Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Article En | MEDLINE | ID: mdl-24362253

The lack of toxicological information on many of the compounds that humans use or are exposed to, intentionally or unintentionally, poses a big problem in risk assessment. To fill this data gap, more emphasis is given to fast in vitro screening tools that can add toxicologically relevant information regarding the mode(s) of action via which compounds can elicit adverse effects, including genotoxic effects. By use of bioassays that can monitor the activation of specific cellular signalling pathways, many compounds can be screened in a high-throughput manner. We have developed two new specific reporter-gene assays that can monitor the effects of compounds on two pathways of interest: the p53 pathway (p53 CALUX) for genotoxicity and the Nrf2 pathway (Nrf2 CALUX) for oxidative stress. To exclude non-specific effects by compounds influencing the luciferase reporter-gene expression non-specifically, a third assay was developed to monitor changes in luciferase expression by compounds in general (Cytotox CALUX). To facilitate interpretation of the data and to avoid artefacts, all three reporter-gene assays used simple and defined reporter genes and a similar cellular basis, the human U2OS cell line. The three cell lines were validated with a range of reference compounds including genotoxic and non-genotoxic agents. The sensitivity (95%) and specificity (85%) of the p53 CALUX was high, showing that the assay is able to identify various types of genotoxic compound, while avoiding the detection of false positives. The Nrf2 CALUX showed specific responses to oxidants only, enabling the identification of compounds that elicit part of their genotoxicity via oxidative stress. All reporter-gene assays can be used in a high-throughput screening format and can be supplemented with other U2OS-based reporter-gene assays that can profile nuclear receptor activity, and several other signalling pathways.


DNA Damage , Luciferases/metabolism , Mutagenicity Tests/methods , Oxidative Stress , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Genes, Reporter/genetics , Humans , Luciferases/genetics , Luminescent Measurements , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reproducibility of Results , Response Elements/genetics , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenobiotics/classification , Xenobiotics/pharmacology
2.
Haematologica ; 96(10): 1478-87, 2011 Oct.
Article En | MEDLINE | ID: mdl-21791472

BACKGROUND: Several studies of pediatric acute myeloid leukemia have described the various type-I or type-II aberrations and their relationship with clinical outcome. However, there has been no recent comprehensive overview of these genetic aberrations in one large pediatric acute myeloid leukemia cohort. DESIGN AND METHODS: We studied the different genetic aberrations, their associations and their impact on prognosis in a large pediatric acute myeloid leukemia series (n=506). Karyotypes were studied, and hotspot regions of NPM1, CEPBA, MLL, WT1, FLT3, N-RAS, K-RAS, PTPN11 and KIT were screened for mutations of available samples. The mutational status of all type-I and type-II aberrations was available in 330 and 263 cases, respectively. Survival analysis was performed in a subset (n=385) treated on consecutive acute myeloid leukemia Berlin-Frankfurt-Munster Study Group and Dutch Childhood Oncology Group treatment protocols. RESULTS: Genetic aberrations were associated with specific clinical characteristics, e.g. significantly higher diagnostic white blood cell counts in MLL-rearranged, WT1-mutated and FLT3-ITD-positive acute myeloid leukemia. Furthermore, there was a significant difference in the distribution of these aberrations between children below and above the age of two years. Non-random associations, e.g. KIT mutations with core-binding factor acute myeloid leukemia, and FLT3-ITD with t(15;17)(q22;q21), NPM1- and WT1-mutated acute myeloid leukemia, respectively, were observed. Multivariate analysis revealed a 'favorable karyotype', i.e. t(15;17)(q22;q21), t(8;21)(q22;q22) and inv(16)(p13q22)/t(16;16)(p13;q22). NPM1 and CEBPA double mutations were independent factors for favorable event-free survival. WT1 mutations combined with FLT3-ITD showed the worst outcome for 5-year overall survival (22±14%) and 5-year event-free survival (20±13%), although it was not an independent factor in multivariate analysis. CONCLUSIONS: Integrative analysis of type-I and type-II aberrations provides an insight into the frequencies, non-random associations and prognostic impact of the various aberrations, reflecting the heterogeneity of pediatric acute myeloid leukemia. These aberrations are likely to guide the stratification of pediatric acute myeloid leukemia and may direct the development of targeted therapies.


