44 results on '"McClure BJ"'
Search Results
2. Outcomes for Australian children with relapsed/refractory acute lymphoblastic leukaemia treated with blinatumomab.
- Author
-
Sutton, R, Pozza, LD, Khaw, SL, Fraser, C, Revesz, T, Chamberlain, J, Mitchell, R, Trahair, TN, Bateman, CM, Venn, NC, Law, T, Ong, E, Heatley, SL, McClure, BJ, Meyer, C, Marschalek, R, Henderson, MJ, Cross, S, White, DL, Kotecha, RS, Sutton, R, Pozza, LD, Khaw, SL, Fraser, C, Revesz, T, Chamberlain, J, Mitchell, R, Trahair, TN, Bateman, CM, Venn, NC, Law, T, Ong, E, Heatley, SL, McClure, BJ, Meyer, C, Marschalek, R, Henderson, MJ, Cross, S, White, DL, and Kotecha, RS
- Abstract
We report on the Australian experience of blinatumomab for treatment of 24 children with relapsed/refractory precursor B-cell acute lymphoblastic leukaemia (B-ALL) and high-risk genetics, resulting in a minimal residual disease (MRD) response rate of 58%, 2-year progression-free survival (PFS) of 39% and 2-year overall survival of 63%. In total, 83% (n = 20/24) proceeded to haematopoietic stem cell transplant, directly after blinatumomab (n = 12) or following additional salvage therapy (n = 8). Four patients successfully received CD19-directed chimeric antigen receptor T-cell therapy despite prior blinatumomab exposure. Inferior 2-year PFS was associated with MRD positivity (20%, n = 15) and in KMT2A-rearranged infants (15%, n = 9). Our findings highlight that not all children with relapsed/refractory B-ALL respond to blinatumomab and factors such as blast genotype may affect prognosis.
- Published
- 2021
3. New approaches in the treatment of asthma
- Author
-
Ramshaw, Hs, Woodcock, Jm, Bagley, Cj, McClure, Bj, Hercus, Tr, and Lopez, Af
- Published
- 2001
4. Pre-B acute lymphoblastic leukaemia recurrent fusion, EP300-ZNF384, is associated with a distinct gene expression
- Author
-
McClure, BJ, Heatley, SL, Kok, CH, Sadras, T, An, J, Hughes, TP, Lock, RB, Yeung, D, Sutton, R, White, DL, McClure, BJ, Heatley, SL, Kok, CH, Sadras, T, An, J, Hughes, TP, Lock, RB, Yeung, D, Sutton, R, and White, DL
- Abstract
Background: Zinc-finger protein 384 (ZNF384) fusions are an emerging subtype of precursor B-cell acute lymphoblastic leukaemia (pre-B-ALL) and here we further characterised their prevalence, survival outcomes and transcriptome. Methods: Bone marrow mononuclear cells from 274 BCR-ABL1-negative pre-B-ALL patients were immunophenotyped and transcriptome molecularly characterised. Transcriptomic data was analysed by principal component analysis and gene-set enrichment analysis to identify gene and pathway expression changes. Results: We exclusively detect E1A-associated protein p300 (EP300)-ZNF384 in 5.7% of BCR-ABL1-negative adolescent/young adult (AYA)/adult pre-B-ALL patients. EP300-ZNF384 patients do not appear to be a high-risk subgroup. Transcriptomic analysis revealed that EP300-ZNF384 samples have a distinct gene expression profile that results in the up-regulation of Janus kinase/signal transducers and activators of transcription (JAK/STAT) and cell adhesion pathways and down-regulation of cell cycle and DNA repair pathways. Conclusions: Importantly, this report contributes to a better overview of the incidence of EP300-ZNF384 patients and show that they have a distinct gene signature with concurrent up-regulation of JAK-STAT pathway, reduced expression of B-cell regulators and reduced DNA repair capacity.
- Published
- 2018
5. The mechanism of GM-CSF inhibition by human GM-CSF auto-antibodies suggests novel therapeutic opportunities
- Author
-
Dhagat, U, Hercus, TR, Broughton, SE, Nero, TL, Shing, KSCT, Barry, EF, Thomson, CA, Bryson, S, Pai, EF, McClure, BJ, Schrader, JW, Lopez, AF, Parker, MW, Dhagat, U, Hercus, TR, Broughton, SE, Nero, TL, Shing, KSCT, Barry, EF, Thomson, CA, Bryson, S, Pai, EF, McClure, BJ, Schrader, JW, Lopez, AF, and Parker, MW
- Abstract
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.
- Published
- 2018
6. High Yield Production of a Soluble Human Interleukin-3 Variant from E. coli with Wild-Type Bioactivity and Improved Radiolabeling Properties
- Author
-
van Raaij, MJ, Hercus, TR, Barry, EF, Dottore, M, McClure, BJ, Webb, AI, Lopez, AF, Young, IG, Murphy, JM, van Raaij, MJ, Hercus, TR, Barry, EF, Dottore, M, McClure, BJ, Webb, AI, Lopez, AF, Young, IG, and Murphy, JM
- Abstract
Human interleukin-3 (hIL-3) is a polypeptide growth factor that regulates the proliferation, differentiation, survival and function of hematopoietic progenitors and many mature blood cell lineages. Although recombinant hIL-3 is a widely used laboratory reagent in hematology, standard methods for its preparation, including those employed by commercial suppliers, remain arduous owing to a reliance on refolding insoluble protein expressed in E. coli. In addition, wild-type hIL-3 is a poor substrate for radio-iodination, which has been a long-standing hindrance to its use in receptor binding assays. To overcome these problems, we developed a method for expression of hIL-3 in E. coli as a soluble protein, with typical yields of >3mg of purified hIL-3 per litre of shaking microbial culture. Additionally, we introduced a non-native tyrosine residue into our hIL-3 analog, which allowed radio-iodination to high specific activities for receptor binding studies whilst not compromising bioactivity. The method presented herein provides a cost-effective and convenient route to milligram quantities of a hIL-3 analog with wild-type bioactivity that, unlike wild-type hIL‑3, can be efficiently radio-iodinated for receptor binding studies.
- Published
- 2013
7. New approaches in the treatment of asthma.
- Author
-
Lopez, AF, Ramshaw, Hs, Woodcock, Jm, Bagley, Cj, McClure, Bj, Hercus, Tr, and Lopez, Af
- Subjects
ASTHMA treatment ,EOSINOPHILIA - Abstract
Summary Asthma is a common and complex inflammatory disease of the airways that remains incurable. Current forms of therapy are long term and may exhibit associated side-effect problems. Major participants in the development of an asthma phenotype include the triggering stimuli such as the allergens themselves, cells such as T cells, epithelial cells and mast cells that produce a variety of cytokines including IL-5, GM-CSF, IL-3, IL-4 and IL-13 and chemokines such as eotaxin. Significantly, the eosinophil, a specialized blood cell type, is invariably associated with this disease. The eosinophil has long been incriminated in the pathology of asthma due to its ability to release preformed and unique toxic substances as well as newly formed pro-inflammatory mediators. The regulation of eosinophil production and function is carried out by soluble peptides or factors. Of these IL-5, GM-CSF and IL-3 are of paramount importance as they control eosinophil functional activity and are the only known eosinophilopoietic factors. In addition they regulate the eosinophil life span by inhibiting apoptosis. While one therapeutic approach in asthma is directed at inhibiting single eosinophil products such as leukotrienes or single eosinophil regulators such as IL-5, we believe that the simultaneous inhibition of more than one component is preferable. This may be particularly important with eosinophil regulators in that not only IL-5, but also GM-CSF has been repeatedly implicated in clinical studies of asthma. The fact that GM-CSF is produced by many cells in the body and in copious amounts by lung epithelial cells highlights this need further. Our approach takes advantage of the fact that the IL-5 and GM-CSF receptors (as well as IL-3 receptors) utilize a shared subunit to bind, with high affinity, to these cytokines and the same common subunit mediates signal transduction culminating in all the biological activities mentioned. By generating the monoclonal antibody BION-1 to... [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
8. Asciminib is a novel inhibitor of ABL1 and ABL2 gene fusions in ALL but requires the ABL SH3 domain for efficacy.
- Author
-
Eadie LN, Lagonik E, Page EC, Schutz CE, Heatley SL, McClure BJ, Forgione MO, Yeung DT, Hughes TP, and White DL
- Subjects
- Humans, src Homology Domains genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl antagonists & inhibitors, Niacinamide analogs & derivatives, Pyrazoles, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
9. The 10 th Barossa meeting: Cell Signalling to Cancer Medicine.
- Author
-
Kan WL, McClure BJ, Hahn CN, and Powell JA
- Subjects
- Humans, Signal Transduction, Neoplasms genetics, Medicine
- Published
- 2024
- Full Text
- View/download PDF
10. Desmoglein-2 as a cancer modulator: friend or foe?
- Author
-
Myo Min KK, Ffrench CB, McClure BJ, Ortiz M, Dorward EL, Samuel MS, Ebert LM, Mahoney MG, and Bonder CS
- Abstract
Desmoglein-2 (DSG2) is a calcium-binding single pass transmembrane glycoprotein and a member of the large cadherin family. Until recently, DSG2 was thought to only function as a cell adhesion protein embedded within desmosome junctions designed to enable cells to better tolerate mechanical stress. However, additional roles for DSG2 outside of desmosomes are continuing to emerge, particularly in cancer. Herein, we review the current literature on DSG2 in cancer and detail its impact on biological functions such as cell adhesion, proliferation, migration, invasion, intracellular signaling, extracellular vesicle release and vasculogenic mimicry. An increased understanding of the diverse repertoire of the biological functions of DSG2 holds promise to exploit this cell surface protein as a potential prognostic biomarker and/or target for better patient outcomes. This review explores the canonical and non-canonical functions of DSG2, as well as the context-dependent impacts of DSG2 in the realm of cancer., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Myo Min, Ffrench, McClure, Ortiz, Dorward, Samuel, Ebert, Mahoney and Bonder.)
- Published
- 2023
- Full Text
- View/download PDF
11. Reproducible Bioinformatics Analysis Workflows for Detecting IGH Gene Fusions in B-Cell Acute Lymphoblastic Leukaemia Patients.
- Author
-
Thomson AJ, Rehn JA, Heatley SL, Eadie LN, Page EC, Schutz C, McClure BJ, Sutton R, Dalla-Pozza L, Moore AS, Greenwood M, Kotecha RS, Fong CY, Yong ASM, Yeung DT, Breen J, and White DL
- Abstract
B-cell acute lymphoblastic leukaemia (B-ALL) is characterised by diverse genomic alterations, the most frequent being gene fusions detected via transcriptomic analysis (mRNA-seq). Due to its hypervariable nature, gene fusions involving the Immunoglobulin Heavy Chain ( IGH ) locus can be difficult to detect with standard gene fusion calling algorithms and significant computational resources and analysis times are required. We aimed to optimize a gene fusion calling workflow to achieve best-case sensitivity for IGH gene fusion detection. Using Nextflow, we developed a simplified workflow containing the algorithms FusionCatcher, Arriba, and STAR-Fusion. We analysed samples from 35 patients harbouring IGH fusions ( IGH::CRLF2 n = 17, IGH::DUX4 n = 15, IGH::EPOR n = 3) and assessed the detection rates for each caller, before optimizing the parameters to enhance sensitivity for IGH fusions. Initial results showed that FusionCatcher and Arriba outperformed STAR-Fusion (85-89% vs. 29% of IGH fusions reported). We found that extensive filtering in STAR-Fusion hindered IGH reporting. By adjusting specific filtering steps (e.g., read support, fusion fragments per million total reads), we achieved a 94% reporting rate for IGH fusions with STAR-Fusion. This analysis highlights the importance of filtering optimization for IGH gene fusion events, offering alternative workflows for difficult-to-detect high-risk B-ALL subtypes.
- Published
- 2023
- Full Text
- View/download PDF
12. Case Report: Rare IKZF1 Gene Fusions Identified in Neonate with Congenital KMT2A -Rearranged Acute Lymphoblastic Leukemia.
- Author
-
Eadie LN, Rehn JA, Breen J, Osborn MP, Jessop S, Downes CEJ, Heatley SL, McClure BJ, Yeung DT, Revesz T, Saxon B, and White DL
- Subjects
- Infant, Infant, Newborn, Humans, Transcription Factors genetics, Gene Fusion, Chromosome Aberrations, Genomics, Ikaros Transcription Factor genetics, Jumonji Domain-Containing Histone Demethylases genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, F-Box Proteins genetics
- Abstract
Chromosomal rearrangements involving the KMT2A gene occur frequently in acute lymphoblastic leukaemia (ALL). KMT2A -rearranged ALL ( KMT2Ar ALL) has poor long-term survival rates and is the most common ALL subtype in infants less than 1 year of age. KMT2Ar ALL frequently occurs with additional chromosomal abnormalities including disruption of the IKZF1 gene, usually by exon deletion. Typically, KMT2Ar ALL in infants is accompanied by a limited number of cooperative le-sions. Here we report a case of aggressive infant KMT2Ar ALL harbouring additional rare IKZF1 gene fusions. Comprehensive genomic and transcriptomic analyses were performed on sequential samples. This report highlights the genomic complexity of this particular disease and describes the novel gene fusions IKZF1::TUT1 and KDM2A::IKZF1 .
- Published
- 2023
- Full Text
- View/download PDF
13. RaScALL: Rapid (Ra) screening (Sc) of RNA-seq data for prognostically significant genomic alterations in acute lymphoblastic leukaemia (ALL).
- Author
-
Rehn J, Mayoh C, Heatley SL, McClure BJ, Eadie LN, Schutz C, Yeung DT, Cowley MJ, Breen J, and White DL
- Subjects
- Humans, RNA-Seq, Australia, Genomics methods, RNA, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
RNA-sequencing (RNA-seq) efforts in acute lymphoblastic leukaemia (ALL) have identified numerous prognostically significant genomic alterations which can guide diagnostic risk stratification and treatment choices when detected early. However, integrating RNA-seq in a clinical setting requires rapid detection and accurate reporting of clinically relevant alterations. Here we present RaScALL, an implementation of the k-mer based variant detection tool km, capable of identifying more than 100 prognostically significant lesions observed in ALL, including gene fusions, single nucleotide variants and focal gene deletions. We compared genomic alterations detected by RaScALL and those reported by alignment-based de novo variant detection tools in a study cohort of 180 Australian patient samples. Results were validated using 100 patient samples from a published North American cohort. RaScALL demonstrated a high degree of accuracy for reporting subtype defining genomic alterations. Gene fusions, including difficult to detect fusions involving EPOR and DUX4, were accurately identified in 98% of reported cases in the study cohort (n = 164) and 95% of samples (n = 63) in the validation cohort. Pathogenic sequence variants were correctly identified in 75% of tested samples, including all cases involving subtype defining variants PAX5 p.P80R (n = 12) and IKZF1 p.N159Y (n = 4). Intragenic IKZF1 deletions resulting in aberrant transcript isoforms were also detectable with 98% accuracy. Importantly, the median analysis time for detection of all targeted alterations averaged 22 minutes per sample, significantly shorter than standard alignment-based approaches. The application of RaScALL enables rapid identification and reporting of previously identified genomic alterations of known clinical relevance., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
14. JAK2 Alterations in Acute Lymphoblastic Leukemia: Molecular Insights for Superior Precision Medicine Strategies.
- Author
-
Downes CE, McClure BJ, McDougal DP, Heatley SL, Bruning JB, Thomas D, Yeung DT, and White DL
- Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, arising from immature lymphocytes that show uncontrolled proliferation and arrested differentiation. Genomic alterations affecting Janus kinase 2 ( JAK2 ) correlate with some of the poorest outcomes within the Philadelphia-like subtype of ALL. Given the success of kinase inhibitors in the treatment of chronic myeloid leukemia, the discovery of activating JAK2 point mutations and JAK2 fusion genes in ALL, was a breakthrough for potential targeted therapies. However, the molecular mechanisms by which these alterations activate JAK2 and promote downstream signaling is poorly understood. Furthermore, as clinical data regarding the limitations of approved JAK inhibitors in myeloproliferative disorders matures, there is a growing awareness of the need for alternative precision medicine approaches for specific JAK2 lesions. This review focuses on the molecular mechanisms behind ALL-associated JAK2 mutations and JAK2 fusion genes, known and potential causes of JAK-inhibitor resistance, and how JAK2 alterations could be targeted using alternative and novel rationally designed therapies to guide precision medicine approaches for these high-risk subtypes of ALL., Competing Interests: DW receives research support from BMS, Honoraria from Amgen. DY receives research support from Novartis, Ariad and BMS, Honoraria from Novartis, BMS, Amgen and Pfizer. None of these agencies have had a role in the preparation of this manuscript. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Downes, McClure, McDougal, Heatley, Bruning, Thomas, Yeung and White.)
