Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(1): e23355, 2024 01.
Article in English | MEDLINE | ID: mdl-38071609

ABSTRACT

Drosophila melanogaster (fruit fly) insulin receptor (D-IR) is highly homologous to the human counterpart. Like the human pathway, D-IR responds to numerous insulin-like peptides to activate cellular signals that regulate growth, development, and lipid metabolism in fruit flies. Allelic mutations in the D-IR kinase domain elevate life expectancy in fruit flies. We developed a robust heterologous expression system to express and purify wild-type and longevity-associated mutant D-IR kinase domains to investigate enzyme kinetics and substrate specificities. D-IR exhibits remarkable similarities to the human insulin receptor kinase domain but diverges in substrate preferences. We show that longevity-associated mutations reduce D-IR catalytic activity. Deletion of the unique kinase insert domain portion or mutations proximal to activating tyrosines do not influence kinase activity, suggesting their potential role in substrate recruitment and downstream signaling. Through biochemical investigations, this study enhances our comprehension of D-IR's role in Drosophila physiology, complementing genetic studies and expanding our knowledge on the catalytic functions of this conserved signaling pathway.


Subject(s)
Drosophila Proteins , Drosophila , Humans , Animals , Drosophila/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Drosophila melanogaster/metabolism , Longevity/genetics , Signal Transduction/physiology , Insulin/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
2.
Cell Chem Biol ; 30(6): 618-631.e12, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37290440

ABSTRACT

Recurrent JAK2 alterations are observed in myeloproliferative neoplasms, B-cell acute lymphoblastic leukemia, and other hematologic malignancies. Currently available type I JAK2 inhibitors have limited activity in these diseases. Preclinical data support the improved efficacy of type II JAK2 inhibitors, which lock the kinase in the inactive conformation. By screening small molecule libraries, we identified a lead compound with JAK2 selectivity. We highlight analogs with on-target biochemical and cellular activity and demonstrate in vivo activity using a mouse model of polycythemia vera. We present a co-crystal structure that confirms the type II binding mode of our compounds with the "DFG-out" conformation of the JAK2 activation loop. Finally, we identify a JAK2 G993A mutation that confers resistance to the type II JAK2 inhibitor CHZ868 but not to our analogs. These data provide a template for identifying novel type II kinase inhibitors and inform further development of agents targeting JAK2 that overcome resistance.


Subject(s)
Myeloproliferative Disorders , Humans , Mutation , Myeloproliferative Disorders/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism
3.
Cell Rep ; 42(3): 112201, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36867534

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins , Janus Kinases , Animals , Mice , Janus Kinases/metabolism , DNA-Binding Proteins/metabolism , Cryoelectron Microscopy , Trans-Activators/metabolism , Janus Kinase 1/metabolism , Signal Transduction , Phosphorylation , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism
4.
Pharmaceuticals (Basel) ; 16(1)2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36678572

ABSTRACT

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.

5.
EBioMedicine ; 78: 103944, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35465948

ABSTRACT

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: 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: 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: The work was funded by grants from the NIH to N.R.L. (DA046100, AI122390 and AI120898) and 55 to M.J.M. (UM1AI148574).


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Spike Glycoprotein, Coronavirus/genetics , Vaccination
6.
Science ; 376(6589): 139-140, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35389811

ABSTRACT

The full-length structure of a Janus kinase provides insights for drug development.


Subject(s)
Drug Development , Enzyme Activation , Janus Kinase 2 , Janus Kinases
7.
Science ; 367(6478): 643-652, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32029621

ABSTRACT

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.


Subject(s)
Carcinogenesis/genetics , Cell Membrane/chemistry , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Protein Multimerization , Receptors, Erythropoietin/chemistry , Receptors, Somatotropin/chemistry , Receptors, Thrombopoietin/chemistry , Amino Acid Substitution/genetics , HeLa Cells , Humans , Janus Kinase 2/antagonists & inhibitors , Ligands , Microscopy, Fluorescence , Models, Molecular , Mutation , Nitriles , Phenylalanine/genetics , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction , Single Molecule Imaging , Valine/genetics
8.
Nature ; 576(7787): 482-486, 2019 12.
Article in English | MEDLINE | ID: mdl-31827279

ABSTRACT

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.


