Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Genet Med ; 26(5): 101101, 2024 May.
Article in English | MEDLINE | ID: mdl-38362852

ABSTRACT

PURPOSE: Females with biallelic CHEK2 germline pathogenic variants (gPVs) more often develop multiple breast cancers than individuals with monoallelic CHEK2 gPVs. This study is aimed at expanding the knowledge on the occurrence of other malignancies. METHODS: Exome sequencing of individuals who developed multiple primary malignancies identified 3 individuals with the CHEK2 (NM_007194.4) c.1100del p.(Thr367MetfsTer15) loss-of-function gPV in a biallelic state. We collected the phenotypes of an additional cohort of individuals with CHEK2 biallelic gPVs (n = 291). RESULTS: In total, 157 individuals (53.4%; 157/294 individuals) developed ≥1 (pre)malignancy. The most common (pre)malignancies next to breast cancer were colorectal- (n = 19), thyroid- (n = 19), and prostate (pre)malignancies (n = 12). Females with biallelic CHEK2 loss-of-function gPVs more frequently developed ≥2 (pre)malignancies and at an earlier age compared with females biallelic for the CHEK2 c.470T>C p.(Ile157Thr) missense variant. Furthermore, 26 males (31%; 26/84 males) with CHEK2 biallelic gPVs developed ≥1 (pre)malignancies of 15 origins. CONCLUSION: Our study suggests that CHEK2 biallelic gPVs likely increase the susceptibility to develop multiple malignancies in various tissues, both in females and males. However, it is possible that a substantial proportion of individuals with CHEK2 biallelic gPVs is missed as diagnostic testing for CHEK2 often is limited to individuals who developed breast cancer.


Subject(s)
Checkpoint Kinase 2 , Genetic Predisposition to Disease , Germ-Line Mutation , Neoplasms , Adult , Female , Humans , Male , Middle Aged , Alleles , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Checkpoint Kinase 2/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Exome Sequencing/methods , Germ-Line Mutation/genetics , Neoplasms/genetics , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
2.
Am J Hum Genet ; 109(5): 953-960, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35460607

ABSTRACT

We report an autosomal recessive, multi-organ tumor predisposition syndrome, caused by bi-allelic loss-of-function germline variants in the base excision repair (BER) gene MBD4. We identified five individuals with bi-allelic MBD4 variants within four families and these individuals had a personal and/or family history of adenomatous colorectal polyposis, acute myeloid leukemia, and uveal melanoma. MBD4 encodes a glycosylase involved in repair of G:T mismatches resulting from deamination of 5'-methylcytosine. The colorectal adenomas from MBD4-deficient individuals showed a mutator phenotype attributable to mutational signature SBS1, consistent with the function of MBD4. MBD4-deficient polyps harbored somatic mutations in similar driver genes to sporadic colorectal tumors, although AMER1 mutations were more common and KRAS mutations less frequent. Our findings expand the role of BER deficiencies in tumor predisposition. Inclusion of MBD4 in genetic testing for polyposis and multi-tumor phenotypes is warranted to improve disease management.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Uveal Neoplasms , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Endodeoxyribonucleases/genetics , Genetic Predisposition to Disease , Germ Cells/pathology , Germ-Line Mutation/genetics , Humans , Uveal Neoplasms/genetics
3.
Mol Cancer ; 17(1): 23, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29448935

ABSTRACT

Germline mutations in BUB1 and BUB3 have been reported to increase the risk of developing colorectal cancer (CRC) at young age, in presence of variegated aneuploidy and reminiscent dysmorphic traits of mosaic variegated aneuploidy syndrome. We performed a mutational analysis of BUB1 and BUB3 in 456 uncharacterized mismatch repair-proficient hereditary non-polyposis CRC families and 88 polyposis cases. Four novel or rare germline variants, one splice-site and three missense, were identified in four families. Neither variegated aneuploidy nor dysmorphic traits were observed in carriers. Evident functional effects in the heterozygous form were observed for c.1965-1G>A, but not for c.2296G>A (p.E766K), in spite of the positive co-segregation in the family. BUB1 c.2473C>T (p.P825S) and BUB3 c.77C>T (p.T26I) remained as variants of uncertain significance. As of today, the rarity of functionally relevant mutations identified in familial and/or early onset series does not support the inclusion of BUB1 and BUB3 testing in routine genetic diagnostics of familial CRC.


Subject(s)
Adenomatous Polyposis Coli/genetics , Cell Cycle Proteins/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Germ-Line Mutation , Poly-ADP-Ribose Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/genetics , Cell Cycle Proteins/chemistry , Humans , Models, Molecular , Pedigree , Poly-ADP-Ribose Binding Proteins/chemistry , Protein Conformation , Protein Serine-Threonine Kinases/chemistry
4.
Genes Chromosomes Cancer ; 55(11): 855-63, 2016 11.
Article in English | MEDLINE | ID: mdl-27239782

ABSTRACT

Germline mutations in BUB1B, encoding BUBR1, one of the crucial components of the spindle assembly checkpoint (SAC), have been shown to cause variable phenotypes, including the recessive mosaic variegated aneuploidy (MVA) syndrome, which predisposes to cancer. Reduced levels of the wild-type BUBR1 protein have been linked to the development of gastrointestinal neoplasms. To determine whether mutations in BUB1B are enriched in individuals with colorectal cancer (CRC), we performed amplicon-based targeted next-generation sequencing of BUB1B on germline DNA of 192 individuals with early-onset CRC (≤50 years). None of the individuals was found to be homozygous or compound heterozygous for mutations in BUB1B. However, we did identify two rare heterozygous variants, p.Glu390del and p.Cys945Tyr, in patients who developed CRC at the ages of 41 and 43 years, respectively. Both variants were shown not to affect BUBR1 protein expression levels and protein localization. Since the p.Glu390del variant is located in the BUB3-binding domain, we also performed immunoprecipitation to examine whether this variant affects the binding of BUB1 or BUB3 to BUBR1 but, compared to wild-type BUBR1, no difference was observed. Our data suggest that mutations in BUB1B do not occur frequently in the germline of individuals with CRC and that BUB1B unlikely plays a major role in the predisposition to early-onset CRC. Whether carriers of pathogenic BUB1B mutations, such as the parents of MVA syndrome patients, have an increased risk for cancer remains of interest, as studies in mice have suggested that haploinsufficiency of BUB1B may cause an increase in carcinogen-induced tumors. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Germ-Line Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Adolescent , Adult , Age of Onset , Alleles , Animals , Colorectal Neoplasms/pathology , Female , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , M Phase Cell Cycle Checkpoints/genetics , Male , Mice , Middle Aged
5.
PLoS Genet ; 12(2): e1005880, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26901136

ABSTRACT

Approximately 25-30% of colorectal cancer (CRC) cases are expected to result from a genetic predisposition, but in only 5-10% of these cases highly penetrant germline mutations are found. The remaining CRC heritability is still unexplained, and may be caused by a hitherto-undefined set of rare variants with a moderately penetrant risk. Here we aimed to identify novel risk factors for early-onset CRC using whole-exome sequencing, which was performed on a cohort of CRC individuals (n = 55) with a disease onset before 45 years of age. We searched for genes that were recurrently affected by rare variants (minor allele frequency ≤ 0.001) with potentially damaging effects and, subsequently, re-sequenced the candidate genes in a replication cohort of 174 early-onset or familial CRC individuals. Two functionally relevant genes with low frequency variants with potentially damaging effects, PTPN12 and LRP6, were found in at least three individuals. The protein tyrosine phosphatase PTP-PEST, encoded by PTPN12, is a regulator of cell motility and LRP6 is a component of the WNT-FZD-LRP5-LRP6 complex that triggers WNT signaling. All variants in LRP6 were identified in individuals with an extremely early-onset of the disease (≤30 years of age), and two of the three variants showed increased WNT signaling activity in vitro. In conclusion, we present PTPN12 and LRP6 as novel candidates contributing to the heterogeneous susceptibility to CRC.


Subject(s)
Colorectal Neoplasms/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Age of Onset , Amino Acid Sequence , Chromosome Segregation/genetics , Cohort Studies , Colorectal Neoplasms/enzymology , DNA Mismatch Repair/genetics , Exome/genetics , Genes, Neoplasm , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Sequence Analysis, DNA , Signal Transduction/genetics , Wnt Proteins/metabolism
6.
J Pathol ; 236(2): 155-64, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25712196

