Network


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

Hotspot


Dive into the research topics where Heidi J.A. Wallweber is active.

Publication


Featured researches published by Heidi J.A. Wallweber.


Cell | 2007

IAP Antagonists Induce Autoubiquitination of c-IAPs, NF-κB Activation, and TNFα-Dependent Apoptosis

Eugene Varfolomeev; John W. Blankenship; Sarah M. Wayson; Anna V. Fedorova; Nobuhiko Kayagaki; Parie Garg; Kerry Zobel; Jasmin N. Dynek; Linda O. Elliott; Heidi J.A. Wallweber; John A. Flygare; Wayne J. Fairbrother; Kurt Deshayes; Vishva M. Dixit; Domagoj Vucic

Inhibitor of apoptosis (IAP) proteins are antiapoptotic regulators that block cell death in response to diverse stimuli. They are expressed at elevated levels in human malignancies and are attractive targets for the development of novel cancer therapeutics. Herein, we demonstrate that small-molecule IAP antagonists bind to select baculovirus IAP repeat (BIR) domains resulting in dramatic induction of auto-ubiquitination activity and rapid proteasomal degradation of c-IAPs. The IAP antagonists also induce cell death that is dependent on TNF signaling and de novo protein biosynthesis. Additionally, the c-IAP proteins were found to function as regulators of NF-kappaB signaling. Through their ubiquitin E3 ligase activities c-IAP1 and c-IAP2 promote proteasomal degradation of NIK, the central ser/thr kinase in the noncanonical NF-kappaB pathway.


Science | 2017

T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition

Enfu Hui; Jeanne Cheung; Jing Zhu; Xiaolei Su; Marcus J. Taylor; Heidi J.A. Wallweber; Dibyendu K. Sasmal; Jun Huang; Jeong M. Kim; Ira Mellman; Ronald D. Vale

Immunotherapeutic PD-1–targeted therapies require CD28 to promote cancer cell killing. CD28 is a critical target for PD-1 blockade PD-1–targeted therapies have been a breakthrough for treating certain tumors and can rejuvenate T cells to unleash the anticancer immune response (see the Perspective by Clouthier and Ohashi). It is widely believed that PD-1 suppresses signaling through the T cell receptor (TCR). However, Hui et al. find instead that the TCR costimulatory receptor, CD28, is the primary target of PD-1 signaling. Independently, Kamphorst et al. show that CD28 is required for PD-1 therapies to kill cancer cells efficiently and eliminate chronic viral infections in mice. Lung cancer patients that responded to PD-1 therapy had more CD28+ T cells, which suggests that CD28 may predict treatment response. Science, this issue p. 1428, p. 1423; see also p. 1373 Programmed cell death–1 (PD-1) is a coinhibitory receptor that suppresses T cell activation and is an important cancer immunotherapy target. Upon activation by its ligand PD-L1, PD-1 is thought to suppress signaling through the T cell receptor (TCR). By titrating PD-1 signaling in a biochemical reconstitution system, we demonstrate that the co-receptor CD28 is strongly preferred over the TCR as a target for dephosphorylation by PD-1–recruited Shp2 phosphatase. We also show that CD28, but not the TCR, is preferentially dephosphorylated in response to PD-1 activation by PD-L1 in an intact cell system. These results reveal that PD-1 suppresses T cell function primarily by inactivating CD28 signaling, suggesting that costimulatory pathways play key roles in regulating effector T cell function and responses to anti–PD-L1/PD-1 therapy.


Journal of Biological Chemistry | 2005

Structures of APRIL-Receptor Complexes LIKE BCMA, TACI EMPLOYS ONLY A SINGLE CYSTEINE-RICH DOMAIN FOR HIGH AFFINITY LIGAND BINDING

Sarah G. Hymowitz; Darshana Ramesh Patel; Heidi J.A. Wallweber; Steven T. Runyon; Minhong Yan; JianPing Yin; Stephanie Shriver; Nathaniel C. Gordon; Borlan Pan; Nicholas J. Skelton; Robert F. Kelley; Melissa A. Starovasnik

TACI is a member of the tumor necrosis factor receptor superfamily and serves as a key regulator of B cell function. TACI binds two ligands, APRIL and BAFF, with high affinity and contains two cysteine-rich domains (CRDs) in its extracellular region; in contrast, BCMA and BR3, the other known high affinity receptors for APRIL and BAFF, respectively, contain only a single or partial CRD. However, another form of TACI exists wherein the N-terminal CRD is removed by alternative splicing. We find that this shorter form is capable of ligand-induced cell signaling and that the second CRD alone (TACI_d2) contains full affinity for both ligands. Furthermore, we report the solution structure and alanine-scanning mutagenesis of TACI_d2 along with co-crystal structures of APRIL·TACI_d2 and APRIL·BCMA complexes that together reveal the mechanism by which TACI engages high affinity ligand binding through a single CRD, and we highlight sources of ligand-receptor specificity within the APRIL/BAFF system.