Chromosome Aberrations , Genetic Heterogeneity , Leukemia, Myeloid, Acute/genetics , Adolescent , Age Factors , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Karyotype , Leukemia, Myeloid, Acute/mortality , Male , Mutation , Nucleophosmin , Prognosis , Sex Factors , Survival Analysis
3.
Am J Med Genet A ; 149A(7): 1468-75, 2009 Jul.
Article En | MEDLINE | ID: mdl-19449434

We present a family with multiple cytogenetic abnormalities, identified through a girl with several dysmorphic features and cardiac problems, suspected for Jacobsen syndrome. Cytogenetic analysis showed a 46,XX,del(11)(qter) karyotype, which was confirmed by fluorescence in situ hybridization (FISH). Cytogenetic investigation of the parents showed a chromosome aberration in both: the father had a t(11;12)(p13;q22) translocation and the mother was carrier of an ins(4;11)(p14;q24q25). FISH analysis with an 11q-subtelomeric probe from the second-generation telomere clone set and BACs from 11q24-q25 suggested a complex maternal rearrangement. However, subsequent array analysis showed a single interstitial deletion in the proband, derived from the maternal insertion. The aberrant karyotypes in both parents implicated an increased risk of unbalanced fetal chromosome composition, thus high risk for a child with multiple congenital abnormalities. Therefore, during the next pregnancy, the couple opted for prenatal diagnosis by means of amniocentesis. An interphase FISH strategy for uncultured amniotic fluid cells predicted two possible unbalanced fetal chromosome constitutions. Karyotyping of cultured amniotic cells confirmed one of the predicted unbalanced cytogenetic options, demonstrating the value of a fast interphase strategy for parents who both are carriers of a chromosomal abnormality. In addition, we present an overview of patients with Jacobsen syndrome and an interstitial 11q deletion reported thus far in literature.


Chromosome Deletion , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 4 , Mothers , Translocation, Genetic , Child, Preschool , Family , Female , Humans , Infant, Newborn , Inheritance Patterns , Jacobsen Distal 11q Deletion Syndrome/diagnosis , Jacobsen Distal 11q Deletion Syndrome/genetics , Male
4.
Genes Chromosomes Cancer ; 45(11): 1041-9, 2006 Nov.
Article En | MEDLINE | ID: mdl-16897742

The majority of translocations that involve the long arms of chromosomes 11 and 17 in acute myeloid leukemia appear identical on the cytogenetic level. Nevertheless, they are diverse on the molecular level. At present, two genes are known in 11q23 and four in 17q12-25 that generate five distinct fusion genes: MLL-MLLT6/AF17, MLL-LASP1, MLL-ACACA or MLL-SEPT9/MSF, and ZBTB16/PLZF-RARA. We analyzed 14 cases with a t(11;17) by fluorescence in situ hybridization and molecular genetic techniques and determined the molecular characteristics of their fusion genes. We identified six different gene fusions that comprised seven cases with a MLL-MLLT6/AF17, three with a MLL-SEPT9/MSF, and one each with MLL-LASP1, MLL-ACACA, and ZBTB16/PLZF-RARA fusions. In the remaining case, a MLL-SEPT6/Xq24 fusion suggested a complex rearrangement. The MLL-MLLT6/AF17 transcripts were extremely heterogeneous and the detection of seven different in-frame transcript and splice variants enabled us to predict the protein domains relevant for leukemogenesis. The putative MLL-MLLT6 consensus chimeric protein consists of the AT-hook DNA-binding, the methyltransferase, and the CXXC zinc-finger domains of MLL and the highly conserved octapeptide and the leucine-zipper dimerization motifs of MLLT6. The MLL-SEPT9 transcripts showed a similar high degree of variability. These analyses prove that the diverse types of t(11;17)-associated fusion genes can be reliably identified and delineated with a proper combination of cytogenetic and molecular genetic techniques. The heterogeneity of transcripts encountered in cases with MLL-MLLT6/AF17 and MLL-SEPT9/MSF fusions clearly demonstrates that thorough attention has to be paid to the appropriate selection of primers to cover all these hitherto unrecognized fusion variants.


