Network


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

Hotspot


Dive into the research topics where Thomas H. Scheuermann is active.

Publication


Featured researches published by Thomas H. Scheuermann.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Artificial ligand binding within the HIF2α PAS-B domain of the HIF2 transcription factor

Thomas H. Scheuermann; Diana R. Tomchick; Mischa Machius; Yan Guo; Richard K. Bruick; Kevin H. Gardner

The hypoxia-inducible factor (HIF) basic helix–loop–helix Per-aryl hydrocarbon receptor nuclear translocator (ARNT)-Sim (bHLH-PAS) transcription factors are master regulators of the conserved molecular mechanism by which metazoans sense and respond to reductions in local oxygen concentrations. In humans, HIF is critically important for the sustained growth and metastasis of solid tumors. Here, we describe crystal structures of the heterodimer formed by the C-terminal PAS domains from the HIF2α and ARNT subunits of the HIF2 transcription factor, both in the absence and presence of an artificial ligand. Unexpectedly, the HIF2α PAS-B domain contains a large internal cavity that accommodates ligands identified from a small-molecule screen. Binding one of these ligands to HIF2α PAS-B modulates the affinity of the HIF2α:ARNT PAS-B heterodimer in vitro. Given the essential role of PAS domains in forming active HIF heterodimers, these results suggest a presently uncharacterized ligand-mediated mechanism for regulating HIF2 activity in endogenous and clinical settings.


Nature Chemical Biology | 2013

Allosteric inhibition of hypoxia inducible factor-2 with small molecules

Thomas H. Scheuermann; Qiming Li; He-Wen Ma; Jason Key; Lei Zhang; Rui-rui Chen; Joseph A. Garcia; Jacinth Naidoo; Jamie Longgood; Doug E. Frantz; Uttam K. Tambar; Kevin H. Gardner; Richard K. Bruick

Hypoxia Inducible Factors (HIFs) are heterodimeric transcription factors induced in many cancers where they frequently promote the expression of many protumorigenic pathways. Though transcription factors are typically considered “undruggable”, the PAS-B domain of the HIF-2α subunit contains a large cavity within its hydrophobic core that offers a unique foothold for small-molecule regulation. Here we identify artificial ligands that bind within this pocket and characterize the resulting structural and functional changes caused by binding. Notably, these ligands antagonize HIF-2 heterodimerization and DNA-binding activity in vitro and in cultured cells, reducing HIF-2 target gene expression. Despite the high identity between HIF-2α and HIF-1α, these ligands are highly selective and do not affect HIF-1 function. These chemical tools establish the molecular basis for selective regulation of HIF-2, providing potential therapeutic opportunities to intervene in HIF-2-driven tumors such as renal cell carcinomas.


Journal of the American Chemical Society | 2009

Principles of Ligand Binding within a Completely Buried Cavity in HIF2α PAS-B

Jason Key; Thomas H. Scheuermann; Peter C. Anderson; Valerie Daggett; Kevin H. Gardner

Hypoxia-inducible factors (HIFs) are heterodimeric transcription factors responsible for the metazoan hypoxia response and promote tumor growth, metastasis, and resistance to cancer treatment. The C-terminal Per-ARNT-Sim (PAS) domain of HIF2alpha (HIF2alpha PAS-B) contains a preformed solvent-inaccessible cavity that binds artificial ligands that allosterically perturb the formation of the HIF heterodimer. To better understand how small molecules bind within this domain, we examined the structures and equilibrium and transition-state thermodynamics of HIF2alpha PAS-B with several artificial ligands using isothermal titration calorimetry, NMR exchange spectroscopy, and X-ray crystallography. Rapid association rates reveal that ligand binding is not dependent upon a slow conformational change in the protein to permit ligand access, despite the closed conformation observed in the NMR and crystal structures. Compensating enthalpic and entropic contributions to the thermodynamic barrier for ligand binding suggest a binding-competent transition state characterized by increased structural disorder. Finally, molecular dynamics simulations reveal conversion between open and closed conformations of the protein and pathways of ligand entry into the binding pocket.


Journal of Medicinal Chemistry | 2013

Development of inhibitors of the PAS-B domain of the HIF-2α transcription factor

Jamie L. Rogers; Liela Bayeh; Thomas H. Scheuermann; Jamie Longgood; Jason Key; Jacinth Naidoo; Lisa Melito; Cameron Shokri; Doug E. Frantz; Richard K. Bruick; Kevin H. Gardner; John B. MacMillan; Uttam K. Tambar

Hypoxia inducible factors (HIFs) are heterodimeric transcription factors induced in a variety of pathophysiological settings, including cancer. We describe the first detailed structure-activity relationship study of small molecules designed to inhibit HIF-2α-ARNT heterodimerization by binding an internal cavity of the HIF-2α PAS-B domain. Through a series of biophysical characterizations of inhibitor-protein interactions (NMR and X-ray crystallography), we have established the structural requirements for artificial inhibitors of the HIF-2α-ARNT PAS-B interaction. These results may serve as a foundation for discovering therapeutic agents that function by a novel mode of action.