- Published
- 2022
- Full Text
- View/download PDF
15. TP53 loss‑of‑function mutations reduce sensitivity of acute leukaemia to the curaxin CBL0137.
- Author
-
Forgione MO, McClure BJ, Page EC, Yeung DT, Eadie LN, and White DL
- Subjects
- Carbazoles pharmacology, Humans, Mutation, Tumor Suppressor Protein p53 genetics, Glioblastoma drug therapy, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
The presence of a TP53 mutation is a predictor of poor outcome in leukaemia, and efficacious targeted therapies for these patients are lacking. The curaxin CBL0137 has demonstrated promising antitumour activities in multiple cancers such as glioblastoma, acting through p53 activation, NF‑κB inhibition and chromatin remodelling. In the present study, it was revealed using Annexin‑V/7‑AAD apoptosis assays that CBL0137 has efficacy across several human acute leukaemia cell lines with wild‑type TP53 , but sensitivity is reduced in TP53 ‑mutated subtypes. A heterozygous TP53 loss‑of‑function mutation in the KMT2A‑AFF1 human RS4;11 cell line was generated, and it was demonstrated that heterozygous TP53 loss‑of‑function is sufficient to cause a significant reduction in CBL0137 sensitivity. To the best of our knowledge, this is the first evidence to suggest a clinically significant role for functional p53 in the efficacy of CBL0137 in acute leukaemia. Future CBL0137 clinical trials should include TP53 mutation screening, to establish the clinical relevance of TP53 mutations in CBL0137 efficacy.
- Published
- 2022
- Full Text
- View/download PDF
16. Case Report: Precision Medicine Target Revealed by In Vitro Modeling of Relapsed, Refractory Acute Lymphoblastic Leukemia From a Child With Neurofibromatosis.
- Author
-
Heatley SL, Page EC, Eadie LN, McClure BJ, Rehn J, Yeung DT, Osborn M, Revesz T, Kirby M, and White DL
- Abstract
Children with neurofibromatosis have a higher risk of developing juvenile myelomonocytic leukemia and acute myeloid leukemia, but rarely develop B-cell acute lymphoblastic leukemia (B-ALL). Through in-vitro modeling, a novel NF1 p.L2467 frameshift (fs) mutation identified in a relapsed/refractory Ph-like B-ALL patient with neurofibromatosis demonstrated cytokine independence and increased RAS signaling, indicative of leukemic transformation. Furthermore, these cells were sensitive to the MEK inhibitors trametinib and mirdametinib. Bi-allelic NF1 loss of function may be a contributing factor to relapse and with sensitivity to MEK inhibitors, suggests a novel precision medicine target in the setting of neurofibromatosis patients with B-ALL., Competing Interests: DTY received research support from Novartis, Ariad and BMS, Honoraria and Advisory role Novartis. DLW received Research support from Novartis and BMS, and Honoraria from BMS. MO received non-financial support from Amgen. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Heatley, Page, Eadie, McClure, Rehn, Yeung, Osborn, Revesz, Kirby and White.)
- Published
- 2022
- Full Text
- View/download PDF
17. Targeting the Human β c Receptor Inhibits Contact Dermatitis in a Transgenic Mouse Model.
- Author
-
Yip KH, McKenzie D, Ramshaw HS, Chao J, McClure BJ, Raquet E, Kraushaar T, Röder J, Maxwell M, Alhamdoosh M, Hammet A, Fong JH, Zeglinski K, Monaghan K, Pant H, Grimbaldeston MA, Vairo G, Wilson NJ, Owczarek CM, Hercus TR, Lopez AF, and Tumes DJ
- Subjects
- Animals, Cytokines, Eosinophils, Humans, Interleukin-3 metabolism, Interleukin-5 metabolism, Mice, Mice, Transgenic, Dermatitis, Contact, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
Allergic contact dermatitis (ACD) is a prevalent and poorly controlled inflammatory disease caused by skin infiltration of T cells and granulocytes. The beta common (β
c ) cytokines GM-CSF, IL-3, and IL-5 are powerful regulators of granulocyte function that signal through their common receptor subunit βc , a property that has made βc an attractive target to simultaneously inhibit these cytokines. However, the species specificity of βc has precluded testing of inhibitors of human βc in mouse models. To overcome this problem, we developed a human βc receptor transgenic mouse strain with a hematopoietic cell‒specific expression of human βc instead of mouse βc . Human βc receptor transgenic cells responded to mouse GM-CSF and IL-5 but not to IL-3 in vitro and developed tissue pathology and cellular inflammation comparable with those in wild-type mice in a model of ACD. Similarly, Il3-/- mice developed ACD pathology comparable with that of wild-type mice. Importantly, the blocking anti-human βc antibody CSL311 strongly suppressed ear pinna thickening and histopathological changes typical of ACD and reduced accumulation of neutrophils, mast cells, and eosinophils in the skin. These results show that GM-CSF and IL-5 but not IL-3 are major mediators of ACD and define the human βc receptor transgenic mouse as a unique platform to test the inhibitors of βc in vivo., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
18. Two novel cases of NUTM1-rearranged B-cell acute lymphoblastic leukaemia presenting with high-risk features.
- Author
-
McClure BJ, Pal M, Heatley SL, Rehn J, Schutz C, Breen J, Venn NC, Sutton R, Khaw SL, Yeung DT, and White DL
- Subjects
- Gene Rearrangement, Humans, Neoplasm Proteins, Nuclear Proteins, Receptors, Cytokine genetics, Transcription Factors, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2022
- Full Text
- View/download PDF
19. Identification of a novel GOLGA4-JAK2 fusion gene in B-cell acute lymphoblastic leukaemia.
- Author
-
Downes CEJ, Rehn J, Heatley SL, Yeung D, McClure BJ, and White DL
- Subjects
- Animals, Biopsy, Bone Marrow pathology, Cell Line, Tumor, Cell Survival drug effects, DNA Mutational Analysis, Disease Models, Animal, Gene Expression Regulation, Neoplastic drug effects, Gene Rearrangement, Genetic Predisposition to Disease, Humans, Male, Mice, Middle Aged, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Prognosis, Protein Kinase Inhibitors, Signal Transduction, Autoantigens genetics, Biomarkers, Tumor, Janus Kinase 2 genetics, Oncogene Proteins, Fusion genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Rearrangements of Janus kinase 2 (JAK2r) form a subtype of acute lymphoblastic leukaemia (ALL) associated with poor patient outcomes. We present a high-risk case of B-cell ALL (B-ALL) where retrospective mRNA sequencing identified a novel GOLGA4-JAK2 fusion gene. Expression of GOLGA4-JAK2 in murine pro-B cells promoted factor-independent growth, implicating GOLGA4-JAK2 as an oncogenic driver. Cells expressing GOLGA4-JAK2 demonstrated constitutive activation of JAK/STAT signalling and were sensitive to JAK inhibitors. This study contributes to the diverse collection of JAK2 fusion genes identified in B-ALL and supports the incorporation of JAK inhibitors into treatment strategies to improve outcomes for this subtype., (© 2021 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
20. HMGN1 plays a significant role in CRLF2 driven Down Syndrome leukemia and provides a potential therapeutic target in this high-risk cohort.
- Author
-
Page EC, Heatley SL, Eadie LN, McClure BJ, de Bock CE, Omari S, Yeung DT, Hughes TP, Thomas PQ, and White DL
- Subjects
- Animals, Female, Humans, Mice, Cell Line, Tumor, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, HMGN1 Protein, Down Syndrome complications, Down Syndrome genetics, Down Syndrome metabolism, Receptors, Cytokine genetics, Receptors, Cytokine metabolism
- Abstract
The genetic basis of the predisposition for Down Syndrome (DS) patients to develop cytokine receptor-like factor 2 rearranged (CRLF2r) acute lymphoblastic leukemia (ALL) is currently unknown. Genes located on chromosome 21 and expressed in hematopoietic cells are likely candidates for investigation of CRLF2r DS-ALL pathogenesis. We explored the high-mobility group nucleosome-binding protein 1 (HMGN1), located in the DS critical region, in an inducible CRISPR/Cas9 knockout (KO) xenograft model to assess the effect of HMGN1 loss of function on the leukemic burden. We demonstrated HMGN1 KO-mitigated leukemic phenotypes including hepatosplenomegaly, thrombocytopenia, and anemia, commonly observed in leukemia patients, and significantly increased survival in vivo. HMGN1 overexpression in murine stem cells and Ba/F3 cells in vitro, in combination with P2RY8-CRLF2, resulted in cytokine-independent transformation and upregulation of cell signaling pathways associated with leukemic development. Finally, in vitro screening demonstrated successful targeting of P2RY8-CRLF2 and HMGN1 co-expressing cell lines and patient samples with fedratinib (JAK2 inhibitor), and GSK-J4 (demethylase inhibitor) in combination. Together, these data provide critical insight into the development and persistence of CRLF2r DS-ALL and identify HMGN1 as a potential therapeutic target to improve outcomes and reduce toxicity in this high-risk cohort of young patients., (© 2021. Crown.)