Subject(s)
Hexokinase/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Allosteric Regulation , Animals , Cell Line, Tumor , Enzyme Activation , Glycolysis , Guanosine Triphosphate/metabolism , Hexokinase/chemistry , Humans , In Vitro Techniques , Isoenzymes/metabolism , Lipoylation , Male , Mice , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Neoplasms/enzymology , Neoplasms/metabolism , Protein Binding , Protein Transport
9.
J Allergy Clin Immunol ; 143(4): 1549-1559.e6, 2019 04.
Article in English | MEDLINE | ID: mdl-30092288

ABSTRACT

BACKGROUND: Janus kinases (JAKs; JAK1 to JAK3 and tyrosine kinase 2) 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 Janus kinase homology (JH) 2. Current clinical JAK inhibitors target the tyrosine kinase domain and lack mutation and pathway selectivity. OBJECTIVE: We sought to characterize mechanisms and differences for pathogenic and cytokine-induced JAK2 activation to enable design of novel selective JAK inhibitors. METHODS: We performed a systematic analysis of JAK2 activation requirements using structure-guided mutagenesis, cell-signaling assays, microscopy, and biochemical analysis. RESULTS: Distinct structural requirements were 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], Src homology 2-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, whereas JH2 αC mutations reduce homomeric (JAK2-JAK2) erythropoietin signaling and almost completely abrogate heteromeric (JAK2-JAK1) IFN-γ 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 has the potential for reduced side effects by retaining erythropoietin and IFN-γ functions. Simultaneously, however, we identified the JH2 αC interface as a potential target for pathway-selective JAK inhibitors in patients with diseases with unmutated JAK2, thus providing new insights into the development of novel pharmacologic interventions.


Subject(s)
Enzyme Activation/physiology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , DNA Mutational Analysis , Drug Discovery , Enzyme Inhibitors/pharmacology , Humans , Janus Kinase 2/chemistry , Janus Kinase Inhibitors , Models, Molecular , Protein Conformation , Protein Domains
10.
PLoS One ; 13(10): e0205540, 2018.
Article in English | MEDLINE | ID: mdl-30286164

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0199942.].

11.
PLoS One ; 13(6): e0199942, 2018.
Article in English | MEDLINE | ID: mdl-29953543

ABSTRACT

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.


Subject(s)
Copper/chemistry , Intermediate-Conductance Calcium-Activated Potassium Channels/chemistry , Crystallography, X-Ray , Humans , Protein Domains , Protein Structure, Secondary
12.
Article in English | MEDLINE | ID: mdl-29379470

ABSTRACT

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.

13.
J Clin Invest ; 126(11): 4361-4371, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27760050

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a risk factor for type 2 diabetes (T2D), but whether NAFLD plays a causal role in the pathogenesis of T2D is uncertain. One proposed mechanism linking NAFLD to hepatic insulin resistance involves diacylglycerol-mediated (DAG-mediated) activation of protein kinase C-ε (PKCε) and the consequent inhibition of insulin receptor (INSR) kinase activity. However, the molecular mechanism underlying PKCε inhibition of INSR kinase activity is unknown. Here, we used mass spectrometry to identify the phosphorylation site Thr1160 as a PKCε substrate in the functionally critical INSR kinase activation loop. We hypothesized that Thr1160 phosphorylation impairs INSR kinase activity by destabilizing the active configuration of the INSR kinase, and our results confirmed this prediction by demonstrating severely impaired INSR kinase activity in phosphomimetic T1160E mutants. Conversely, the INSR T1160A mutant was not inhibited by PKCε in vitro. Furthermore, mice with a threonine-to-alanine mutation at the homologous residue Thr1150 (InsrT1150A mice) were protected from high fat diet-induced hepatic insulin resistance. InsrT1150A mice also displayed increased insulin signaling, suppression of hepatic glucose production, and increased hepatic glycogen synthesis compared with WT controls during hyperinsulinemic clamp studies. These data reveal a critical pathophysiological role for INSR Thr1160 phosphorylation and provide further mechanistic links between PKCε and INSR in mediating NAFLD-induced hepatic insulin resistance.


Subject(s)
Dietary Fats/adverse effects , Insulin Resistance , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Amino Acid Substitution , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Dietary Fats/pharmacology , Glycogen/biosynthesis , Glycogen/genetics , Liver/pathology , Mice , Mice, Mutant Strains , Mutation, Missense , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Phosphorylation , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Receptor, Insulin/genetics
14.
Elife ; 52016 08 19.
Article in English | MEDLINE | ID: mdl-27542194

ABSTRACT

KCa2.1, KCa2.2, KCa2.3 and KCa3.1 constitute a family of mammalian small- to intermediate-conductance potassium channels that are activated by calcium-calmodulin. KCa3.1 is unique among these four channels in that activation requires, in addition to calcium, phosphorylation of a single histidine residue (His358) in the cytoplasmic region, by nucleoside diphosphate kinase-B (NDPK-B). The mechanism by which KCa3.1 is activated by histidine phosphorylation is unknown. Histidine phosphorylation is well characterized in prokaryotes but poorly understood in eukaryotes. Here, we demonstrate that phosphorylation of His358 activates KCa3.1 by antagonizing copper-mediated inhibition of the channel. Furthermore, we show that activated CD4(+) T cells deficient in intracellular copper exhibit increased KCa3.1 histidine phosphorylation and channel activity, leading to increased calcium flux and cytokine production. These findings reveal a novel regulatory mechanism for a mammalian potassium channel and for T-cell activation, and highlight a unique feature of histidine versus serine/threonine and tyrosine as a regulatory phosphorylation site.


Subject(s)
Copper/metabolism , Enzyme Inhibitors/metabolism , Histidine/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Mice , Nucleoside-Diphosphate Kinase/metabolism , Patch-Clamp Techniques , Phosphorylation
15.
Cancer Cell ; 28(1): 1-2, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26175407

ABSTRACT

Activating JAK2 mutants cause hematological malignancies. Current clinical type I JAK2 inhibitors effectively relieve symptoms but fail to resolve the disease. In this issue of Cancer Cell, two articles by Wu and colleagues and Meyer and colleagues characterize a type II JAK2 inhibitor that is effective in preclinical models of JAK2-dependent myeloproliferative neoplasms and B cell acute lymphoblastic leukemia.


Subject(s)
Aminopyridines/administration & dosage , Antineoplastic Agents/administration & dosage , Benzimidazoles/administration & dosage , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm/drug effects , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Myeloproliferative Disorders/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/administration & dosage , Animals , Humans
16.
Blood ; 125(22): 3388-92, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25824690

ABSTRACT

The critical role of Janus kinase-2 (JAK2) in regulation of myelopoiesis was established 2 decades ago, but identification of mutations in the pseudokinase domain of JAK2 in myeloproliferative neoplasms (MPNs) and in other hematologic malignancies highlighted the role of JAK2 in human disease. These findings have revolutionized the diagnostics of MPNs and led to development of novel JAK2 therapeutics. However, the molecular mechanisms by which mutations in the pseudokinase domain lead to hyperactivation of JAK2 and clinical disease have been unclear. Here, we describe recent advances in the molecular characterization of the JAK2 pseudokinase domain and how pathogenic mutations lead to constitutive activation of JAK2.


Subject(s)
Hematologic Neoplasms/genetics , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Animals , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Homeostasis/genetics , Humans , Janus Kinase 2/chemistry , Models, Molecular , Mutation , Protein Structure, Tertiary/genetics
17.
Proc Natl Acad Sci U S A ; 112(15): 4642-7, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25825724

ABSTRACT

Pseudokinases lack conserved motifs typically required for kinase activity. Nearly half of pseudokinases bind ATP, but only few retain phosphotransfer activity, leaving the functional role of nucleotide binding in most cases unknown. Janus kinases (JAKs) are nonreceptor tyrosine kinases with a tandem pseudokinase-kinase domain configuration, where the pseudokinase domain (JAK homology 2, JH2) has important regulatory functions and harbors mutations underlying hematological and immunological diseases. JH2 of JAK1, JAK2, and TYK2 all bind ATP, but the significance of this is unclear. We characterize the role of nucleotide binding in normal and pathogenic JAK signaling using comprehensive structure-based mutagenesis. Disruption of JH2 ATP binding in wild-type JAK2 has only minor effects, and in the presence of type I cytokine receptors, the mutations do not affect JAK2 activation. However, JH2 mutants devoid of ATP binding ameliorate the hyperactivation of JAK2 V617F. Disrupting ATP binding in JH2 also inhibits the hyperactivity of other pathogenic JAK2 mutants, as well as of JAK1 V658F, and prevents induction of erythrocytosis in a JAK2 V617F myeloproliferative neoplasm mouse model. Molecular dynamic simulations and thermal-shift analysis indicate that ATP binding stabilizes JH2, with a pronounced effect on the C helix region, which plays a critical role in pathogenic activation of JAK2. Taken together, our results suggest that ATP binding to JH2 serves a structural role in JAKs, which is required for aberrant activity of pathogenic JAK mutants. The inhibitory effect of abrogating JH2 ATP binding in pathogenic JAK mutants may warrant novel therapeutic approaches.


