Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 24(3): 384-399, 2022 03.
Article in English | MEDLINE | ID: mdl-35210568

ABSTRACT

Canonically, EZH2 serves as the catalytic subunit of PRC2, which mediates H3K27me3 deposition and transcriptional repression. Here, we report that in acute leukaemias, EZH2 has additional noncanonical functions by binding cMyc at non-PRC2 targets and uses a hidden transactivation domain (TAD) for (co)activator recruitment and gene activation. Both canonical (EZH2-PRC2) and noncanonical (EZH2-TAD-cMyc-coactivators) activities of EZH2 promote oncogenesis, which explains the slow and ineffective antitumour effect of inhibitors of the catalytic function of EZH2. To suppress the multifaceted activities of EZH2, we used proteolysis-targeting chimera (PROTAC) to develop a degrader, MS177, which achieved effective, on-target depletion of EZH2 and interacting partners (that is, both canonical EZH2-PRC2 and noncanonical EZH2-cMyc complexes). Compared with inhibitors of the enzymatic function of EZH2, MS177 is fast-acting and more potent in suppressing cancer growth. This study reveals noncanonical oncogenic roles of EZH2, reports a PROTAC for targeting the multifaceted tumorigenic functions of EZH2 and presents an attractive strategy for treating EZH2-dependent cancers.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Neoplasms , Carcinogenesis/genetics , Cytoskeletal Proteins/metabolism , E1A-Associated p300 Protein , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Proteolysis , Transcriptional Activation
2.
Nat Genet ; 52(12): 1384-1396, 2020 12.
Article in English | MEDLINE | ID: mdl-33139953

ABSTRACT

Trimethylated histone H3 lysine 27 (H3K27me3) regulates gene repression, cell-fate determination and differentiation. We report that a conserved bromo-adjacent homology (BAH) module of BAHCC1 (BAHCC1BAH) 'recognizes' H3K27me3 specifically and enforces silencing of H3K27me3-demarcated genes in mammalian cells. Biochemical, structural and integrated chromatin immunoprecipitation-sequencing-based analyses demonstrate that direct readout of H3K27me3 by BAHCC1 is achieved through a hydrophobic trimethyl-L-lysine-binding 'cage' formed by BAHCC1BAH, mediating colocalization of BAHCC1 and H3K27me3-marked genes. BAHCC1 is highly expressed in human acute leukemia and interacts with transcriptional corepressors. In leukemia, depletion of BAHCC1, or disruption of the BAHCC1BAH-H3K27me3 interaction, causes derepression of H3K27me3-targeted genes that are involved in tumor suppression and cell differentiation, leading to suppression of oncogenesis. In mice, introduction of a germline mutation at Bahcc1 to disrupt its H3K27me3 engagement causes partial postnatal lethality, supporting a role in development. This study identifies an H3K27me3-directed transduction pathway in mammals that relies on a conserved BAH 'reader'.


Subject(s)
Carcinogenesis/genetics , Histone Code/genetics , Histones/metabolism , Leukemia/genetics , Proteins/genetics , Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Expression Regulation/genetics , Gene Silencing/physiology , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells , Leukemia/pathology , Methylation , Mice , Mice, Transgenic , Neoplasm Transplantation , Protein Processing, Post-Translational/genetics , Transplantation, Heterologous
3.
Blood ; 134(14): 1176-1189, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31383640

ABSTRACT

Dysregulation of polycomb repressive complex 2 (PRC2) promotes oncogenesis partly through its enzymatic function for inducing trimethylation of histone H3 lysine 27 (H3K27me3). However, it remains to be determined how PRC2 activity is regulated in normal and diseased settings. We here report a PRC2-associated cofactor, PHD finger protein 19 (PHF19; also known as polycomb-like 3), as a crucial mediator of tumorigenicity in multiple myeloma (MM). Overexpression and/or genomic amplification of PHF19 is found associated with malignant progression of MM and plasma cell leukemia, correlating to worse treatment outcomes. Using various MM models, we demonstrated a critical requirement of PHF19 for tumor growth in vitro and in vivo. Mechanistically, PHF19-mediated oncogenic effect relies on its PRC2-interacting and chromatin-binding functions. Chromatin immunoprecipitation followed by sequencing profiling showed a critical role for PHF19 in maintaining the H3K27me3 landscape. PHF19 depletion led to loss of broad H3K27me3 domains, possibly due to impaired H3K27me3 spreading from cytosine guanine dinucleotide islands, which is reminiscent to the reported effect of an "onco"-histone mutation, H3K27 to methionine (H3K27M). RNA-sequencing-based transcriptome profiling in MM lines also demonstrated a requirement of PHF19 for optimal silencing of PRC2 targets, which include cell cycle inhibitors and interferon-JAK-STAT signaling genes critically involved in tumor suppression. Correlation studies using patient sample data sets further support a clinical relevance of the PHF19-regulated pathways. Lastly, we show that MM cells are generally sensitive to PRC2 inhibitors. Collectively, this study demonstrates that PHF19 promotes MM tumorigenesis through enhancing H3K27me3 deposition and PRC2's gene-regulatory functions, lending support for PRC2 blockade as a means for MM therapeutics.


Subject(s)
Carcinogenesis/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Multiple Myeloma/metabolism , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Humans , Methylation , Mice , Multiple Myeloma/pathology
4.
Cell Stress ; 3(2): 38-46, 2019 Jan 21.
Article in English | MEDLINE | ID: mdl-31225499

ABSTRACT

Exposure of genomic, single-stranded DNA (ssDNA) during transcription and replication creates opportunities for the formation of inappropriate secondary structures. Cells manage this exposure by using topoisomerases and helicases to reduce the inherent topological stress that arises from unwinding the double helix and by coating ssDNA with protective protein complexes. Interestingly, specific DNA-RNA hybrids, known as R-loops, form during transcription and exist in homeostasis throughout the genomes of prokaryotes and eukaryotes. These hybrids nucleate from guanine rich clusters in the template strand and extend across GC rich spans of transcribed genes. In vivo regulatory functions have evolved from R-loops, including regulation of gene expression and telomere lengthening. However, they also exist as a form of stress, particularly when replication forks collide with the transcription machinery. New methodologies and models are being developed to delineate the biology of R-loops, including those related to cell stress-based diseases like cancer. As accumulation of R-loops is associated with disease, targeting molecular pathways that regulate their formation or removal could provide new avenues for therapeutic intervention. This review covers recent understandings of the molecular basis for R-loop formation, removal, and biological outcomes in the context of cellular stress.

5.
Cell Commun Signal ; 17(1): 24, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30885209

ABSTRACT

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) results in changes that promote de-differentiation, migration, and invasion in non-small cell lung cancer (NSCLC). While it is recognized that EMT promotes altered energy utilization, identification of metabolic pathways that link EMT with cancer progression is needed. Work presented here indicates that mesenchymal NSCLC upregulates glutamine-fructose-6-phosphate transaminase 2 (GFPT2). GFPT2 is the rate-limiting enzyme in the synthesis of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc). UDP-GlcNAc is the obligate activator of O-linked N-acetylglucosamine transferase (OGT). METHODS: Analysis of our transcriptomic data indicates that GFPT2 is one of the most significantly upregulated metabolic genes in mesenchymal NSCLC. Ectopic GFPT2 expression, as well as gene silencing strategies were used to determine the importance of this metabolic enzyme in regulating EMT-driven processes of cell motility and invasion. RESULTS: Our work demonstrates that GFPT2 is transcriptionally upregulated by NF-κB and repressed by the NAD+-dependent deacetylase SIRT6. Depletion of GFPT2 expression in NSCLC highlights its importance in regulating cell migration and invasion during EMT. CONCLUSIONS: Consistent with GFPT2 promoting cancer progression, we find that elevated GFPT2 expression correlates with poor clinical outcome in NSCLC. Modulation of GFPT2 activity offers a potentially important therapeutic target to combat NSCLC disease progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Lung Neoplasms/pathology , NF-kappa B/metabolism , Sirtuins/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung/genetics , Cell Movement , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Lung Neoplasms/genetics , Signal Transduction , Transcriptional Activation
6.
Stem Cell Reports ; 10(3): 675-683, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29456179