- Published
- 2022
- Full Text
- View/download PDF
21. Acquired JAK2 mutations confer resistance to JAK inhibitors in cell models of acute lymphoblastic leukemia.
- Author
-
Downes CEJ, McClure BJ, Bruning JB, Page E, Breen J, Rehn J, Yeung DT, and White DL
- Abstract
Ruxolitinib (rux) Phase II clinical trials are underway for the treatment of high-risk JAK2-rearranged (JAK2r) B-cell acute lymphoblastic leukemia (B-ALL). Treatment resistance to targeted inhibitors in other settings is common; elucidating potential mechanisms of rux resistance in JAK2r B-ALL will enable development of therapeutic strategies to overcome or avert resistance. We generated a murine pro-B cell model of ATF7IP-JAK2 with acquired resistance to multiple type-I JAK inhibitors. Resistance was associated with mutations within the JAK2 ATP/rux binding site, including a JAK2 p.G993A mutation. Using in vitro models of JAK2r B-ALL, JAK2 p.G993A conferred resistance to six type-I JAK inhibitors and the type-II JAK inhibitor, CHZ-868. Using computational modeling, we postulate that JAK2 p.G993A enabled JAK2 activation in the presence of drug binding through a unique resistance mechanism that modulates the mobility of the conserved JAK2 activation loop. This study highlights the importance of monitoring mutation emergence and may inform future drug design and the development of therapeutic strategies for this high-risk patient cohort., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
22. Outcomes for Australian children with relapsed/refractory acute lymphoblastic leukaemia treated with blinatumomab.
- Author
-
Sutton R, Pozza LD, Khaw SL, Fraser C, Revesz T, Chamberlain J, Mitchell R, Trahair TN, Bateman CM, Venn NC, Law T, Ong E, Heatley SL, McClure BJ, Meyer C, Marschalek R, Henderson MJ, Cross S, White DL, and Kotecha RS
- Subjects
- Australia, Child, Female, Humans, Male, Neoplasm Recurrence, Local drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Retrospective Studies, Treatment Outcome, Antibodies, Bispecific therapeutic use, Antineoplastic Agents therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
We report on the Australian experience of blinatumomab for treatment of 24 children with relapsed/refractory precursor B-cell acute lymphoblastic leukaemia (B-ALL) and high-risk genetics, resulting in a minimal residual disease (MRD) response rate of 58%, 2-year progression-free survival (PFS) of 39% and 2-year overall survival of 63%. In total, 83% (n = 20/24) proceeded to haematopoietic stem cell transplant, directly after blinatumomab (n = 12) or following additional salvage therapy (n = 8). Four patients successfully received CD19-directed chimeric antigen receptor T-cell therapy despite prior blinatumomab exposure. Inferior 2-year PFS was associated with MRD positivity (20%, n = 15) and in KMT2A-rearranged infants (15%, n = 9). Our findings highlight that not all children with relapsed/refractory B-ALL respond to blinatumomab and factors such as blast genotype may affect prognosis., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
23. In-vitro modeling of TKI resistance in the high-risk B-cell acute lymphoblastic leukemia fusion gene RANBP2-ABL1 - implications for targeted therapy.
- Author
-
Heatley SL, Asari K, Schutz CE, Leclercq TM, McClure BJ, Eadie LN, Hughes TP, Yeung DT, and White DL
- Subjects
- B-Lymphocytes, Child, Drug Resistance, Neoplasm genetics, Fusion Proteins, bcr-abl genetics, Humans, Molecular Chaperones, Mutation, Nuclear Pore Complex Proteins, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Abstract
Acute lymphoblastic leukemia remains a leading cause of cancer-related death in children. Furthermore, subtypes such as Ph-like ALL remain at high-risk of relapse, and treatment resistance remains a significant clinical issue. The patient-derived Ph-like ALL RANBP2-ABL1 fusion gene was transduced into Ba/F3 cells and allowed to become resistant to the tyrosine kinase inhibitors (TKIs) imatinib or dasatinib, followed by secondary resistance to ponatinib. RANBP2-ABL1 Ba/F3 cells developed the clinically relevant ABL1 p.T315I mutation and upon secondary resistance to ponatinib, developed compound mutations, including a novel ABL1 p.L302H mutation. Significantly, compound mutations were targetable with a combination of asciminib and ponatinib. In-vitro modeling of Ph-like ALL RANBP2-ABL1 has identified kinase domain mutations in response to TKI treatment, that may have important clinical ramifications. Early detection of mutations is paramount to guide treatment strategies and improve survival in this high-risk group of patients.
- Published
- 2021
- Full Text
- View/download PDF
24. High-risk B-cell acute lymphoblastic leukaemia presenting with hypereosinophilia and acquiring a novel PAX5 fusion on relapse.
- Author
-
McClure BJ, Heatley SL, Rehn J, Breen J, Sutton R, Hughes TP, Suppiah R, Revesz T, Osborn M, White D, Yeung DT, and White DL
- Subjects
- Adolescent, Humans, Male, Hypereosinophilic Syndrome genetics, Hypereosinophilic Syndrome metabolism, Hypereosinophilic Syndrome pathology, Immunoglobulin Heavy Chains genetics, Immunoglobulin Heavy Chains metabolism, Interleukin-3 genetics, Interleukin-3 metabolism, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, PAX5 Transcription Factor genetics, PAX5 Transcription Factor metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Published
- 2020
- Full Text
- View/download PDF
25. MLLT10 rearranged acute leukemia: Incidence, prognosis, and possible therapeutic strategies.
- Author
-
Forgione MO, McClure BJ, Yeung DT, Eadie LN, and White DL
- Abstract
Rearrangements of the MLLT10 gene occur in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), most commonly T-lineage ALL (T-ALL), in patients of all ages. MLLT10 rearranged (MLLT10r) acute leukemia presents a complex diagnostic and therapeutic challenge due to frequent presentation of immature or mixed phenotype, and a lack of consensus regarding optimal therapy. Cases of MLLT10r AML or T-ALL bearing immature phenotype are at high risk of poor outcome, but the underlying molecular mechanisms and sensitivity to targeted therapies remain poorly characterized. This review addresses the incidence and prognostic significance of MLLT10r in acute leukemia, and how the aberrant gene expression profile of this disease can inform potential targeted therapeutic strategies. Understanding the underlying genomics of MLLT10r acute leukemia, both clinically and molecularly, will improve prognostic stratification and accelerate the development of targeted therapeutic strategies, to improve patient outcomes., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
26. KMT2A rearranged acute lymphoblastic leukaemia: Unravelling the genomic complexity and heterogeneity of this high-risk disease.
- Author
-
Forgione MO, McClure BJ, Eadie LN, Yeung DT, and White DL
- Subjects
- Animals, Gene Rearrangement genetics, Genomics, High-Throughput Nucleotide Sequencing, Humans, Mice, Mutation genetics, Neoplasm Recurrence, Local pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Survival Rate, Genetic Heterogeneity, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Neoplasm Recurrence, Local genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
KMT2A rearranged (KMT2Ar) acute lymphoblastic leukaemia (ALL) is a high-risk genomic subtype, with long-term survival rates of less than 60% across all age groups. These cases present a complex clinical challenge, with a high incidence in infants, high-risk clinical features and propensity for aggressive relapse. KMT2A rearrangements are highly pathogenic leukaemic drivers, reflected by the high incidence of KMT2Ar ALL in infants, who carry few leukaemia-associated cooperative mutations. However, transgenic murine models of KMT2Ar ALL typically exhibit long latency and mature or mixed phenotype, and fail to recapitulate the aggressive disease observed clinically. Next-generation sequencing has revealed that KMT2Ar ALL also occurs in adolescents and adults, and potentially cooperative genomic lesions such as PI3K-RAS pathway variants are present in KMT2Ar patients of all ages. This review addresses the aetiology of KMT2Ar ALL, with a focus on the cell of origin and mutational landscape, and how genomic profiling of KMT2Ar ALL patients in the era of next-generation sequencing demonstrates that KMT2Ar ALL is a complex heterogenous disease. Ultimately, understanding the underlying biology of KMT2Ar ALL will be important in improving long-term outcomes for these high-risk patients., (Crown Copyright © 2019. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. The mechanism of GM-CSF inhibition by human GM-CSF auto-antibodies suggests novel therapeutic opportunities.