Subject(s)
Adenosine Triphosphate/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mutation, Missense , Adenosine Triphosphate/chemistry , Animals , Binding Sites/genetics , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Activation/genetics , Female , Humans , Immunoblotting , Janus Kinase 2/chemistry , Mice, Inbred C57BL , Molecular Dynamics Simulation , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Erythropoietin/metabolism
18.
Mol Cell ; 57(6): 984-994, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25728768

ABSTRACT

One of several roles of the Mycobacterium tuberculosis proteasome is to defend against host-produced nitric oxide (NO), a free radical that can damage numerous biological macromolecules. Mutations that inactivate proteasomal degradation in Mycobacterium tuberculosis result in bacteria that are hypersensitive to NO and attenuated for growth in vivo, but it was not known why. To elucidate the link between proteasome function, NO resistance, and pathogenesis, we screened for suppressors of NO hypersensitivity in a mycobacterial proteasome ATPase mutant and identified mutations in Rv1205. We determined that Rv1205 encodes a pupylated proteasome substrate. Rv1205 is a homolog of the plant enzyme LONELY GUY, which catalyzes the production of hormones called cytokinins. Remarkably, we report that an obligate human pathogen secretes several cytokinins. Finally, we determined that the Rv1205-dependent accumulation of cytokinin breakdown products is likely responsible for the sensitization of Mycobacterium tuberculosis proteasome-associated mutants to NO.


Subject(s)
Aminohydrolases/metabolism , Cytokinins/biosynthesis , Mycobacterium tuberculosis/metabolism , Nitric Oxide/metabolism , Proteasome Endopeptidase Complex/metabolism , Aldehydes/metabolism , Aminohydrolases/genetics , Animals , Arabidopsis Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cytokinins/metabolism , Host-Pathogen Interactions , Mice, Inbred C57BL , Mutation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Nitric Oxide/pharmacology , Suppression, Genetic
19.
Nat Commun ; 6: 6406, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25758790

ABSTRACT

The insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R) are highly related receptor tyrosine kinases with a disulfide-linked homodimeric architecture. Ligand binding to the receptor ectodomain triggers tyrosine autophosphorylation of the cytoplasmic domains, which stimulates catalytic activity and creates recruitment sites for downstream signalling proteins. Whether the two phosphorylated tyrosine kinase domains within the receptor dimer function independently or cooperatively to phosphorylate protein substrates is not known. Here we provide crystallographic, biophysical and biochemical evidence demonstrating that the phosphorylated kinase domains of IR and IGF1R form a specific dimeric arrangement involving an exchange of the juxtamembrane region proximal to the kinase domain. In this dimer, the active position of α-helix C in the kinase N lobe is stabilized, which promotes downstream substrate phosphorylation. These studies afford a novel strategy for the design of small-molecule IR agonists as potential therapeutic agents for type 2 diabetes.


Subject(s)
Antigens, CD/chemistry , Insulin/chemistry , Receptor, Insulin/chemistry , Receptors, Somatomedin/chemistry , Animals , Antigens, CD/genetics , Baculoviridae/genetics , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Models, Molecular , Phosphorylation , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Receptor, IGF Type 1 , Receptor, Insulin/genetics , Receptors, Somatomedin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sf9 Cells , Spodoptera , Unilamellar Liposomes/chemistry
20.
Elife ; 32014 Oct 28.
Article in English | MEDLINE | ID: mdl-25350976

ABSTRACT

When insulin-like growth factor-1 (IGF1) binds to its receptor, a physical constraint is released that allows the two transmembrane helices to come together to facilitate activation of the receptor.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Amino Acid Substitution , Humans , Insulin/metabolism , Models, Biological , Receptor, Insulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...