Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Cell Proteomics ; 20: 100132, 2021.
Article in English | MEDLINE | ID: mdl-34389466

ABSTRACT

Structural analysis of host-pathogen protein complexes remains challenging, largely due to their structural heterogeneity. Here, we describe a pipeline for the structural characterization of these complexes using integrative structure modeling based on chemical cross-links and residue-protein contacts inferred from mutagenesis studies. We used this approach on the HIV-1 Vif protein bound to restriction factor APOBEC3G (A3G), the Cullin-5 E3 ring ligase (CRL5), and the cellular transcription factor Core Binding Factor Beta (CBFß) to determine the structure of the (A3G-Vif-CRL5-CBFß) complex. Using the MS-cleavable DSSO cross-linker to obtain a set of 132 cross-links within this reconstituted complex along with the atomic structures of the subunits and mutagenesis data, we computed an integrative structure model of the heptameric A3G-Vif-CRL5-CBFß complex. The structure, which was validated using a series of tests, reveals that A3G is bound to Vif mostly through its N-terminal domain. Moreover, the model ensemble quantifies the dynamic heterogeneity of the A3G C-terminal domain and Cul5 positions. Finally, the model was used to rationalize previous structural, mutagenesis and functional data not used for modeling, including information related to the A3G-bound and unbound structures as well as mapping functional mutations to the A3G-Vif interface. The experimental and computational approach described here is generally applicable to other challenging host-pathogen protein complexes.


Subject(s)
APOBEC-3G Deaminase/chemistry , Core Binding Factor beta Subunit/chemistry , Cullin Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , vif Gene Products, Human Immunodeficiency Virus/chemistry , Mass Spectrometry , Models, Molecular
2.
Structure ; 29(5): 404-412.e4, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33275876

ABSTRACT

ETS family transcription factors of ERG and FLI1 play a key role in oncogenesis of prostate cancer and Ewing sarcoma by binding regulatory DNA sites and interfering with function of other factors. Mithramycin (MTM) is an anti-cancer, DNA binding natural product that functions as a potent antagonist of ERG and FLI1 by an unknown mechanism. We present a series of crystal structures of the DNA binding domain (DBD) of ERG/FLI1 culminating in a structure of a high-order complex of the ERG/FLI1 DBD, transcription factor Runx2, core-binding factor beta (Cbfß), and MTM on a DNA enhancer site, along with supporting DNA binding studies using MTM and its analogues. Taken together, these data provide insight into allosteric mechanisms underlying ERG and FLI1 transactions and their disruption by MTM analogues.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Plicamycin/pharmacology , Proto-Oncogene Protein c-fli-1/chemistry , Allosteric Regulation/drug effects , Antibiotics, Antineoplastic/chemistry , Binding Sites , Core Binding Factor Alpha 1 Subunit/chemistry , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/metabolism , Humans , Molecular Docking Simulation , Plicamycin/chemistry , Protein Binding , Proto-Oncogene Protein c-fli-1/metabolism , Transcriptional Regulator ERG/chemistry , Transcriptional Regulator ERG/metabolism
3.
Nat Struct Mol Biol ; 26(12): 1176-1183, 2019 12.
Article in English | MEDLINE | ID: mdl-31792451

ABSTRACT

HIV-1 virion infectivity factor (Vif) promotes degradation of the antiviral APOBEC3 (A3) proteins through the host ubiquitin-proteasome pathway to enable viral immune evasion. Disrupting Vif-A3 interactions to reinstate the A3-catalyzed suppression of human immunodeficiency virus type 1 (HIV-1) replication is a potential approach for antiviral therapeutics. However, the molecular mechanisms by which Vif recognizes A3 proteins remain elusive. Here we report a cryo-EM structure of the Vif-targeted C-terminal domain of human A3F in complex with HIV-1 Vif and the cellular cofactor core-binding factor beta (CBFß) at 3.9-Å resolution. The structure shows that Vif and CBFß form a platform to recruit A3F, revealing a direct A3F-recruiting role of CBFß beyond Vif stabilization, and captures multiple independent A3F-Vif interfaces. Together with our biochemical and cellular studies, our structural findings establish the molecular determinants that are critical for Vif-mediated neutralization of A3F and provide a comprehensive framework of how HIV-1 Vif hijacks the host protein degradation machinery to counteract viral restriction by A3F.