- Author
-
Dhagat U, Hercus TR, Broughton SE, Nero TL, Cheung Tung Shing KS, Barry EF, Thomson CA, Bryson S, Pai EF, McClure BJ, Schrader JW, Lopez AF, and Parker MW
- Subjects
- 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, Antibodies, Neutralizing chemistry, Antigen-Antibody Complex chemistry, Arthritis, Rheumatoid therapy, Autoantibodies chemistry, Epitopes chemistry, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Immunotherapy methods
- Abstract
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.
- Published
- 2018
- Full Text
- View/download PDF
28. Pre-B acute lymphoblastic leukaemia recurrent fusion, EP300-ZNF384, is associated with a distinct gene expression.
- Author
-
McClure BJ, Heatley SL, Kok CH, Sadras T, An J, Hughes TP, Lock RB, Yeung D, Sutton R, and White DL
- Subjects
- Adolescent, Adult, Child, Female, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Gene Frequency, Genes, abl genetics, Humans, Janus Kinases metabolism, Male, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Recurrence, STAT Transcription Factors metabolism, Signal Transduction genetics, Survival Analysis, Young Adult, E1A-Associated p300 Protein genetics, Oncogene Proteins, Fusion genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma epidemiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Trans-Activators genetics, Transcriptome
- Abstract
Background: Zinc-finger protein 384 (ZNF384) fusions are an emerging subtype of precursor B-cell acute lymphoblastic leukaemia (pre-B-ALL) and here we further characterised their prevalence, survival outcomes and transcriptome., Methods: Bone marrow mononuclear cells from 274 BCR-ABL1-negative pre-B-ALL patients were immunophenotyped and transcriptome molecularly characterised. Transcriptomic data was analysed by principal component analysis and gene-set enrichment analysis to identify gene and pathway expression changes., Results: We exclusively detect E1A-associated protein p300 (EP300)-ZNF384 in 5.7% of BCR-ABL1-negative adolescent/young adult (AYA)/adult pre-B-ALL patients. EP300-ZNF384 patients do not appear to be a high-risk subgroup. Transcriptomic analysis revealed that EP300-ZNF384 samples have a distinct gene expression profile that results in the up-regulation of Janus kinase/signal transducers and activators of transcription (JAK/STAT) and cell adhesion pathways and down-regulation of cell cycle and DNA repair pathways., Conclusions: Importantly, this report contributes to a better overview of the incidence of EP300-ZNF384 patients and show that they have a distinct gene signature with concurrent up-regulation of JAK-STAT pathway, reduced expression of B-cell regulators and reduced DNA repair capacity.
- Published
- 2018
- Full Text
- View/download PDF
29. Differential expression of MUC4, GPR110 and IL2RA defines two groups of CRLF2-rearranged acute lymphoblastic leukemia patients with distinct secondary lesions.
- Author
-
Sadras T, Heatley SL, Kok CH, Dang P, Galbraith KM, McClure BJ, Muskovic W, Venn NC, Moore S, Osborn M, Revesz T, Moore AS, Hughes TP, Yeung D, Sutton R, and White DL
- Subjects
- Female, Humans, Interleukin-2 Receptor alpha Subunit genetics, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Mucin-4 genetics, Mutation genetics, Oncogene Proteins genetics, Philadelphia Chromosome, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Prognosis, Receptors, G-Protein-Coupled genetics, Tumor Cells, Cultured, Gene Expression Regulation, Leukemic, Gene Rearrangement, Interleukin-2 Receptor alpha Subunit metabolism, Mucin-4 metabolism, Oncogene Proteins metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Receptors, Cytokine genetics, Receptors, G-Protein-Coupled metabolism
- Abstract
CRLF2-rearrangements (CRLF2-r) occur frequently in Ph-like B-ALL, a high-risk ALL sub-type characterized by a signaling profile similar to Ph + ALL, however accumulating evidence indicates genetic heterogeneity within CRLF2-r ALL. We performed thorough genomic characterization of 35 CRLF2-r cases (P2RY8-CRLF2 n = 18; IGH-CRLF2 n = 17). Activating JAK2 mutations were present in 34% of patients, and a CRLF2-F232C mutation was identified in an additional 17%. IKZF1 deletions were detected in 63% of cases. The majority of patients (26/35) classified as Ph-like, and these were characterized by significantly higher levels of MUC4, GPR110 and IL2RA/CD25. In addition, Ph-like CRLF2-r samples were significantly enriched for IKZF1 deletions, JAK2/CRLF2 mutations and increased expression of JAK/STAT target genes (CISH, SOCS1), suggesting that mutation-driven CRLF2/JAK2 activation is more frequent in this sub-group. Less is known about the genomics of CRLF2-r cases lacking JAK2-pathway mutations, but KRAS/NRAS mutations were identified in 4/9 non-Ph-like samples. This work highlights the heterogeneity of secondary lesions which may arise and influence intracellular-pathway activation in CRLF2-r patients, and importantly presents distinct therapeutic targets within a group of patients harboring identical primary translocations, for whom efficient directed therapies are currently lacking., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. A novel somatic JAK2 kinase-domain mutation in pediatric acute lymphoblastic leukemia with rapid on-treatment development of LOH.
- Author
-
Sadras T, Heatley SL, Kok CH, McClure BJ, Yeung D, Hughes TP, Sutton R, Ziegler DS, and White DL
- Subjects
- Base Sequence, Child, Female, Gene Rearrangement genetics, Humans, Protein Domains, Recurrence, Janus Kinase 2 chemistry, Janus Kinase 2 genetics, Loss of Heterozygosity genetics, Mutation genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
We report a novel somatic mutation in the kinase domain of JAK2 (R938Q) in a high-risk pediatric case of B-cell acute lymphoblastic leukemia (ALL). The patient developed on-therapy relapse at 12 months, and interestingly, the JAK2 locus acquired loss of heterozygosity during treatment resulting in 100% mutation load. Furthermore, we show that primary ALL mononuclear cells harboring the JAK2 R938Q mutation display reduced sensitivity to the JAK1/2 ATP-competitive inhibitor ruxolitinib in vitro, compared to ALL cells that carry a more common JAK2 pseudokinase domain mutation. Our findings are in line with previous reports that demonstrate that mutations within the kinase domain of JAK2 are associated with resistance to type I JAK inhibitors. Importantly, given the recent inclusion of ruxolitinib in trial protocols for children with JAK pathway alterations, we predict that inter-patient genetic variability may result in suboptimal responses to JAK inhibitor therapy in a subset of cases. The need for alternate targeted and/or combination therapies for patients who display inherent or developed resistance to JAK inhibitor therapy will be warranted, and we propose that kinase-mutants less sensitive to type I JAK inhibitors may present a currently unexplored platform for investigation of improved therapies., (Copyright © 2017. Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
31. Conformational Changes in the GM-CSF Receptor Suggest a Molecular Mechanism for Affinity Conversion and Receptor Signaling.
- Author
-
Broughton SE, Hercus TR, Nero TL, Dottore M, McClure BJ, Dhagat U, Taing H, Gorman MA, King-Scott J, Lopez AF, and Parker MW
- Subjects
- 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, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Protein Subunits chemistry, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor chemistry
- Abstract
The GM-CSF, IL-3, and IL-5 receptors constitute the βc 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 βc 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., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
32. CSL311, a novel, potent, therapeutic monoclonal antibody for the treatment of diseases mediated by the common β chain of the IL-3, GM-CSF and IL-5 receptors.