Alternative Splicing , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 17 , Leukemia, Monocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Translocation, Genetic , Adolescent , Adult , Aged , Child , Child, Preschool , DNA-Binding Proteins/genetics , Female , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Infant , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Genes Chromosomes Cancer ; 45(8): 731-9, 2006 Aug.
Article En | MEDLINE | ID: mdl-16646086

The t(7;12)(q36;p13) is a recurrent translocation involving the ETV6/TEL gene (12p13) and a heterogeneous breakpoint at 7q36. A fusion transcript between HLXB9 and ETV6 in AML with t(7;12) is occasionally found. To study the incidence of t(7;12) in infant and childhood acute leukemia, we screened 320 cases <36 months using FISH. Additionally, 28 pediatric cases >36 months with cytogenetic breakpoints at 12p and 7q were investigated. We studied the presence of an HXLB9-ETV6 fusion transcript and quantified the expression of various genes located in the 7q36 breakpoint region. In total, six AML patients carried the t(7;12) of which five were infants and one child of 18 months. Only one out of 99 infant ALL patients harbored the t(7;12). No t(7;12) was found in older children with AML or ALL. AML patients carrying a t(7;12) had a poor outcome with a 3-year EFS of 0%. A fusion of HLXB9 to ETV6 was found in four AML cases with t(7;12). The 7q36 genes NOM1, LMBR1, RNF32, and SHH were equally expressed among t(7;12)-positive AML versus t(7;12)-negative AML, t(7;12)-negative ALL, or normal bone marrow. However, the HLXB9 expression was highly increased in t(7;12)-positive cases, including those with an HLXB9-ETV6 fusion. We conclude that the t(7;12) is almost exclusively present in infant AML and covers 30% of infant AML, while it is extremely rare in infant ALL and older children. The t(7;12) is associated with a poor outcome and an ectopic expression of HLXB9 is commonly involved in this genetic subtype of leukemia.


Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 7 , Homeodomain Proteins/genetics , Leukemia, Myeloid/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics , Translocation, Genetic , Acute Disease , Child, Preschool , Chromosome Breakage , Cohort Studies , Female , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Models, Genetic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Transcription Factors/metabolism , ETS Translocation Variant 6 Protein
6.
Genes Chromosomes Cancer ; 45(5): 437-46, 2006 May.
Article En | MEDLINE | ID: mdl-16419055

Chromosome rearrangements are found in many acute leukemias. As a result, genes at the breakpoints can be disrupted, forming fusion genes. One of the genes involved in several chromosome aberrations in hematological malignancies is NUP98 (11p15). As NUP98 is close to the 11p telomere, small translocations might easily be missed. Using a NUP98-specific split-signal fluorescence in situ hybridization (FISH) probe combination, we analyzed 84 patients with acute myeloid leukemia (AML), acute lymphoblastic leukemia, or myelodysplastic syndrome with either normal karyotypes or 11p abnormalities to investigate whether there are unidentified 11p15 rearrangements. Neither NUP98 translocations nor deletions were identified in cases with normal karyotypes, indicating these aberrations may be very rare in this group. However, NUP98 deletions were observed in four cases with unbalanced 11p aberrations, indicating that the breakpoint is centromeric of NUP98. Rearrangements of NUP98 were identified in two patients, both showing 11p abnormalities in the diagnostic karyotype: a t(4;11)(q1?3;p15) with expression of the NUP98-RAP1GDS1 fusion product detected in a 60-year-old woman with AML-M0, and an add(11)(p15) with a der(21)t(11;21)(p15;p13) observed cytogenetically in a 1-year-old boy with AML-M7. JARID1A was identified as the fusion partner of NUP98 using 3' RACE, RT-PCR, and FISH. JARID1A, at 12p13, codes for retinoblastoma binding protein 2, a protein implicated in transcriptional regulation. This is the first report of JARID1A as a partner gene in leukemia.


Chromosomes, Human, Pair 12 , Leukemia/genetics , Nuclear Pore Complex Proteins/genetics , Acute Disease , Adolescent , Adult , Aged , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , DNA Primers , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Karyotyping , Male , Middle Aged , Molecular Sequence Data
7.
Blood ; 106(5): 1817-23, 2005 Sep 01.
Article En | MEDLINE | ID: mdl-15899912

Drug resistance in childhood acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) is associated with impaired ability to induce apoptosis. To elucidate causes of apoptotic defects, we studied the protein expression of Apaf-1, procaspases-2, -3, -6, -7, -8, -10, and poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) in cells from children with acute lymphoblastic leukemia (ALL; n = 43) and acute myeloid leukemia (AML; n = 10). PARP expression was present in all B-lineage samples, but absent in 4 of 15 T-lineage ALL samples and 3 of 10 AML cases, which was not caused by genomic deletions. PARP expression was a median 7-fold lower in T-lineage ALL (P < .001) and 10-fold lower in AML (P < .001) compared with B-lineage ALL. PARP expression was 4-fold lower in prednisolone, vincristine and L-asparaginase (PVA)-resistant compared with PVA-sensitive ALL patients (P < .001). Procaspase-2 expression was 3-fold lower in T-lineage ALL (P = .022) and AML (P = .014) compared with B-lineage ALL. In addition, procaspase-2 expression was 2-fold lower in PVA-resistant compared to PVA-sensitive ALL patients (P = .042). No relation between apoptotic protease-activating factor 1 (Apaf-1), procaspases-3, -6, -7, -8, -10, and drug resistance was found. In conclusion, low baseline expression of PARP and procaspase-2 is related to cellular drug resistance in childhood acute lymphoblastic leukemia.


Caspases/metabolism , Drug Resistance, Neoplasm/physiology , Poly(ADP-ribose) Polymerases/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Apoptosis/physiology , Caspase 2 , Caspases/genetics , Cell Line, Tumor , Child , Drug Screening Assays, Antitumor , Gene Expression Regulation, Enzymologic , Humans , Poly(ADP-ribose) Polymerases/genetics , RNA, Messenger/genetics , RNA, Messenger/physiology
8.
Genes Chromosomes Cancer ; 39(4): 324-34, 2004 Apr.
Article En | MEDLINE | ID: mdl-14978793

The t(9;11) has been described in patients with acute myeloid leukemia (AML), and two genes [AF9 (at 9p21) and FBP17 (at 9q34)] have been cloned as fusion partners of the MLL gene. From an AML-M5 with a t(9;11)(q34;q23), we identified a novel MLL fusion partner, AF9Q34. The AF9Q34 protein shows high homology with nGAP, a RAS GTPase-activating protein (RASGAP), and contains the highly conserved GRD and FLR motifs characteristic of RASGAPs. Recently, the rat homologue (DAB2IP) also was identified and reported to act as a RASGAP both in vivo and in vitro. RASGAPs negatively regulate the activity of RAS proteins that modulate diverse cellular processes by cycling between an inactive GDP-bound and an active GTP-bound state. In addition, the NH(2) terminus harbors an amino acid stretch with homology to the pleckstrin homology (PH) domain implicated in regulating the interaction between RAS and the catalytic domain of RASGAP. As a result of the breakpoint in the AF9Q34-MLL fusion protein, this PH domain is disrupted. This suggests that because of the translocation, the normal function of the AF9Q34 gene is aborted. Thus, AF9Q34 encodes a novel RASGAP gene that appears to be deregulated as a result of the translocation. The identification of this RASGAP protein in a novel MLL fusion implies that an indirect RAS-deregulating mechanism could be involved in leukemic transformation.


DNA-Binding Proteins/genetics , Leukemia, Monocytic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogenes , Transcription Factors , ras GTPase-Activating Proteins/genetics , Amino Acid Sequence/genetics , Base Sequence/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Chromosome Breakage/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 9/genetics , Fatty Acid-Binding Proteins , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , HL-60 Cells/chemistry , HL-60 Cells/metabolism , HeLa Cells/chemistry , HeLa Cells/metabolism , Histone-Lysine N-Methyltransferase , Humans , Jurkat Cells/chemistry , Jurkat Cells/metabolism , K562 Cells/chemistry , K562 Cells/metabolism , Male , Middle Aged , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Organ Specificity/genetics , Translocation, Genetic/genetics , U937 Cells/chemistry , U937 Cells/metabolism
9.
Mol Hum Reprod ; 10(2): 77-83, 2004 Feb.
Article En | MEDLINE | ID: mdl-14742691

Anti-Müllerian hormone (AMH) is a member of the transforming growth factor-beta superfamily, which plays an important role in both ovarian primordial follicle recruitment and dominant follicle selection in mice. However, the role of AMH in folliculogenesis in humans has not been investigated in detail. In the present study, AMH expression was assessed using immunohistochemistry in ovarian sections, obtained from healthy regularly cycling women. To this end, a novel monoclonal antibody to human AMH was developed. AMH expression was not observed in primordial follicles, whereas 74% of the primary follicles showed at least a weak signal in the granulosa cells. The highest level of AMH expression was present in the granulosa cells of secondary, preantral and small antral follicles

Glycoproteins/metabolism , Ovarian Follicle/metabolism , Testicular Hormones/metabolism , Adolescent , Adult , Animals , Anti-Mullerian Hormone , Antibodies, Monoclonal , Blotting, Western , Female , Glycoproteins/analysis , Glycoproteins/immunology , Granulosa Cells/cytology , Granulosa Cells/metabolism , Humans , Immunohistochemistry , Male , Mice , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Staining and Labeling , Testicular Hormones/analysis , Testicular Hormones/immunology
11.
Genes Chromosomes Cancer ; 35(1): 92-6, 2002 Sep.
Article En | MEDLINE | ID: mdl-12203795

Infant acute lymphoblastic leukemia (ALL) with MLL gene rearrangements is characterized by a proB phenotype and a poor clinical outcome. We analyzed an infant proB ALL with t(2;11)(p15;p14) and an MLL rearrangement on Southern blot analysis. Rapid amplification of cDNA ends-polymerase chain reaction (PCR) and reverse transcriptase-PCR identified the LAF4 gene mapped on chromosome region 2q11.2-q12 as a fusion partner of the MLL gene. The LAF4 gene was identified previously by its high sequence homology to the AF4 protein and encodes a protein of 1,227 amino acids. The t(4;11)(q21;q23), creating the MLL-AF4 chimeric transcripts, is the predominant 11q23 chromosome translocation in infant ALL and is associated with an extremely poor prognosis. Our findings further suggest that fusion of MLL to one of the AF4 family members (AF4/LAF4/AF5Q31) might determine a proB-cell phenotype in infant leukemia.


DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogenes , Transcription Factors , Amino Acid Sequence/genetics , Base Sequence/genetics , Chromosome Mapping , Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 4/genetics , Female , Histone-Lysine N-Methyltransferase , Humans , Infant , Karyotyping , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Sequence Homology, Nucleic Acid , Transcriptional Elongation Factors , Translocation, Genetic/genetics
...