Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Cell Biochem Suppl ; Suppl 36: 129-43, 2001.
Article in English | MEDLINE | ID: mdl-11455578

ABSTRACT

Polycomb group proteins are involved in the maintenance of cellular identity. As multimeric complexes they repress cell type-specific sets of target genes. One model predicts that the composition of Polycomb group complexes determines the specificity for their target genes. To study this hypothesis, we analyzed the expression of Polycomb group genes in various human tissues using Northern blotting and immunohistochemistry. We found that Polycomb group expression varies greatly among tissues and even among specific cell types within a particular tissue. Variations in mRNA expression ranged from expression of all analyzed Polycomb group genes in the heart and testis to no detectable Polycomb group expression at all in bone marrow. Furthermore, each Polycomb group gene was expressed in a different number of tissues. RING1 was expressed in practically all tissues, while HPH1 was expressed in only a few tissues. Also within one tissue the level of Polycomb group expression varied greatly. Cell type-specific Polycomb group expression patterns were observed in thyroid, pancreas, and kidney. Finally, in various developmental stages of fetal kidney, different Polycomb group expression patterns were observed. We conclude that Polycomb group expression can vary depending on the tissue, cell type, and development stage. Polycomb group complexes can only be composed of the Polycomb group proteins that are expressed. This implies that with cell type-specific Polycomb group expression patterns, cell type-specific Polycomb group complexes exist. The fact that there are cell type-specific Polycomb group targets and cell type-specific Polycomb group complexes fits well with the hypothesis that the composition of Polycomb group complexes may determine their target specificity. J. Cell. Biochem. Suppl. 36: 129-143, 2001.


Subject(s)
Repressor Proteins/metabolism , Blotting, Northern , Fetus , Humans , Immunohistochemistry , Organ Specificity , Polycomb-Group Proteins , RNA, Messenger/metabolism , Repressor Proteins/genetics , Transcription, Genetic
2.
Blood ; 97(12): 3896-901, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11389032

ABSTRACT

Polycomb-group (PcG) proteins, such as BMI-1 and EZH2, form multimeric gene-repressing complexes involved in axial patterning, hematopoiesis, and cell cycle regulation. In addition, BMI-1 is involved in experimental lymphomagenesis. Little is known about its role in human lymphomagenesis. Here, BMI-1 and EZH2 expression patterns are analyzed in a variety of B-cell non-Hodgkin lymphomas (B-NHLs), including small lymphocytic lymphoma, follicular lymphoma, large B-cell lymphoma, mantle-cell lymphoma, and Burkitt lymphoma. In contrast to the mutually exclusive pattern of BMI-1 and EZH2 in reactive follicles, the neoplastic cells in B-NHLs of intermediate- and high-grade malignancy showed strong coexpression of BMI-1 and EZH2. This pattern overlapped with the expression of Mib-1/Ki-67, a marker for proliferation. Neoplastic cells in B-NHL of low-grade malignancy were either BMI-1(low)/EZH2(+) (neoplastic centroblasts) or BMI-1(low)EZH2(-) (neoplastic centrocytes). These observations show that low-, intermediate-, and high grade B-NHLs are associated with increased coexpression of the BMI-1 and EZH2 PcG proteins, whose normal expression pattern is mutually exclusive. This expression pattern is probably caused by a failure to down-regulate BMI-1 in dividing neoplastic cells, because BMI-1 expression is absent from normal dividing B cells. These observations are in agreement with findings in studies of Bmi-1 transgenic mice. The extent of BMI-1/EZH2 coexpression correlated with clinical grade and the presence of Mib-1/Ki-67 expression, suggesting that the irregular expression of BMI-1 and EZH2 is an early event in the formation of B-NHL. This points to a role for abnormal PcG expression in human lymphomagenesis. (Blood. 2001;97:3896-3901)


