Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
BMC Pregnancy Childbirth ; 22(1): 134, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35180852

ABSTRACT

BACKGROUND: The Mothers Autonomy in Decision Making Scale (MADM) assesses women's autonomy and role in decision making. The Mothers on Respect Index (MORi) asseses women's experiences of respect when interacting with their healthcare providers. The Childbirth Experience Questionnaire 2.0 assesses the overall experience of childbirth (CEQ2.0). There are no validated Dutch measures of the quality of women's experiences in the intrapartum period. Therefore, the aim of this study was to evaluate the psychometric properties of these measures in their Dutch translations. METHODS: The available Dutch versions of the MADM and MORi were adapted to assess experiences in the intrapartum period. The CEQ2.0 was translated by using forward-backward procedures. The three measures were included in an online survey including items on individual characteristics (i.e. maternal, birth, birth interventions). Reliability was assessed by calculating Cronbach's alphas. Mann-Whitney, Kruskal Wallis or Student T-tests were applied where appropriate, to assess discrimination between women who differed on individual characteristics (known group validity). We hypothesized that women who experienced pregnancy complications and birth interventions would have statistically lower scores on the MADM, MORi and CEQ2.0, compared with women who had healthy pregnancies and physiological births. Convergent validity was assessed using Spearman Rank correlations between the MADM, MORi and/or CEQ2.0. We hypothesized moderate to strong correlations between these measures. Women's uptake of and feedback on the measures were tracked to assess acceptability and clarity. RESULTS: In total 621 women were included in the cross sectional study. The calculated Cronbach's alphas for the MADM, MORi and CEQ, were ≥ 0.77. Knowngroup validity was confirmed through significant differences on all relevant individual characteristics, except for vaginal laceration repair. Spearman Rank correlations ranged from 0.46-0.80. In total 98% of the included women out of the eligible population completed the MADM and MORi for each healthcare professional they encountered during childbirth. The proportions of MADM and MORi-items which were difficult to complete ranged from 0.0-10.8%, 0.6-2.7%, respectively. CONCLUSIONS: The results of our study showed that the Dutch version of the MADM, MORi and CEQ2.0 in Dutch are valid instruments that can be used to assess women's experiences in the intrapartum period.


Subject(s)
Labor, Obstetric/psychology , Parturition/psychology , Perinatal Care , Peripartum Period/psychology , Psychometrics , Surveys and Questionnaires , Adult , Cross-Sectional Studies , Decision Making , Female , Humans , Netherlands , Personal Autonomy , Pregnancy , Reproducibility of Results , Respect , Translations
2.
Oncogene ; 35(11): 1341-52, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26050622

ABSTRACT

Polycomb Group (PcG) proteins form Polycomb Repressive Complexes (PRCs) that function as epigenetic repressors of gene expression. The large variety of PcG proteins, in addition to the high number of paralogs, allows for the formation of diverse PRCs with different properties, providing fine-tuned control over cell specification. Initially identified as being oncogenes, a small number of PcG genes are involved in tumor development in part through the repression of the CDKN2A locus. Therefore, enhanced PcG-mediated repression has long been assumed to be cancer promoting. However, recent data have revealed that for some cancers, PcG proteins act as tumor suppressors, indicating that this traditional view is oversimplified. In this review, we present an overview of the roles of PcG genes in oncogenesis and how the nature of their role is context dependent.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epigenetic Repression/genetics , Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 2/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Neoplasms/pathology , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Proteins/genetics
3.
Med Teach ; 31(11): e494-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19909026

ABSTRACT

BACKGROUND: Previous research revealed relationships between learning strategies and knowledge acquisition. During clerkships, however, students' focus widens beyond mere knowledge acquisition as they further develop overall competence. This shift in focus can influence learning strategy use. AIM: We explored which learning strategies were used during clerkships and their relationship to clinical performance. METHODS: Participants were 113 (78%) clerks at the university hospital or one of six affiliated hospitals. Learning strategies were assessed using the 'Approaches to Learning at Work Questionnaire' (deep, surface-rational and surface-disorganised learning). Clinical performance was calculated by taking the mean of clinical assessment marks. The relationship between learning strategies and clinical performance was explored using regression analysis. RESULTS: Most students (89%) did not clearly prefer a single learning strategy. No relationship was found between learning strategies and clinical performance. DISCUSSION: Since overall competence comprises integration of knowledge, skills and professional behaviour, we assume that students without a clear preference use more than one learning strategy. Finding no relationship between learning strategies and clinical performance reflects the complexity of clinical learning. Depending on circumstances it may be important to obtain relevant information quickly (surface-rational) or understand material thoroughly (deep). In future research we will examine when and why students use different learning strategies.