ABSTRACT

Heritable genetic variants can significantly affect the lifetime risk of developing cancer, including polyposis and colorectal cancer (CRC). Variants in genes currently known to be associated with a high risk for polyposis or CRC, however, explain only a limited number of hereditary cases. The identification of additional genetic causes is, therefore, crucial to improve CRC prevention, detection and treatment. We have performed genome-wide and targeted DNA copy number profiling and resequencing in early-onset and familial polyposis/CRC patients, and show that deletions affecting the open reading frame of the tumour suppressor gene FOCAD are recurrent and significantly enriched in CRC patients compared with unaffected controls. All patients carrying FOCAD deletions exhibited a personal or family history of polyposis. RNA in situ hybridization revealed FOCAD expression in epithelial cells in the colonic crypt, the site of tumour initiation, as well as in colonic tumours and organoids. Our data suggest that monoallelic germline deletions in the tumour suppressor gene FOCAD underlie moderate genetic predisposition to the development of polyposis and CRC.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , Gene Deletion , Germ-Line Mutation/genetics , Tumor Suppressor Proteins/genetics , Adenomatous Polyposis Coli/metabolism , Adult , Case-Control Studies , Chromosomes, Human, Pair 9/genetics , Colorectal Neoplasms/metabolism , DNA Copy Number Variations/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Heterozygote , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Open Reading Frames/genetics , Tumor Suppressor Proteins/metabolism
7.
Gastroenterology ; 145(3): 544-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23747338

ABSTRACT

The spindle assembly checkpoint controls proper chromosome segregation during mitosis and prevents aneuploidy-an important feature of cancer cells. We performed genome-wide and targeted copy number and mutation analyses of germline DNA from 208 patients with familial or early-onset (40 years of age or younger) colorectal cancer; we identified haploinsufficiency or heterozygous mutations in the spindle assembly checkpoint genes BUB1 and BUB3 in 2.9% of them. Besides colorectal cancer, these patients had variegated aneuploidies in multiple tissues and variable dysmorphic features. These results indicate that mutations in BUB1 and BUB3 cause mosaic variegated aneuploidy and increase the risk of colorectal cancer at a young age.


Subject(s)
Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Germ-Line Mutation , Protein Serine-Threonine Kinases/genetics , Adult , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/metabolism , Genome-Wide Association Study , Humans , Poly-ADP-Ribose Binding Proteins , Protein Serine-Threonine Kinases/metabolism , Risk Factors
8.
PLoS One ; 5(11): e15128, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21152103

ABSTRACT

BACKGROUND: Although the mitotic arrest deficient protein MAD2B (MAD2L2) is thought to inhibit the anaphase promoting complex (APC) by binding to CDC20 and/or CDH1 (FZR1), its exact role in cell cycle control still remains to be established. METHODOLOGY/PRINCIPAL FINDINGS: Using a yeast two-hybrid interaction trap we identified the human clathrin light chain A (CLTA) as a novel MAD2B binding protein. A direct interaction was established in mammalian cells via GST pull-down and endogenous co-immunoprecipitation during the G2/M phase of the cell cycle. Through subsequent confocal laser scanning microscopy we found that MAD2B and CLTA co-localize at the mitotic spindle. Clathrin forms a trimeric structure, i.e., the clathrin triskelion, consisting of three heavy chains (CLTC), each with an associated light chain. This clathrin structure has previously been shown to be required for the function of the mitotic spindle through stabilization of kinetochore fibers. Upon siRNA-mediated MAD2B depletion, we found that CLTA was no longer concentrated at the mitotic spindle but, instead, diffusely distributed throughout the cell. In addition, we found a marked increase in the percentage of misaligned chromosomes. CONCLUSIONS/SIGNIFICANCE: Previously, we identified MAD2B as an interactor of the renal cell carcinoma (RCC)-associated protein PRCC. In addition, we found that fusion of PRCC with the transcription factor TFE3 in t(X;1)(p11;q21)-positive RCCs results in an impairment of this interaction and a concomitant failure to shuttle MAD2B to the nucleus. Our current data show that MAD2B interacts with CLTA during the G2/M phase of the cell cycle and that depletion of MAD2B leads to a marked increase in the percentage of misaligned chromosomes and a redistribution of CLTA during mitosis.


Subject(s)
Clathrin Light Chains/metabolism , Mitosis , Proteins/metabolism , Spindle Apparatus/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Clathrin Light Chains/genetics , G2 Phase , HEK293 Cells , Humans , Immunoprecipitation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mad2 Proteins , Microscopy, Confocal , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Binding , Proteins/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
10.
Cancer Genet Cytogenet ; 195(2): 105-11, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19963109

ABSTRACT

FBXW7 (alias CDC4) is a p53-dependent tumor suppressor gene that exhibits mutations or deletions in a variety of human tumors. Mutation or deletion of the FBXW7 gene has been associated with an increase in chromosomal instability and cell cycle progression. In addition, the FBXW7 protein has been found to act as a component of the ubiquitin proteasome system and to degrade several oncogenic proteins that function in cellular growth regulatory pathways. By using a rapid breakpoint cloning procedure in a case of renal cell cancer (RCC), we found that the FBXW7 gene was disrupted by a constitutional t(3;4)(q21;q31). Subsequent analysis of the tumor tissue revealed the presence of several anomalies, including loss of the derivative chromosome 3. Upon screening of a cohort of 29 independent primary RCCs, we identified one novel pathogenic mutation, suggesting that the FBXW7 gene may also play a role in the development of sporadic RCCs. In addition, we screened a cohort of 48 unrelated familial RCC cases with unknown etiology. Except for several known or benign sequence variants such as single nucleotide polymorphisms (SNPs), no additional pathogenic variants were found. Previous mouse models have suggested that the FBXW7 gene may play a role in the predisposition to tumor development. Here we report that disruption of this gene may predispose to the development of human RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Cycle Proteins/genetics , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 4 , F-Box Proteins/genetics , Kidney Neoplasms/genetics , Translocation, Genetic , Ubiquitin-Protein Ligases/genetics , Base Sequence , DNA Primers , F-Box-WD Repeat-Containing Protein 7 , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mutation , Polymerase Chain Reaction
11.
PLoS One ; 4(9): e7020, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19753112

ABSTRACT

BACKGROUND: Previously, we identified the mitotic arrest deficient protein MAD2B (MAD2L2) as a bona fide interactor of the renal cell carcinoma (RCC)-associated protein PRCC. In addition, we found that fusion of PRCC with the transcription factor TFE3 in t(X;1)(p11;q21)-positive RCCs results in an impairment of this interaction and, concomitantly, an abrogation of cell cycle progression. Although MAD2B is thought to inhibit the anaphase promoting complex (APC) by binding to CDC20 and/or CDH1(FZR1), its exact role in cell cycle control still remains to be established. METHODOLOGY/PRINCIPAL FINDINGS: Using a yeast two-hybrid interaction trap we identified the small GTPase RAN, a well-known cell cycle regulator, as a novel MAD2B binding protein. Endogenous interaction was established in mammalian cells via co-localization and co-immunoprecipitation of the respective proteins. The interaction domain of RAN could be assigned to a C-terminal moiety of 60 amino acids, whereas MAD2B had to be present in its full-length conformation. The MAD2B-RAN interaction was found to persist throughout the cell cycle. During mitosis, co-localization at the spindle was observed. CONCLUSIONS/SIGNIFICANCE: The small GTPase RAN is a novel MAD2B binding protein. This novel protein-protein interaction may play a role in (i) the control over the spindle checkpoint during mitosis and (ii) the regulation of nucleocytoplasmic trafficking during interphase.


Subject(s)
Mitosis , Proteins/metabolism , Saccharomyces cerevisiae/metabolism , ran GTP-Binding Protein/chemistry , Animals , COS Cells , Cdc20 Proteins , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Humans , Mad2 Proteins , Models, Biological , Protein Interaction Mapping , Two-Hybrid System Techniques
12.
Exp Cell Res ; 315(14): 2399-409, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19422821

ABSTRACT

Previously, we found that in t(X;1)(p11;q21)-positive renal cell carcinomas the bHLH-LZ transcription factor TFE3 is fused to a novel protein designated PRCC. In addition, we found that the PRCCTFE3 fusion protein, which has retained all known functional domains of TFE3, acts as a more potent transcriptional activator than wild type TFE3. We also found that PRCCTFE3 expression confers in vitro and in vivo transformation onto various cell types, including those of the kidney. Here we show that de novo expression of the PRCCTFE3 fusion protein provokes cell cycle delay. This delay, which is mediated by induction of the cyclin-dependent kinase inhibitor p21((WAF1/CIP1)), affects both the G1/S and the G2/M phases of the cell cycle and prevents the cells from undergoing polyploidization. We also show that the PRCCTFE3 fusion protein binds directly to the p21((WAF1/CIP1)) promoter and that the PRCCTFE3-induced up-regulation of p21((WAF1/CIP1)) leads to activation of the pRB pathway. Finally, we show that in t(X;1)(p11;q21)-positive renal tumor cells several processes that link PRCCTFE3 expression to p21((WAF1/CIP1))-mediated cell cycle delay are abrogated. Our data suggest a scenario in which, during the course of renal cell carcinoma development, an initial PRCCTFE3-induced cell cycle delay must be numbed, thus permitting continued proliferation and progression towards full-blown malignancy.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Carcinoma, Renal Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , HeLa Cells , Humans , Kidney Neoplasms/pathology , Nocodazole/pharmacology , Polyploidy , Promoter Regions, Genetic/physiology
13.
Breast Cancer Res Treat ; 114(1): 23-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18351453

ABSTRACT

Endocrine treatment of breast cancer is widely applied and effective. However, in advanced disease cases, the tumors will eventually progress into an estrogen-independent and therapy-resistant phenotype. To elucidate the molecular mechanisms underlying this endocrine therapy failure, we applied retroviral insertion mutagenesis to identify the main genes conferring estrogen independence to human breast cancer cells. Estrogen-dependent ZR-75-1 cells were infected with replication-defective retroviruses followed by selection with the anti-estrogen 4-hydroxy-tamoxifen. In the resulting panel of 79 tamoxifen-resistant cell lines, the viral integrations were mapped within the human genome. Genes located in the immediate proximity of the retroviral integration sites were characterized for altered expression and their capacity to confer anti-estrogen resistance when transfected into breast cancer cells. Out of 15 candidate BCAR (breast cancer anti-estrogen resistance) genes, seven (AKT1, AKT2, BCAR1, BCAR3, EGFR, GRB7, and TRERF1/BCAR2) were shown to directly underlie estrogen independence. Our results show that insertion mutagenesis is a powerful tool to identify BCAR loci, which may provide insights into the molecular and cellular mechanisms of breast tumor progression and therapy resistance thereby offering novel targets for the development of tailor-made therapeutical and prevention strategies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Estrogens/physiology , Cell Line, Tumor , Estrogens/genetics , Female , Humans , Mutagenesis, Insertional , Retroviridae , Virus Integration
14.
Cancer Genet Cytogenet ; 179(1): 11-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17981209

ABSTRACT

Our group and others had previously developed a high throughput procedure to map translocation breakpoints using chromosome flow sorting in conjunction with microarray-based comparative genomic hybridization (arrayCGH). Here we applied both conventional positional cloning and integrated arrayCGH procedures to the mapping of constitutional chromosome anomalies in four patients with renal cell cancer (RCC), three with a chromosome 3 translocation, and one with an insertion involving chromosome 3. In one of these patients, who was carrying a t(3;4)(p13;p15), the KCNIP4 gene was found to be disrupted. KCNIP4 belongs to a family of potassium channel-interacting proteins and is highly expressed in normal kidney cells. In addition, KCNIP4 splice variants have specifically been encountered in RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Kv Channel-Interacting Proteins/genetics , Translocation, Genetic , Cell Line, Tumor , Chromosome Breakage , Chromosome Mapping , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 4 , Cloning, Molecular , Humans , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Mutagenesis, Insertional
15.
Hum Mol Genet ; 14(14): 1955-63, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15917269

ABSTRACT

Recently, we identified a patient with an infantile sacrococcygeal teratoma and a constitutional t(12;15)(q13;q25). Here, we show that, as a result of this chromosomal translocation, the SUMO/Sentrin-specific protease 1 gene (SENP1) on chromosome 12 and the embryonic polarity-related mesoderm development gene (MESDC2) on chromosome 15 are disrupted and fused. Both reciprocal SENP1-MESDC2 (SEME) and MESDC2-SENP1 (MESE) fusion genes are transcribed in tumor-derived cells and their open reading frames encode aberrant proteins. As a consequence of this, and in contrast to wild-type (WT) MESDC2, the translocation-associated SEME protein is no longer targeted to the endoplasmatic reticulum, leading to a presumed loss-of-function as a chaperone for the WNT co-receptors LRP5 and/or LRP6. Ultimately, this might lead to abnormal development and/or routing of germ cell tumor precursor cells. SUMO, a post-translational modifier, plays an important role in several cellular key processes and is cleaved from its substrates by WT SENP1. Using a PML desumoylation assay, we found that translocation-associated MESE proteins exhibit desumoylation capacities similar to those observed for WT SENP1. We speculate that spatio-temporal disturbances in desumoylating activities during critical stages of embryonic development might have predisposed the patient. Together, the constitutional t(12;15)(q13;q25) translocation revealed two novel candidate genes for neonatal/infantile GCT development: MESDC2 and SENP1.


Subject(s)
Cell Polarity , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Endopeptidases/genetics , Molecular Chaperones/genetics , Teratoma/genetics , Translocation, Genetic , Animals , Blotting, Southern , Cell Line , Cricetinae , Cysteine Endopeptidases , DNA Primers , Humans , In Situ Hybridization, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...