Subject(s)
Drosophila Proteins , Lymphoma, B-Cell/chemistry , Lymphoma, B-Cell/etiology , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Repressor Proteins/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, Nuclear , Biomarkers, Tumor/metabolism , Cell Cycle/physiology , Cell Transformation, Neoplastic/metabolism , Child , Disease Progression , Frozen Sections , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Lymph Nodes/pathology , Lymphoma, B-Cell/pathology , Middle Aged , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2
3.
J Immunol ; 166(10): 5925-34, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342607

ABSTRACT

BMI-1 and EZH2 Polycomb-group (PcG) proteins belong to two distinct protein complexes involved in the regulation of hematopoiesis. Using unique PcG-specific antisera and triple immunofluorescence, we found that mature resting peripheral T cells expressed BMI-1, whereas dividing blasts were EZH2(+). By contrast, subcapsular immature double-negative (DN) (CD4(-)/CD8(-)) T cells in the thymus coexpressed BMI-1 and EZH2 or were BMI-1 single positive. Their descendants, double-positive (DP; CD4(+)/CD8(+)) cortical thymocytes, expressed EZH2 without BMI-1. Most EZH2(+) DN and DP thymocytes were dividing, while DN BMI-1(+)/EZH2(-) thymocytes were resting and proliferation was occasionally noted in DN BMI-1(+)/EZH2(+) cells. Maturation of DP cortical thymocytes to single-positive (CD4(+)/CD8(-) or CD8(+)/CD4(-)) medullar thymocytes correlated with decreased detectability of EZH2 and continued relative absence of BMI-1. Our data show that BMI-1 and EZH2 expression in mature peripheral T cells is mutually exclusive and linked to proliferation status, and that this pattern is not yet established in thymocytes of the cortex and medulla. T cell stage-specific PcG expression profiles suggest that PcG genes contribute to regulation of T cell differentiation. They probably reflect stabilization of cell type-specific gene expression and irreversibility of lineage choice. The difference in PcG expression between medullar thymocytes and mature interfollicular T cells indicates that additional maturation processes occur after thymocyte transportation from the thymus.


Subject(s)
Drosophila Proteins , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation/immunology , Humans , Immunophenotyping , Lymph Nodes/cytology , Lymph Nodes/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/physiology , T-Lymphocyte Subsets/chemistry , Thymus Gland/chemistry
4.
Mol Cell Biol ; 21(4): 1360-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158321

ABSTRACT

Polycomb group (PcG) proteins form multimeric protein complexes which are involved in the heritable stable repression of genes. Previously, we identified two distinct human PcG protein complexes. The EED-EZH protein complex contains the EED and EZH2 PcG proteins, and the HPC-HPH PcG complex contains the HPC, HPH, BMI1, and RING1 PcG proteins. Here we show that YY1, a homolog of the Drosophila PcG protein pleiohomeotic (Pho), interacts specificially with the human PcG protein EED but not with proteins of the HPC-HPH PcG complex. Since YY1 and Pho are DNA-binding proteins, the interaction between YY1 and EED provides a direct link between the chromatin-associated EED-EZH PcG complex and the DNA of target genes. To study the functional significance of the interaction, we expressed the Xenopus homologs of EED and YY1 in Xenopus embryos. Both Xeed and XYY1 induce an ectopic neural axis but do not induce mesodermal tissues. In contrast, members of the HPC-HPH PcG complex do not induce neural tissue. The exclusive, direct neuralizing activity of both the Xeed and XYY1 proteins underlines the significance of the interaction between the two proteins. Our data also indicate a role for chromatin-associated proteins, such as PcG proteins, in Xenopus neural induction.