ABSTRACT

Self-renewal and differentiation of adult stem cells are tightly regulated partly through configuration of chromatin structure by chromatin remodelers. Using knockout mice, we here demonstrate that bromodomain PHD finger transcription factor (BPTF), a component of the nucleosome remodeling factor (NURF) chromatin-remodeling complex, is essential for maintaining the population size of hematopoietic stem/progenitor cells (HSPCs), including long-term hematopoietic stem cells (HSCs). Bptf-deficient HSCs are defective in reconstituted hematopoiesis, and hematopoietic-specific knockout of Bptf caused profound defects including bone marrow failure and anemia. Genome-wide transcriptome profiling revealed that BPTF loss caused downregulation of HSC-specific gene-expression programs, which contain several master transcription factors (Meis1, Pbx1, Mn1, and Lmo2) required for HSC maintenance and self-renewal. Furthermore, we show that BPTF potentiates the chromatin accessibility of key HSC "stemness" genes. These results demonstrate an essential requirement of the chromatin remodeler BPTF and NURF for activation of "stemness" gene-expression programs and proper function of adult HSCs.


Subject(s)
Adult Stem Cells/physiology , Antigens, Nuclear/genetics , Chromatin/genetics , Hematopoietic Stem Cells/physiology , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Chromatin Assembly and Disassembly/physiology , Down-Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Hematopoiesis/genetics , Mice , Mice, Knockout
7.
Cell Rep ; 12(9): 1400-6, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299963

ABSTRACT

The protein stability and chromatin functions of UHRF1 (ubiquitin-like, containing PHD and RING finger domains, 1) are regulated in a cell-cycle-dependent manner. We report a structural characterization of the complex between UHRF1 and the deubiquitinase USP7. The first two UBL domains of USP7 bind to the polybasic region (PBR) of UHRF1, and this interaction is required for the USP7-mediated deubiquitination of UHRF1. Importantly, we find that the USP7-binding site of the UHRF1 PBR overlaps with the region engaging in an intramolecular interaction with the N-terminal tandem Tudor domain (TTD). We show that the USP7-UHRF1 interaction perturbs the TTD-PBR interaction of UHRF1, thereby shifting the conformation of UHRF1 from a TTD-"occluded" state to a state open for multivalent histone binding. Consistently, introduction of a USP7-interaction-defective mutation to UHRF1 significantly reduces its chromatin association. Together, these results link USP7 interaction to the dynamic deubiquitination and chromatin association of UHRF1.


Subject(s)
Allosteric Site , CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination , Allosteric Regulation , Amino Acid Sequence , CCAAT-Enhancer-Binding Proteins/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Protein Binding , Protein Transport , Ubiquitin Thiolesterase/chemistry , Ubiquitin-Protein Ligases , Ubiquitin-Specific Peptidase 7
8.
Blood ; 125(2): 346-57, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25395428

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) and related EZH1 control gene expression and promote tumorigenesis via methylating histone H3 at lysine 27 (H3K27). These methyltransferases are ideal therapeutic targets due to their frequent hyperactive mutations and overexpression found in cancer, including hematopoietic malignancies. Here, we characterized a set of small molecules that allow pharmacologic manipulation of EZH2 and EZH1, which include UNC1999, a selective inhibitor of both enzymes, and UNC2400, an inactive analog compound useful for assessment of off-target effect. UNC1999 suppresses global H3K27 trimethylation/dimethylation (H3K27me3/2) and inhibits growth of mixed lineage leukemia (MLL)-rearranged leukemia cells. UNC1999-induced transcriptome alterations overlap those following knockdown of embryonic ectoderm development, a common cofactor of EZH2 and EZH1, demonstrating UNC1999's on-target inhibition. Mechanistically, UNC1999 preferentially affects distal regulatory elements such as enhancers, leading to derepression of polycomb targets including Cdkn2a. Gene derepression correlates with a decrease in H3K27me3 and concurrent gain in H3K27 acetylation. UNC2400 does not induce such effects. Oral administration of UNC1999 prolongs survival of a well-defined murine leukemia model bearing MLL-AF9. Collectively, our study provides the detailed profiling for a set of chemicals to manipulate EZH2 and EZH1 and establishes specific enzymatic inhibition of polycomb repressive complex 2 (PRC2)-EZH2 and PRC2-EZH1 by small-molecule compounds as a novel therapeutics for MLL-rearranged leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Biphenotypic, Acute/enzymology , Polycomb Repressive Complex 2/antagonists & inhibitors , Animals , Chromatin Immunoprecipitation , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/pharmacology , Immunoblotting , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
9.
Cancer Res ; 75(2): 426-35, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25432175

ABSTRACT

Soluble growth factors and cytokines within the tumor microenvironment aid in the induction of the epithelial-to-mesenchymal transition (EMT). Although EMT promotes the development of cancer-initiating cells (CIC), cellular mechanisms by which cancer cells maintain mesenchymal phenotypes remain poorly understood. Work presented here indicates that induction of EMT stimulates non-small cell lung cancer (NSCLC) to secrete soluble factors that function in an autocrine fashion. Using gene expression profiling of all annotated and predicted secreted gene products, we find that NF-κB activity is required to upregulate INHBA/Activin, a morphogen in the TGFß superfamily. INHBA is capable of inducing and maintaining mesenchymal phenotypes, including the expression of EMT master-switch regulators and self-renewal factors that sustain CIC phenotypes and promote lung metastasis. Our work demonstrates that INHBA mRNA and protein expression are commonly elevated in primary human NSCLC and provide evidence that INHBA is a critical autocrine factor that maintains mesenchymal properties of CICs to promote metastasis in NSCLC.


Subject(s)
Activins/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , NF-kappa B/metabolism , Neoplastic Stem Cells/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Inhibin-beta Subunits/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mesoderm/metabolism , Mesoderm/pathology , Neoplastic Stem Cells/metabolism , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Spheroids, Cellular , Up-Regulation
10.
Epigenetics Chromatin ; 6(1): 28, 2013 Sep 02.
Article in English | MEDLINE | ID: mdl-24004852

ABSTRACT

BACKGROUND: The epithelial-mesenchymal transition (EMT) is a de-differentiation process required for wound healing and development. In tumors of epithelial origin aberrant induction of EMT contributes to cancer progression and metastasis. Studies have begun to implicate epigenetic reprogramming in EMT; however, the relationship between reprogramming and the coordination of cellular processes is largely unexplored. We have previously developed a system to study EMT in a canonical non-small cell lung cancer (NSCLC) model. In this system we have shown that the induction of EMT results in constitutive NF-κB activity. We hypothesized a role for chromatin remodeling in the sustained deregulation of cellular signaling pathways. RESULTS: We mapped sixteen histone modifications and two variants for epithelial and mesenchymal states. Combinatorial patterns of epigenetic changes were quantified at gene and enhancer loci. We found a distinct chromatin signature among genes in well-established EMT pathways. Strikingly, these genes are only a small minority of those that are differentially expressed. At putative enhancers of genes with the 'EMT-signature' we observed highly coordinated epigenetic activation or repression. Furthermore, enhancers that are activated are bound by a set of transcription factors that is distinct from those that bind repressed enhancers. Upregulated genes with the 'EMT-signature' are upstream regulators of NF-κB, but are also bound by NF-κB at their promoters and enhancers. These results suggest a chromatin-mediated positive feedback as a likely mechanism for sustained NF-κB activation. CONCLUSIONS: There is highly specific epigenetic regulation at genes and enhancers across several pathways critical to EMT. The sites of these changes in chromatin state implicate several inducible transcription factors with critical roles in EMT (NF-κB, AP-1 and MYC) as targets of this reprogramming. Furthermore, we find evidence that suggests that these transcription factors are in chromatin-mediated transcriptional feedback loops that regulate critical EMT genes. In sum, we establish an important link between chromatin remodeling and shifts in cellular reprogramming.

11.
PLoS One ; 8(7): e68597, 2013.
Article in English | MEDLINE | ID: mdl-23935876

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a de-differentiation process that has been implicated in metastasis and the generation of cancer initiating cells (CICs) in solid tumors. To examine EMT in non-small cell lung cancer (NSCLC), we utilized a three dimensional (3D) cell culture system in which cells were co-stimulated with tumor necrosis factor alpha (TNF) and transforming growth factor beta (TGFß). NSCLC spheroid cultures display elevated expression of EMT master-switch transcription factors, TWIST1, SNAI1/Snail1, SNAI2/Slug and ZEB2/Sip1, and are highly invasive. Mesenchymal NSCLC cultures show CIC characteristics, displaying elevated expression of transcription factors KLF4, SOX2, POU5F1/Oct4, MYCN, and KIT. As a result, these putative CIC display a cancer "stem-like" phenotype by forming lung metastases under limiting cell dilution. The pleiotropic transcription factor, NF-κB, has been implicated in EMT and metastasis. Thus, we set out to develop a NSCLC model to further characterize the role of NF-κB activation in the development of CICs. Here, we demonstrate that induction of EMT in 3D cultures results in constitutive NF-κB activity. Furthermore, inhibition of NF-κB resulted in the loss of TWIST1, SNAI2, and ZEB2 induction, and a failure of cells to invade and metastasize. Our work indicates that NF-κB is required for NSCLC metastasis, in part, by transcriptionally upregulating master-switch transcription factors required for EMT.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4 , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , NF-kappa B/genetics , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Phenotype , Spheroids, Cellular , Tumor Cells, Cultured
12.
Proc Natl Acad Sci U S A ; 109(42): 16888-93, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027940

ABSTRACT

The molecular mechanisms linking glucose metabolism with active transcription remain undercharacterized in mammalian cells. Using nuclear factor-κB (NF-κB) as a glucose-responsive transcription factor, we show that cells use the hexosamine biosynthesis pathway and O-linked ß-N-acetylglucosamine (O-GlcNAc) transferase (OGT) to potentiate gene expression in response to tumor necrosis factor (TNF) or etoposide. Chromatin immunoprecipitation assays demonstrate that, upon induction, OGT localizes to NF-κB-regulated promoters to enhance RelA acetylation. Knockdown of OGT abolishes p300-mediated acetylation of RelA on K310, a posttranslational mark required for full NF-κB transcription. Mapping studies reveal T305 as an important residue required for attachment of the O-GlcNAc moiety on RelA. Furthermore, p300 fails to acetylate a full-length RelA(T305A) mutant, linking O-GlcNAc and acetylation events on NF-κB. Reconstitution of RelA null cells with the RelA(T305A) mutant illustrates the importance of this residue for NF-κB-dependent gene expression and cell survival. Our work provides evidence for a unique regulation where attachment of the O-GlcNAc moiety to RelA potentiates p300 acetylation and NF-κB transcription.


Subject(s)
Acetylglucosamine/metabolism , Gene Expression Regulation/physiology , Glucose/metabolism , Metabolic Networks and Pathways/physiology , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Acetylation , Chromatin Immunoprecipitation , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Etoposide/metabolism , HEK293 Cells , Hexosamines/biosynthesis , Humans , Immunoblotting , Immunoprecipitation , Luciferases , Metabolic Networks and Pathways/genetics , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Mol Cell ; 37(4): 453-4, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20188663

ABSTRACT

Although IKK-related kinases are known to augment immune pathways, their importance to DNA-damage response has not been previously elucidated; in this issue of Molecular Cell, Renner et al. (2010) show that genotoxic stress requires SUMOylated IKKvarepsilon to regulate NF-kappaB transcription and cell survival.


Subject(s)
Apoptosis , DNA Damage , I-kappa B Kinase/metabolism , Signal Transduction , I-kappa B Kinase/genetics , Protein Binding , SUMO-1 Protein/metabolism
14.
J Cell Biol ; 163(3): 535-45, 2003 Nov 10.
Article in English | MEDLINE | ID: mdl-14610056

ABSTRACT

The mechanisms by which catenins regulate cadherin function are not fully understood, and the precise function of p120 catenin (p120ctn) has remained particularly elusive. In microvascular endothelial cells, p120ctn colocalized extensively with cell surface VE-cadherin, but failed to colocalize with VE-cadherin that had entered intracellular degradative compartments. To test the possibility that p120ctn binding to VE-cadherin regulates VE-cadherin internalization, a series of approaches were undertaken to manipulate p120ctn availability to endogenous VE-cadherin. Expression of VE-cadherin mutants that competed for p120ctn binding triggered the degradation of endogenous VE-cadherin. Similarly, reducing levels of p120ctn using siRNA caused a dramatic and dose-related reduction in cellular levels of VE-cadherin. In contrast, overexpression of p120ctn increased VE-cadherin cell surface levels and inhibited entry of cell surface VE-cadherin into degradative compartments. These results demonstrate that cellular levels of p120ctn function as a set point mechanism that regulates cadherin expression levels, and that a major function of p120ctn is to control cadherin internalization and degradation.


Subject(s)
Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion/genetics , Endothelium, Vascular/metabolism , Phosphoproteins/metabolism , Protein Transport/genetics , Antigens, CD , Cadherins/genetics , Catenins , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Line , Endocytosis/genetics , Feedback, Physiological/genetics , Humans , Mutation/genetics , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Protein Binding/drug effects , Protein Binding/genetics , RNA, Small Interfering/pharmacology , Delta Catenin
15.
J Biol Chem ; 278(21): 19199-208, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12626512

ABSTRACT

VE-cadherin is an endothelial-specific cadherin that plays important roles in vascular morphogenesis and growth control. To investigate the mechanisms by which endothelial cells regulate cadherin cell surface levels, a VE-cadherin mutant containing the non-adhesive interleukin-2 (IL-2) receptor extracellular domain and the VE-cadherin cytoplasmic tail (IL-2R-VE-cadcyto) was expressed in microvascular endothelial cells. Expression of the IL-2R-VE-cadcyto mutant resulted in the internalization of endogenous VE-cadherin and in a dramatic decrease in endogenous VE-cadherin levels. The internalized VE-cadherin co-localized with early endosomes, and the lysosomal inhibitor chloroquine dramatically inhibited the down-regulation of VE-cadherin in cells expressing the IL-2R-VE-cadcyto mutant. Chloroquine treatment also resulted in the accumulation of a VE-cadherin fragment lacking the beta-catenin binding domain of the VE-cadherin cytoplasmic tail. The formation of the VE-cadherin fragment could be prevented by treating endothelial cells with proteasome inhibitors. Furthermore, inhibition of the proteasome prevented VE-cadherin internalization and inhibited the disruption of endothelial intercellular junctions by the IL-2RVE-cadcyto mutant. These results provide new insights into the mechanisms of VE-cadherin processing and degradation in microvascular endothelial cells.


Subject(s)
Cadherins/metabolism , Endothelium, Vascular/metabolism , Adenoviridae/genetics , Animals , Antigens, CD , Blotting, Western , COS Cells , Cadherins/genetics , Cell Line , Cells, Cultured , Chloroquine/pharmacology , Cysteine Endopeptidases , Endosomes/metabolism , Fluorescent Antibody Technique , Gene Deletion , Gene Expression , Genetic Vectors , Humans , Intercellular Junctions/drug effects , Kidney , Lysosomes/drug effects , Lysosomes/metabolism , Male , Microcirculation , Multienzyme Complexes/antagonists & inhibitors , Mutagenesis , Proteasome Endopeptidase Complex , Receptors, Interleukin-2/genetics , Recombinant Fusion Proteins , Recombinant Proteins , Skin/blood supply , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...