Subject(s)
Cytosine Deaminase/chemistry , HIV-1/chemistry , vif Gene Products, Human Immunodeficiency Virus/chemistry , Core Binding Factor beta Subunit/chemistry , Cryoelectron Microscopy , Cytosine Deaminase/antagonists & inhibitors , Cytosine Deaminase/ultrastructure , Humans , Immune Evasion , Models, Molecular , Protein Conformation , Protein Domains , Protein Interaction Mapping , Proteolysis , Structure-Activity Relationship , vif Gene Products, Human Immunodeficiency Virus/pharmacology , vif Gene Products, Human Immunodeficiency Virus/ultrastructure
4.
Biophys J ; 116(8): 1432-1445, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30961890

ABSTRACT

Human immunodeficiency virus-1 viral infectivity factor (Vif) is an intrinsically disordered protein responsible for the ubiquitination of the APOBEC3 (A3) antiviral proteins. Vif folds when it binds Cullin-RING E3 ligase 5 and the transcription cofactor CBF-ß. A five-protein complex containing the substrate receptor (Vif, CBF-ß, Elongin-B, Elongin-C (VCBC)) and Cullin5 (CUL5) has a published crystal structure, but dynamics of this VCBC-CUL5 complex have not been characterized. Here, we use molecular dynamics (MD) simulations and NMR to characterize the dynamics of the VCBC complex with and without CUL5 and an A3 protein bound. Our simulations show that the VCBC complex undergoes global dynamics involving twisting and clamshell opening of the complex, whereas VCBC-CUL5 maintains a more static conformation, similar to the crystal structure. This observation from MD is supported by methyl-transverse relaxation-optimized spectroscopy NMR data, which indicates that the VCBC complex without CUL5 is dynamic on the µs-ms timescale. Our NMR data also show that the VCBC complex is more conformationally restricted when bound to the antiviral APOBEC3F (one of the A3 proteins), consistent with our MD simulations. Vif contains a flexible linker region located at the hinge of the VCBC complex, which changes conformation in conjunction with the global dynamics of the complex. Like other substrate receptors, VCBC can exist alone or in complex with CUL5 and other proteins in cells. Accordingly, the VCBC complex could be a good target for therapeutics that would inhibit full assembly of the ubiquitination complex by stabilizing an alternate VCBC conformation.


Subject(s)
Cullin Proteins/chemistry , Cytidine Deaminase/chemistry , Molecular Dynamics Simulation , vif Gene Products, Human Immunodeficiency Virus/chemistry , APOBEC Deaminases , Core Binding Factor beta Subunit/chemistry , Crystallization , Elongin/chemistry , Humans , Kinetics , Protein Binding , Protein Conformation , Protein Folding , Structure-Activity Relationship , Ubiquitination
5.
EBioMedicine ; 8: 117-131, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27428424

ABSTRACT

Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFß binding partner. CBFß enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFß are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFß and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor beta Subunit/metabolism , Neoplasms/metabolism , Allosteric Regulation/drug effects , Antineoplastic Agents/chemistry , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/genetics , Drug Discovery , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Leukemia , Models, Molecular , Molecular Conformation , Mutation , Neoplasms/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Multimerization , Signal Transduction/drug effects , Structure-Activity Relationship
6.
Arch Biochem Biophys ; 567: 1-12, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25575784

ABSTRACT

The CBFß gene encodes a transcription factor that, in combination with CBFα (also called Runx, runt-related transcription factor) regulates expression of several target genes. CBFß interacts with all Runx family members, such as RUNX2, a regulator of bone-related gene transcription that contains a conserved DNA-binding domain. CBFß stimulates DNA binding of the Runt domain, and is essential for most of the known functions of RUNX2. A comparative analysis of the zebrafish cbfß gene and protein, and of its orthologous identified homologous proteins in different species indicates a highly conserved function. We cloned eleven zebrafish cbfß gene transcripts, one resulting in the known Cbfß protein (with 187 aa), and three additional variants resulting from skipping exon 5a (resulting in a protein with 174 aa) or exon 5b (resulting in a protein with 201 aa), both observed for the first time in zebrafish, and a completely novel isoform containing both exon 5a and 5b (resulting in a protein with 188 aa). Functional analysis of these isoforms provides insight into their role in regulating gene transcription. From the other variants two are premature termination Cbfß forms, while the others show in-frame exon-skipping causing changes in the Cbfß domain that may affect its function.


Subject(s)
Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Transcription, Genetic , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Amino Acid Sequence , Animals , Chromosomes/genetics , Cloning, Molecular , Conserved Sequence , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor beta Subunit/biosynthesis , Core Binding Factor beta Subunit/chemistry , Gene Expression Regulation, Developmental , Molecular Sequence Data , Protein Biosynthesis , Protein Isoforms/biosynthesis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/chemistry
7.
J Mol Biol ; 427(8): 1655-69, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25083921

ABSTRACT

Cooperative assemblies of transcription factors (TFs) on target gene enhancers coordinate cell proliferation, fate specification, and differentiation through precise and complicated transcriptional mechanisms. Chemical modifications, such as phosphorylation, of TFs induced by cell signaling further modulate the dynamic cooperativity of TFs. In this study, we found that various Ets1-containing TF-DNA complexes respond differently to calcium-induced phosphorylation of Ets1, which is known to inhibit Ets1-DNA binding. Crystallographic analysis of a complex comprising Ets1, Runx1, and CBFß at the TCRα enhancer revealed that Ets1 acquires robust binding stability in the Runx1 and DNA-complexed state, via allosteric mechanisms. This allows phosphorylated Ets1 to be retained at the TCRα enhancer with Runx1, in contrast to other Ets1 target gene enhancers including mb-1 and stromelysin-1. This study provides a structure-based model for cell-signaling-dependent regulation of target genes, mediated via chemical modification of TFs.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , DNA/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Animals , Base Sequence , Cell Line , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor beta Subunit/chemistry , Crystallography, X-Ray , DNA/chemistry , Humans , Mice , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Proto-Oncogene Protein c-ets-1/chemistry , Transcriptional Activation
9.
Nature ; 505(7482): 229-33, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24402281

ABSTRACT

The human immunodeficiency virus (HIV)-1 protein Vif has a central role in the neutralization of host innate defences by hijacking cellular proteasomal degradation pathways to subvert the antiviral activity of host restriction factors; however, the underlying mechanism by which Vif achieves this remains unclear. Here we report a crystal structure of the Vif-CBF-ß-CUL5-ELOB-ELOC complex. The structure reveals that Vif, by means of two domains, organizes formation of the pentameric complex by interacting with CBF-ß, CUL5 and ELOC. The larger domain (α/ß domain) of Vif binds to the same side of CBF-ß as RUNX1, indicating that Vif and RUNX1 are exclusive for CBF-ß binding. Interactions of the smaller domain (α-domain) of Vif with ELOC and CUL5 are cooperative and mimic those of SOCS2 with the latter two proteins. A unique zinc-finger motif of Vif, which is located between the two Vif domains, makes no contacts with the other proteins but stabilizes the conformation of the α-domain, which may be important for Vif-CUL5 interaction. Together, our data reveal the structural basis for Vif hijacking of the CBF-ß and CUL5 E3 ligase complex, laying a foundation for rational design of novel anti-HIV drugs.


Subject(s)
Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/metabolism , Cullin Proteins/chemistry , Cullin Proteins/metabolism , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Core Binding Factor Alpha 2 Subunit/metabolism , Crystallography, X-Ray , Elongin , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Stability , Protein Structure, Tertiary , Suppressor of Cytokine Signaling Proteins , Transcription Factors/chemistry , Transcription Factors/metabolism
10.
J Virol ; 88(6): 3320-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390335

