Try a new search

Format these results:

Searched for:

person:hubbas01

in-biosketch:yes

Total Results:

46


Structural basis of Janus kinase trans-activation

Caveney, Nathanael A; Saxton, Robert A; Waghray, Deepa; Glassman, Caleb R; Tsutsumi, Naotaka; Hubbard, Stevan R; Garcia, K Christopher
Janus kinases (JAKs) mediate signal transduction downstream of cytokine receptors. Cytokine-dependent dimerization is conveyed across the cell membrane to drive JAK dimerization, trans-phosphorylation, and activation. Activated JAKs in turn phosphorylate receptor intracellular domains (ICDs), resulting in the recruitment, phosphorylation, and activation of signal transducer and activator of transcription (STAT)-family transcription factors. The structural arrangement of a JAK1 dimer complex with IFNλR1 ICD was recently elucidated while bound by stabilizing nanobodies. While this revealed insights into the dimerization-dependent activation of JAKs and the role of oncogenic mutations in this process, the tyrosine kinase (TK) domains were separated by a distance not compatible with the trans-phosphorylation events between the TK domains. Here, we report the cryoelectron microscopy structure of a mouse JAK1 complex in a putative trans-activation state and expand these insights to other physiologically relevant JAK complexes, providing mechanistic insight into the crucial trans-activation step of JAK signaling and allosteric mechanisms of JAK inhibition.
PMID: 36867534
ISSN: 2211-1247
CID: 5466782

Identification of Novel Small Molecule Ligands for JAK2 Pseudokinase Domain

Virtanen, Anniina T.; Haikarainen, Teemu; Sampathkumar, Parthasarathy; Palmroth, Maaria; Liukkonen, Sanna; Liu, Jianping; Nekhotiaeva, Natalia; Hubbard, Stevan R.; Silvennoinen, Olli
Hyperactive mutation V617F in the JAK2 regulatory pseudokinase domain (JH2) is prevalent in patients with myeloproliferative neoplasms. Here, we identified novel small molecules that target JH2 of JAK2 V617F and characterized binding via biochemical and structural approaches. Screening of 107,600 small molecules resulted in identification of 55 binders to the ATP-binding pocket of recombinant JAK2 JH2 V617F protein at a low hit rate of 0.05%, which indicates unique structural characteristics of the JAK2 JH2 ATP-binding pocket. Selected hits and structural analogs were further assessed for binding to JH2 and JH1 (kinase) domains of JAK family members (JAK1-3, TYK2) and for effects on MPN model cell viability. Crystal structures were determined with JAK2 JH2 wild-type and V617F. The JH2-selective binders were identified in diaminotriazole, diaminotriazine, and phenylpyrazolo-pyrimidone chemical entities, but they showed low-affinity, and no inhibition of MPN cells was detected, while compounds binding to both JAK2 JH1 and JH2 domains inhibited MPN cell viability. X-ray crystal structures of protein-ligand complexes indicated generally similar binding modes between the ligands and V617F or wild-type JAK2. Ligands of JAK2 JH2 V617F are applicable as probes in JAK-STAT research, and SAR optimization combined with structural insights may yield higher-affinity inhibitors with biological activity.
SCOPUS:85146766468
ISSN: 1424-8247
CID: 5423862

Unlocking the secrets to Janus kinase activation

Levine, Ross L; Hubbard, Stevan R
The full-length structure of a Janus kinase provides insights for drug development.
PMID: 35389811
ISSN: 1095-9203
CID: 5201682

Increased resistance of SARS-CoV-2 Omicron variant to neutralization by vaccine-elicited and therapeutic antibodies

Tada, Takuya; Zhou, Hao; Dcosta, Belinda M; Samanovic, Marie I; Chivukula, Vidya; Herati, Ramin S; Hubbard, Stevan R; Mulligan, Mark J; Landau, Nathaniel R
BACKGROUND:SARS-CoV-2 vaccines currently authorized for emergency use have been highly successful in preventing infection and lessening disease severity. The vaccines maintain effectiveness against earlier SARS-CoV-2 Variants of Concern but the heavily mutated, highly transmissible Omicron variant presents an obstacle both to vaccine protection and monoclonal antibody therapies. METHODS:Pseudotyped lentiviruses were incubated with serum from vaccinated and boosted donors or therapeutic monoclonal antibody and then applied to target cells. After 2 days, luciferase activity was measured in a microplate luminometer. Resistance mutations of the Omicron spike were identified using point-mutated spike protein pseudotypes and mapped onto the three-dimensional spike protein structure. FINDINGS/RESULTS:Virus with the Omicron spike protein was 26-fold resistant to neutralization by recovered donor sera and 26-34-fold resistance to Pfizer BNT162b2 and Moderna vaccine-elicited antibodies following two immunizations. A booster immunization increased neutralizing titres against Omicron. Neutralizing titres against Omicron were increased in the sera with a history of prior SARS-CoV-2 infection. Analysis of the therapeutic monoclonal antibodies showed that the Regeneron and Eli Lilly monoclonal antibodies were ineffective against the Omicron pseudotype while Sotrovimab and Evusheld were partially effective. INTERPRETATION/CONCLUSIONS:The results highlight the benefit of a booster immunization to protect against the Omicron variant and demonstrate the challenge to monoclonal antibody therapy. The decrease in neutralizing titres against Omicron suggest that much of the vaccine efficacy may rely on T cells. FUNDING/BACKGROUND:The work was funded by grants from the NIH to N.R.L. (DA046100, AI122390 and AI120898) and 55 to M.J.M. (UM1AI148574).
PMCID:9021600
PMID: 35465948
ISSN: 2352-3964
CID: 5205452

Mechanism of homodimeric cytokine receptor activation and dysregulation by oncogenic mutations

Wilmes, Stephan; Hafer, Maximillian; Vuorio, Joni; Tucker, Julie A; Winkelmann, Hauke; Löchte, Sara; Stanly, Tess A; Pulgar Prieto, Katiuska D; Poojari, Chetan; Sharma, Vivek; Richter, Christian P; Kurre, Rainer; Hubbard, Stevan R; Garcia, K Christopher; Moraga, Ignacio; Vattulainen, Ilpo; Hitchcock, Ian S; Piehler, Jacob
Homodimeric class I cytokine receptors are assumed to exist as preformed dimers that are activated by ligand-induced conformational changes. We quantified the dimerization of three prototypic class I cytokine receptors in the plasma membrane of living cells by single-molecule fluorescence microscopy. Spatial and spatiotemporal correlation of individual receptor subunits showed ligand-induced dimerization and revealed that the associated Janus kinase 2 (JAK2) dimerizes through its pseudokinase domain. Oncogenic receptor and hyperactive JAK2 mutants promoted ligand-independent dimerization, highlighting the formation of receptor dimers as the switch responsible for signal activation. Atomistic modeling and molecular dynamics simulations based on a detailed energetic analysis of the interactions involved in dimerization yielded a mechanistic blueprint for homodimeric class I cytokine receptor activation and its dysregulation by individual mutations.
PMID: 32029621
ISSN: 1095-9203
CID: 4301542

KRAS4A directly regulates hexokinase 1

Amendola, Caroline R; Mahaffey, James P; Parker, Seth J; Ahearn, Ian M; Chen, Wei-Ching; Zhou, Mo; Court, Helen; Shi, Jie; Mendoza, Sebastian L; Morten, Michael J; Rothenberg, Eli; Gottlieb, Eyal; Wadghiri, Youssef Z; Possemato, Richard; Hubbard, Stevan R; Balmain, Allan; Kimmelman, Alec C; Philips, Mark R
The most frequently mutated oncogene in cancer is KRAS, which uses alternative fourth exons to generate two gene products (KRAS4A and KRAS4B) that differ only in their C-terminal membrane-targeting region1. Because oncogenic mutations occur in exons 2 or 3, two constitutively active KRAS proteins-each capable of transforming cells-are encoded when KRAS is activated by mutation2. No functional distinctions among the splice variants have so far been established. Oncogenic KRAS alters the metabolism of tumour cells3 in several ways, including increased glucose uptake and glycolysis even in the presence of abundant oxygen4 (the Warburg effect). Whereas these metabolic effects of oncogenic KRAS have been explained by transcriptional upregulation of glucose transporters and glycolytic enzymes3-5, it is not known whether there is direct regulation of metabolic enzymes. Here we report a direct, GTP-dependent interaction between KRAS4A and hexokinase 1 (HK1) that alters the activity of the kinase, and thereby establish that HK1 is an effector of KRAS4A. This interaction is unique to KRAS4A because the palmitoylation-depalmitoylation cycle of this RAS isoform enables colocalization with HK1 on the outer mitochondrial membrane. The expression of KRAS4A in cancer may drive unique metabolic vulnerabilities that can be exploited therapeutically.
PMID: 31827279
ISSN: 1476-4687
CID: 4234582

New Paradigms for the Mechanisms of Thrombopoietin Receptor Activation and Dysregulation By the JAK2XX Mutation [Meeting Abstract]

Wilmes, S; Hafer, M; Stanly, T A; Moraga, I; Vuorio, J; Poojari, C; Sharma, V; Lochte, S; Pulgar, Prieto K; Tucker, J A; Hubbard, S R; Garcia, K C; Vattulainen, I; Hitchcock, I; Piehler, J
Janus kinase (JAK2)V617F is the most common mutation found in patients with Philadelphia chromosome negative myeloproliferative neoplasms (Ph- MPNs). The discovery of this mutation over 15 years ago revolutionised MPN diagnosis and inspired the development of JAK inhibitors as new therapeutic interventions. However, despite extensive structural and biophysical studies using JAK2 domains in isolation, the exact molecular mechanisms of JAK2XX activation remains elusive. We have previously demonstrated that expression of the thrombopoietin (TPO) receptor, MPL, which interacts directly with JAK2, is essential for disease development in a mouse model of a JAK2XX-positiveMPN (Blood 2014 124:3956-3963). Using total internal reflection fluorescence (TIRF) microscopy, we visualized MPL interaction dynamics in live cells on single molecule level. Effective cell surface MPL fluorescence labelling and dual-color imaging allowed us to determine the level of MPL dimerization under various experimental conditions. Using this assay, we clearly established that MPL is monomeric at physiologically relevant receptor densities. However, TPO stimulation results in significant dimerization of MPL (>50%) and an equilibrium between monomers and dimers. This counters the current dogma that MPL exists at the membrane as a pre-formed dimer. Strikingly, we found that JAK2XX shifts this monomer-dimer equilibrium leading to significant TPO-independent MPL dimerization providing a novel mechanistic model of oncogenic JAK2 activation. To highlight the role of ligand-independent receptor dimerization in JAK2 activation, we compared three groups of autoactivating mutations in the PK domain covering the FERM-SH2 (FS2)-PK linker region (Group I), residues in the proximity of the alphaC helix (Group II) and at the autoinhibitory PK-TK interface (Group III). Consistent MPL dimerization was only observed for mutations in groups I and II. Mutations in these groups both localize to a potential homomeric PK/PK interface that has been implicated as a switch of JAK activation. Using MD simulations, we also found that the FERM domain of JAK2 strongly interacts with the inner leaflet of the lipid bilayer of the plasma membrane via a single hydrophobic residue (L224) surrounded by several positively charged residues that allows the region to act as a membrane anchor. This tight coupling to the membrane enforces an appropriate orientation between the JAKs within the receptor dimers required for optimal intermolecular PK/PK interaction that is critical for receptor dimerization. To interfere with membrane anchoring, we introduced a negative charge in this position (L224E). Strikingly, ligand-independent MPL dimerization and activation by JAK2XX was dramatically reduced upon introducing L224E, supporting the vital importance of L224 for orienting JAK2 at the membrane to allow productive PK-PK interactions. Here, we demonstrate that JAK2XX mutation acts by altering and strengthening the intermolecular interactions involving the PK/PK dimerization interface. In essence, these mutations drive cytoplasmic stabilization of receptor-JAK dimers, bypassing extracellular stabilization of dimers via cytokine binding. These results provide critical and entirely novel mechanistic insights into signal initiation in MPNs and readdress the roles of receptor-associated proteins. Disclosures: Hubbard: Ajax Therapeutics, Inc.: Membership on an entity's Board of Directors or advisory committees, Other: Co-Founder.XXCopyright
EMBASE:2013267819
ISSN: 0006-4971
CID: 4928162

Janus kinase 2 activation mechanisms revealed by analysis of suppressing mutations

Hammarén, Henrik M; Virtanen, Anniina T; Abraham, Bobin George; Peussa, Heidi; Hubbard, Stevan R; Silvennoinen, Olli
BACKGROUND:Janus kinases (JAK1-3, TYK2) mediate cytokine signals in the regulation of hematopoiesis and immunity. JAK2 clinical mutations cause myeloproliferative neoplasms and leukemia and the mutations strongly concentrate in the regulatory pseudokinase domain, JAK homology 2, JH2. Current clinical JAK inhibitors target the tyrosine kinase domain and lack mutation- and pathway-selectivity. OBJECTIVE:To characterize mechanisms and differences for pathogenic and cytokine-induced JAK2 activation to enable design of novel selective JAK inhibitors. METHODS:Systematic analysis of JAK2 activation requirements using structure-guided mutagenesis, cell signaling assays, microscopy, and biochemical analysis. RESULTS:Distinct structural requirements identified for activation of different pathogenic mutations. Specifically, the predominant JAK2 mutation V617F is the most sensitive to structural perturbations in multiple JH2 elements (C helix (αC), SH2-JH2 linker and ATP-binding site). In contrast, activation of K539L is resistant to most perturbations. Normal cytokine signaling shows distinct differences in activation requirements: JH2 ATP-binding site mutations have only a minor effect on signaling, while JH2 αC mutations reduce homomeric (JAK2-JAK2) EPO signaling, and almost completely abrogate heteromeric (JAK2-JAK1) IFNγ signaling, potentially by disrupting a dimerization interface on JH2. CONCLUSIONS:These results suggest that therapeutic approaches targeting the JH2 ATP-binding site and αC could be effective in inhibiting most pathogenic mutations. JH2 ATP-site targeting have potential for reduced side-effects by retaining EPO and IFNγ functions. Simultaneously, however, we identify the JH2 αC interface as a potential target for pathway-selective JAK inhibitors in diseases with unmutated JAK2, thus providing new insights for the development of novel pharmacological interventions.
PMID: 30092288
ISSN: 1097-6825
CID: 3226652

Crystal structure of the C-terminal four-helix bundle of the potassium channel KCa3.1

Ji, Tianyang; Corbalán-García, Senena; Hubbard, Stevan R
KCa3.1 (also known as SK4 or IK1) is a mammalian intermediate-conductance potassium channel that plays a critical role in the activation of T cells, B cells, and mast cells, effluxing potassium ions to maintain a negative membrane potential for influxing calcium ions. KCa3.1 shares primary sequence similarity with three other (low-conductance) potassium channels: KCa2.1, KCa2.2, and KCa2.3 (also known as SK1-3). These four homotetrameric channels bind calmodulin (CaM) in the cytoplasmic region, and calcium binding to CaM triggers channel activation. Unique to KCa3.1, activation also requires phosphorylation of a single histidine residue, His358, in the cytoplasmic region, which relieves copper-mediated inhibition of the channel. Near the cytoplasmic C-terminus of KCa3.1 (and KCa2.1-2.3), secondary-structure analysis predicts the presence of a coiled-coil/heptad repeat. Here, we report the crystal structure of the C-terminal coiled-coil region of KCa3.1, which forms a parallel four-helix bundle, consistent with the tetrameric nature of the channel. Interestingly, the four copies of a histidine residue, His389, in an 'a' position within the heptad repeat, are observed to bind a copper ion along the four-fold axis of the bundle. These results suggest that His358, the inhibitory histidine in KCa3.1, might coordinate a copper ion through a similar binding mode.
PMCID:6023178
PMID: 29953543
ISSN: 1932-6203
CID: 3161962

Mechanistic Insights into Regulation of JAK2 Tyrosine Kinase

Hubbard, Stevan R
JAK2 is a member of the Janus kinase (JAKs) family of non-receptor protein tyrosine kinases, which includes JAK1-3 and TYK2. JAKs serve as the cytoplasmic signaling components of cytokine receptors and are activated through cytokine-mediated trans-phosphorylation, which leads to receptor phosphorylation and recruitment and phosphorylation of signal transducer and activator of transcription (STAT) proteins. JAKs are unique among tyrosine kinases in that they possess a pseudokinase domain, which is just upstream of the C-terminal tyrosine kinase domain. A wealth of biochemical and clinical data have established that the pseudokinase domain of JAKs is crucial for maintaining a low basal (absence of cytokine) level of tyrosine kinase activity. In particular, gain-of-function mutations in the JAK genes, most frequently, V617F in the pseudokinase domain of JAK2, have been mapped in patients with blood disorders, including myeloproliferative neoplasms and leukemias. Recent structural and biochemical studies have begun to decipher the molecular mechanisms that maintain the basal, low-activity state of JAKs and that, via mutation, lead to constitutive activity and disease. This review will examine these mechanisms and describe how this knowledge could potentially inform drug development efforts aimed at obtaining a mutant (V617F)-selective inhibitor of JAK2.
PMCID:5770812
PMID: 29379470
ISSN: 1664-2392
CID: 2933322