Nature Structural & Molecular Biology | 2016

Structure and ligand-binding mechanism of the human OX1 and OX2 orexin receptors

Jie Yin; Kerim Babaoglu; Chad A. Brautigam; Lindsay Clark; Zhenhua Shao; Thomas H. Scheuermann; Charles M. Harrell; Anthony L. Gotter; Anthony J. Roecker; Christopher J. Winrow; John J. Renger; Paul J. Coleman; Daniel M. Rosenbaum

The orexin (also known as hypocretin) G protein–coupled receptors (GPCRs) regulate sleep and other behavioral functions in mammals, and are therapeutic targets for sleep and wake disorders. The human receptors hOX1R and hOX2R, which are 64% identical in sequence, have overlapping but distinct physiological functions and potential therapeutic profiles. We determined structures of hOX1R bound to the OX1R-selective antagonist SB-674042 and the dual antagonist suvorexant at 2.8-Å and 2.75-Å resolution, respectively, and used molecular modeling to illuminate mechanisms of antagonist subtype selectivity between hOX1R and hOX2R. The hOX1R structures also reveal a conserved amphipathic α-helix, in the extracellular N-terminal region, that interacts with orexin-A and is essential for high-potency neuropeptide activation at both receptors. The orexin-receptor crystal structures are valuable tools for the design and development of selective orexin-receptor antagonists and agonists.


Methods | 2015

High-precision, automated integration of multiple isothermal titration calorimetric thermograms: new features of NITPIC.

Thomas H. Scheuermann; Chad A. Brautigam

Isothermal titration calorimetry (ITC) has become a standard and widely available tool to measure the thermodynamic parameters of macromolecular associations. Modern applications of the method, including global analysis and drug screening, require the acquisition of multiple sets of data; sometimes these data sets number in the hundreds. Therefore, there is a need for quick, precise, and automated means to process the data, particularly at the first step of data analysis, which is commonly the integration of the raw data to yield an interpretable isotherm. Herein, we describe enhancements to an algorithm that previously has been shown to provide an automated, unbiased, and high-precision means to integrate ITC data. These improvements allow for the speedy and precise serial integration of an unlimited number of ITC data sets, and they have been implemented in the freeware program NITPIC, version 1.1.0. We present a comprehensive comparison of the performance of this software against an older version of NITPIC and a current version of Origin, which is commonly used for integration. The new methods recapitulate the excellent performance of the previous versions of NITPIC while speeding it up substantially, and their precision is significantly better than that of Origin. This new version of NITPIC is therefore well suited to the serial integration of many ITC data sets.


Analytical Biochemistry | 2016

On the acquisition and analysis of microscale thermophoresis data.

Thomas H. Scheuermann; Shae B. Padrick; Kevin H. Gardner; Chad A. Brautigam

A comprehensive understanding of the molecular mechanisms underpinning cellular functions is dependent on a detailed characterization of the energetics of macromolecular binding, often quantified by the equilibrium dissociation constant, KD. While many biophysical methods may be used to obtain KD, the focus of this report is a relatively new method called microscale thermophoresis (MST). In an MST experiment, a capillary tube filled with a solution containing a dye-labeled solute is illuminated with an infrared laser, rapidly creating a temperature gradient. Molecules will migrate along this gradient, causing changes in the observed fluorescence. Because the net migration of the labeled molecules will depend on their liganded state, a binding curve as a function of ligand concentration can be constructed from MST data and analyzed to determine KD. Herein, simulations demonstrate the limits of KD that can be measured in current instrumentation. They also show that binding kinetics is a major concern in planning and executing MST experiments. Additionally, studies of two protein-protein interactions illustrate challenges encountered in acquiring and analyzing MST data. Combined, these approaches indicate a set of best practices for performing and analyzing MST experiments. Software for rigorous data analysis is also introduced.