ABSTRACT

UNLABELLED: The human immunodeficiency virus type 1 (HIV-1)-encoded virion infectivity factor (Vif) is required to inactivate the host restriction factor APOBEC3 by engaging Cullin 5 (Cul5)-RING ubiquitin ligase (CRL5). Core binding factor beta (CBF-ß) is a novel regulator of Vif-CRL5 function; as yet, its mechanism of regulation remains unclear. In the present study, we demonstrate that CBF-ß promotion of Vif-CRL5 assembly is independent of its influence on Vif stability and is also a conserved feature of primate lentiviral Vif proteins. Furthermore, CBF-ß is critical for the formation of the Vif-ElonginB/ElonginC-Cul5 core E3 ubiquitin ligase complex in vitro. CBF-ß from diverse vertebrate species supported HIV-1 Vif function, indicating the conserved nature of Vif-CBF-ß interfaces. Considering the importance of the interaction between Vif and CBF-ß in viral CRL5 function, disrupting this interaction represents an attractive pharmacological intervention against HIV-1. IMPORTANCE: HIV-1 encodes virion infectivity factor (Vif) to inactivate its host's antiviral APOBEC3 proteins. Vif triggers APOBEC3 degradation by forming Vif-Cullin 5 (Cul5)-RING ubiquitin ligase (CRL5). Core binding factor beta (CBF-ß) is a novel regulator of Vif-CRL5 function whose mechanism of regulation remains poorly defined. In the present study, we demonstrate that the promotion of Vif-CRL5 assembly by CBF-ß can be separated from its influence on Vif stability. The promotion of Vif-CRL5 assembly, but not the influence on Vif stability, is conserved among primate lentiviral Vif proteins: we found that CBF-ß from diverse vertebrate species supported HIV-1 Vif function. Considering the importance of the interaction between Vif and CBF-ß in viral CRL5 function and HIV-1 replication, disrupting this interaction is an attractive strategy against HIV-1.


Subject(s)
Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Cullin Proteins/metabolism , Evolution, Molecular , HIV Infections/metabolism , HIV-1/metabolism , Simian Immunodeficiency Virus/metabolism , Ubiquitin-Protein Ligases/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Core Binding Factor beta Subunit/chemistry , Cullin Proteins/genetics , Elongin , Gene Products, vif/chemistry , Gene Products, vif/genetics , Gene Products, vif/metabolism , HIV Infections/enzymology , HIV Infections/genetics , HIV Infections/virology , HIV-1/chemistry , HIV-1/genetics , Humans , Molecular Sequence Data , Protein Binding , Sequence Alignment , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/genetics
11.
Nature ; 481(7381): 376-9, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22190036

ABSTRACT

The human APOBEC3 cytidine deaminases are potent inhibitors of diverse retroviruses, including human immunodeficiency virus-1 (HIV-1). HIV-1 Vif forms an E3 ubiquitin ligase complex with cullin 5 (CUL5), elongin B and elongin C , which promotes the polyubiquitination and degradation of APOBEC3 substrates. Here we demonstrate in human T cells that core binding factor ß (CBF-ß) is a key regulator of the evasion of HIV-1 from the host defence mediated by APOBEC3. CBF-ß, the non-DNA-binding subunit of a heterodimeric transcription factor, regulates the folding and DNA-binding activity of partner RUNX family proteins, which have important roles in the development and differentiation of diverse cell types, including T lymphocytes. In our study, knockdown of endogenous CBF-ß blocked Vif-induced APOBEC3G polyubiquitination and degradation. CBF-ß was not required for the interaction between Vif and APOBEC3G, yet was essential for the assembly of the Vif-CUL5 E3-ubiquitin-ligase complex. CBF-ß proved to be a unique regulator of primate lentiviral Vif and not a general component of the CUL5 E3 ubiquitin ligase. We show that Vif and CBF-ß physically interact, and that the amino-terminal region of Vif is required for this interaction. Furthermore, interactions with Vif required regions in CBF-ß that are not involved in RUNX protein binding. Considering the importance of the interaction between Vif and CBF-ß, disrupting this interaction represents an attractive pharmacological intervention against HIV-1.


Subject(s)
Cell Differentiation , Core Binding Factor beta Subunit/metabolism , HIV-1/physiology , Host-Pathogen Interactions , Immune Evasion , T-Lymphocytes/cytology , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , Cell Line , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/genetics , Cullin Proteins/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Gene Knockdown Techniques , HEK293 Cells , HIV-1/genetics , HIV-1/immunology , Humans , Immunoprecipitation , Models, Molecular , Protein Binding , Proteolysis , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/genetics
12.
J Cell Sci ; 123(Pt 15): 2613-20, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20605922

