Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Cancer Discov ; 13(8): 1922-1947, 2023 08 04.
Article En | MEDLINE | ID: mdl-37191437

Leukemia stem cells (LSC) possess distinct self-renewal and arrested differentiation properties that are responsible for disease emergence, therapy failure, and recurrence in acute myeloid leukemia (AML). Despite AML displaying extensive biological and clinical heterogeneity, LSC with high interleukin-3 receptor (IL3R) levels are a constant yet puzzling feature, as this receptor lacks tyrosine kinase activity. Here, we show that the heterodimeric IL3Rα/ßc receptor assembles into hexamers and dodecamers through a unique interface in the 3D structure, where high IL3Rα/ßc ratios bias hexamer formation. Importantly, receptor stoichiometry is clinically relevant as it varies across the individual cells in the AML hierarchy, in which high IL3Rα/ßc ratios in LSCs drive hexamer-mediated stemness programs and poor patient survival, while low ratios mediate differentiation. Our study establishes a new paradigm in which alternative cytokine receptor stoichiometries differentially regulate cell fate, a signaling mechanism that may be generalizable to other transformed cellular hierarchies and of potential therapeutic significance. SIGNIFICANCE: Stemness is a hallmark of many cancers and is largely responsible for disease emergence, progression, and relapse. Our finding that clinically significant stemness programs in AML are directly regulated by different stoichiometries of cytokine receptors represents a hitherto unexplained mechanism underlying cell-fate decisions in cancer stem cell hierarchies. This article is highlighted in the In This Issue feature, p. 1749.


Leukemia, Myeloid, Acute , Receptors, Cytokine , Humans , Receptors, Cytokine/therapeutic use , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Phosphorylation , Signal Transduction , Cell Proliferation , Neoplastic Stem Cells
2.
J Vet Intern Med ; 37(1): 80-91, 2023 Jan.
Article En | MEDLINE | ID: mdl-36645022

BACKGROUND: Ionized calcium concentration ([iCa]) is more sensitive for detecting calcium disturbances than serum total calcium concentration but literature on ionized hypercalcemia in cats is limited. Urolithiasis is a possible adverse consequence of hypercalcemia. HYPOTHESIS/OBJECTIVES: To describe clinical details of diagnoses associated with ionized hypercalcemia in cats and association with urolithiasis. ANIMALS: Cats (238) seen between 2009 and 2019 at a referral hospital with [iCa] above the normal reference interval. METHODS: Observational cross-sectional study. Signalment, serum biochemical and imaging findings were reviewed for cats with ionized hypercalcemia considered to be clinically relevant (>1.41 mmol/L). Data were summarized by cause of hypercalcemia (i.e., diagnosis). RESULTS: Diagnoses for the 238 cats with [iCa] >1.41 mmol/L included: acute kidney injury (AKI; 13%), malignancy-associated (10.1%), idiopathic hypercalcemia (IHC; 10.1%), chronic kidney disease/renal diet-associated (8.4%), iatrogenic (5.5%), primary hyperparathyroidism (2.1%), vitamin D toxicity (2.1%) and granulomatous disease (1.7%). In 112 cases (47.1%), no cause for ionized hypercalcemia could be determined (n = 95), hypercalcemia was transient (n = 12), or the cat was juvenile (<1 year; n = 5). Urolithiasis was identified in 83.3% of AKI, 72.7% of iatrogenic, 61.1% of CKD/renal diet-associated and 50% of IHC cases that were imaged (<50% for other diagnoses). Diagnoses with a high proportion of concurrent total hypercalcemia included primary hyperparathyroidism (100%), vitamin D toxicity (100%), malignancy-associated (71.4%), granulomatous disease (66.7%) and IHC (65.2%). CONCLUSIONS AND CLINICAL IMPORTANCE: Ionized hypercalcemia was most commonly associated with kidney diseases, neoplasia or IHC. The proportion of urolithiasis cases varied by diagnosis.


Acute Kidney Injury , Cat Diseases , Hypercalcemia , Hyperparathyroidism, Primary , Neoplasms , Renal Insufficiency, Chronic , Urolithiasis , Cats , Animals , Hypercalcemia/etiology , Hypercalcemia/veterinary , Calcium , Hyperparathyroidism, Primary/veterinary , Cross-Sectional Studies , Renal Insufficiency, Chronic/veterinary , Neoplasms/veterinary , Urolithiasis/complications , Urolithiasis/diagnosis , Urolithiasis/veterinary , Acute Kidney Injury/complications , Acute Kidney Injury/veterinary , Iatrogenic Disease/veterinary , Vitamin D , Cat Diseases/diagnosis , Cat Diseases/etiology
3.
Sci Rep ; 9(1): 7851, 2019 May 21.
Article En | MEDLINE | ID: mdl-31110193

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

4.
Sci Rep ; 8(1): 12457, 2018 08 20.
Article En | MEDLINE | ID: mdl-30127368

A direct interaction between the erythropoietin (EPOR) and the beta-common (ßc) receptors to form an Innate Repair Receptor (IRR) is controversial. On one hand, studies have shown a functional link between EPOR and ßc receptor in tissue protection while others have shown no involvement of the ßc receptor in tissue repair. To date there is no biophysical evidence to confirm a direct association of the two receptors either in vitro or in vivo. We investigated the existence of an interaction between the extracellular regions of EPOR and the ßc receptor in silico and in vitro (either in the presence or absence of EPO or EPO-derived peptide ARA290). Although a possible interaction between EPOR and ßc was suggested by our computational and genomic studies, our in vitro biophysical analysis demonstrates that the extracellular regions of the two receptors do not specifically associate. We also explored the involvement of the ßc receptor gene (Csf2rb) under anaemic stress conditions and found no requirement for the ßc receptor in mice. In light of these studies, we conclude that the extracellular regions of the EPOR and the ßc receptor do not directly interact and that the IRR is not involved in anaemic stress.

5.
MAbs ; 10(7): 1018-1029, 2018 10.
Article En | MEDLINE | ID: mdl-29969365

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that can stimulate a variety of cells, but its overexpression leads to excessive production and activation of granulocytes and macrophages with many pathogenic effects. This cytokine is a therapeutic target in inflammatory diseases, and several anti-GM-CSF antibodies have advanced to Phase 2 clinical trials in patients with such diseases, e.g., rheumatoid arthritis. GM-CSF is also an essential factor in preventing pulmonary alveolar proteinosis (PAP), a disease associated with GM-CSF malfunction arising most typically through the presence of GM-CSF neutralizing auto-antibodies. Understanding the mechanism of action for neutralizing antibodies that target GM-CSF is important for improving their specificity and affinity as therapeutics and, conversely, in devising strategies to reduce the effects of GM-CSF auto-antibodies in PAP. We have solved the crystal structures of human GM-CSF bound to antigen-binding fragments of two neutralizing antibodies, the human auto-antibody F1 and the mouse monoclonal antibody 4D4. Coordinates and structure factors of the crystal structures of the GM-CSF:F1 Fab and the GM-CSF:4D4 Fab complexes have been deposited in the RCSB Protein Data Bank under the accession numbers 6BFQ and 6BFS, respectively. The structures show that these antibodies bind to mutually exclusive epitopes on GM-CSF; however, both prevent the cytokine from interacting with its alpha receptor subunit and hence prevent receptor activation. Importantly, identification of the F1 epitope together with functional analyses highlighted modifications to GM-CSF that would abolish auto-antibody recognition whilst retaining GM-CSF function. These results provide a framework for developing novel GM-CSF molecules for PAP treatment and for optimizing current anti-GM-CSF antibodies for use in treating inflammatory disorders.


Antibodies, Neutralizing/chemistry , Antigen-Antibody Complex/chemistry , Arthritis, Rheumatoid/therapy , Autoantibodies/chemistry , Epitopes/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunotherapy/methods , Antibodies, Neutralizing/metabolism , Arthritis, Rheumatoid/immunology , Autoantibodies/metabolism , Autoantibodies/pharmacology , Crystallography, X-Ray , Cytokines/metabolism , Epitopes/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Molecular Structure , Protein Binding , Protein Conformation
6.
Nat Commun ; 9(1): 386, 2018 01 26.
Article En | MEDLINE | ID: mdl-29374162

The interleukin-3 (IL-3) receptor is a cell-surface heterodimer that links the haemopoietic, vascular and immune systems and is overexpressed in acute and chronic myeloid leukaemia progenitor cells. It belongs to the type I cytokine receptor family in which the α-subunits consist of two fibronectin III-like domains that bind cytokine, and a third, evolutionarily unrelated and topologically conserved, N-terminal domain (NTD) with unknown function. Here we show by crystallography that, while the NTD of IL3Rα is highly mobile in the presence of IL-3, it becomes surprisingly rigid in the presence of IL-3 K116W. Mutagenesis, biochemical and functional studies show that the NTD of IL3Rα regulates IL-3 binding and signalling and reveal an unexpected role in preventing spontaneous receptor dimerisation. Our work identifies a dual role for the NTD in this cytokine receptor family, protecting against inappropriate signalling and dynamically regulating cytokine receptor binding and function.