- Author
-
Panousis C, Dhagat U, Edwards KM, Rayzman V, Hardy MP, Braley H, Gauvreau GM, Hercus TR, Smith S, Sehmi R, McMillan L, Dottore M, McClure BJ, Fabri LJ, Vairo G, Lopez AF, Parker MW, Nash AD, Wilson NJ, Wilson MJ, and Owczarek CM
- Subjects
- Animals, Antigen-Antibody Complex chemistry, Antigen-Antibody Complex immunology, Asthma drug therapy, Asthma immunology, Asthma pathology, Crystallography, X-Ray, Eosinophils immunology, Eosinophils pathology, Humans, Mice, Antibodies, Monoclonal, Murine-Derived chemistry, Antibodies, Monoclonal, Murine-Derived immunology, Antibodies, Monoclonal, Murine-Derived therapeutic use, Cytokine Receptor Common beta Subunit chemistry, Cytokine Receptor Common beta Subunit immunology, Epitopes chemistry, Epitopes immunology, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Interleukin-3 antagonists & inhibitors, Interleukin-3 immunology, Interleukin-5 antagonists & inhibitors, Interleukin-5 immunology
- Abstract
The β common-signaling cytokines interleukin (IL)-3, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-5 stimulate pro-inflammatory activities of haematopoietic cells via a receptor complex incorporating cytokine-specific α and shared β common (βc, CD131) receptor. Evidence from animal models and recent clinical trials demonstrate that these cytokines are critical mediators of the pathogenesis of inflammatory airway disease such as asthma. However, no therapeutic agents, other than steroids, that specifically and effectively target inflammation mediated by all 3 of these cytokines exist. We employed phage display technology to identify and optimize a novel, human monoclonal antibody (CSL311) that binds to a unique epitope that is specific to the cytokine-binding site of the human βc receptor. The binding epitope of CSL311 on the βc receptor was defined by X-ray crystallography and site-directed mutagenesis. CSL311 has picomolar binding affinity for the human βc receptor, and at therapeutic concentrations is a highly potent antagonist of the combined activities of IL-3, GM-CSF and IL-5 on primary eosinophil survival in vitro. Importantly, CSL311 inhibited the survival of inflammatory cells present in induced sputum from human allergic asthmatic subjects undergoing allergen bronchoprovocation. Due to its high potency and ability to simultaneously suppress the activity of all 3 β common cytokines, CSL311 may provide a new strategy for the treatment of chronic inflammatory diseases where the human βc receptor is central to pathogenesis. The coordinates for the βc/CSL311 Fab complex structure have been deposited with the RCSB Protein Data Bank (PDB 5DWU).
- Published
- 2016
- Full Text
- View/download PDF
33. Dual mechanism of interleukin-3 receptor blockade by an anti-cancer antibody.
- Author
-
Broughton SE, Hercus TR, Hardy MP, McClure BJ, Nero TL, Dottore M, Huynh H, Braley H, Barry EF, Kan WL, Dhagat U, Scotney P, Hartman D, Busfield SJ, Owczarek CM, Nash AD, Wilson NJ, Parker MW, and Lopez AF
- Subjects
- 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, Antibodies, Monoclonal, Humanized chemistry, Antineoplastic Agents chemistry, Interleukin-3 Receptor alpha Subunit chemistry
- Abstract
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., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. High yield production of a soluble human interleukin-3 variant from E. coli with wild-type bioactivity and improved radiolabeling properties.
- Author
-
Hercus TR, Barry EF, Dottore M, McClure BJ, Webb AI, Lopez AF, Young IG, and Murphy JM
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Chromatography, Reverse-Phase, DNA Primers, Humans, Interleukin-3 chemistry, Interleukin-3 genetics, Interleukin-3 physiology, Mass Spectrometry, Mice, Molecular Sequence Data, Sequence Homology, Amino Acid, Solubility, Escherichia coli genetics, Interleukin-3 biosynthesis
- Abstract
Human interleukin-3 (hIL-3) is a polypeptide growth factor that regulates the proliferation, differentiation, survival and function of hematopoietic progenitors and many mature blood cell lineages. Although recombinant hIL-3 is a widely used laboratory reagent in hematology, standard methods for its preparation, including those employed by commercial suppliers, remain arduous owing to a reliance on refolding insoluble protein expressed in E. coli. In addition, wild-type hIL-3 is a poor substrate for radio-iodination, which has been a long-standing hindrance to its use in receptor binding assays. To overcome these problems, we developed a method for expression of hIL-3 in E. coli as a soluble protein, with typical yields of >3mg of purified hIL-3 per litre of shaking microbial culture. Additionally, we introduced a non-native tyrosine residue into our hIL-3 analog, which allowed radio-iodination to high specific activities for receptor binding studies whilst not compromising bioactivity. The method presented herein provides a cost-effective and convenient route to milligram quantities of a hIL-3 analog with wild-type bioactivity that, unlike wild-type hIL‑3, can be efficiently radio-iodinated for receptor binding studies.
- Published
- 2013
- Full Text
- View/download PDF
35. Expression profiling of a hemopoietic cell survival transcriptome implicates osteopontin as a functional prognostic factor in AML.
- Author
-
Powell JA, Thomas D, Barry EF, Kok CH, McClure BJ, Tsykin A, To LB, Brown A, Lewis ID, Herbert K, Goodall GJ, Speed TP, Asou N, Jacob B, Osato M, Haylock DN, Nilsson SK, D'Andrea RJ, Lopez AF, and Guthridge MA
- Subjects
- Adult, Aged, Cell Survival, Cytokine Receptor Common beta Subunit metabolism, Female, Gene Expression Regulation, Leukemic, Gene Knockdown Techniques, Gene Regulatory Networks, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells pathology, Humans, Leukemia, Myeloid metabolism, Leukemia, Myeloid mortality, Leukemia, Myeloid pathology, Male, Middle Aged, Neoplasm Proteins genetics, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Osteopontin biosynthesis, Osteopontin genetics, Phosphatidylinositol 3-Kinases physiology, Phosphoinositide-3 Kinase Inhibitors, Phosphoserine metabolism, Prognosis, RNA, Messenger biosynthesis, RNA, Messenger genetics, RNA, Neoplasm biosynthesis, RNA, Neoplasm genetics, RNA, Small Interfering pharmacology, Signal Transduction genetics, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured metabolism, Tumor Cells, Cultured pathology, Gene Expression Profiling, Hematopoietic Stem Cells metabolism, Leukemia, Myeloid genetics, Neoplasm Proteins physiology, Osteopontin physiology
- Abstract
Deregulated cell survival programs are a classic hallmark of cancer. We have previously identified a serine residue (Ser585) in the betac subunit of the granulocyte-macrophage colony-stimulating factor receptor that selectively and independently promotes cell survival. We now show that Ser585 phosphorylation is constitutive in 20 (87%) of 23 acute myeloid leukemia (AML) patient samples, indicating that this survival-only pathway is frequently deregulated in leukemia. We performed a global expression screen to identify gene targets of this survival pathway and report a 138-gene betac Ser585-regulated transcriptome. Pathway analysis defines a gene network enriched for PI3-kinase target genes and a cluster of genes involved in cancer and cell survival. We show that one such gene, osteopontin (OPN), is a functionally relevant target of the Ser585-survival pathway as shown by siRNA-mediated knockdown of OPN expression that induces cell death in both AML blasts and CD34(+)CD38(-)CD123(+) leukemic progenitors. Increased expression of OPN at diagnosis is associated with poor prognosis with multivariate analysis indicating that it is an independent predictor of overall patient survival in normal karyotype AML (n = 60; HR = 2.2; P = .01). These results delineate a novel cytokine-regulated Ser585/PI3-kinase signaling network that is deregulated in AML and identify OPN as a potential prognostic and therapeutic target.
- Published
- 2009
- Full Text
- View/download PDF
36. The structure of the GM-CSF receptor complex reveals a distinct mode of cytokine receptor activation.
- Author
-
Hansen G, Hercus TR, McClure BJ, Stomski FC, Dottore M, Powell J, Ramshaw H, Woodcock JM, Xu Y, Guthridge M, McKinstry WJ, Lopez AF, and Parker MW
- Subjects
- Amino Acid Sequence, Crystallography, Humans, Models, Molecular, Molecular Sequence Data, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a pleiotropic cytokine that controls the production and function of blood cells, is deregulated in clinical conditions such as rheumatoid arthritis and leukemia, yet offers therapeutic value for other diseases. Its receptors are heterodimers consisting of a ligand-specific alpha subunit and a betac subunit that is shared with the interleukin (IL)-3 and IL-5 receptors. How signaling is initiated remains an enigma. We report here the crystal structure of the human GM-CSF/GM-CSF receptor ternary complex and its assembly into an unexpected dodecamer or higher-order complex. Importantly, mutagenesis of the GM-CSF receptor at the dodecamer interface and functional studies reveal that dodecamer formation is required for receptor activation and signaling. This unusual form of receptor assembly likely applies also to IL-3 and IL-5 receptors, providing a structural basis for understanding their mechanism of activation and for the development of therapeutics.
- Published
- 2008
- Full Text
- View/download PDF
37. Growth factor pleiotropy is controlled by a receptor Tyr/Ser motif that acts as a binary switch.
- Author
-
Guthridge MA, Powell JA, Barry EF, Stomski FC, McClure BJ, Ramshaw H, Felquer FA, Dottore M, Thomas DT, To B, Begley CG, and Lopez AF
- Subjects
- 14-3-3 Proteins metabolism, Amino Acid Motifs, Animals, Binding Sites, CD11b Antigen metabolism, Cell Differentiation, Cell Line, Cell Survival, Cyclic AMP-Dependent Protein Kinases metabolism, Humans, Leukemia, Myeloid metabolism, Mice, Mice, Knockout, Mutation, Phosphorylation, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Serine metabolism, Signal Transduction, Tyrosine metabolism, Cell Proliferation, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor physiology
- Abstract
Pleiotropism is a hallmark of cytokines and growth factors; yet, the underlying mechanisms are not clearly understood. We have identified a motif in the granulocyte macrophage-colony-stimulating factor receptor composed of a tyrosine and a serine residue that functions as a binary switch for the independent regulation of multiple biological activities. Signalling occurs either through Ser585 at lower cytokine concentrations, leading to cell survival only, or through Tyr577 at higher cytokine concentrations, leading to cell survival as well as proliferation, differentiation or functional activation. The phosphorylation of Ser585 and Tyr577 is mutually exclusive and occurs via a unidirectional mechanism that involves protein kinase A and tyrosine kinases, respectively, and is deregulated in at least some leukemias. We have identified similar Tyr/Ser motifs in other cell surface receptors, suggesting that such signalling switches may play important roles in generating specificity and pleiotropy in other biological systems.
- Published
- 2006
- Full Text
- View/download PDF
38. The phosphoserine-585-dependent pathway of the GM-CSF/IL-3/IL-5 receptors mediates hematopoietic cell survival through activation of NF-kappaB and induction of bcl-2.
- Author
-
Guthridge MA, Barry EF, Felquer FA, McClure BJ, Stomski FC, Ramshaw H, and Lopez AF
- Subjects
- Animals, Cell Division, Cell Line, Cell Survival, Gene Expression Regulation, Humans, Mice, Mutagenesis, Site-Directed, Phosphatidylinositol 3-Kinases metabolism, Phosphoserine chemistry, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Receptors, Interleukin chemistry, Receptors, Interleukin genetics, Receptors, Interleukin-3 chemistry, Receptors, Interleukin-3 genetics, Receptors, Interleukin-5, Signal Transduction, Genes, bcl-2, NF-kappa B metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Interleukin metabolism, Receptors, Interleukin-3 metabolism, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic metabolism
- Abstract
We have recently identified a novel mechanism of hematopoietic cell survival that involves site-specific serine phosphorylation of the common beta subunit (beta(c)) of the granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3), and IL-5 receptors. However, the downstream components of this pathway are not known, nor is its relationship to survival signals triggered by tyrosine phosphorylation of the receptor clear. We have now found that phosphorylation of Ser585 of beta(c) in response to GM-CSF recruited 14-3-3 and phosphatidyl inositol 3-OH kinase (PI 3-kinase) to the receptor, while phosphorylation of the neighboring Tyr577 within this "viability domain" promoted the activation of both Src homology and collagen (Shc) and Ras. These are independent processes as demonstrated by the intact reactivity of phosphospecific anti-Ser585 and anti-Tyr577 antibodies on the cytotoxic T-lymphocyte-ecotrophic retroviral receptor neomycin (CTL-EN) mutants beta(c)Tyr577Phe and beta(c)Ser585Gly, respectively. Importantly, while mutants in which either Ser585 (beta(c)Ser585Gly) or all tyrosines (beta(c)F8) were substituted showed a defect in Akt phosphorylation, nuclear factor kappaB (NF-kappaB) activation, bcl-2 induction, and cell survival, the mutant beta(c)Tyr577Phe was defective in Shc, Ras, and extracellular signal-related kinase (ERK) activation, but supported CTL-EN cell survival in response to GM-CSF. These results demonstrate that both serine and tyrosine phosphorylation pathways play a role in hematopoietic cell survival, are initially independent of each other, and converge on NF-kappaB to promote bcl-2 expression.
- Published
- 2004
- Full Text
- View/download PDF
39. Molecular assembly of the ternary granulocyte-macrophage colony-stimulating factor receptor complex.
- Author
-
McClure BJ, Hercus TR, Cambareri BA, Woodcock JM, Bagley CJ, Howlett GJ, and Lopez AF
- Subjects
- Animals, Baculoviridae genetics, Binding Sites, Chromatography, Gel, Cloning, Molecular, DNA, Complementary genetics, Dimerization, Electrophoresis, Polyacrylamide Gel, Humans, Isotope Labeling, Molecular Structure, Phosphorus Radioisotopes, Polymerase Chain Reaction, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Recombinant Proteins biosynthesis, Solubility, Spectrometry, Mass, Electrospray Ionization, Spodoptera metabolism, Transfection, Ultracentrifugation, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics
- Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic cytokine that stimulates the production and functional activity of granulocytes and macrophages, properties that have encouraged its clinical use in bone marrow transplantation and in certain infectious diseases. Despite the importance of GM-CSF in regulating myeloid cell numbers and function, little is known about the exact composition and mechanism of assembly of the GM-CSF receptor complex. We have now produced soluble forms of the GM-CSF receptor alpha chain (sGMRalpha) and beta chain (sbetac) and utilized GM-CSF, the GM-CSF antagonist E21R (Glu21Arg), and the betac-blocking monoclonal antibody BION-1 to define the molecular assembly of the GM-CSF receptor complex. We found that GM-CSF and E21R were able to form low-affinity, binary complexes with sGMRalpha, each having a stoichiometry of 1:1. Importantly, GM-CSF but not E21R formed a ternary complex with sGMRalpha and sbetac, and this complex could be disrupted by E21R. Significantly, size-exclusion chromatography, analytical ultracentrifugation, and radioactive tracer experiments indicated that the ternary complex is composed of one sbetac dimer with a single molecule each of sGMRalpha and of GM-CSF. In addition, a hitherto unrecognized direct interaction between betac and GM-CSF was detected that was absent with E21R and was abolished by BION-1. These results demonstrate a novel mechanism of cytokine receptor assembly likely to apply also to interleukin-3 (IL-3) and IL-5 and have implications for our molecular understanding and potential manipulation of GM-CSF activation of its receptor.
- Published
- 2003
- Full Text
- View/download PDF
40. GM-CSF binding to its receptor induces oligomerisation of the common beta-subunit.
- Author
-
McClure BJ, Woodcock JM, Harrison-Findik D, Lopez AF, and D'Andrea RJ
- Subjects
- Animals, Cell Line, Cytokine Receptor Common beta Subunit, Dimerization, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Granulocyte-Macrophage Colony-Stimulating Factor physiology, Humans, Mice, Protein Binding physiology, Receptors, Cell Surface genetics, Receptors, Cell Surface physiology, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor physiology, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins physiology, Signal Transduction physiology, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Cell Surface metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
The stoichiometry of the granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor complex is still unresolved. We have utilised a sensitive, functional assay for receptor homodimerisation to show that GM-CSF induces dimerisation of the common signalling subunit, hbeta(c). We generated a chimeric cytokine receptor in which the extracellular and transmembrane domains of hbeta(c)are fused to the cytoplasmic domain of erythropoietin receptor (EPO-R). Given that to induce EPO-R activation and mitogenic signalling there is a requirement for formation of a specific homodimeric complex, we reasoned that the cytoplasmic domain of EPO-R could be utilised as a highly sensitive reporter for functional homodimer formation. We show that, in the presence of a cytoplasmically truncated GM-CSF alpha-subunit, the hbetac-EPO receptor chimera transduces a mitogenic signal in BaF-B03 in response to GM-CSF. This is consistent with formation of a hbeta(c)homodimer following GM-CSF binding and implies that ligand stimulation induces formation of a higher order complex that contains the hbeta(c)homodimer., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
41. Site-specific serine phosphorylation of the IL-3 receptor is required for hemopoietic cell survival.
- Author
-
Guthridge MA, Stomski FC, Barry EF, Winnall W, Woodcock JM, McClure BJ, Dottore M, Berndt MC, and Lopez AF
- Subjects
- 14-3-3 Proteins, Amino Acid Sequence, Animals, Binding Sites, Cell Division, Cell Line, Cell Survival, Cyclic AMP-Dependent Protein Kinases metabolism, Hematopoietic System metabolism, Interleukin-3 pharmacology, Mice, Mutagenesis, Site-Directed, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Proteins metabolism, Receptors, Interleukin-3 chemistry, Receptors, Interleukin-3 genetics, Serine metabolism, Signal Transduction, Hematopoietic System cytology, Hematopoietic System immunology, Receptors, Interleukin-3 metabolism, Tyrosine 3-Monooxygenase
- Abstract
In the hemopoietic compartment, IL-3, GM-CSF, and IL-5 receptors are major transducers of survival signals; however, the receptor-proximal events that determine this vital function have not been defined. We have found that IL-3 stimulation induces phosphorylation of Ser-585 of beta(c). This promotes the association of phospho-Ser-585 of beta(c) with 14-3-3 and the p85 subunit of PI 3-K. Mutation of Ser-585 specifically impairs the PI 3-K signaling pathway and reduces cell survival in response to IL-3. These results define a distinct IL-3 receptor-mediated survival pathway regulated by site-specific receptor serine phosphorylation and 14-3-3 binding and suggest that this novel mode of signaling may be utilized by disparate transmembrane receptors that have as a common theme the transduction of survival signals.
- Published
- 2000
42. Structure of the activation domain of the GM-CSF/IL-3/IL-5 receptor common beta-chain bound to an antagonist.
- Author
-
Rossjohn J, McKinstry WJ, Woodcock JM, McClure BJ, Hercus TR, Parker MW, Lopez AF, and Bagley CJ
- Subjects
- Antibodies, Monoclonal metabolism, Antibodies, Monoclonal pharmacology, Binding Sites, Cell Line, Epitope Mapping, Humans, Ligands, Molecular Sequence Data, Protein Binding, Protein Conformation, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Receptors, Interleukin antagonists & inhibitors, Receptors, Interleukin metabolism, Receptors, Interleukin-3 antagonists & inhibitors, Receptors, Interleukin-3 metabolism, Receptors, Interleukin-5, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Receptors, Interleukin chemistry, Receptors, Interleukin-3 chemistry
- Abstract
Heterodimeric cytokine receptors generally consist of a major cytokine-binding subunit and a signaling subunit. The latter can transduce signals by more than 1 cytokine, as exemplified by the granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2 (IL-2), and IL-6 receptor systems. However, often the signaling subunits in isolation are unable to bind cytokines, a fact that has made it more difficult to obtain structural definition of their ligand-binding sites. This report details the crystal structure of the ligand-binding domain of the GM-CSF/IL-3/IL-5 receptor beta-chain (beta(c)) signaling subunit in complex with the Fab fragment of the antagonistic monoclonal antibody, BION-1. This is the first single antagonist of all 3 known eosinophil-producing cytokines, and it is therefore capable of regulating eosinophil-related diseases such as asthma. The structure reveals a fibronectin type III domain, and the antagonist-binding site involves major contributions from the loop between the B and C strands and overlaps the cytokine-binding site. Furthermore, tyrosine(421) (Tyr(421)), a key residue involved in receptor activation, lies in the neighboring loop between the F and G strands, although it is not immediately adjacent to the cytokine-binding residues in the B-C loop. Interestingly, functional experiments using receptors mutated across these loops demonstrate that they are cooperatively involved in full receptor activation. The experiments, however, reveal subtle differences between the B-C loop and Tyr(421), which is suggestive of distinct functional roles. The elucidation of the structure of the ligand-binding domain of beta(c) also suggests how different cytokines recognize a single receptor subunit, which may have implications for homologous receptor systems. (Blood. 2000;95:2491-2498)
- Published
- 2000
43. The human granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor exists as a preformed receptor complex that can be activated by GM-CSF, interleukin-3, or interleukin-5.
- Author
-
Woodcock JM, McClure BJ, Stomski FC, Elliott MJ, Bagley CJ, and Lopez AF
- Subjects
- Animals, CHO Cells, Cricetinae, Humans, Janus Kinase 2, Kinetics, Models, Molecular, Molecular Weight, Phosphorylation, Protein Conformation, Protein-Tyrosine Kinases metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor chemistry, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor drug effects, Solubility, Surface Properties, Tumor Cells, Cultured, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Interleukin-3 pharmacology, Interleukin-5 pharmacology, Proto-Oncogene Proteins, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism
- Abstract
The granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor is expressed on normal and malignant hematopoietic cells as well as on cells from other organs in which it transduces a variety of functions. Despite the widespread expression and pleiotropic nature of the GM-CSF receptor, little is known about its assembly and activation mechanism. Using a combination of biochemical and functional approaches, we have found that the human GM-CSF receptor exists as an inducible complex, analogous to the interleukin-3 (IL-3) receptor, and also as a preformed complex, unlike the IL-3 receptor or indeed other members of the cytokine receptor superfamily. We found that monoclonal antibodies to the GM-CSF receptor alpha chain (GMR alpha) and to the common beta chain of the GM-CSF, IL-3, and IL-5 receptors (beta(c)) immunoprecipitated both GMR alpha and beta(c) from the surface of primary myeloid cells, myeloid cell lines, and transfected cells in the absence of GM-CSF. Further association of the two chains could be induced by the addition of GM-CSF. The preformed complex required only the extracellular regions of GMR alpha and beta(c), as shown by the ability of soluble beta(c) to associate with membrane-anchored GMR alpha or soluble GMR alpha. Kinetic experiments on eosinophils and monocytes with radiolabeled GM-CSF, IL-3, and IL-5 showed association characteristics unique to GM-CSF. Significantly, receptor phosphorylation experiments showed that not only GM-CSF but also IL-3 and IL-5 stimulated the phosphorylation of GMR alpha-associated beta(c). These results indicate a pattern of assembly of the heterodimeric GM-CSF receptor that is unique among receptors of the cytokine receptor superfamily. These results also suggest that the preformed GM-CSF receptor complex mediates the instantaneous binding of GM-CSF and is a target of phosphorylation by IL-3 and IL-5, raising the possibility that some of the biologic activities of IL-3 and IL-5 are mediated through the GM-CSF receptor complex.
- Published
- 1997
44. Increased levels of megakaryocyte progenitors in peripheral blood mobilised by chemotherapy and/or haemopoietic growth factor protocols.
- Author
-
Dyson PG, Jackson KA, McClure BJ, Rawling TP, and To LB
- Subjects
- Adult, Antineoplastic Agents, Alkylating administration & dosage, Blood Cell Count, Clinical Protocols, Colony-Forming Units Assay, Cyclophosphamide administration & dosage, Female, Hematopoietic Cell Growth Factors administration & dosage, Hematopoietic Stem Cells cytology, Humans, Male, Megakaryocytes cytology, Neoplasms blood, Neoplasms therapy, Platelet Count, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells drug effects, Megakaryocytes drug effects
- Abstract
We have quantitated colony-forming unit megakaryocyte, (CFU-Mk), burst-forming unit megakaryocyte, (BFU-Mk), colony-forming unit granulocyte-macrophage (CFU-GM), and CD34+ cells in 98 mobilised PB samples from 53 patients mobilised by one of six protocols, including myelosuppressive chemotherapy alone (n = 22), or in combination with recombinant haemopoietic growth factors (n = 32), and growth factors alone (n = 17) or in combination (n = 27). The frequency of megakaryocyte progenitors (total Mk = CFU-Mk + BFU-Mk) in mobilised PB (mean 356, range 0-3240/10(6)) was similar to that in steady-state BM (mean 429, range 0-3315/10(6) n = 45). The levels of total Mk in mobilised PB (mean 1509, range 0-36 099/ml) showed a mean 75-fold increase compared with steady state PB (mean 20, range 0-86/ml, n = 15). In mobilised PB the levels of CFU-Mk were significantly correlated with levels of BFU-Mk (rs = 0.71, P < 0.0001) and the levels of megakaryocyte progenitors correlated significantly with those of myeloid progenitors (rs = 0.59, P < 0.0001) and CD34+ cells (rs = 0.69, P < 0.0001). The mobilisation of megakaryocyte progenitors into the circulation in response to high-dose chemotherapy and/or haemopoietic growth factors contributes to an understanding of the rapid platelet recovery following PBSC transplantation and suggests that the measurement of megakaryocyte progenitors may be a useful indicator for platelet reconstitutive capacity.
- Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.