Journal of Medicinal Chemistry | 2015

Isoform-Selective and Stereoselective Inhibition of Hypoxia Inducible Factor-2

Thomas H. Scheuermann; Daniel Stroud; Christopher E. Sleet; Liela Bayeh; Cameron Shokri; Hanzhi Wang; Charles G. Caldwell; Jamie Longgood; John B. MacMillan; Richard K. Bruick; Kevin H. Gardner; Uttam K. Tambar

Hypoxia inducible factor (HIF) transcription factors reside at the center of signaling pathways used by mammalian cells to sense and respond to low oxygen levels. While essential to maintain oxygen homeostasis, misregulation of HIF protein activity correlates with tumor development and metastasis. To provide artificial routes to target misregulated HIF activity, we identified small molecule antagonists of the HIF-2 transcription factor that bind an internal cavity within the C-terminal PAS domain of the HIF-2α subunit. Here we describe a new class of chiral small molecule ligands that provide the highest affinity binding, the most effective, isoform-selective inhibition of HIF-2 in cells, and trigger the largest protein conformation changes reported to date. The current results further illuminate the molecular mechanism of HIF-2 antagonism and suggest additional routes to develop higher affinity and potency HIF-2 antagonists.


Methods in Enzymology | 2007

Hypoxia‐Inducible Factors Per/ARNT/Sim Domains: Structure and Function

Thomas H. Scheuermann; Jinsong Yang; Lei Zhang; Kevin H. Gardner; Richard K. Bruick

Hypoxia-inducible factors (HIFs) are key transcriptional regulators of genes involved in cellular adaptation to reduced oxygen availability through effects on anaerobic metabolism, oxygen delivery, angiogenesis, and cellular survival and proliferation. As such, HIFs contribute to the pathogenesis of diseases in which oxygen availability is compromised, notably ischemia and tumorigenesis. Though tremendous progress has been made in elucidating the mechanisms underlying O(2)-dependent regulation of HIF by Fe(II)- and 2-oxoglutarate-dependent dioxygenases, HIF induction can be uncoupled from these modes of regulation in diseases such as cancer. Consequently, renewed interest has developed in understanding the structure/function relationships of individual P(er)/ARNT/S(im) (PAS) domains that are important for maintaining transcriptionally active HIF complexes, regardless of the manner by which HIF is induced. This review highlights strategies for the biophysical and biochemical characterization of the PAS domains found within both HIF subunits and provides a platform for future efforts to exploit these domains in therapeutic settings.


Journal of Biological Chemistry | 2015

Coiled-coil Coactivators Play a Structural Role Mediating Interactions in Hypoxia Inducible Factor Heterodimerization

Yirui Guo; Thomas H. Scheuermann; Carrie L. Partch; Diana R. Tomchick; Kevin H. Gardner

Background: Coiled-coil coactivators can enhance HIF-dependent gene transcription via direct interaction with the HIF/ARNT heterodimer. Results: ARNT uses the β-sheet of the PAS-B domain to recruit coiled-coil coactivators. Conclusion: Coiled-coil coactivators bridge HIF and ARNT via the PAS-B domain β-sheet contacts to both proteins to form a ternary structure. Significance: This work reveals the mechanism for assembling a coiled-coil coactivator complex with the HIF-2 transcription factor heterodimer. The hypoxia-inducible factor complex (HIF-α·aryl hydrocarbon receptor nuclear translocator (ARNT)) requires association with several transcription coactivators for a successful cellular response to hypoxic stress. In addition to the conventional global transcription coactivator CREB-binding protein/p300 (CBP/p300) that binds to the HIF-α transactivation domain, a new group of transcription coactivators called the coiled-coil coactivators (CCCs) interact directly with the second PER-ARNT-SIM (PAS) domain of ARNT (ARNT PAS-B). These less studied transcription coactivators play essential roles in the HIF-dependent hypoxia response, and CCC misregulation is associated with several forms of cancer. To better understand CCC protein recruitment by the heterodimeric HIF transcription factor, we used x-ray crystallography, NMR spectroscopy, and biochemical methods to investigate the structure of the ARNT PAS-B domain in complex with the C-terminal fragment of a coiled-coil coactivator protein, transforming acidic coiled-coil coactivator 3 (TACC3). We found that the HIF-2α PAS-B domain also directly interacts with TACC3, motivating an NMR data-derived model suggesting a means by which TACC3 could form a ternary complex with HIF-2α PAS-B and ARNT PAS-B via β-sheet/coiled-coil interactions. These findings suggest that TACC3 could be recruited as a bridge to cooperatively mediate between the HIF-2α PAS-B·ARNT PAS-B complex, thereby participating more directly in HIF-dependent gene transcription than previously anticipated.

Collaboration


Dive into the Thomas H. Scheuermann's collaboration.

Top Co-Authors

Avatar

Kevin H. Gardner

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Richard K. Bruick

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Uttam K. Tambar

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Doug E. Frantz

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Chad A. Brautigam

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Charles G. Caldwell

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jamie Longgood

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jason Key

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lei Zhang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Cameron Shokri

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge