Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ted R. Hupp is active.

Publication


Featured researches published by Ted R. Hupp.


Cell | 1992

Regulation of the specific DNA binding function of p53

Ted R. Hupp; David W. Meek; Carol Midgley; David P. Lane

The DNA binding activity of p53 is required for its tumor suppressor function; we show here that this activity is cryptic but can be activated by cellular factors acting on a C-terminal regulatory domain of p53. A gel mobility shift assay demonstrated that recombinant wild-type human p53 binds DNA sequence specifically only weakly, but a monoclonal antibody binding near the C terminus activated the cryptic DNA binding activity stoichiometrically. p53 DNA binding could be activated by a C-terminal deletion of p53, mild proteolysis of full-length p53, E. coli dnaK (which disrupts protein-protein complexes), or casein kinase II (and coincident phosphorylation of a C-terminal site on p53). Activation of p53 DNA binding may be critical in regulation of its ability to arrest cell growth and thus its tumor suppressor function.


Cell | 1995

Small peptides activate the latent sequence-specific DNA binding function of p53

Ted R. Hupp; Alison Sparks; David P. Lane

Normal cells contain p53 protein in a latent state that can be activated for sequence-specific transcription by low levels of UV radiation without an increase in protein levels. Microinjection of cells with an antibody specific to the C-terminal negative regulatory domain can activate the function of p53 as a specific transcription factor in the absence of irradiation damage, suggesting that posttranslational modification of a negative regulatory domain in vivo is a rate-limiting step for p53 activation. Small peptides derived from the negative regulatory domain of p53 have been used as biochemical tools to distinguish between allosteric and steric mechanisms of negative regulation of p53 tetramer activity. Presented is the development of a highly specific peptide activation system that is consistent with an allosteric mechanism of negative regulation and that forms a precedent for the synthesis of novel low molecular mass modifiers of the p53 response.


Current Biology | 1994

Allosteric activation of latent p53 tetramers

Ted R. Hupp; David P. Lane

BACKGROUND The DNA-binding activity of p53 is essential to its function as a tumour suppressor. Point mutations that abolish this activity have been found to occur frequently in the p53 genes of human cancer cells. Wild-type p53 protein assembles into oligomers with latent DNA-binding activity that can be activated in vitro by phosphorylation of a carboxy-terminal regulatory region, catalyzed by protein kinase C or casein kinase II. We have investigated the mechanism underlying this post-translational regulation of p53. Specifically, we have asked the following questions. First, whether the carboxy-terminal regulatory site contributes to p53s ability to form tetramers. Second, whether the latent DNA-binding activity of p53 can be activated in vivo. And third, whether the activation of p53 is reversible. RESULTS Biophysical molecular-sizing analysis shows that both latent and activated forms of p53 are tetramers. Using a novel method, we have further established that p53 remains tetrameric when bound to DNA. We have also found that p53 can indeed be activated in vivo: p53 prepared from cells can be separated into activated and latent forms. Finally, we generated a monoclonal antibody specific for the casein kinase II target site in the carboxy-terminal regulatory region of p53, and used it to demonstrate the allosteric inhibition of in vitro and in vivo activated forms of p53. CONCLUSIONS p53 protein assembles naturally as a tetramer that can be converted between latent and activated forms by a concerted, allosteric transition. The highly purified, reconstituted system that we have developed, in which the DNA-binding activity of p53 can be reversibly regulated, should facilitate the discovery of agents that can modulate the DNA-binding activity of p53--particularly those that can activate mutant p53 proteins and that may have potential in the design of anti-cancer drugs.


Biochemical Journal | 1999

Novel phosphorylation sites of human tumour suppressor protein p53 at Ser20 and Thr18 that disrupt the binding of mdm2 (mouse double minute 2) protein are modified in human cancers

Ashley L. Craig; Lindsay Burch; Borek Vojtesek; Jaroslava Mikutowska; Alastair M. Thompson; Ted R. Hupp

The ability to separate the isoforms of human tumour suppressor protein p53 expressed in insect cells using heparin-Sepharose correlates with differences in the isoelectric point of p53, demonstrating that p53 can be heterogeneously modified and providing support for the use of insect cells as a model system for identifying novel signalling pathways that target p53. One p53 isoform that was reduced in its binding to the monoclonal antibody DO-1 could be stimulated in its binding to DO-1 by prior incubation with protein phosphatases, suggesting the presence of a previously unidentified N-terminal phosphorylation site capable of masking the DO-1 epitope. A synthetic peptide from the N-terminal domain of p53 containing phosphate at Ser(20) inhibited DO-1 binding, thus identifying the phosphorylation site responsible for DO-1 epitope masking. Monoclonal antibodies overlapping the DO-1 epitope were developed that are specific for phospho-Thr(18) (adjacent to the DO-1 epitope) and phospho-Ser(20) (within the DO-1 epitope) to determine whether direct evidence could be obtained for novel phosphorylation sites in human p53. A monoclonal antibody highly specific for phospho-Ser(20) detected significant phosphorylation of human p53 expressed in insect cells, whereas the relative proportion of p53 modified at Thr(18) was substantially lower. The relevance of these two novel phosphorylation sites to p53 regulation in human cells was made evident by the extensive phosphorylation of human p53 at Thr(18) and Ser(20) in a panel of human breast cancers with a wild-type p53 status. Phospho-Ser(20) or phospho-Thr(18) containing p53 peptides are as effective as the phospho-Ser(15) peptide at reducing mdm2 (mouse double minute 2) protein binding, indicating that the functional effects of these phosphorylation events might be to regulate the binding of heterologous proteins to p53. These results provide evidence in vivo for two novel phosphorylation sites within p53 at Ser(20) and Thr(18) that can affect p53 protein-protein interactions and indicate that some human cancers might have amplified one or more Ser(20) and Thr(18) kinase signalling cascades to modulate p53 activity.


Journal of Biological Chemistry | 2002

The conformationally flexible S9-S10 linker region in the core domain of p53 contains a novel MDM2 binding site whose mutation increases ubiquitination of p53 in vivo.

Harumi Shimizu; Lindsay Burch; Amanda J. Smith; David Dornan; Maura Wallace; Kathryn L. Ball; Ted R. Hupp

Although the N-terminal BOX-I domain of the tumor suppressor protein p53 contains the primary docking site for MDM2, previous studies demonstrated that RNA stabilizes the MDM2·p53 complex using a p53 mutant lacking theBOX-I motif. In vitro assays measuring the specific activity of MDM2 in the ligand-free and RNA-bound state identified a novel MDM2 interaction site in the core domain of p53. As defined using phage-peptide display, the RNA·MDM2 isoform exhibited a notable switch in peptide binding specificity, with enhanced affinity for novel peptide sequences in either p53 or small nuclear ribonucleoprotein-U (snRNP-U) and substantially reduced affinity for the primary p53 binding site in the BOX-I domain. The consensus binding site for the RNA·MDM2 complex within p53 is SGXLLGESXF, which links the S9–S10 β-sheets flanking the BOX-IV and BOX-V motifs in the core domain and which is a site of reversible conformational flexibility in p53. Mutation of conserved amino acids in the linker at Ser261 and Leu264, which bridges the S9–S10 β-sheets, stimulated p53 activity from reporter templates and increased MDM2-dependent ubiquitination of p53. Furthermore, mutation of the conserved Phe270 within the S10 β-sheet resulted in a mutant p53, which binds more stably to RNA·MDM2 complexes in vitro and which is strikingly hyper-ubiquitinated in vivo. Introducing an Ala19 mutation into the p53F270A protein abolished both RNA·MDM2 complex binding and hyper-ubiquitinationin vivo, thus indicating that p53F270A protein hyper-ubiquitination depends upon MDM2 binding to its primary site in the BOX-I domain. Together, these data identify a novel MDM2 binding interface within the S9–S10 β-sheet region of p53 that plays a regulatory role in modulating the rate of MDM2-dependent ubiquitination of p53 in cells.


EMBO Reports | 2001

Inhibition of p53‐dependent transcription by BOX‐I phospho‐peptide mimetics that bind to p300

David Dornan; Ted R. Hupp

The N‐terminal BOX‐I domain of p53 containing a docking site for the negative regulator MDM2 and the positive effector p300, harbours two recently identified phosphorylation sites at Thr18 or Ser20 whose affect on p300 is undefined. Biochemical assays demonstrate that although MDM2 binding is inhibited by these phosphorylations, p300 binding is strikingly stabilized by Thr18 or Ser20 phosphorylation. Introducing EGFP‐BOX‐I domain peptides with an aspartate substitution at Thr18 or Ser20 induced a significant inhibition of endogenous p53‐dependent transcription in cycling cells, in irradiated cells, as well as in cells transiently co‐transfected with p300 and p53. In contrast an EGFP‐wild‐type BOX‐I domain peptide stimulated p53 activity via inhibition of MDM2 protein binding. These results suggest that phosphorylation of p53 at Thr18 or Ser20 can activate p53 by stabilizing the p300–p53 complex and also identify a class of small molecular weight ligands capable of selective discrimination between MDM2‐ and p300‐dependent activities.


Drug Discovery Today | 2003

Drug discovery and p53

David P. Lane; Ted R. Hupp

In the past two decades, the identification of commonly mutated oncogenes and tumour suppressor genes has driven an unprecedented growth in our understanding of the genetic basis of human cancer. Although oncogenes can clearly serve as classically defined drug targets whose inactivation by small molecules could place a brake on cancer cell proliferation, the restoration of mutated tumour suppressor gene activity by small molecules might appear on the surface to be unrealistic. However, there is a growing realization that many eukaryotic regulatory proteins are partially unfolded and such intrinsically disordered proteins acquire a folded structure after binding to their biological target. Molecular characterization of the p53 protein has shown that its conformational flexibility and intrinsic thermodynamic instability provide a foundation from which its conformation can be quickly post-translationally modified.


Journal of Biological Chemistry | 2001

Stoichiometric phosphorylation of human p53 at Ser315 stimulates p53-dependent transcription

Jeremy P. Blaydes; M. Gloria Luciani; Sarka Pospisilova; Helen Mei-Ling Ball; Borek Vojtesek; Ted R. Hupp

p53 protein activity as a transcription factor can be activated in vivo by antibodies that target its C-terminal negative regulatory domain suggesting that cellular enzymes that target this domain may play a role in stimulating p53-dependent gene expression. A phospho-specific monoclonal antibody to the C-terminal Ser315phospho-epitope was used to determine whether phosphorylation of endogenous p53 at Ser315 can be detected in vivo, whether steady-state Ser315 phosphorylation increases or decreases in an irradiated cell, and whether this phosphorylation event activates or inhibits p53 in vivo. A native phospho-specific IgG binding assay was developed for quantitating the extent of p53 phosphorylation at Ser315 where one, two, three, or four phosphates/tetramer could be defined after in vitro phosphorylation by cyclin-dependent protein kinases. Using this assay, near-stoichiometric Ser315 phosphorylation of endogenous p53 protein was detected in vivo after UV irradiation of MCF7 and A375 cells, coinciding with elevated p53-dependent transcription. Transfection of the p53 gene with an alanine mutation at the Ser315 site into Saos-2 cells gave rise to a form of p53 protein with a substantially reduced specific activity as a transcription factor. The treatment of cells with the cyclin-dependent protein kinase inhibitor Roscovitine promoted a reduction in the specific activity of endogenous p53 or ectopically expressed p53. These results indicate that the majority of p53 protein has been phosphorylated at Ser315 after irradiation damage and identify a cyclin-dependent kinase pathway that plays a role in stimulating p53 function.


FEBS Journal | 2010

Death‐associated protein kinase (DAPK) and signal transduction: additional roles beyond cell death

Yao Lin; Ted R. Hupp; Craig Stevens

Death‐associated protein kinase (DAPK) is a stress‐regulated protein kinase that mediates a range of processes, including signal‐induced cell death and autophagy. Although the kinase domain of DAPK has a range of substrates that mediate its signalling, the additional protein interaction domains of DAPK are relatively ill defined. This review will summarize our current knowledge of the DAPK interactome, the use of peptide aptamers to define novel protein–protein interaction motifs, and how these new protein–protein interactions give insight into DAPK functions in diverse cellular processes, including growth factor signalling, the regulation of autophagy, and its emerging role in the regulation of immune responses.


Proceedings of the National Academy of Sciences of the United States of America | 2013

M2 pyruvate kinase provides a mechanism for nutrient sensing and regulation of cell proliferation

Hugh P. Morgan; Francis J. O’Reilly; Martin A. Wear; J. Robert O’Neill; Linda A. Fothergill-Gilmore; Ted R. Hupp; Malcolm D. Walkinshaw

We show that the M2 isoform of pyruvate kinase (M2PYK) exists in equilibrium between monomers and tetramers regulated by allosteric binding of naturally occurring small-molecule metabolites. Phenylalanine stabilizes an inactive T-state tetrameric conformer and inhibits M2PYK with an IC50 value of 0.24 mM, whereas thyroid hormone (triiodo-l-thyronine, T3) stabilizes an inactive monomeric form of M2PYK with an IC50 of 78 nM. The allosteric activator fructose-1,6-bisphosphate [F16BP, AC50 (concentration that gives 50% activation) of 7 μM] shifts the equilibrium to the tetrameric active R-state, which has a similar activity to that of the constitutively fully active isoform M1PYK. Proliferation assays using HCT-116 cells showed that addition of inhibitors phenylalanine and T3 both increased cell proliferation, whereas addition of the activator F16BP reduced proliferation. F16BP abrogates the inhibitory effect of both phenylalanine and T3, highlighting a dominant role of M2PYK allosteric activation in the regulation of cancer proliferation. X-ray structures show constitutively fully active M1PYK and F16BP-bound M2PYK in an R-state conformation with a lysine at the dimer-interface acting as a peg in a hole, locking the active tetramer conformation. Binding of phenylalanine in an allosteric pocket induces a 13° rotation of the protomers, destroying the peg-in-hole R-state interface. This distinct T-state tetramer is stabilized by flipped out Trp/Arg side chains that stack across the dimer interface. X-ray structures and biophysical binding data of M2PYK complexes explain how, at a molecular level, fluctuations in concentrations of amino acids, thyroid hormone, and glucose metabolites switch M2PYK on and off to provide the cell with a nutrient sensing and growth signaling mechanism.

Collaboration


Dive into the Ted R. Hupp's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Euan Murray

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alastair M. Thompson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Borivoj Vojtesek

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge