Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Proc Natl Acad Sci U S A ; 117(28): 16292-16301, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601208

ABSTRACT

Notch pathway signaling is implicated in several human cancers. Aberrant activation and mutations of Notch signaling components are linked to tumor initiation, maintenance, and resistance to cancer therapy. Several strategies, such as monoclonal antibodies against Notch ligands and receptors, as well as small-molecule γ-secretase inhibitors (GSIs), have been developed to interfere with Notch receptor activation at proximal points in the pathway. However, the use of drug-like small molecules to target the downstream mediators of Notch signaling, the Notch transcription activation complex, remains largely unexplored. Here, we report the discovery of an orally active small-molecule inhibitor (termed CB-103) of the Notch transcription activation complex. We show that CB-103 inhibits Notch signaling in primary human T cell acute lymphoblastic leukemia and other Notch-dependent human tumor cell lines, and concomitantly induces cell cycle arrest and apoptosis, thereby impairing proliferation, including in GSI-resistant human tumor cell lines with chromosomal translocations and rearrangements in Notch genes. CB-103 produces Notch loss-of-function phenotypes in flies and mice and inhibits the growth of human breast cancer and leukemia xenografts, notably without causing the dose-limiting intestinal toxicity associated with other Notch inhibitors. Thus, we describe a pharmacological strategy that interferes with Notch signaling by disrupting the Notch transcription complex and shows therapeutic potential for treating Notch-driven cancers.


Subject(s)
Receptors, Notch/metabolism , Small Molecule Libraries/pharmacology , Transcriptional Activation/drug effects , Animals , Apoptosis/drug effects , Binding Sites , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drosophila , Drug Resistance, Neoplasm/drug effects , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Mice , Mutation , Phenotype , Protein Multimerization , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
2.
Cell Rep ; 26(4): 845-854.e6, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30673607

ABSTRACT

Notch is a conserved signaling pathway that is essential for metazoan development and homeostasis; dysregulated signaling underlies the pathophysiology of numerous human diseases. Receptor-ligand interactions result in gene expression changes, which are regulated by the transcription factor RBPJ. RBPJ forms a complex with the intracellular domain of the Notch receptor and the coactivator Mastermind to activate transcription, but it can also function as a repressor by interacting with corepressor proteins. Here, we determine the structure of RBPJ bound to the corepressor SHARP and DNA, revealing its mode of binding to RBPJ. We tested structure-based mutants in biophysical and biochemical-cellular assays to characterize the role of RBPJ as a repressor, clearly demonstrating that RBPJ mutants deficient for SHARP binding are incapable of repressing transcription of genes responsive to Notch signaling in cells. Altogether, our structure-function studies provide significant insights into the repressor function of RBPJ.


Subject(s)
DNA-Binding Proteins , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Multiprotein Complexes , RNA-Binding Proteins , Signal Transduction , Transcription, Genetic , Animals , Binding Sites , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Structure, Quaternary , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Notch/chemistry , Receptors, Notch/genetics , Receptors, Notch/metabolism
3.
Adv Exp Med Biol ; 1066: 279-295, 2018.
Article in English | MEDLINE | ID: mdl-30030832

ABSTRACT

The highly conserved Notch signal transduction pathway orchestrates fundamental cellular processes including, differentiation, proliferation, and apoptosis during embryonic development and in the adult organism. Dysregulated Notch signaling underlies the etiology of a variety of human diseases, such as certain types of cancers, developmental disorders and cardiovascular disease. Ligand binding induces proteolytic cleavage of the Notch receptor and nuclear translocation of the Notch intracellular domain (NICD), which forms a ternary complex with the transcription factor CSL and the coactivator MAML to upregulate transcription of Notch target genes. The DNA-binding protein CSL is the centrepiece of transcriptional regulation in the Notch pathway, acting as a molecular hub for interactions with either corepressors or coactivators to repress or activate, respectively, transcription. Here we review previous structure-function studies of CSL-associated coregulator complexes and discuss the molecular insights gleaned from this research. We discuss the functional consequences of both activating and repressing binding partners using the same interaction platforms on CSL. We also emphasize that although there has been a significant uptick in structural information over the past decade, it is still under debate how the molecular switch from repression to activation mediated by CSL occurs at Notch target genes and whether it will be possible to manipulate these transcription complexes therapeutically in the future.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Multiprotein Complexes/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription, Genetic/physiology , Animals , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Protein Domains , Receptors, Notch/chemistry , Receptors, Notch/genetics , Structure-Activity Relationship
4.
J Biol Chem ; 292(25): 10549-10563, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28487372