ABSTRACT

In muscle cells, a complex network of Z-disc proteins allows proper reception, transduction and transmission of mechanical and biochemical signals. Mutations in genes encoding different Z-disc proteins such as integrin-linked kinase (ILK) and nexilin have recently been shown to cause heart failure by distinct mechanisms such as disturbed mechanosensing, altered mechanotransduction or mechanical Z-disc destabilization. We identified core-binding factor ß (CBFß) as an essential component for maintaining sarcomeric Z-disc and myofilament organization in heart and skeletal muscle. In CBFß-deficient cardiomyocytes and skeletal-muscle cells, myofilaments are thinned and Z-discs are misaligned, leading to progressive impairment of heart and skeletal-muscle function. Transcription of the gene encoding CBFß mainly depends on JunB activity. In JunB-morphant zebrafish, which show a heart-failure phenotype similar to that of CBFß-deficient zebrafish, transcript and protein levels of CBFß are severely reduced. Accordingly, ectopic expression of CBFß can reconstitute cardiomyocyte function and rescue heart failure in JunB morphants, demonstrating for the first time an essential role of JunB-CBFß signaling for maintaining sarcomere architecture and function.


Subject(s)
Core Binding Factor beta Subunit/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Sarcomeres/metabolism , Amino Acid Sequence , Animals , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/genetics , Heart Failure/genetics , Heart Failure/metabolism , Humans , Immunoblotting , In Situ Hybridization , Mice , Microscopy, Electron, Transmission , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myocardium/metabolism , Myocardium/ultrastructure , Myocytes, Cardiac/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-jun/genetics , Sarcomeres/diagnostic imaging , Sarcomeres/genetics , Sequence Homology, Amino Acid , Ultrasonography , Zebrafish
13.
Nucleic Acids Res ; 38(18): 6124-34, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20483917

ABSTRACT

Runx1 is a developmentally regulated transcription factor that is essential for haemopoiesis. Runx1 can bind as a monomer to the core consensus sequence TGTGG, but binds more efficiently as a hetero-dimer together with the non-DNA binding protein CBFß as a complex termed core binding factor (CBF). Here, we demonstrated that CBF can also assemble as a dimeric complex on two overlapping Runx1 sites within the palindromic sequence TGTGGCTGCCCACA in the human granulocyte macrophage colony-stimulating factor enhancer. Furthermore, we demonstrated that binding of Runx1 to the enhancer is rigidly controlled at the level of chromatin accessibility, and is dependent upon prior induction of NFAT and AP-1, which disrupt a positioned nucleosome in this region. We employed in vivo footprinting to demonstrate that, upon activation of the enhancer, both sites are efficiently occupied. In vitro binding assays confirmed that two CBF complexes can bind this site simultaneously, and transfection assays demonstrated that both sites contribute significantly to enhancer function. Computer modelling based on the Runx1/CBFß/DNA crystal structure further revealed that two molecules of CBF could potentially bind to this class of palindromic sequence as a dimeric complex in a conformation whereby both Runx1 and CBFß within the two CBF complexes are closely aligned.


Subject(s)
Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor beta Subunit/chemistry , Enhancer Elements, Genetic , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Inverted Repeat Sequences , Animals , Binding Sites , Cell Line , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Dimerization , Humans , Mice , Models, Molecular
14.
J Cell Biochem ; 110(1): 97-103, 2010 May.
Article in English | MEDLINE | ID: mdl-20225274

ABSTRACT

Cleidocranial dysplasia (CCD) is caused by haploinsufficiency in RUNX2 function. We have previously identified a series of RUNX2 mutations in Korean CCD patients, including a novel R131G missense mutation in the Runt-homology domain. Here, we examine the functional consequences of the RUNX2(R131G) mutation, which could potentially affect DNA binding, nuclear localization signal, and/or heterodimerization with core-binding factor-beta (CBF-beta). Immunofluorescence microscopy and western blot analysis with subcellular fractions show that RUNX2(R131G) is localized in the nucleus. Immunoprecipitation analysis reveals that heterodimerization with CBF-beta is retained. However, precipitation assays with biotinylated oligonucleotides and reporter gene assays with RUNX2 responsive promoters together reveal that DNA-binding activity and consequently the transactivation of potential of RUNX2(R131G) is abrogated. We conclude that loss of DNA binding, but not nuclear localization or CBF-beta heterodimerization, causes RUNX2 haploinsufficiency in patients with the RUNX2(R131G) mutation. Retention of specific functions including nuclear localization and binding to CBF-beta of the RUNX2(R131G) mutation may render the mutant protein an effective competitor that interferes with wild-type function.


Subject(s)
Amino Acid Substitution/genetics , Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , DNA/metabolism , Mutation/drug effects , Amino Acid Motifs , Animals , CHO Cells , Cell Nucleus/metabolism , Core Binding Factor Alpha 1 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/chemistry , Cricetinae , Cricetulus , HeLa Cells , Humans , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Protein Transport , Transcriptional Activation/genetics
15.
Cancer Treat Res ; 145: 127-47, 2010.
Article in English | MEDLINE | ID: mdl-20306249

ABSTRACT

The existence of non-random mutations in critical regulators of cell growth and differentiation is a recurring theme in cancer pathogenesis and provides the basis for our modern, molecular approach to the study and treatment of malignant diseases. Nowhere is this more true than in the study of leukemogenesis, where research has converged upon a critical group of genes involved in hematopoietic stem and progenitor cell self-renewal and fate specification. Prominent among these is the heterodimeric transcriptional regulator, RUNX1/CBFbeta. RUNX1 is a site-specific DNA-binding protein whose consensus response element is found in the promoters of many hematopoietically relevant genes. CBFbeta interacts with RUNX1, stabilizing its interaction with DNA to promote the actions of RUNX1/CBFbeta in transcriptional control. Both the RUNX1 and the CBFbeta genes participate in proleukemic chromosomal alterations. Together they contribute to approximately one-third of acute myelogenous leukemia (AML) and one-quarter of acute lymphoblastic leukemia (ALL) cases, making RUNX1 and CBFbeta the most frequently affected genes known in the pathogenesis of acute leukemia. Investigating the mechanisms by which RUNX1, CBFbeta, and their proleukemic fusion proteins influence leukemogenesis has contributed greatly to our understanding of both normal and malignant hematopoiesis. Here we present an overview of the structural features of RUNX1/CBFbeta and their derivatives, their roles in transcriptional control, and their contributions to normal and malignant hematopoiesis.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/genetics , Gene Expression Regulation, Leukemic/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/physiology , Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/physiology , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/pathology , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/genetics , Transcription, Genetic , Translocation, Genetic
16.
Biochim Biophys Acta ; 1789(11-12): 702-8, 2009.
Article in English | MEDLINE | ID: mdl-19735751

ABSTRACT

Transcriptional function of a novel Crlz1 protein was examined by using the CBF site-containing IgJ enhancer, because it was originally cloned due to its ability to bind CBFbeta, a subunit of CBF heterodimer, of which Runx is the other subunit. In a cotransfection experiment, Crlz1 was shown to increase the IgJ enhancer activity due to its CBF sites, as verified by both the absence of Crlz1 effect on the CBF-site mutated IgJ enhancer and the presence of transcriptional synergy between Crlz1 and CBFbeta. Most significantly, the cytoplasmic CBFbeta was shown to be mobilized into the nucleus when it was coexpressed with the nuclear Crlz1. This mobilized nuclear CBFbeta could then heterodimerize with the nuclear Runx to bind to its target DNA site with a high affinity. Furthermore, in our coimmunoprecipitation and chromatin immunoprecipitation experiments, Crlz1 was found to be bound to the resulting CBF heterodimer in a form of ternary complex and to remain in that ternary complex even when CBF bound to its target DNA site such as IgJ enhancer.


Subject(s)
Cell Nucleus/metabolism , Core Binding Factor beta Subunit/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Active Transport, Cell Nucleus , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Immunoglobulin J-Chains/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Confocal , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Binding , Protein Multimerization , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection
18.
Blood ; 113(13): 3070-9, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19179469

ABSTRACT

AML1-ETO and TEL-AML1 are chimeric proteins resulting from the t(8;21)(q22;q22) in acute myeloid leukemia, and the t(12;21)(p13;q22) in pre-B-cell leukemia, respectively. The Runt domain of AML1 in both proteins mediates DNA binding and heterodimerization with the core binding factor beta (CBFbeta) subunit. To determine whether CBFbeta is required for AML1-ETO and TEL-AML1 activity, we introduced amino acid substitutions into the Runt domain that disrupt heterodimerization with CBFbeta but not DNA binding. We show that CBFbeta contributes to AML1-ETO's inhibition of granulocyte differentiation, is essential for its ability to enhance the clonogenic potential of primary mouse bone marrow cells, and is indispensable for its cooperativity with the activated receptor tyrosine kinase TEL-PDGFbetaR in generating acute myeloid leukemia in mice. Similarly, CBFbeta is essential for TEL-AML1's ability to promote self-renewal of B cell precursors in vitro. These studies validate the Runt domain/CBFbeta interaction as a therapeutic target in core binding factor leukemias.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/physiology , Oncogene Proteins, Fusion/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Granulocytes/metabolism , Granulocytes/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological , Models, Molecular , Mutation/physiology , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Protein Binding , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , RUNX1 Translocation Partner 1 Protein , Transfection
19.
Blood ; 112(9): 3777-87, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18695000

ABSTRACT

The heterodimeric transcription factor RUNX1/PEBP2-beta (also known as AML1/CBF-beta) is essential for definitive hematopoiesis. Here, we show that interaction with PEBP2-beta leads to the phosphorylation of RUNX1, which in turn induces p300 phosphorylation. This is mediated by homeodomain interacting kinase 2 (HIPK2), targeting Ser(249), Ser(273), and Thr(276) in RUNX1, in a manner that is also dependent on the RUNX1 PY motif. Importantly, we observed the in vitro disruption of this phosphorylation cascade by multiple leukemogenic genetic defects targeting RUNX1/CBFB. In particular, the oncogenic protein PEBP2-beta-SMMHC prevents RUNX1/p300 phosphorylation by sequestering HIPK2 to mislocalized RUNX1/beta-SMMHC complexes. Therefore, phosphorylation of RUNX1 appears a critical step in its association with and phosphorylation of p300, and its disruption may be a common theme in RUNX1-associated leukemogenesis.


Subject(s)
CCAAT-Binding Factor/metabolism , Carrier Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Leukemia/etiology , Protein Serine-Threonine Kinases/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Base Sequence , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/chemistry , DNA Primers/genetics , Humans , K562 Cells , Leukemia/genetics , Leukemia/metabolism , Mice , Multiprotein Complexes , Mutagenesis, Site-Directed , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , p300-CBP Transcription Factors/chemistry
20.
BMC Evol Biol ; 8: 228, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18681949

ABSTRACT

BACKGROUND: Members of the Runx family of transcriptional regulators, which bind DNA as heterodimers with CBFbeta, are known to play critical roles in embryonic development in many triploblastic animals such as mammals and insects. They are known to regulate basic developmental processes such as cell fate determination and cellular potency in multiple stem-cell types, including the sensory nerve cell progenitors of ganglia in mammals. RESULTS: In this study, we detect and characterize the hitherto unexplored Runx/CBFbeta genes of cnidarians and sponges, two basal animal lineages that are well known for their extensive regenerative capacity. Comparative structural modeling indicates that the Runx-CBFbeta-DNA complex from most cnidarians and sponges is highly similar to that found in humans, with changes in the residues involved in Runx-CBFbeta dimerization in either of the proteins mirrored by compensatory changes in the binding partner. In situ hybridization studies reveal that Nematostella Runx and CBFbeta are expressed predominantly in small isolated foci at the base of the ectoderm of the tentacles in adult animals, possibly representing neurons or their progenitors. CONCLUSION: These results reveal that Runx and CBFbeta likely functioned together to regulate transcription in the common ancestor of all metazoans, and the structure of the Runx-CBFbeta-DNA complex has remained extremely conserved since the human-sponge divergence. The expression data suggest a hypothesis that these genes may have played a role in nerve cell differentiation or maintenance in the common ancestor of cnidarians and bilaterians.


Subject(s)
Cnidaria/genetics , Core Binding Factor alpha Subunits/genetics , Core Binding Factor beta Subunit/genetics , Porifera/genetics , Transcription Factors/genetics , Animals , Cnidaria/classification , Contig Mapping , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/chemistry , Evolution, Molecular , Expressed Sequence Tags , Models, Molecular , Phylogeny , Porifera/classification , Protein Conformation , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...