and Western and immunohistochemistry analyses were done in order to characterize the signaling properties of two FMTC-associated RET mutations, Y791F and S891A, respectively, both affecting the tyrosine kinase domain of the receptor. We show that these RET-FMTC mutants are monomeric receptors which are autophosphorylated and activated independently of glial cell line-derived neurotrophic factor. Moreover, we show that the dysfunctional signaling properties of these mutants, when compared with wild-type RET, involve constitutive activation of signal transducers and activators of transcription 3 (STAT3). Furthermore, we show that STAT3 activation is mediated by a signaling pathway involving Src, JAK1, and JAK2, differing from STAT3 activation promoted by RET(C634R) which was previously found to be independent of Src and JAKs. Three-dimensional modeling of the RET catalytic domain suggested that the structural changes promoted by the respective amino acids substitutions lead to a more accessible substrate and ATP-binding monomeric conformation. Finally, immunohistochemical analysis of FMTC tumor samples support the in vitro data, because nuclear localized, Y705-phosphorylated STAT3, as well as a high degree of RET expression at the plasma membrane was observed.


Subject(s)
Carcinoma, Medullary , Mutation/genetics , Nerve Growth Factors/metabolism , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Thyroid Neoplasms , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Animals , Blotting, Western , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Enzyme Activation , Glial Cell Line-Derived Neurotrophic Factor , Humans , Immunoenzyme Techniques , Immunoprecipitation , Janus Kinase 1 , Janus Kinase 2 , Luciferases/metabolism , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Oncogene Proteins/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Proto-Oncogene Proteins pp60(c-src) , Receptor Protein-Tyrosine Kinases/genetics , STAT3 Transcription Factor , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Trans-Activators/metabolism
17.
Eur J Hum Genet ; 12(8): 604-12, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15138456

ABSTRACT

Hirschsprung disease (HSCR), a congenital disorder characterized by intestinal obstruction due to absence of enteric ganglia along variable lengths of the intestinal tract, occurs both in familial and sporadic cases. RET mutations have been found in approximately 50% of the families, but explains only a minority of sporadic cases. This study aims at investigating a possible role of RET in sporadic HSCR patients. Haplotypes of 13 DNA markers, within and flanking RET, have been determined for 117 sporadic HSCR patients and their parents. Strong association was observed for six markers in the 5' region of RET. The largest distortions in allele transmission were found at the same markers. One single haplotype composed of these six markers was present in 55.6% of patients versus 16.2% of controls. Odds ratios (ORs) revealed a highly increased risk of homozygotes for this haplotype to develop HSCR (OR>20). These results allowed us to conclude that RET plays a crucial role in HSCR even when no RET mutations are found. An unknown functional disease variant(s) with a dosage-dependent effect in HSCR is likely located between the promoter region and exon 2 of RET.


Subject(s)
Hirschsprung Disease/genetics , Mutation/genetics , Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adult , Child , DNA Primers , Exons/genetics , Gene Components , Gene Frequency , Genetic Predisposition to Disease/genetics , Haplotypes/genetics , Humans , Linkage Disequilibrium , Microsatellite Repeats/genetics , Netherlands , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-ret
18.
Am J Hum Genet ; 71(4): 969-74, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12214285

ABSTRACT

Hirschsprung disease (HSCR) is a common genetic disorder characterized by intestinal obstruction secondary to enteric aganglionosis. HSCR demonstrates a complex pattern of inheritance, with the RET proto-oncogene acting as a major gene and with several additional susceptibility loci related to the Ret-signaling pathway or to other developmental programs of neural crest cells. To test how the HSCR phenotype may be affected by the presence of genetic variants, we investigated the role of a single-nucleotide polymorphism (SNP), 2508C-->T (S836S), in exon 14 of the RET gene, characterized by low frequency among patients with HSCR and overrepresentation in individuals affected by sporadic medullary thyroid carcinoma. Typing of several different markers across the RET gene demonstrated that a whole conserved haplotype displayed anomalous distribution and nonrandom segregation in families with HSCR. We provide genetic evidence about a protective role of this low-penetrant haplotype in the pathogenesis of HSCR and demonstrate a possible functional effect linked to RET messenger RNA expression.


Subject(s)
Drosophila Proteins , Hirschsprung Disease/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Alleles , Gene Expression , Haplotypes , Humans , Pedigree , Proto-Oncogene Mas , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-ret , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis
19.
Am J Med Genet ; 108(3): 223-5, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11891690

ABSTRACT

ABCD syndrome is an autosomal recessive syndrome characterized by albinism, black lock, cell migration disorder of the neurocytes of the gut (Hirschsprung disease [HSCR]), and deafness. This phenotype clearly overlaps with the features of the Shah-Waardenburg syndrome, comprising sensorineural deafness; hypopigmentation of skin, hair, and irides; and HSCR. Therefore, we screened DNA of the index patient of the ABCD syndrome family for mutations in the endothelin B receptor (EDNRB) gene, a gene known to be involved in Shah-Waardenburg syndrome. A homozygous nonsense mutation in exon 3 (R201X) of the EDNRB gene was found. We therefore suggest that ABCD syndrome is not a separate entity, but an expression of Shah-Waardenburg syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Albinism/pathology , Deafness/pathology , Receptors, Endothelin/genetics , Abnormalities, Multiple/pathology , Base Sequence , Consanguinity , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Endothelin-3/genetics , Fatal Outcome , Female , High Mobility Group Proteins/genetics , Hirschsprung Disease/pathology , Homozygote , Humans , Hypopigmentation/pathology , Infant , Mutation , Phenotype , Receptor, Endothelin B , SOXE Transcription Factors , Syndrome , Transcription Factors , Waardenburg Syndrome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...