Subject(s)
Clinical Clerkship , Clinical Competence/standards , Learning , Humans , Students, Medical , Surveys and Questionnaires
4.
Oncogene ; 27(54): 6806-16, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18762810

ABSTRACT

c-Myc drives uncontrolled cell proliferation in various human cancers. However, in mouse embryo fibroblasts (MEFs), c-Myc also induces apoptosis by activating the p19Arf tumor suppressor pathway. Tbx2, a transcriptional repressor of p19Arf, can collaborate with c-Myc by suppressing apoptosis. MEFs overexpressing c-Myc and Tbx2 are immortal but not transformed. We have performed an unbiased genetic screen, which identified 12 oncogenes that collaborate with c-Myc and Tbx2 to transform MEFs in vitro. One of them encodes the LPA2 receptor for the lipid growth factor lysophosphatidic acid (LPA). We find that LPA1 and LPA4, but not LPA3, can reproduce the transforming effect of LPA2. Using pharmacological inhibitors, we show that the in vitro cell transformation induced by LPA receptors is dependent on the Gi-linked ERK and PI3K signaling pathways. The transforming ability of LPA1, LPA2 and LPA4 was confirmed by tumor formation assays in vivo and correlated with prolonged ERK1/2 activation in response to LPA. Our results reveal a direct role for LPA receptor signaling in cell transformation and tumorigenesis in conjunction with c-Myc and reduced p19Arf expression.


Subject(s)
Cell Transformation, Neoplastic , Genes, myc , Lysophospholipids/physiology , Receptors, Lysophosphatidic Acid/physiology , Animals , Cell Division/physiology , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Embryo, Mammalian/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic , Genetic Testing/methods , Humans , Mice , Neoplasms/pathology
5.
J Clin Pathol ; 61(6): 744-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18326020

ABSTRACT

BACKGROUND: The prognosis of adenoid cystic carcinoma (ACC), a malignant salivary gland tumour, depends on clinicopathological parameters. To decipher the biological behaviour of ACC, and to identify patients at risk of developing metastases, additional markers are needed. METHODS: Expression of the cell cycle proteins p53, cyclin D1, p16(INK4a), E2F1 and Ki-67, together with the Polycomb group (PcG) proteins BMI-1, MEL-18, EZH2 and EED was investigated immunohistochemically 21 formalin-fixed, paraffin-embedded primary ACCs in relation to tumour characteristics. RESULTS: ACC revealed significantly increased expression of the cell cycle proteins compared to normal salivary tissue (n = 17). Members of the two PcG complexes displayed mutually exclusive expression in normal salivary gland tissue, with BMI-1 and MEL-18 being abundantly present. In ACC, this expression pattern was disturbed, with EZH2 and EED showing significantly increased expression levels. In univariate analysis, presence of recurrence, poor differentiation and high EZH2 levels (>25% immunopositivity) significantly correlated with unfavourable outcome. ACCs with high proliferative rate (>25% Ki-67 immunopositivity) significantly correlated with high levels of EZH2 and p16. Only the development of recurrence was an independent prognostic factor of survival in multivariate analysis. CONCLUSIONS: Expression of PcG complexes and of essential cell cycle proteins is highly deregulated in ACC. Also, EZH2 expression has prognostic relevance in this malignancy.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Adenoid Cystic/chemistry , DNA-Binding Proteins/analysis , Salivary Gland Neoplasms/chemistry , Transcription Factors/analysis , Adult , Aged , Aged, 80 and over , Carcinoma, Adenoid Cystic/mortality , Carcinoma, Adenoid Cystic/pathology , Cell Cycle Proteins/analysis , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Female , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Polycomb Repressive Complex 2 , Prognosis , Proportional Hazards Models , Salivary Gland Neoplasms/mortality , Salivary Gland Neoplasms/pathology , Survival Analysis
6.
Br J Cancer ; 95(9): 1202-11, 2006 Nov 06.
Article in English | MEDLINE | ID: mdl-17024127

ABSTRACT

We used SEREX technology to identify novel tumour-associated antigens in patients with primary hepatocellular carcinoma and found serological responses to the polycomb group (PcG) protein BMI-1, which is overexpressed in a range of different tumour types. Further studies identified T-cell responses to both BMI-1 and another PcG protein, EZH2, in cancer patients and at relatively lower levels in some normal donors. We next identified several CD8+ T-cell epitopes derived from BMI-1 and EZH2 and demonstrated that EZH2-derived peptides elicited more significant interferon-gamma (IFN-gamma) release than BMI-1-derived peptides. That CD8(+) T cells were responsible for the observed responses was confirmed for EZH2 by both IFN-gamma capture assays and tetramer staining using an HLA-A0201-restricted, EZH2-derived YMSCSFLFNL (aa 666-674) epitope. The ability of YMSCSFLFNL (aa 666-674) to stimulate the in vitro expansion of specific T cells from peripheral blood lymphocytes was greatly enhanced when the CD25(+) T-cell population was depleted. EZH2-specific cytotoxic T lymphocyte clones specific for two HLA-A0201 epitopes were generated and found to recognise endogenously processed EZH2 in both HLA-matched fibroblasts and tumour cell lines. Given the widespread overexpression of PcG proteins in cancer and their critical role in oncogenesis, these data suggest that they may be useful targets for cancer immunotherapy.


Subject(s)
DNA-Binding Proteins/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , DNA-Binding Proteins/analysis , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Interferon-gamma/biosynthesis , Interleukin-2 Receptor alpha Subunit/analysis , Leukocytes, Mononuclear/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/analysis , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/analysis
7.
Oncogene ; 24(52): 7656-72, 2005 Nov 21.
Article in English | MEDLINE | ID: mdl-16299527

ABSTRACT

Retroviral insertion mutagenesis screens in mice are powerful tools for efficient identification of oncogenic mutations in an in vivo setting. Many oncogenes identified in these screens have also been shown to play a causal role in the development of human cancers. Sequencing and annotation of the mouse genome, along with recent improvements in insertion site cloning has greatly facilitated identification of oncogenic events in retrovirus-induced tumours. In this review, we discuss the features of retroviral insertion mutagenesis screens, covering the mechanisms by which retroviral insertions mutate cellular genes, the practical aspects of insertion site cloning, the identification and analysis of common insertion sites, and finally we address the potential for use of somatic insertional mutagens in the study of nonhaematopoietic and nonmammary tumour types.


Subject(s)
Mutagenesis, Insertional , Retroviridae/genetics , Cloning, Molecular , DNA Transposable Elements , Genes, Tumor Suppressor , Genetic Testing , Humans , Neoplasms/genetics , Proto-Oncogenes
9.
Br J Cancer ; 84(10): 1372-6, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11355949

ABSTRACT

Genes of the polycomb group function by silencing homeotic selector genes that regulate embryogenesis. In mice, downregulation of one of the polycomb genes, bmi-1, leads to neurological alterations and severe proliferative defects in lymphoid cells, whilst bmi-1 overexpression, together with upregulation of myc-1, induces lymphoma. An oncogenic function has been further supported in primary fibroblast studies where bmi-1 overexpression induces immortalization due to repression of p16/p19ARF, and where together with H-ras, it readily transforms MEFs. It was the aim of this study to assess the expression of bmi-1 in resectable non-small cell lung cancer (NSCLC) in association with p16 and p14ARF (=human p19ARF). Tumours (48 resectable NSCLC (32 squamous, 9 adeno-, 2 large cell, 4 undifferentiated carcinomas and 1 carcinoid); stage I, 29, II, 7, III, 12; T1, 18, T2, 30; differentiation: G1 12, G2 19, G3 17) were studied by immunohistochemistry for protein expression and by comparative multiplex PCR for gene amplification analysis. In tumour-free, normal lung tissue from patients, weak - moderate bmi-1 staining was seen in some epithelial cells, lymphocytes, glandular cells and in fibroblasts, whereas blood, endothelial, chondrocytes, muscle cells and adipocytes did not exhibit any bmi-1 expression. In tumours, malignant cells were negative/weakly, moderately and strongly positive in 20, 22 and 6 cases, respectively. As assessed by multiplex PCR, bmi-1 gene amplification was not the reason for high-level bmi-1 expression. Tumours with moderate or strong bmi-1 expression were more likely to have low levels of p16 and p14ARF (P = 0.02). Similarly, tumours negative for both, p16 and p14ARF, exhibit moderate-strong bmi-1 staining. 58% of resectable NSCLC exhibit moderate-high levels of bmi-1 protein. The inverse correlation of bmi-1 and the INK4 locus proteins expression (p16/p14ARF) supports a possible role for bmi-1 misregulation in lung carcinogenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins , Aged , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Cell Line , Female , Humans , Lung/cytology , Lung/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Nuclear Proteins/analysis , Organ Specificity , Polycomb Repressive Complex 1 , Polymerase Chain Reaction , Proto-Oncogene Proteins/analysis , Survival Rate , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF , Zinc Fingers
10.
Cancer Res ; 61(6): 2409-12, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11289106