Science | 2011

Antagonists Induce a Conformational Change in cIAP1 That Promotes Autoubiquitination

Erin C. Dueber; Allyn J. Schoeffler; Andreas Lingel; J. Michael Elliott; Anna V. Fedorova; Anthony M. Giannetti; Kerry Zobel; Brigitte Maurer; Eugene Varfolomeev; Ping Wu; Heidi J.A. Wallweber; Sarah G. Hymowitz; Kurt Deshayes; Domagoj Vucic; Wayne J. Fairbrother

Antagonist binding to an apoptosis inhibitor releases inhibition by promoting dimerization required for autoubiquitination. Inhibitor of apoptosis (IAP) proteins are negative regulators of cell death. IAP family members contain RING domains that impart E3 ubiquitin ligase activity. Binding of endogenous or small-molecule antagonists to select baculovirus IAP repeat (BIR) domains within cellular IAP (cIAP) proteins promotes autoubiquitination and proteasomal degradation and so releases inhibition of apoptosis mediated by cIAP. Although the molecular details of antagonist–BIR domain interactions are well understood, it is not clear how this binding event influences the activity of the RING domain. Here biochemical and structural studies reveal that the unliganded, multidomain cIAP1 sequesters the RING domain within a compact, monomeric structure that prevents RING dimerization. Antagonist binding induces conformational rearrangements that enable RING dimerization and formation of the active E3 ligase.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Structure of the pseudokinase–kinase domains from protein kinase TYK2 reveals a mechanism for Janus kinase (JAK) autoinhibition

Patrick J. Lupardus; Mark Ultsch; Heidi J.A. Wallweber; Pawan Bir Kohli; Adam R. Johnson; Charles Eigenbrot

Significance Cytokine signaling is essential for cell growth, hematopoiesis, and immune system function. Cytokine-mediated receptor dimerization induces intracellular activation of receptor-bound Janus kinases (JAKs), which then induce downstream transcriptional responses. We have determined a two-domain crystal structure containing the pseudokinase and kinase domains from the JAK family member TYK2, which identifies an inhibitory interaction interface between the two domains. Cancer-associated mutations found in other JAK family members map to this inhibitory interaction site, whereas analogous mutations in TYK2 cause in vitro activation of the kinase. This study identifies a mechanism for pseudokinase-mediated autoinhibition of the TYK2 kinase domain and suggests a means by which cancer-associated JAK mutations induce aberrant kinase activity. Janus kinases (JAKs) are receptor-associated multidomain tyrosine kinases that act downstream of many cytokines and interferons. JAK kinase activity is regulated by the adjacent pseudokinase domain via an unknown mechanism. Here, we report the 2.8-Å structure of the two-domain pseudokinase–kinase module from the JAK family member TYK2 in its autoinhibited form. We find that the pseudokinase and kinase interact near the kinase active site and that most reported mutations in cancer-associated JAK alleles cluster in or near this interface. Mutation of residues near the TYK2 interface that are analogous to those in cancer-associated JAK alleles, including the V617F and “exon 12” JAK2 mutations, results in increased kinase activity in vitro. These data indicate that JAK pseudokinases are autoinhibitory domains that hold the kinase domain inactive until receptor dimerization stimulates transition to an active state.


Neuron | 2016

Positive Allosteric Modulators of GluN2A-Containing NMDARs with Distinct Modes of Action and Impacts on Circuit Function

David H. Hackos; Patrick Lupardus; Teddy Grand; Yelin Chen; Tzu-Ming Wang; Paul Reynen; Amy Gustafson; Heidi J.A. Wallweber; Matthew Volgraf; Benjamin D. Sellers; Jacob B. Schwarz; Pierre Paoletti; Morgan Sheng; Qiang Zhou; Jesse E. Hanson

To enhance physiological function of NMDA receptors (NMDARs), we identified positive allosteric modulators (PAMs) of NMDARs with selectivity for GluN2A subunit-containing receptors. X-ray crystallography revealed a binding site at the GluN1-GluN2A dimer interface of the extracellular ligand-binding domains (LBDs). Despite the similarity between the LBDs of NMDARs and AMPA receptors (AMPARs), GluN2A PAMs with good selectivity against AMPARs were identified. Potentiation was observed with recombinant triheteromeric GluN1/GluN2A/GluN2B NMDARs and with synaptically activated NMDARs in brain slices from wild-type (WT), but not GluN2A knockout (KO), mice. Individual GluN2A PAMs exhibited variable degrees of glutamate (Glu) dependence, impact on NMDAR Glu EC50, and slowing of channel deactivation. These distinct PAMs also exhibited differential impacts during synaptic plasticity induction. The identification of a new NMDAR modulatory site and characterization of GluN2A-selective PAMs provide powerful molecular tools to dissect NMDAR function and demonstrate the feasibility of a therapeutically desirable type of NMDAR enhancement.


ACS Medicinal Chemistry Letters | 2015

Minor Structural Change to Tertiary Sulfonamide RORc Ligands Led to Opposite Mechanisms of Action.