Interleukin-3 Receptor alpha Subunit/chemistry , Interleukin-3 Receptor alpha Subunit/metabolism , Protein Domains , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Crystallography, X-Ray , HEK293 Cells , Humans , Interleukin-3/chemistry , Interleukin-3/genetics , Interleukin-3/metabolism , Interleukin-3 Receptor alpha Subunit/genetics , Molecular Dynamics Simulation , Mutation , Protein Binding
7.
Article En | MEDLINE | ID: mdl-28716883

The ß common ([ßc]/CD131) family of cytokines comprises granulocyte macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-3, and IL-5, all of which use ßc as their key signaling receptor subunit. This is a prototypic signaling subunit-sharing cytokine family that has unveiled many biological paradigms and structural principles applicable to the IL-2, IL-4, and IL-6 receptor families, all of which also share one or more signaling subunits. Originally identified for their functions in the hematopoietic system, the ßc cytokines are now known to be truly pleiotropic, impacting on multiple cell types, organs, and biological systems, and thereby controlling the balance between health and disease. This review will focus on the emerging biological roles for the ßc cytokines, our progress toward understanding the mechanisms of receptor assembly and signaling, and the application of this knowledge to develop exciting new therapeutic approaches against human disease.


Cytokines/classification , Cytokines/metabolism , Cytokines/genetics , Gene Expression Regulation/physiology , Humans , Inflammation/metabolism , Sepsis/metabolism , Signal Transduction
8.
Structure ; 24(8): 1271-1281, 2016 08 02.
Article En | MEDLINE | ID: mdl-27396825

The GM-CSF, IL-3, and IL-5 receptors constitute the ßc family, playing important roles in inflammation, autoimmunity, and cancer. Typical of heterodimeric type I cytokine receptors, signaling requires recruitment of the shared subunit to the initial cytokine:α subunit binary complex through an affinity conversion mechanism. This critical process is poorly understood due to the paucity of crystal structures of both binary and ternary receptor complexes for the same cytokine. We have now solved the structure of the binary GM-CSF:GMRα complex at 2.8-Å resolution and compared it with the structure of the ternary complex, revealing distinct conformational changes. Guided by these differences we performed mutational and functional studies that, importantly, show GMRα interactions playing a major role in receptor signaling while ßc interactions control high-affinity binding. These results support the notion that conformational changes underlie the mechanism of GM-CSF receptor activation and also suggest how related type I cytokine receptors signal.


Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Protein Subunits/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Amino Acid Motifs , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
9.
Cytokine ; 74(2): 247-58, 2015 Aug.
Article En | MEDLINE | ID: mdl-25982846

Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and IL-5 are members of a small family of cytokines that share a beta receptor subunit (ßc). These cytokines regulate the growth, differentiation, migration and effector function activities of many hematopoietic cells in bone marrow, blood and sites of inflammation. Excessive or aberrant signaling can result in chronic inflammatory conditions and myeloid leukemias. The crystal structures of the GM-CSF ternary complex, the IL-5 binary complex and the very recent IL-3 receptor alpha subunit build upon decades of structure-function studies, giving new insights into cytokine-receptor specificity and signal transduction. Selective modulation of receptor function is now a real possibility and the structures of the ßc receptor family are being used to discover novel and disease-specific therapeutics.


Cytokine Receptor Common beta Subunit , Cytokines , Signal Transduction/immunology , Animals , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/immunology , Cytokines/chemistry , Cytokines/immunology , Humans , Protein Structure, Quaternary , Structure-Activity Relationship
10.
Cell Rep ; 8(2): 410-9, 2014 Jul 24.
Article En | MEDLINE | ID: mdl-25043189

Interleukin-3 (IL-3) is an activated T cell product that bridges innate and adaptive immunity and contributes to several immunopathologies. Here, we report the crystal structure of the IL-3 receptor α chain (IL3Rα) in complex with the anti-leukemia antibody CSL362 that reveals the N-terminal domain (NTD), a domain also present in the granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-5, and IL-13 receptors, adopting unique "open" and classical "closed" conformations. Although extensive mutational analyses of the NTD epitope of CSL362 show minor overlap with the IL-3 binding site, CSL362 only inhibits IL-3 binding to the closed conformation, indicating alternative mechanisms for blocking IL-3 signaling. Significantly, whereas "open-like" IL3Rα mutants can simultaneously bind IL-3 and CSL362, CSL362 still prevents the assembly of a higher-order IL-3 receptor-signaling complex. The discovery of open forms of cytokine receptors provides the framework for development of potent antibodies that can achieve a "double hit" cytokine receptor blockade.


Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/chemistry , Interleukin-3 Receptor alpha Subunit/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized/immunology , Antineoplastic Agents/metabolism , Binding Sites, Antibody , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Interleukin-3 Receptor alpha Subunit/immunology , Molecular Sequence Data , Protein Binding
11.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 3): 358-61, 2014 Mar.
Article En | MEDLINE | ID: mdl-24598927

Interleukin-3 (IL-3) is a member of the beta common family of cytokines that regulate multiple functions of myeloid cells. The IL-3 receptor-specific alpha subunit (IL3Rα) is overexpressed on stem cells/progenitor cells of patients with acute myeloid leukaemia, where elevated receptor expression correlates clinically with a reduced patient survival rate. The monoclonal antibody (MAb) CSL362 is a humanized MAb derived from the murine MAb 7G3, originally identified for its ability to specifically recognize the human IL-3 receptor and for blocking the signalling of IL-3 in myeloid and endothelial cells. In order to elucidate the molecular mechanism of CSL362 antagonism, a preliminary structure of human IL3Rα in complex with the MAb CSL362 has been determined.


Antibodies, Monoclonal, Humanized/chemistry , Interleukin-3 Receptor alpha Subunit/chemistry , Amino Acid Sequence , Crystallization , Humans , Molecular Sequence Data , Protein Binding , X-Ray Diffraction
12.
Mol Cell Oncol ; 1(4): e969129, 2014.
Article En | MEDLINE | ID: mdl-27308368

CSL362 is a humanized interleukin-3 (IL-3)-neutralizing monoclonal antibody with enhanced effector function that binds the α subunit of the IL-3 receptor (IL3Rα). The crystal structure of an IL3Rα:CSL362 complex shows that IL3Rα adopts "open" and "closed" conformations. CSL362 blocks IL-3 function through both IL3Rα conformations but via distinct and unexpected mechanisms.

13.
Cytokine Growth Factor Rev ; 24(3): 189-201, 2013 Jun.
Article En | MEDLINE | ID: mdl-23535386

The GM-CSF, IL-3 and IL-5 family of cytokines, also known as the ßc family due to their receptors sharing the signalling subunit ßc, regulates multiple biological processes such as native and adaptive immunity, inflammation, normal and malignant hemopoieis, and autoimmunity. Australian scientists played a major role in the discovery and biological characterisation of the ßc cytokines and their recent work is revealing unique features of cytokine receptor assembly and signalling. Furthermore, specific antibodies have been generated to modulate their function. Characterisation of the structural and dynamic requirements for the activation of the ßc receptor family and the molecular definition of downstream signalling pathways are providing new insights into cytokine receptor signalling as well as new therapeutic opportunities.


Cytokines/physiology , Signal Transduction/physiology , Animals , Autoimmunity/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Inflammation/physiopathology , Interleukin-3/physiology , Interleukin-5/physiology , Janus Kinases/metabolism , NF-kappa B/physiology , Receptors, Cytokine/physiology
14.
Immunol Rev ; 250(1): 277-302, 2012 Nov.
Article En | MEDLINE | ID: mdl-23046136

Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3), and IL-5 are members of a discrete family of cytokines that regulates the growth, differentiation, migration and effector function activities of many hematopoietic cells and immunocytes. These cytokines are involved in normal responses to infectious agents, bridging innate and adaptive immunity. However, in certain cases, the overexpression of these cytokines or their receptors can lead to excessive or aberrant initiation of signaling resulting in pathological conditions, with chronic inflammatory diseases and myeloid leukemias the most notable examples. Recent crystal structures of the GM-CSF receptor ternary complex and the IL-5 binary complex have revealed new paradigms of cytokine receptor activation. Together with a wealth of associated structure-function studies, they have significantly enhanced our understanding of how these receptors recognize cytokines and initiate signals across cell membranes. Importantly, these structures provide opportunities for structure-based approaches for the discovery of novel and disease-specific therapeutics. In addition, recent biochemical evidence has suggested that the GM-CSF/IL-3/IL-5 receptor family is capable of interacting productively with other membrane proteins at the cell surface. Such interactions may afford additional or unique biological activities and might be harnessed for selective modulation of the function of these receptors in disease.


Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Interleukin-3/chemistry , Interleukin-5/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Interleukin-3/chemistry , Receptors, Interleukin-5/chemistry , Crystallography, X-Ray , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-3/immunology , Interleukin-3/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Leukemia, Myeloid/immunology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Models, Molecular , Protein Binding , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Interleukin-3/immunology , Receptors, Interleukin-3/metabolism , Receptors, Interleukin-5/immunology , Receptors, Interleukin-5/metabolism , Signal Transduction , Structure-Activity Relationship
15.
Immunity ; 37(4): 611-21, 2012 Oct 19.
Article En | MEDLINE | ID: mdl-23063329

Celiac disease is a human leukocyte antigen (HLA)-DQ2- and/or DQ8-associated T cell-mediated disorder that is induced by dietary gluten. Although it is established how gluten peptides bind HLA-DQ8 and HLA-DQ2, it is unclear how such peptide-HLA complexes are engaged by the T cell receptor (TCR), a recognition event that triggers disease pathology. We show that biased TCR usage (TRBV9(∗)01) underpins the recognition of HLA-DQ8-α-I-gliadin. The structure of a prototypical TRBV9(∗)01-TCR-HLA-DQ8-α-I-gliadin complex shows that the TCR docks centrally above HLA-DQ8-α-I-gliadin, in which all complementarity-determining region-ß (CDRß) loops interact with the gliadin peptide. Mutagenesis at the TRBV9(∗)01-TCR-HLA-DQ8-α-I-gliadin interface provides an energetic basis for the Vß bias. Moreover, CDR3 diversity accounts for TRBV9(∗)01(+) TCRs exhibiting differing reactivities toward the gliadin epitopes at various deamidation states. Accordingly, biased TCR usage is an important factor in the pathogenesis of DQ8-mediated celiac disease.


Celiac Disease/immunology , Gliadin/immunology , HLA-DQ Antigens/immunology , Receptors, Antigen, T-Cell/immunology , Amino Acid Sequence , Epitopes, T-Lymphocyte/immunology , HLA-DQ Antigens/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Peptide Fragments/immunology , Protein Interaction Domains and Motifs , Receptors, Antigen, T-Cell/chemistry
16.
Curr Opin Struct Biol ; 22(3): 350-9, 2012 Jun.
Article En | MEDLINE | ID: mdl-22521507

Cytokines are well recognized for the pleiotropic nature of their signaling and biological activities on many cell types and their role in health and disease. Recent years have seen a steady stream of new cytokine receptor crystal structures including those that are activated by GM-CSF, type I interferon, and a variety of interleukins. Highlights include the observation of a dodecameric signaling complex for the GM-CSF receptor, electron microscopy imaging of an intact gp130/IL-6/IL-6Rα ternary receptor complex bound to its signal transducing Janus kinase and visualization of novel cytokine recognition mechanisms in the interleukin-17 and type I interferon families. This increasing knowledge in cytokine structural biology is driving new opportunities for developing novel therapies to modulate cytokine function in a diverse range of diseases including malignancies and chronic inflammation.


Cell Membrane/chemistry , Receptors, Cytokine/chemistry , Animals , Cell Membrane/metabolism , Cytokine Receptor gp130/chemistry , Cytokine Receptor gp130/metabolism , Cytokines/chemistry , Cytokines/metabolism , Glycosylation , Humans , Janus Kinase 1/chemistry , Janus Kinase 1/metabolism , Models, Biological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, Cytokine/metabolism , Signal Transduction
17.
Growth Factors ; 30(2): 63-75, 2012 Apr.
Article En | MEDLINE | ID: mdl-22257375

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a pluripotent cytokine produced by many cells in the body, which regulates normal and malignant hemopoiesis as well as innate and adaptive immunity. GM-CSF assembles and activates its heterodimeric receptor complex on the surface of myeloid cells, initiating multiple signaling pathways that control key functions such as cell survival, cell proliferation, and functional activation. Understanding the molecular composition of these pathways, the interaction of the various components as well as the kinetics and dose-dependent mechanics of receptor activation provides valuable insights into the function of GM-CSF as well as the related cytokines, interleukin-3 and interleukin-5. This knowledge provides opportunities for the development of new therapies to block the action of these cytokines in hematological malignancy and chronic inflammation.


Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematologic Neoplasms/metabolism , Inflammation/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Chronic Disease , Hematologic Neoplasms/pathology , Humans , Inflammation/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Signal Transduction
18.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 63(Pt 12): 1021-5, 2007 Dec 01.
Article En | MEDLINE | ID: mdl-18084083

The major histocompatibility complex (MHC) class II molecules HLA-DQ2 and HLA-DQ8 are key risk factors in coeliac disease, as they bind deamidated gluten peptides that are subsequently recognized by CD4+ T cells. Here, the production and crystallization of both HLA-DQ2 and HLA-DQ8 in complex with the deamidated gliadin peptides DQ2 alpha-I (PQPELPYPQ) and DQ8 alpha-I (EGSFQPSQE), respectively, are reported.


Celiac Disease/metabolism , Gliadin/metabolism , HLA-DQ Antigens/metabolism , Leukocytes/metabolism , Peptides/metabolism , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Deamination , Gliadin/chemistry , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/genetics , HLA-DQ Antigens/isolation & purification , Humans , Leukocytes/chemistry , Peptides/chemistry , Protein Binding
19.
J Mol Biol ; 367(3): 839-47, 2007 Mar 30.
Article En | MEDLINE | ID: mdl-17292918

c-Fms, a member of the Platelet-derived Growth Factor (PDGF) receptor family of receptor tyrosine kinases (RTKs), is the receptor for macrophage colony stimulating factor (CSF-1) that regulates proliferation, differentiation and survival of cells of the mononuclear phagocyte lineage. Abnormal expression of c-fms proto-oncogene is associated with a significant number of human pathologies, including a variety of cancers and rheumatoid arthritis. Accordingly, c-Fms represents an attractive therapeutic target. To further understand the regulation of c-Fms, we determined the 2.7 A resolution crystal structure of the cytosolic domain of c-Fms that comprised the kinase domain and the juxtamembrane domain. The structure reveals the crucial inhibitory role of the juxtamembrane domain (JM) that binds to a hydrophobic site immediately adjacent to the ATP binding pocket. This interaction prevents the activation loop from adopting an active conformation thereby locking the c-Fms kinase into an autoinhibited state. As observed for other members of the PDGF receptor family, namely c-Kit and Flt3, three JM-derived tyrosine residues primarily drive the mechanism for autoinhibition in c-Fms, therefore defining a common autoinhibitory mechanism within this family. Moreover the structure provides an understanding of c-Fms inhibition by Gleevec as well as providing a platform for the development of more selective inhibitors that target the inactive conformation of c-Fms kinase.


Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Amino Acid Sequence , Benzamides , Crystallography, X-Ray , Humans , Imatinib Mesylate , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Mas , Pyrimidines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Homology, Amino Acid
20.
Blood ; 107(1): 176-83, 2006 Jan 01.
Article En | MEDLINE | ID: mdl-16174768

JAK2, a member of the Janus kinase (JAK) family of protein tyrosine kinases (PTKs), is an important intracellular mediator of cytokine signaling. Mutations of the JAK2 gene are associated with hematologic cancers, and aberrant JAK activity is also associated with a number of immune diseases, including rheumatoid arthritis. Accordingly, the development of JAK2-specific inhibitors has tremendous clinical relevance. Critical to the function of JAK2 is its PTK domain. We report the 2.0 A crystal structure of the active conformation of the JAK2 PTK domain in complex with a high-affinity, pan-JAK inhibitor that appears to bind via an induced fit mechanism. This inhibitor, the tetracyclic pyridone 2-tert-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-1, was buried deep within a constricted ATP-binding site, in which extensive interactions, including residues that are unique to JAK2 and the JAK family, are made with the inhibitor. We present a structural basis of high-affinity JAK-specific inhibition that will undoubtedly provide an invaluable tool for the further design of novel, potent, and specific therapeutics against the JAK family.


Enzyme Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/chemistry , Adenosine Triphosphate , Binding Sites , Crystallography, X-Ray , Humans , Janus Kinase 2 , Molecular Structure , Protein Binding , Protein Conformation
...