ABSTRACT

The BMI-1 gene is a putative oncogene belonging to the Polycomb group family that cooperates with c-myc in the generation of mouse lymphomas and seems to participate in cell cycle regulation and senescence by acting as a transcriptional repressor of the INK4a/ARF locus. The BMI-1 gene has been located on chromosome 10p13, a region involved in chromosomal translocations in infant leukemias, and amplified in occasional non-Hodgkin's lymphomas (NHLs) and solid tumors. To determine the possible alterations of this gene in human malignancies, we have examined 160 lymphoproliferative disorders, 13 myeloid leukemias, and 89 carcinomas by Southern blot analysis and detected BMI-1 gene amplification (3- to 7-fold) in 4 of 36 (11%) mantle cell lymphomas (MCLs) with no alterations in the INK4a/ARF locus. BMI-1 and p16INK4a mRNA and protein expression were also studied by real-time quantitative reverse transcription-PCR and Western blot, respectively, in a subset of NHLs. BMI-1 expression was significantly higher in chronic lymphocytic leukemia and MCL than in follicular lymphoma and large B cell lymphoma. The four tumors with gene amplification showed significantly higher mRNA levels than other MCLs and NHLs with the BMI-1 gene in germline configuration. Five additional MCLs also showed very high mRNA levels without gene amplification. A good correlation between BMI-1 mRNA levels and protein expression was observed in all types of lymphomas. No relationship was detected between BMI-1 and p16INK4a mRNA levels. These findings suggest that BMI-1 gene alterations in human neoplasms are uncommon, but they may contribute to the pathogenesis in a subset of malignant lymphomas, particularly of mantle cell type.


Subject(s)
Gene Amplification , Lymphoma, Mantle-Cell/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression , Genes, Tumor Suppressor , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Lymphoma, Mantle-Cell/metabolism , Nuclear Proteins/biosynthesis , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Development ; 128(9): 1587-97, 2001 May.
Article in English | MEDLINE | ID: mdl-11290297

ABSTRACT

Polycomb group genes were identified as a conserved group of genes whose products are required in multimeric complexes to maintain spatially restricted expression of Hox cluster genes. Unlike in Drosophila, in mammals Polycomb group (PcG) genes are represented as highly related gene pairs, indicative of duplication during metazoan evolution. Mel18 and Bmi1 are mammalian homologs of Drosophila Posterior sex combs. Mice deficient for Mel18 or Bmi1 exhibit similar posterior transformations of the axial skeleton and display severe immune deficiency, suggesting that their gene products act on overlapping pathways/target genes. However unique phenotypes upon loss of either Mel18 or Bmi1 are also observed. We show using embryos doubly deficient for Mel18 and Bmi1 that Mel18 and Bmi1 act in synergy and in a dose-dependent and cell type-specific manner to repress Hox cluster genes and mediate cell survival of embryos during development. In addition, we demonstrate that Mel18 and Bmi1, although essential for maintenance of the appropriate expression domains of Hox cluster genes, are not required for the initial establishment of Hox gene expression. Furthermore, we show an unexpected requirement for Mel18 and Bmi1 gene products to maintain stable expression of Hox cluster genes in regions caudal to the prospective anterior expression boundaries during subsequent development.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Homeobox , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Animals , Bone and Bones/embryology , Embryonic and Fetal Development/genetics , Gene Dosage , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Mice , Mice, Mutant Strains , Polycomb Repressive Complex 1 , Polycomb-Group Proteins
12.
Curr Opin Genet Dev ; 11(2): 175-81, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250141

ABSTRACT

Polycomb group (PcG) proteins maintain silencing at target loci in higher eukaryotes but recent evidence suggests that about half of these proteins are also required for maintenance of activation at homeotic loci. We suggest that PcG and trithorax group response elements should acquire a new name, 'maintenance elements', to reflect the dual function of regulatory elements that bind both groups of proteins. New data suggest that there might be a functional link between PcG repression and cell-cycle regulation.


Subject(s)
Drosophila Proteins , Insect Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors , Animals , Binding Sites , Cell Cycle , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , Insect Proteins/genetics , Mice , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Repressor Proteins/genetics , Response Elements
13.
Nat Genet ; 26(3): 291-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062467

ABSTRACT

To identify new immortalizing genes with potential roles in tumorigenesis, we performed a genetic screen aimed to bypass the rapid and tight senescence arrest of primary fibroblasts deficient for the oncogene Bmi1. We identified the T-box member TBX2 as a potent immortalizing gene that acts by downregulating Cdkn2a (p19(ARF)). TBX2 represses the Cdkn2a (p19(ARF)) promoter and attenuates E2F1, Myc or HRAS-mediated induction of Cdkn2a (p19(ARF)). We found TBX2 to be amplified in a subset of primary human breast cancers, indicating that it might contribute to breast cancer development.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , Cell Cycle Proteins/physiology , Cellular Senescence/genetics , Chromosomes, Human, Pair 17/genetics , DNA-Binding Proteins , Gene Amplification , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Protein Biosynthesis , Repressor Proteins/physiology , T-Box Domain Proteins/physiology , Adenocarcinoma/metabolism , Animals , Breast Neoplasms/metabolism , COS Cells , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/isolation & purification , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , E2F Transcription Factors , E2F1 Transcription Factor , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Genes, BRCA1 , Humans , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/isolation & purification , Neoplastic Syndromes, Hereditary/genetics , Nuclear Proteins/genetics , Oncogenes , Polycomb Repressive Complex 1 , Promoter Regions, Genetic , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/isolation & purification , Retinoblastoma-Binding Protein 1 , T-Box Domain Proteins/genetics , T-Box Domain Proteins/isolation & purification , Transcription Factor DP1 , Transcription Factors/antagonists & inhibitors , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF
14.
EMBO J ; 19(18): 4986-96, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10990462

ABSTRACT

Locus control regions (LCRs) alleviate chromatin-mediated transcriptional repression. Incomplete LCRs partially lose this property when integrated in transcriptionally restrictive genomic regions such as centromeres. This frequently results in position effect variegation (PEV), i.e. the suppression of expression in a proportion of the cells. Here we show that this PEV is influenced by the heterochromatic protein SUV39H1 and by the Polycomb group proteins M33 and BMI-1. A concentration variation of these proteins modulates the proportion of cells expressing human globins in a locus-dependent manner. Similarly, the transcription factors Sp1 or erythroid Krüppel-like factor (EKLF) also influence PEV, characterized by a change in the number of expressing cells and the chromatin structure of the locus. However, in contrast to results obtained in a euchromatic locus, EKLF influences the expression of the gamma- more than the beta-globin genes, suggesting that the relief of silencing is caused by the binding of EKLF to the LCR and that genes at an LCR proximal position are more likely to be in an open chromatin state than genes at a distal position.


Subject(s)
Chromatin/metabolism , Globins/genetics , Suppression, Genetic , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Line , Chromosome Mapping , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Gene Silencing , Globins/biosynthesis , Humans , In Situ Hybridization, Fluorescence , Kruppel-Like Transcription Factors , Liver/embryology , Liver/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Transcriptional Activation
15.
Proc Natl Acad Sci U S A ; 96(25): 14372-7, 1999 Dec 07.
Article in English | MEDLINE | ID: mdl-10588712

ABSTRACT

Control of cell identity during development is specified in large part by the unique expression patterns of multiple homeobox-containing (Hox) genes in specific segments of an embryo. Trithorax and Polycomb-group (Trx-G and Pc-G) proteins in Drosophila maintain Hox expression or repression, respectively. Mixed lineage leukemia (MLL) is frequently involved in chromosomal translocations associated with acute leukemia and is the one established mammalian homologue of Trx. Bmi-1 was first identified as a collaborator in c-myc-induced murine lymphomagenesis and is homologous to the Drosophila Pc-G member Posterior sex combs. Here, we note the axial-skeletal transformations and altered Hox expression patterns of Mll-deficient and Bmi-1-deficient mice were normalized when both Mll and Bmi-1 were deleted, demonstrating their antagonistic role in determining segmental identity. Embryonic fibroblasts from Mll-deficient compared with Bmi-1-deficient mice demonstrate reciprocal regulation of Hox genes as well as an integrated Hoxc8-lacZ reporter construct. Reexpression of MLL was able to overcome repression, rescuing expression of Hoxc8-lacZ in Mll-deficient cells. Consistent with this, MLL and BMI-I display discrete subnuclear colocalization. Although Drosophila Pc-G and Trx-G members have been shown to maintain a previously established transcriptional pattern, we demonstrate that MLL can also dynamically regulate a target Hox gene.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins , Embryonic and Fetal Development , Insect Proteins/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogenes , Repressor Proteins , Transcription Factors , Animals , Bone and Bones/abnormalities , Female , Gene Expression Regulation, Developmental , Genes, Homeobox , Histone-Lysine N-Methyltransferase , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein , Polycomb Repressive Complex 1 , Pregnancy
16.
J Cell Sci ; 112 ( Pt 24): 4627-39, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10574711

ABSTRACT

The human proto-oncogene Bmi1 is a member of the mammalian Polycomb Group (Pc-G) genes. The subnuclear distribution of the BMI1 protein was studied in several primary human and tumor-derived cell lines using immunohistochemical and biochemical methods. In primary and tumor cells, nuclear BMI1 shows a fine-grain distribution over chromatin, usually dense in interphase nuclei and significantly weaker along mitotic chromosomes. In addition, BMI1 preferentially associates with several distinct heterochromatic domains in tumor cell lines. In both primary and tumor cell lines a marked cell cycle-regulation of Pc-G-chromatin interaction is observed: nuclear BMI1-staining dissipates in late S phase and is re-established early in G(1)-phase. Chromatin-association of BMI1 inversely correlates with its phosphorylation status in a cell cycle-dependent fashion: at G(1)/S, hypophosphorylated BMI1 is specifically retained in the chromatin-associated nuclear protein fraction, whereas during G(2)/M, phosphorylated BMI1 is not chromatin-bound. Our findings indicate a strict cell cycle-controlled regulation of Pc-G complex-chromatin association and provide molecular tools for improving our understanding of Pc-G complex regulation and function in mammalian cells.


Subject(s)
Cell Cycle/physiology , Chromatin/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromosomes, Human, Pair 1 , Fluorescent Antibody Technique, Indirect , Humans , In Situ Hybridization, Fluorescence , Nuclear Proteins/genetics , Phosphorylation , Polycomb Repressive Complex 1 , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics
17.
Genes Dev ; 13(20): 2678-90, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10541554

ABSTRACT

The bmi-1 and myc oncogenes collaborate strongly in murine lymphomagenesis, but the basis for this collaboration was not understood. We recently identified the ink4a-ARF tumor suppressor locus as a critical downstream target of the Polycomb-group transcriptional repressor Bmi-1. Others have shown that part of Myc's ability to induce apoptosis depends on induction of p19arf. Here we demonstrate that down-regulation of ink4a-ARF by Bmi-1 underlies its ability to cooperate with Myc in tumorigenesis. Heterozygosity for bmi-1 inhibits lymphomagenesis in Emu-myc mice by enhancing c-Myc-induced apoptosis. We observe increased apoptosis in bmi-1(-/-) lymphoid organs, which can be rescued by deletion of ink4a-ARF or overexpression of bcl2. Furthermore, Bmi-1 collaborates with Myc in enhancing proliferation and transformation of primary embryo fibroblasts (MEFs) in an ink4a-ARF dependent manner, by prohibiting Myc-mediated induction of p19arf and apoptosis. We observe strong collaboration between the Emu-myc transgene and heterozygosity for ink4a-ARF, which is accompanied by loss of the wild-type ink4a-ARF allele and formation of highly aggressive B-cell lymphomas. Together, these results reinforce the critical role of Bmi-1 as a dose-dependent regulator of ink4a-ARF, which on its turn acts to prevent tumorigenesis on activation of oncogenes such as c-myc.


Subject(s)
Apoptosis/genetics , Genes, myc , Genes, p16 , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/genetics , Nuclear Proteins/genetics , Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins , Animals , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cocarcinogenesis , Down-Regulation , Female , Gene Expression , Genes, bcl-2 , Heterozygote , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Polycomb Repressive Complex 1 , Tumor Suppressor Protein p14ARF
18.
Genes Dev ; 13(20): 2691-703, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10541555

ABSTRACT

The murine Polycomb-Group (PcG) proteins Eed and Bmi1 govern axial patterning during embryonic development by segment-specific repression of Hox gene expression. The two proteins engage in distinct multimeric complexes that are thought to use a common molecular mechanism to render the regulatory regions of Hox and other downstream target genes inaccessible to transcriptional activators. Beyond axial patterning, Bmi1 is also involved in hemopoiesis because a loss-of-function allele causes a profound decrease in bone marrow progenitor cells. Here, evidence is presented that is consistent with an antagonistic function of eed and Bmi1 in hemopoietic cell proliferation. Heterozygosity for an eed null allele causes marked myelo- and lymphoproliferative defects, indicating that eed is involved in the negative regulation of the pool size of lymphoid and myeloid progenitor cells. This antiproliferative function of eed does not appear to be mediated by Hox genes or the tumor suppressor locus p16(INK4a)/p19(ARF) because expression of these genes was not altered in eed mutants. Intercross experiments between eed and Bmi1 mutant mice revealed that Bmi1 is epistatic to eed in the control of primitive bone marrow cell proliferation. However, the genetic interaction between the two genes is cell-type specific as the presence of one or two mutant alleles of eed trans-complements the Bmi1-deficiency in pre-B bone marrow cells. These studies thus suggest that hemopoietic cell proliferation is regulated by the relative contribution of repressive (Eed-containing) and enhancing (Bmi1-containing) PcG gene complexes.


Subject(s)
Hematopoiesis/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Aging/genetics , Aging/pathology , Animals , Cell Division/genetics , Colony-Forming Units Assay , Female , Gene Expression Regulation, Developmental , Genes, Homeobox , Genes, Regulator , Genes, p16 , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Polycomb-Group Proteins
19.
Semin Cell Dev Biol ; 10(2): 227-35, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10441076

ABSTRACT

The Polycomb-group constitutes an important, widely conserved group of transcriptional repressors best known for their function in stably maintaining the inactive expression patterns of key developmental regulators, including homeotic genes. Together with the counteracting trithorax-group proteins, they establish a form of cellular memory by faithfully maintaining transcription states determined early in embryogenesis. Besides being crucial for the correct execution of developmental programs, Polycomb-group mediated silencing also appears to be involved in controlling cell proliferation. Here we discuss several aspects of Pc-G function: target gene specificity and recognition as well as propagation of inactive chromatin states to subsequent cell generations.


Subject(s)
Chromatin/ultrastructure , Drosophila Proteins , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental/physiology , Insect Proteins/physiology , Repressor Proteins/physiology , Transcription Factors , Transcription, Genetic/physiology , Animals , Cell Division , Chromatin/genetics , DNA-Binding Proteins/physiology , Drosophila melanogaster/metabolism , Embryonic and Fetal Development/genetics , Female , Genes, Homeobox , Genes, Insect , Male , Mice , Models, Genetic , Multigene Family , Phenotype , Polycomb Repressive Complex 1 , Polycomb-Group Proteins
20.
Curr Opin Genet Dev ; 9(3): 355-61, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10377289

ABSTRACT

The chromatin structure associated transcriptional memory function by Polycomb-group and trithorax-group protein complexes integrate normal development with control of cellular proliferation. This is illustrated by recent mechanistic insights that link Polycomb repression with histone deacetylases and the identification of new target genes that provide a connection to cell cycle control and tumorigenesis.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins , Insect Proteins/physiology , Leukemia/genetics , Transcription Factors , Animals , Cell Cycle , DNA-Binding Proteins/genetics , Humans , Insect Proteins/genetics , Polycomb Repressive Complex 1
SELECTION OF CITATIONS
SEARCH DETAIL
...