Subject(s)
DNA-Binding Proteins/physiology , Nerve Tissue/embryology , Repressor Proteins/physiology , Transcription Factors/physiology , Xenopus Proteins , Xenopus/embryology , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , DNA-Binding Proteins/genetics , Drosophila/genetics , Drosophila Proteins , Erythroid-Specific DNA-Binding Factors , Humans , Molecular Sequence Data , Neural Tube Defects/embryology , Neural Tube Defects/genetics , Phenotype , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Sequence Homology, Amino Acid , Transcription Factors/genetics , Two-Hybrid System Techniques , Xenopus/genetics , YY1 Transcription Factor
5.
Am J Pathol ; 157(3): 709-15, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980109

ABSTRACT

The human BMI-1 and EZH2 polycomb group (PcG) proteins are constituents of two distinct complexes of PcG proteins with gene regulatory activity. PcG proteins ensure correct embryonic development by suppressing homeobox genes, and they also contribute to regulation of lymphopoiesis. The two PcG complexes are thought to regulate different target genes and probably have different tissue distributions. Altered expression of PcG genes is linked to transformation in cell lines and induction of tumors in mutant mice, but the role of PcG genes in human cancers is relatively unexplored. Using antisera specific for human PcG proteins, we used immunohistochemistry and immunofluorescence to detect BMI-1 and EZH2 PcG proteins in Reed-Sternberg cells of Hodgkin's disease (HRS). The expression patterns were compared to those in follicular lymphocytes of the lymph node, the normal counterparts of HRS cells. In the germinal center, expression of BMI-1 is restricted to resting Mib-1/Ki-67(-) centrocytes, whereas EZH2 expression is associated with dividing Mib-1/Ki-67(+) centroblasts. By contrast, HRS cells coexpress BMI-1, EZH2, and Mib-1/Ki-67. Because HRS cells are thought to originate from germinal center lymphocytes, these observations suggests that Hodgkin's disease is associated with coexpression of BMI-1 and EZH2 in HRS cells.


Subject(s)
Drosophila Proteins , Hodgkin Disease/metabolism , Lymph Nodes/metabolism , Nuclear Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Reed-Sternberg Cells/metabolism , Repressor Proteins/biosynthesis , Adolescent , Adult , Aged , Female , Gene Expression , Germinal Center/metabolism , Germinal Center/pathology , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Humans , Immunoenzyme Techniques , Lymph Nodes/pathology , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Middle Aged , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins/genetics , Reed-Sternberg Cells/pathology , Repressor Proteins/genetics
6.
J Immunol ; 164(1): 1-4, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10604983

ABSTRACT

Polycomb group (Pc-G) proteins regulate homeotic gene expression in Drosophila, mouse, and humans. Mouse Pc-G proteins are also essential for adult hematopoietic development and contribute to cell cycle regulation. We show that human Pc-G expression patterns correlate with different B cell differentiation stages and that they reflect germinal center (GC) architecture. The transition of resting mantle B cells to rapidly dividing Mib-1(Ki-67)+ follicular centroblasts coincides with loss of BMI-1 and RING1 Pc-G protein detection and appearance of ENX and EED Pc-G protein expression. By contrast, differentiation of centroblasts into centrocytes correlates with reappearance of BMI-1/RING1 and loss of ENX/EED and Mib-1 expression. The mutually exclusive expression of ENX/EED and BMI-1/RING1 reflects the differential composition of two distinct Pc-G complexes. The Pc-G expression profiles in various GC B cell differentiation stages suggest a role for Pc-G proteins in GC development.


Subject(s)
B-Lymphocyte Subsets/metabolism , Gene Expression Regulation, Developmental/immunology , Genes, Homeobox/immunology , Germinal Center/metabolism , Repressor Proteins/genetics , B-Lymphocyte Subsets/cytology , Cell Differentiation/immunology , DNA-Binding Proteins/biosynthesis , Germinal Center/cytology , Humans , Nuclear Proteins/biosynthesis , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Proto-Oncogene Proteins/biosynthesis , Repressor Proteins/biosynthesis
7.
Mol Cell Biol ; 18(6): 3586-95, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9584199