Olivier René; Benjamin P. Fauber; Gladys de Leon Boenig; Brenda Burton; Céline Eidenschenk; Christine Everett; Alberto Gobbi; Sarah G. Hymowitz; Adam R. Johnson; James R. Kiefer; Marya Liimatta; Peter Lockey; Maxine Norman; Wenjun Ouyang; Heidi J.A. Wallweber; Harvey Wong

A minor structural change to tertiary sulfonamide RORc ligands led to distinct mechanisms of action. Co-crystal structures of two compounds revealed mechanistically consistent protein conformational changes. Optimized phenylsulfonamides were identified as RORc agonists while benzylsulfonamides exhibited potent inverse agonist activity. Compounds behaving as agonists in our biochemical assay also gave rise to an increased production of IL-17 in human PBMCs whereas inverse agonists led to significant suppression of IL-17 under the same assay conditions. The most potent inverse agonist compound showed >180-fold selectivity over the ROR isoforms as well as all other nuclear receptors that were profiled.


Structure | 2012

The crystal structure of the catalytic domain of the NF-κB inducing kinase reveals a narrow but flexible active site.

Gladys de Leon-Boenig; Krista K. Bowman; Jianwen A. Feng; Terry D. Crawford; Christine Everett; Yvonne Franke; Angela Oh; Mark S. Stanley; Steven Staben; Melissa A. Starovasnik; Heidi J.A. Wallweber; Jiansheng Wu; Lawren C. Wu; Adam R. Johnson; Sarah G. Hymowitz

The NF-κB inducing kinase (NIK) regulates the non-canonical NF-κB pathway downstream of important clinical targets including BAFF, RANKL, and LTβ. Despite numerous genetic studies associating dysregulation of this pathway with autoimmune diseases and hematological cancers, detailed molecular characterization of this central signaling node has been lacking. We undertook a systematic cloning and expression effort to generate soluble, well-behaved proteins encompassing the kinase domains of human and murine NIK. Structures of the apo NIK kinase domain from both species reveal an active-like conformation in the absence of phosphorylation. ATP consumption and peptide phosphorylation assays confirm that phosphorylation of NIK does not increase enzymatic activity. Structures of murine NIK bound to inhibitors possessing two different chemotypes reveal conformational flexibility in the gatekeeper residue controlling access to a hydrophobic pocket. Finally, a single amino acid difference affects the ability of some inhibitors to bind murine and human NIK with the same affinity.


Journal of Medicinal Chemistry | 2016

Discovery of GluN2A-Selective NMDA Receptor Positive Allosteric Modulators (PAMs): Tuning Deactivation Kinetics via Structure-Based Design.

Matthew Volgraf; Benjamin D. Sellers; Yu Jiang; Guosheng Wu; Cuong Ly; Elisia Villemure; Richard Pastor; Po-wai Yuen; Aijun Lu; Xifeng Luo; Mingcui Liu; Shun Zhang; Liang Sun; Yuhong Fu; Patrick J. Lupardus; Heidi J.A. Wallweber; Bianca M. Liederer; Gauri Deshmukh; Emile Plise; Suzanne Tay; Paul Reynen; James B Herrington; Amy Gustafson; Yichin Liu; Akim Dirksen; Matthias G. A. Dietz; Yanzhou Liu; Tzu-Ming Wang; Jesse E. Hanson; David H. Hackos

The N-methyl-D-aspartate receptor (NMDAR) is a Na(+) and Ca(2+) permeable ionotropic glutamate receptor that is activated by the coagonists glycine and glutamate. NMDARs are critical to synaptic signaling and plasticity, and their dysfunction has been implicated in a number of neurological disorders, including schizophrenia, depression, and Alzheimers disease. Herein we describe the discovery of potent GluN2A-selective NMDAR positive allosteric modulators (PAMs) starting from a high-throughput screening hit. Using structure-based design, we sought to increase potency at the GluN2A subtype, while improving selectivity against related α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). The structure-activity relationship of channel deactivation kinetics was studied using a combination of electrophysiology and protein crystallography. Effective incorporation of these strategies resulted in the discovery of GNE-0723 (46), a highly potent and brain penetrant GluN2A-selective NMDAR PAM suitable for in vivo characterization.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of novel pyrazolo[1,5-a]pyrimidines as potent pan-Pim inhibitors by structure- and property-based drug design.

Xiaojing Wang; Steven Magnuson; Richard Pastor; E Fan; Huiyong Hu; Tsui; W Deng; Jeremy Murray; M Steffek; Heidi J.A. Wallweber; John Moffat; Jason Drummond; G.K Chan; E Harstad; Allen Ebens

Pim kinases are promising targets for the development of cancer therapeutics. Among the three Pim isoforms, Pim-2 is particularly important in multiple myeloma, yet is the most difficult to inhibit due to its high affinity for ATP. We identified compound 1 via high throughput screening. Using property-based drug design and co-crystal structures with Pim-1 kinase to guide analog design, we were able to improve potency against all three Pim isoforms including a significant 10,000-fold gain against Pim-2. Compound 17 is a novel lead with low picomolar potency on all three Pim kinase isoforms.

Collaboration


Dive into the Heidi J.A. Wallweber's collaboration.

Researchain Logo
Decentralizing Knowledge