ABSTRACT

The Notch pathway is a cell-to-cell signaling mechanism that is essential for tissue development and maintenance, and aberrant Notch signaling has been implicated in various cancers, congenital defects, and cardiovascular diseases. Notch signaling activates the expression of target genes, which are regulated by the transcription factor CSL (CBF1/RBP-J, Su(H), Lag-1). CSL interacts with both transcriptional corepressor and coactivator proteins, functioning as both a repressor and activator, respectively. Although Notch activation complexes are relatively well understood at the structural level, less is known about how CSL interacts with corepressors. Recently, a new RBP-J (mammalian CSL ortholog)-interacting protein termed RITA has been identified and shown to export RBP-J out of the nucleus, thereby leading to the down-regulation of Notch target gene expression. However, the molecular details of RBP-J/RITA interactions are unclear. Here, using a combination of biochemical/cellular, structural, and biophysical techniques, we demonstrate that endogenous RBP-J and RITA proteins interact in cells, map the binding regions necessary for RBP-J·RITA complex formation, and determine the X-ray structure of the RBP-J·RITA complex bound to DNA. To validate the structure and glean more insights into function, we tested structure-based RBP-J and RITA mutants with biochemical/cellular assays and isothermal titration calorimetry. Whereas our structural and biophysical studies demonstrate that RITA binds RBP-J similarly to the RAM (RBP-J-associated molecule) domain of Notch, our biochemical and cellular assays suggest that RITA interacts with additional regions in RBP-J. Taken together, these results provide molecular insights into the mechanism of RITA-mediated regulation of Notch signaling, contributing to our understanding of how CSL functions as a transcriptional repressor of Notch target genes.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , A549 Cells , Animals , Crystallography, X-Ray , DNA , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Neoplasm Proteins/genetics , Protein Binding , Protein Domains , Structure-Activity Relationship
5.
Cell Chem Biol ; 24(3): 346-359, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28163017

ABSTRACT

Pharmacological modulation of transcription factors (TFs) has only met little success over the past four decades. This is mostly due to standard drug discovery approaches centered on blocking protein/DNA binding or interfering with post-translational modifications. Recent advances in the field of TF biology have revealed a central role of protein-protein interaction in their mode of action. In an attempt to modulate the activity of SOX18 TF, a known regulator of vascular growth in development and disease, we screened a marine extract library for potential small-molecule inhibitors. We identified two compounds, which inspired a series of synthetic SOX18 inhibitors, able to interfere with the SOX18 HMG DNA-binding domain, and to disrupt HMG-dependent protein-protein interaction with RBPJ. These compounds also perturbed SOX18 transcriptional activity in a cell-based reporter gene system. This approach may prove useful in developing a new class of anti-angiogenic compounds based on the inhibition of TF activity.


Subject(s)
SOXF Transcription Factors/antagonists & inhibitors , Small Molecule Libraries/chemistry , Animals , Binding Sites , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , COS Cells , Chlorocebus aethiops , DNA/chemistry , DNA/metabolism , Drug Design , Genes, Reporter , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Inhibitory Concentration 50 , Mice , Nucleic Acid Conformation , Protein Binding , Protein Interaction Maps , Protein Structure, Tertiary , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Salicylic Acid/chemistry , Salicylic Acid/metabolism , Salicylic Acid/pharmacology , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Transcriptional Activation/drug effects
6.
J Mol Biol ; 427(22): 3587-3597, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26344835

ABSTRACT

Formation of the bivalent interaction between the Notch intracellular domain (NICD) and the transcription factor CBF-1/RBP-j, Su(H), Lag-1 (CSL) is a key event in Notch signaling because it switches Notch-responsive genes from a repressed state to an activated state. Interaction of the intrinsically disordered RBP-j-associated molecule (RAM) region of NICD with CSL is thought to both disrupt binding of corepressor proteins to CSL and anchor NICD to CSL, promoting interaction of the ankyrin domain of NICD with CSL through an effective concentration mechanism. To quantify the role of disorder in the RAM linker region on the effective concentration enhancement of Notch transcriptional activation, we measured the effects of linker length variation on activation. The resulting activation profile has general features of a worm-like chain model for effective concentration. However, deviations from the model for short sequence deletions suggest that RAM contains sequence-specific structural elements that may be important for activation. Structural characterization of the RAM linker with sedimentation velocity analytical ultracentrifugation and NMR spectroscopy reveals that the linker is compact and contains three transient helices and two extended and dynamic regions. To test if these secondary structure elements are important for activation, we made sequence substitutions to change the secondary structure propensities of these elements and measured transcriptional activation of the resulting variants. Substitutions to two of these nonrandom elements (helix 2, extended region 1) have effects on activation, but these effects do not depend on the nature of the substituting residues. Thus, the primary sequences of these elements, but not their secondary structures, are influencing signaling.


Subject(s)
Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Regulatory Elements, Transcriptional/genetics , Amino Acid Sequence , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Receptors, Notch/genetics , Sequence Homology, Amino Acid , Signal Transduction , Transcriptional Activation
7.
Protein Sci ; 24(5): 812-22, 2015 May.
Article in English | MEDLINE | ID: mdl-25650119

ABSTRACT

Notch is an intercellular signaling pathway that is highly conserved in metazoans and is essential for proper cellular specification during development and in the adult organism. Misregulated Notch signaling underlies or contributes to the pathogenesis of many human diseases, most notably cancer. Signaling through the Notch pathway ultimately results in changes in gene expression, which is regulated by the transcription factor CSL. Upon pathway activation, CSL forms a ternary complex with the intracellular domain of the Notch receptor (NICD) and the transcriptional coactivator Mastermind (MAM) that activates transcription from Notch target genes. While detailed in vitro studies have been conducted with mammalian and worm orthologous proteins, less is known regarding the molecular details of the Notch ternary complex in Drosophila. Here we thermodynamically characterize the assembly of the fly ternary complex using isothermal titration calorimetry. Our data reveal striking differences in the way the RAM (RBP-J associated molecule) and ANK (ankyrin) domains of NICD interact with CSL that is specific to the fly. Additional analysis using cross-species experiments suggest that these differences are primarily due to fly CSL, while experiments using point mutants show that the interface between fly CSL and ANK is likely similar to the mammalian and worm interface. Finally, we show that the binding of the fly RAM domain to CSL does not affect interactions of the corepressor Hairless with CSL. Taken together, our data suggest species-specific differences in ternary complex assembly that may be significant in understanding how CSL regulates transcription in different organisms.


Subject(s)
DNA-Binding Proteins/chemistry , Drosophila Proteins/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Protein Structure, Tertiary , Receptors, Notch/chemistry , Transcriptional Activation/genetics , Amino Acid Sequence/genetics , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Models, Molecular , Receptors, Notch/genetics , Signal Transduction , Thermodynamics
8.
Nucleic Acids Res ; 42(16): 10550-63, 2014.
Article in English | MEDLINE | ID: mdl-25114055

ABSTRACT

Regulation of transcription is fundamental to development and physiology, and occurs through binding of transcription factors to specific DNA sequences in the genome. CSL (CBF1/Suppressor of Hairless/LAG-1), a core component of the Notch signaling pathway, is one such transcription factor that acts in concert with co-activators or co-repressors to control the activity of associated target genes. One fundamental question is how CSL can recognize and select among different DNA sequences available in vivo and whether variations between selected sequences can influence its function. We have therefore investigated CSL-DNA recognition using computational approaches to analyze the energetics of CSL bound to different DNAs and tested the in silico predictions with in vitro and in vivo assays. Our results reveal novel aspects of CSL binding that may help explain the range of binding observed in vivo. In addition, using molecular dynamics simulations, we show that domain-domain correlations within CSL differ significantly depending on the DNA sequence bound, suggesting that different DNA sequences may directly influence CSL function. Taken together, our results, based on computational chemistry approaches, provide valuable insights into transcription factor-DNA binding, in this particular case increasing our understanding of CSL-DNA interactions and how these may impact on its transcriptional control.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Regulatory Elements, Transcriptional , Binding Sites , Computer Simulation , Consensus Sequence , Cytosine/analysis , DNA/chemistry , DNA/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Molecular Dynamics Simulation , Nucleotide Motifs , Protein Binding , Software
9.
Structure ; 22(1): 3-5, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24411574

ABSTRACT

In this issue of Structure, Collins and colleagues report the structure of the Notch pathway component CSL bound to the corepressor KyoT2. The structure reveals that KyoT2 interacts with CSL in a configuration similar, but not identical to, the Notch RAM domain and provides insights into how CSL switches from an activator to a repressor.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , LIM Domain Proteins/chemistry , Muscle Proteins/chemistry , Receptors, Notch/chemistry , Animals
10.
Structure ; 22(1): 70-81, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24290140

ABSTRACT

Notch refers to a highly conserved cell-to-cell signaling pathway with essential roles in embryonic development and tissue maintenance. Dysfunctional signaling causes human disease, highlighting the importance of pathway regulation. Notch signaling ultimately results in the activation of target genes, which is regulated by the nuclear effector CSL (CBF-1/RBP-J, Su(H), Lag-1). CSL dually functions as an activator and a repressor of transcription through differential interactions with coactivator or corepressor proteins, respectively. Although the structures of CSL-coactivator complexes have been determined, the structures of CSL-corepressor complexes are unknown. Here, using a combination of structural, biophysical, and cellular approaches, we characterize the structure and function of CSL in complex with the corepressor KyoT2. Collectively, our studies provide molecular insights into how KyoT2 binds CSL with high affinity and competes with coactivators, such as Notch, for binding CSL. These studies are important for understanding how CSL functions as both an activator and a repressor of transcription of Notch target genes.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , LIM Domain Proteins/chemistry , Muscle Proteins/chemistry , Receptors, Notch/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Communication , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Genes, Reporter , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Protein Binding , Receptors, Notch/genetics , Receptors, Notch/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Thermodynamics , Transcription, Genetic
11.
PLoS One ; 7(8): e39093, 2012.
Article in English | MEDLINE | ID: mdl-22876274

ABSTRACT

The Notch signaling pathway is an intercellular communication network vital to metazoan development. Notch activation leads to the nuclear localization of the intracellular portion (NICD) of the Notch receptor. Once in the nucleus, NICD binds the transcription factor CSL through a bivalent interaction involving the high-affinity RAM region and the lower affinity ANK domain, converting CSL from a transcriptionally-repressed to an active state. This interaction is believed to directly displace co-repressor proteins from CSL and recruit co-activator proteins. Here we investigate the consequences of this bivalent organization in converting CSL from the repressed to active form. One proposed function of RAM is to promote the weak ANK:CSL interaction; thus, fusion of CSL-ANK should bypass this function of RAM. We find that a CSL-ANK fusion protein is transcriptionally active in reporter assays, but that the addition of RAM in trans further increases transcriptional activity, suggesting another role of RAM in activation. A single F235L point substitution, which disrupts co-repressor binding to CSL, renders the CSL-ANK fusion fully active and refractory to further stimulation by RAM in trans. These results suggest that in the context of a mammalian CSL-ANK fusion protein, the main role of RAM is to displace co-repressor proteins from CSL.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , RNA-Binding Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Amino Acid Substitution , Ankyrin Repeat , Cell Line , Co-Repressor Proteins/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Molecular Docking Simulation , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , Receptors, Notch/chemistry , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcriptional Activation
12.
Structure ; 20(2): 202-4, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22325769

ABSTRACT

In this issue of Structure, Choi et al. use hydrogen/deuterium exchange mass spectrometry to characterize a Notch transcription complex (NTC). When interpreted in the context of NTC X-ray structures, their findings reveal important molecular insights into the dynamics that underlie complex assembly.