ABSTRACT

In Drosophila melanogaster, the Polycomb-group (PcG) and trithorax-group (trxG) genes have been identified as repressors and activators, respectively, of gene expression. Both groups of genes are required for the stable transmission of gene expression patterns to progeny cells throughout development. Several lines of evidence suggest a functional interaction between the PcG and trxG proteins. For example, genetic evidence indicates that the enhancer of zeste [E(z)] gene can be considered both a PcG and a trxG gene. To better understand the molecular interactions in which the E(z) protein is involved, we performed a two-hybrid screen with Enx1/EZH2, a mammalian homolog of E(z), as the target. We report the identification of the human EED protein, which interacts with Enx1/EZH2. EED is the human homolog of eed, a murine PcG gene which has extensive homology with the Drosophila PcG gene extra sex combs (esc). Enx1/EZH2 and EED coimmunoprecipitate, indicating that they also interact in vivo. However, Enx1/EZH2 and EED do not coimmunoprecipitate with other human PcG proteins, such as HPC2 and BMI1. Furthermore, unlike HPC2 and BMI1, which colocalize in nuclear domains of U-2 OS osteosarcoma cells, Enx1/EZH2 and EED do not colocalize with HPC2 or BMI1. Our findings indicate that Enx1/EZH2 and EED are members of a class of PcG proteins that is distinct from previously described human PcG proteins.


Subject(s)
Apoptosis , Drosophila Proteins , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Gene Library , Humans , Ligases , Macromolecular Substances , Molecular Sequence Data , Peptide Mapping , Point Mutation , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/genetics , Species Specificity , Transcription Factors/genetics , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
8.
Mol Cell Biol ; 17(10): 6076-86, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9315667

ABSTRACT

Polycomb (Pc) is involved in the stable and heritable repression of homeotic gene activity during Drosophila development. Here, we report the identification of a novel human Pc homolog, hPc2. This gene is more closely related to a Xenopus Pc homolog, XPc, than to a previously described human Pc homolog, CBX2 (hPc1). However, the hPc2 and CBX2/hPc1 proteins colocalize in interphase nuclei of human U-2 OS osteosarcoma cells, suggesting that the proteins are part of a common protein complex. To study the functions of the novel human Pc homolog, we generated a mutant protein, delta hPc2, which lacks an evolutionarily conserved C-terminal domain. This C-terminal domain is important for hPc2 function, since the delta hPc2 mutant protein which lacks the C-terminal domain is unable to repress gene activity. Expression of the delta hPc2 protein, but not of the wild-type hPc2 protein, results in cellular transformation of mammalian cell lines as judged by phenotypic changes, altered marker gene expression, and anchorage-independent growth. Specifically in delta hPc2-transformed cells, the expression of the c-myc proto-oncogene is strongly enhanced and serum deprivation results in apoptosis. In contrast, overexpression of the wild-type hPc2 protein results in decreased c-myc expression. Our data suggest that hPc2 is a repressor of proto-oncogene activity and that interference with hPc2 function can lead to derepression of proto-oncogene transcription and subsequently to cellular transformation.


Subject(s)
Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Repressor Proteins/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Nucleus/chemistry , Cloning, Molecular , Genes, myc/genetics , Humans , Ligases , Mammary Neoplasms, Experimental , Mice , Molecular Sequence Data , Organ Specificity , Osteosarcoma/chemistry , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Proto-Oncogene Mas , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Rats , Repressor Proteins/analysis , Repressor Proteins/genetics , Sequence Analysis, DNA , Sequence Deletion , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
9.
Mol Cell Biol ; 17(7): 4105-13, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9199346

ABSTRACT

The Polycomb (Pc) protein is a component of a multimeric, chromatin-associated Polycomb group (PcG) protein complex, which is involved in stable repression of gene activity. The identities of components of the PcG protein complex are largely unknown. In a two-hybrid screen with a vertebrate Pc homolog as a target, we identify the human RING1 protein as interacting with Pc. RING1 is a protein that contains the RING finger motif, a specific zinc-binding domain, which is found in many regulatory proteins. So far, the function of the RING1 protein has remained enigmatic. Here, we show that RING1 coimmunoprecipitates with a human Pc homolog, the vertebrate PcG protein BMI1, and HPH1, a human homolog of the PcG protein Polyhomeotic (Ph). Also, RING1 colocalizes with these vertebrate PcG proteins in nuclear domains of SW480 human colorectal adenocarcinoma and Saos-2 human osteosarcoma cells. Finally, we show that RING1, like Pc, is able to repress gene activity when targeted to a reporter gene. Our findings indicate that RING1 is associated with the human PcG protein complex and that RING1, like PcG proteins, can act as a transcriptional repressor.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins , Insect Proteins/metabolism , Repressor Proteins/physiology , Amino Acid Sequence , Cell Compartmentation , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Humans , Immunologic Techniques , Kinetochores/ultrastructure , Molecular Sequence Data , Nuclear Proteins/metabolism , Nucleoproteins/metabolism , Polycomb Repressive Complex 1 , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic
10.
Mol Cell Biol ; 17(4): 2326-35, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9121482

ABSTRACT

In Drosophila melanogaster, the Polycomb-group (PcG) genes have been identified as repressors of gene expression. They are part of a cellular memory system that is responsible for the stable transmission of gene activity to progeny cells. PcG proteins form a large multimeric, chromatin-associated protein complex, but the identity of its components is largely unknown. Here, we identify two human proteins, HPH1 and HPH2, that are associated with the vertebrate PcG protein BMI1. HPH1 and HPH2 coimmunoprecipitate and cofractionate with each other and with BMI1. They also colocalize with BMI1 in interphase nuclei of U-2 OS human osteosarcoma and SW480 human colorectal adenocarcinoma cells. HPH1 and HPH2 have little sequence homology with each other, except in two highly conserved domains, designated homology domains I and II. They share these homology domains I and II with the Drosophila PcG protein Polyhomeotic (Ph), and we, therefore, have named the novel proteins HPH1 and HPH2. HPH1, HPH2, and BMI1 show distinct, although overlapping expression patterns in different tissues and cell lines. Two-hybrid analysis shows that homology domain II of HPH1 interacts with both homology domains I and II of HPH2. In contrast, homology domain I of HPH1 interacts only with homology domain II of HPH2, but not with homology domain I of HPH2. Furthermore, BMI1 does not interact with the individual homology domains. Instead, both intact homology domains I and II need to be present for interactions with BMI1. These data demonstrate the involvement of homology domains I and II in protein-protein interactions and indicate that HPH1 and HPH2 are able to heterodimerize.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Insect Proteins/metabolism , Nuclear Proteins/metabolism , Nucleoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Amino Acid Sequence , Animals , Binding Sites , Cell Nucleus/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Male , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nucleoproteins/chemistry , Nucleoproteins/genetics , Polycomb Repressive Complex 1 , Protein Conformation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured
11.
Mech Dev ; 53(1): 35-46, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8555110

ABSTRACT

The Polycomb group genes in Drosophila are involved in the stable and inheritable repression of gene expression. The Polycomb group proteins probably operate as multimeric complexes that bind to chromatin. To investigate molecular mechanisms of stable repression of gene activity in vertebrates we have begun to study Xenopus homologs of Polycomb group genes. We identified the Xenopus homologs of the Drosophila Polycomb gene and the bmi-1 gene. bmi-1 is a proto-oncogene which has sequence homology with the Polycomb group gene Posterior Sex Combs. We show that the XPolycomb and Xbmi-1 genes are expressed in overlapping patterns in the central nervous system of Xenopus embryos. However, XPolycomb is also expressed in the somites, whereas Xbmi-1 is not. We further demonstrate that the XPolycomb and Xbmi-1 proteins are able to interact with each other via conserved sequence motifs. These data suggest that also vertebrate Polycomb group proteins form multimeric complexes.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Proto-Oncogenes , Repressor Proteins/physiology , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Base Sequence , Embryo, Nonmammalian/physiology , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Xenopus laevis/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...