Subject(s)
DNA-Binding Proteins/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Receptor, Notch1/chemistry , Transcription Factors/chemistry , Humans
13.
Structure ; 20(2): 340-9, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22325781

ABSTRACT

The Notch intracellular domain (NICD) forms a transcriptional activation complex with the DNA-binding factor CSL and a transcriptional co-activator of the Mastermind family (MAML). The "RAM" region of NICD recruits Notch to CSL, facilitating the binding of MAML at the interface between the ankyrin (ANK) repeat domain of NICD and CSL. Here, we report the X-ray structure of a human MAML1/RAM/ANK/CSL/DNA complex, and probe changes in component dynamics upon stepwise assembly of a MAML1/NICD/CSL complex using HX-MS. Association of CSL with NICD exerts remarkably little effect on the exchange kinetics of the ANK domain, whereas MAML1 binding greatly retards the exchange kinetics of ANK repeats 2-3. These exchange patterns identify critical features contributing to the cooperative assembly of Notch transcription complexes (NTCs), highlight the importance of MAML recruitment in rigidifying the ANK domain and stabilizing its interface with CSL, and rationalize the requirement for MAML1 in driving cooperative dimerization of NTCs on paired-site DNA.


Subject(s)
DNA-Binding Proteins/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Receptor, Notch1/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , DNA/chemistry , Humans , Macromolecular Substances/chemistry , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary
14.
PLoS Pathog ; 8(1): e1002479, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22253595

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is tightly linked to at least two lymphoproliferative disorders, primary effusion lymphoma (PEL) and multicentric Castleman's disease (MCD). However, the development of KSHV-mediated lymphoproliferative disease is not fully understood. Here, we generated two recombinant KSHV viruses deleted for the first RBP-Jκ binding site (RTA(1st)) and all three RBP-Jκ binding sites (RTA(all)) within the RTA promoter. Our results showed that RTA(1st) and RTA(all) recombinant viruses possess increased viral latency and a decreased capability for lytic replication in HEK 293 cells, enhancing colony formation and proliferation of infected cells. Furthermore, recombinant RTA(1st) and RTA(all) viruses showed greater infectivity in human peripheral blood mononuclear cells (PBMCs) relative to wt KSHV. Interestingly, KSHV BAC36 wt, RTA(1st) and RTA(all) recombinant viruses infected both T and B cells and all three viruses efficiently infected T and B cells in a time-dependent manner early after infection. Also, the capability of both RTA(1st) and RTA(all) recombinant viruses to infect CD19+ B cells was significantly enhanced. Surprisingly, RTA(1st) and RTA(all) recombinant viruses showed greater infectivity for CD3+ T cells up to 7 days. Furthermore, studies in Telomerase-immortalized human umbilical vein endothelial (TIVE) cells infected with KSHV corroborated our data that RTA(1st) and RTA(all) recombinant viruses have enhanced ability to persist in latently infected cells with increased proliferation. These recombinant viruses now provide a model to explore early stages of primary infection in human PBMCs and development of KSHV-associated lymphoproliferative diseases.


Subject(s)
Cell Proliferation , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Immediate-Early Proteins/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Virus Latency/genetics , Binding Sites/genetics , Cells, Cultured , Gene Expression Regulation, Viral , HEK293 Cells , Herpesviridae Infections/complications , Herpesviridae Infections/genetics , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Herpesvirus 8, Human/growth & development , Humans , Immediate-Early Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/virology , Promoter Regions, Genetic/genetics , Protein Binding , Trans-Activators/metabolism
15.
Dev Biol ; 358(2): 296-308, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21839069

ABSTRACT

During development, progenitor cells of the retina give rise to six principal classes of neurons and the Müller glial cells found within the adult retina. The pancreas transcription factor 1 subunit a (Ptf1a) encodes a basic-helix-loop-helix transcription factor necessary for the specification of horizontal cells and the majority of amacrine cell subtypes in the mouse retina. The Ptf1a-regulated genes and the regulation of Ptf1a activity by transcription cofactors during retinogenesis have been poorly investigated. Using a retrovirus-mediated gene transfer approach, we reported that Ptf1a was sufficient to promote the fates of amacrine and horizontal cells from retinal progenitors and inhibit retinal ganglion cell and photoreceptor differentiation in the chick retina. Both GABAergic H1 and non-GABAergic H3 horizontal cells were induced following the forced expression of Ptf1a. We describe Ptf1a as a strong, negative regulator of Atoh7 expression. Furthermore, the Rbpj-interacting domains of Ptf1a protein were required for its effects on cell fate specification. Together, these data provide a novel insight into the molecular basis of Ptf1a activity on early cell specification in the chick retina.


Subject(s)
Avian Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Retina/embryology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Transcription Factors/metabolism , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Chick Embryo , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Multiprotein Complexes , Protein Interaction Domains and Motifs , Retina/cytology , Retinal Ganglion Cells/classification , Transcription Factors/chemistry , Transcription Factors/genetics
16.
Mol Biol Cell ; 22(17): 3242-52, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21737682

ABSTRACT

In metazoans, the highly conserved Notch pathway drives cellular specification. On receptor activation, the intracellular domain of Notch assembles a transcriptional activator complex that includes the DNA-binding protein CSL, a composite of human C-promoter binding factor 1, Suppressor of Hairless of Drosophila melanogaster [Su(H)], and lin-12 and Glp-1 phenotype of Caenorhabditis elegans. In the absence of ligand, CSL represses Notch target genes. However, despite the structural similarity of CSL orthologues, repression appears largely diverse between organisms. Here we analyze the Notch repressor complex in Drosophila, consisting of the fly CSL protein, Su(H), and the corepressor Hairless, which recruits general repressor proteins. We show that the C-terminal domain of Su(H) is necessary and sufficient for forming a high-affinity complex with Hairless. Mutations in Su(H) that affect interactions with Notch and Mastermind have no effect on Hairless binding. Nonetheless, we demonstrate that Notch and Hairless compete for CSL in vitro and in cell culture. In addition, we identify a site in Hairless that is crucial for binding Su(H) and subsequently show that this Hairless mutant is strongly impaired, failing to properly assemble the repressor complex in vivo. Finally, we demonstrate Hairless-mediated inhibition of Notch signaling in a cell culture assay, which hints at a potentially similar repression mechanism in mammals that might be exploited for therapeutic purposes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans Proteins/chemistry , Cells, Cultured , DNA-Binding Proteins/chemistry , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Larva/genetics , Larva/growth & development , Larva/metabolism , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Notch/chemistry , Receptors, Notch/genetics , Repressor Proteins/chemistry , Sequence Deletion , Thermodynamics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Two-Hybrid System Techniques
17.
J Virol ; 85(13): 6148-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21507979

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV; or human herpesvirus 8 [HHV8]) is implicated in the pathogenesis of many human malignancies including Kaposi's sarcoma (KS), multicentric Castleman's disease (MCD), and primary effusion lymphoma (PEL). KSHV infection displays two alternative life cycles, referred to as the latent and lytic or productive cycle. Previously, we have reported that the replication and transcription activator (RTA), a major lytic cycle transactivator, contributes to the development of KSHV latency by inducing latency-associated nuclear antigen (LANA) expression during early stages of infection by targeting RBP-Jκ, the master regulator of the Notch signaling pathway. Here, we generated a bacterial artificial chromosome (BAC) KSHV recombinant virus with a deletion of the RBP-Jκ site within the LANA promoter to evaluate the function of the RBP-Jκ cognate site in establishing primary latent infection. The results showed that genetic disruption of the RBP-Jκ binding site within the KSHV LANA promoter led to enhanced expression of the KSHV-encoded immediate early RTA, resulting in an increase in lytic replication during primary infection of human peripheral blood mononuclear cells (PBMCs). This system provides a powerful tool for use in indentifying additional cellular and viral molecules involved in LANA-mediated latency maintenance during the early stages of KSHV infection.


Subject(s)
Antigens, Viral/chemistry , Herpesvirus 8, Human/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Nuclear Proteins/chemistry , Promoter Regions, Genetic/genetics , Virus Latency , Antigens, Viral/genetics , Antigens, Viral/metabolism , Binding Sites , Cell Line , Chromosomes, Artificial, Bacterial , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Immediate-Early Proteins , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Leukocytes, Mononuclear/virology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombination, Genetic , Trans-Activators , Virus Activation , Virus Latency/genetics
18.
J Biol Chem ; 286(17): 14892-902, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21372128

ABSTRACT

The Notch pathway is a conserved cell-to-cell signaling mechanism that mediates cell fate decisions in metazoans. Canonical signaling results in changes in gene expression, which is regulated by the nuclear effector of the pathway CSL (CBF1/RBP-J, Su(H), Lag-1). CSL is a DNA binding protein that functions as either a repressor or an activator of transcription, depending upon whether it is complexed by transcriptional corepressor or coactivator proteins, respectively. In stark contrast to CSL-coactivator complexes, e.g. the transcriptionally active CSL-Notch-Mastermind ternary complex, the structure and function of CSL-corepressor complexes are poorly understood. The corepressor MINT (Msx2-interacting nuclear target protein) has been shown in vivo to antagonize Notch signaling and shown in vitro to biochemically interact with CSL; however, the molecular details of this interaction are only partially defined. Here, we provide a quantitative thermodynamic binding analysis of CSL-MINT complexes. Using isothermal titration calorimetry, we demonstrate that MINT forms a high affinity complex with CSL, and we also delineate the domains of MINT and CSL that are necessary and sufficient for complex formation. Moreover, we show in cultured cells that this region of MINT can inhibit Notch signaling in transcriptional reporter assays. Taken together, our results provide functional insights into how CSL is converted from a repressor to an activator of transcription.


Subject(s)
Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Nuclear Proteins/metabolism , Receptors, Notch/genetics , Thermodynamics , Transcription, Genetic , Animals , Cells, Cultured , DNA-Binding Proteins , Fibroblasts/chemistry , Fibroblasts/cytology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Protein Binding , RNA-Binding Proteins , Receptors, Notch/antagonists & inhibitors , Repressor Proteins , Trans-Activators
19.
Nat Struct Mol Biol ; 17(11): 1312-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20972443

ABSTRACT

Ligand-induced proteolysis of Notch produces an intracellular effector domain that transduces essential signals by regulating the transcription of target genes. This function relies on the formation of transcriptional activation complexes that include intracellular Notch, a Mastermind co-activator and the transcription factor CSL bound to cognate DNA. These complexes form higher-order assemblies on paired, head-to-head CSL recognition sites. Here we report the X-ray structure of a dimeric human Notch1 transcription complex loaded on the paired site from the human HES1 promoter. The small interface between the Notch ankyrin domains could accommodate DNA bending and untwisting to allow a range of spacer lengths between the two sites. Cooperative dimerization occurred on the human and mouse Hes5 promoters at a sequence that diverged from the CSL-binding consensus at one of the sites. These studies reveal how promoter organizational features control cooperativity and, thus, the responsiveness of different promoters to Notch signaling.


Subject(s)
Promoter Regions, Genetic , Receptor, Notch1/chemistry , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Conserved Sequence , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dimerization , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Receptor, Notch1/metabolism , Repressor Proteins/chemistry , Transcription Factor HES-1 , Transcription Factors/chemistry , Transcription Factors/metabolism
20.
Cell Death Differ ; 17(11): 1728-38, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20508646

ABSTRACT

Notch1 signaling has a critical function in maintaining a balance among cell proliferation, differentiation, and apoptosis. Our earlier work showed that the Notch1 intracellular domain interferes with the scaffolding function of c-Jun N-terminal kinase (JNK)-interacting protein-1 (JIP1), yet the effect of JIP1 for Notch1-recombining binding protein suppressor of hairless (RBP-Jk) signaling remains unknown. Here, we show that JIP1 suppresses Notch1 activity. JIP1 was found to physically associate with either intracellular domain of Notch1 or RBP-Jk and interfere with the interaction between them. Furthermore, we ascertained that JIP1 caused the cytoplasmic retention of RBP-Jk through an interaction between the C-terminal region of JIP1 including Src homology 3 domain and the proline-rich domain of RBP-Jk. We also found that RBP-Jk inhibits JIP1-mediated activation of the JNK1 signaling cascade and cell death. Our results suggest that direct protein-protein interactions coordinate cross-talk between the Notch1-RBP-Jk and JIP1-JNK pathways.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Receptor, Notch1/metabolism , Signal Transduction , Animals , Apoptosis , Blotting, Western , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Intercellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Rats , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL