Medical Biology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 10
  • Item
    Thumbnail Image
    Therapeutic Effects of a TANK-Binding Kinase 1 Inhibitor in Germinal Center-Driven Collagen-Induced Arthritis
    Louis, C ; Ngo, D ; D'Silva, DB ; Hansen, J ; Phillipson, L ; Jousset, H ; Novello, P ; Segal, D ; Lawlor, KE ; Burns, CJ ; Wicks, IP (WILEY, 2019-01)
    OBJECTIVE: The production of class-switched high-affinity autoantibodies derived from organized germinal centers (GCs) is a hallmark of many autoimmune inflammatory diseases, including rheumatoid arthritis (RA). TANK-binding kinase 1 (TBK-1) is a serine/threonine kinase involved in the maturation of GC follicular helper T (Tfh) cells downstream of inducible costimulator signaling. We undertook this study to assess the therapeutic potential of TBK-1 inhibition using the small-molecule inhibitor WEHI-112 in antibody-dependent models of inflammatory arthritis. METHODS: Using the models of collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and K/BxN serum-transfer-induced arthritis (STIA), we determined the effectiveness of WEHI-112 at inhibiting clinical and histologic features of arthritis in C57BL/6 and DBA/1 mice. We used immunohistochemistry to characterize GC populations during CIA development, and we used enzyme-linked immunosorbent assays to determine levels of Ig autoantibodies in WEHI-112-treated mice compared to vehicle-treated mice. RESULTS: WEHI-112, a tool compound that is semiselective for TBK-1 but that also has activity against IKKε and JAK2, abolished TBK-1-dependent activation of interferon (IFN) regulatory factor 3 and inhibited type I IFN responses in vitro. In vivo, treatment with WEHI-112 selectively abrogated clinical and histologic features of established, antibody-dependent CIA, but had minimal effects on an antibody-independent model of AIA or on K/BxN STIA. In keeping with these findings, WEHI-112 reduced arthritogenic type II collagen-specific IgG1 and IgG2b antibody production. Furthermore, WEHI-112 altered the GC Tfh cell phenotype and GC B cell function in CIA. CONCLUSION: We report that TBK-1 inhibition using WEHI-112 abrogated antibody-dependent CIA. As WEHI-112 failed to inhibit non-antibody-driven joint inflammation, we conclude that the major effect of this compound was most likely the targeting of TBK-1-mediated mechanisms in the GC reaction. This approach may have therapeutic potential in RA and in other GC-associated autoantibody-driven inflammatory diseases.
  • Item
    Thumbnail Image
    TBK1 and IKKε Act Redundantly to Mediate STING-Induced NF-κB Responses in Myeloid Cells
    Balka, KR ; Louis, C ; Saunders, TL ; Smith, AM ; Calleja, DJ ; D'Silva, DB ; Moghaddas, F ; Tailler, M ; Lawlor, KE ; Zhan, Y ; Burns, CJ ; Wicks, IP ; Miner, JJ ; Kile, BT ; Masters, SL ; De Nardo, D (CELL PRESS, 2020-04-07)
    Stimulator of Interferon Genes (STING) is a critical component of host innate immune defense but can contribute to chronic autoimmune or autoinflammatory disease. Once activated, the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS)-STING pathway induces both type I interferon (IFN) expression and nuclear factor-κB (NF-κB)-mediated cytokine production. Currently, these two signaling arms are thought to be mediated by a single upstream kinase, TANK-binding kinase 1 (TBK1). Here, using genetic and pharmacological approaches, we show that TBK1 alone is dispensable for STING-induced NF-κB responses in human and mouse immune cells, as well as in vivo. We further demonstrate that TBK1 acts redundantly with IκB kinase ε (IKKε) to drive NF-κB upon STING activation. Interestingly, we show that activation of IFN regulatory factor 3 (IRF3) is highly dependent on TBK1 kinase activity, whereas NF-κB is significantly less sensitive to TBK1/IKKε kinase inhibition. Our work redefines signaling events downstream of cGAS-STING. Our findings further suggest that cGAS-STING will need to be targeted directly to effectively ameliorate the inflammation underpinning disorders associated with STING hyperactivity.
  • Item
    Thumbnail Image
    Translation inhibitors induce cell death by multiple mechanisms and Mcl-1 reduction is only a minor contributor
    Lindqvist, LM ; Vikstroem, I ; Chambers, JM ; McArthur, K ; Anderson, MA ; Henley, KJ ; Happo, L ; Cluse, L ; Johnstone, RW ; Roberts, AW ; Kile, BT ; Croker, BA ; Burns, CJ ; Rizzacasa, MA ; Strasser, A ; Huang, DCS (NATURE PUBLISHING GROUP, 2012-10)
    There is significant interest in treating cancers by blocking protein synthesis, to which hematological malignancies seem particularly sensitive. The translation elongation inhibitor homoharringtonine (Omacetaxine mepesuccinate) is undergoing clinical trials for chronic myeloid leukemia, whereas the translation initiation inhibitor silvestrol has shown promise in mouse models of cancer. Precisely how these compounds induce cell death is unclear, but reduction in Mcl-1, a labile pro-survival Bcl-2 family member, has been proposed to constitute the critical event. Moreover, the contribution of translation inhibitors to neutropenia and lymphopenia has not been precisely defined. Herein, we demonstrate that primary B cells and neutrophils are highly sensitive to translation inhibitors, which trigger the Bax/Bak-mediated apoptotic pathway. However, contrary to expectations, reduction of Mcl-1 did not significantly enhance cytotoxicity of these compounds, suggesting that it does not have a principal role and cautions that strong correlations do not always signify causality. On the other hand, the killing of T lymphocytes was less dependent on Bax and Bak, indicating that translation inhibitors can also induce cell death via alternative mechanisms. Indeed, loss of clonogenic survival proved to be independent of the Bax/Bak-mediated apoptosis altogether. Our findings warn of potential toxicity as these translation inhibitors are cytotoxic to many differentiated non-cycling cells.
  • Item
    Thumbnail Image
    BET inhibitors induce apoptosis through a MYC independent mechanism and synergise with CDK inhibitors to kill osteosarcoma cells
    Baker, EK ; Taylor, S ; Gupte, A ; Sharp, PP ; Walia, M ; Walsh, NC ; Zannettino, ACW ; Chalk, AM ; Burns, CJ ; Walkley, CR (NATURE PORTFOLIO, 2015-05-06)
    Osteosarcoma (OS) survival rates have plateaued in part due to a lack of new therapeutic options. Here we demonstrate that bromodomain inhibitors (BETi), JQ1, I-BET151, I-BET762, exert potent anti-tumour activity against primary and established OS cell lines, mediated by inhibition of BRD4. Strikingly, unlike previous observations in long-term established human OS cell lines, the antiproliferative activity of JQ1 in primary OS cells was driven by the induction of apoptosis, not cell cycle arrest. In further contrast, JQ1 activity in OS was mediated independently of MYC downregulation. We identified that JQ1 suppresses the transcription factor FOSL1 by displacement of BRD4 from its locus. Loss of FOSL1 phenocopied the antiproliferative effects of JQ1, identifying FOSL1 suppression as a potential novel therapeutic approach for OS. As a monotherapy JQ1 demonstrated significant anti-tumour activity in vivo in an OS graft model. Further, combinatorial treatment approaches showed that JQ1 increased the sensitivity of OS cells to doxorubicin and induced potent synergistic activity when rationally combined with CDK inhibitors. The greater level of activity achieved with the combination of BETi with CDK inhibitors demonstrates the efficacy of this combination therapy. Taken together, our studies show that BET inhibitors are a promising new therapeutic for OS.
  • Item
    Thumbnail Image
    Defining a therapeutic window for kinase inhibitors in leukemia to avoid neutropenia
    McArthur, K ; D'Cruz, AA ; Segal, D ; Lackovic, K ; Wilks, AF ; O'Donnell, JA ; Nowell, CJ ; Gerlic, M ; Huang, DCS ; Burns, CJ ; Croker, BA (IMPACT JOURNALS LLC, 2017-08-29)
    Neutropenia represents one of the major dose-limiting toxicities of many current cancer therapies. To circumvent the off-target effects of cytotoxic chemotherapeutics, kinase inhibitors are increasingly being used as an adjunct therapy to target leukemia. In this study, we conducted a screen of leukemic cell lines in parallel with primary neutrophils to identify kinase inhibitors with the capacity to induce apoptosis of myeloid and lymphoid cell lines whilst sparing primary mouse and human neutrophils. We have utilized a high-throughput live cell imaging platform to demonstrate that cytotoxic drugs have limited effects on neutrophil viability but are toxic to hematopoietic progenitor cells, with the exception of the topoisomerase I inhibitor SN-38. The parallel screening of kinase inhibitors revealed that mouse and human neutrophil viability is dependent on cyclin-dependent kinase (CDK) activity but surprisingly only partially dependent on PI3 kinase and JAK/STAT signaling, revealing dominant pathways contributing to neutrophil viability. Mcl-1 haploinsufficiency sensitized neutrophils to CDK inhibition, demonstrating that Mcl-1 is a direct target for CDK inhibitors. This study reveals a therapeutic window for the kinase inhibitors BEZ235, BMS-3, AZD7762, and (R)-BI-2536 to induce apoptosis of leukemia cell lines whilst maintaining immunocompetence and hemostasis.
  • Item
    No Preview Available
    HBO1 is required for the maintenance of leukaemia stem cells
    MacPherson, L ; Anokye, J ; Yeung, MM ; Lam, EYN ; Chan, Y-C ; Weng, C-F ; Yeh, P ; Knezevic, K ; Butler, MS ; Hoegl, A ; Chan, K-L ; Burr, ML ; Gearing, LJ ; Willson, T ; Liu, J ; Choi, J ; Yang, Y ; Bilardi, RA ; Falk, H ; Nghi, N ; Stupple, PA ; Peat, TS ; Zhang, M ; de Silva, M ; Carrasco-Pozo, C ; Avery, VM ; Khoo, PS ; Dolezal, O ; Dennis, ML ; Nuttall, S ; Surjadi, R ; Newman, J ; Ren, B ; Leaver, DJ ; Sun, Y ; Baell, JB ; Dovey, O ; Vassiliou, GS ; Grebien, F ; Dawson, S-J ; Street, IP ; Monahan, BJ ; Burns, CJ ; Choudhary, C ; Blewitt, ME ; Voss, AK ; Thomas, T ; Dawson, MA (NATURE PORTFOLIO, 2020-01-09)
    Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by transcriptional dysregulation that results in a block in differentiation and increased malignant self-renewal. Various epigenetic therapies aimed at reversing these hallmarks of AML have progressed into clinical trials, but most show only modest efficacy owing to an inability to effectively eradicate leukaemia stem cells (LSCs)1. Here, to specifically identify novel dependencies in LSCs, we screened a bespoke library of small hairpin RNAs that target chromatin regulators in a unique ex vivo mouse model of LSCs. We identify the MYST acetyltransferase HBO1 (also known as KAT7 or MYST2) and several known members of the HBO1 protein complex as critical regulators of LSC maintenance. Using CRISPR domain screening and quantitative mass spectrometry, we identified the histone acetyltransferase domain of HBO1 as being essential in the acetylation of histone H3 at K14. H3 acetylated at K14 (H3K14ac) facilitates the processivity of RNA polymerase II to maintain the high expression of key genes (including Hoxa9 and Hoxa10) that help to sustain the functional properties of LSCs. To leverage this dependency therapeutically, we developed a highly potent small-molecule inhibitor of HBO1 and demonstrate its mode of activity as a competitive analogue of acetyl-CoA. Inhibition of HBO1 phenocopied our genetic data and showed efficacy in a broad range of human cell lines and primary AML cells from patients. These biological, structural and chemical insights into a therapeutic target in AML will enable the clinical translation of these findings.
  • Item
    Thumbnail Image
    A constricted opening in Kir channels does not impede potassium conduction
    Black, KA ; He, S ; Jin, R ; Miller, DM ; Bolla, JR ; Clarke, OB ; Johnson, P ; Windley, M ; Burns, CJ ; Hill, AP ; Laver, D ; Robinson, C ; Smith, BJ ; Gulbis, JM (NATURE PUBLISHING GROUP, 2020-06-15)
    The canonical mechanistic model explaining potassium channel gating is of a conformational change that alternately dilates and constricts a collar-like intracellular entrance to the pore. It is based on the premise that K+ ions maintain a complete hydration shell while passing between the transmembrane cavity and cytosol, which must be accommodated. To put the canonical model to the test, we locked the conformation of a Kir K+ channel to prevent widening of the narrow collar. Unexpectedly, conduction was unimpaired in the locked channels. In parallel, we employed all-atom molecular dynamics to simulate K+ ions moving along the conduction pathway between the lower cavity and cytosol. During simulations, the constriction did not significantly widen. Instead, transient loss of some water molecules facilitated K+ permeation through the collar. The low free energy barrier to partial dehydration in the absence of conformational change indicates Kir channels are not gated by the canonical mechanism.
  • Item
    No Preview Available
    Biochemical and Structural Insights into Doublecortin-like Kinase Domain 1
    Patel, O ; Dai, W ; Mentzel, M ; Griffin, MDW ; Serindoux, J ; Gay, Y ; Fischer, S ; Sterle, S ; Kropp, A ; Burns, CJ ; Ernst, M ; Buchert, M ; Lucet, IS (CELL PRESS, 2016-09-06)
    Doublecortin-like kinase 1 (DCLK1) is a serine/threonine kinase that belongs to the family of microtubule-associated proteins. Originally identified for its role in neurogenesis, DCLK1 has recently been shown to regulate biological processes outside of the CNS. DCLK1 is among the 15 most common putative driver genes for gastric cancers and is highly mutated across various other human cancers. However, our present understanding of how DCLK1 dysfunction leads to tumorigenesis is limited. Here, we provide evidence that DCLK1 kinase activity negatively regulates microtubule polymerization. We present the crystal structure of the DCLK1 kinase domain at 1.7 Å resolution, providing detailed insight into the ATP-binding site that will serve as a framework for future drug design. This structure also allowed for the mapping of cancer-causing mutations within the kinase domain, suggesting that a loss of kinase function may contribute to tumorigenesis.
  • Item
    Thumbnail Image
    HSP90 Inhibition Suppresses Lipopolysaccharide-Induced Lung Inflammation In Vivo
    Lilja, A ; Weeden, CE ; McArthur, K ; Thao, N ; Donald, A ; Wong, ZX ; Dousha, L ; Bozinovski, S ; Vlahos, R ; Burns, CJ ; Asselin-Labat, M-L ; Anderson, GP ; Ryffel, B (PUBLIC LIBRARY SCIENCE, 2015-01-23)
    Inflammation is an important component of cancer diathesis and treatment-refractory inflammation is a feature of many chronic degenerative lung diseases. HSP90 is a 90kDa protein which functions as an ATP-dependent molecular chaperone that regulates the signalling conformation and expression of multiple protein client proteins especially oncogenic mediators. HSP90 inhibitors are in clinical development as cancer therapies but the myeleosuppressive and neutropenic effect of first generation geldanamycin-class inhibitors has confounded studies on the effects on HSP90 inhibitors on inflammation. To address this we assessed the ability of Ganetespib, a non-geldanamycin HSP90 blocker, to suppress lipopolysaccharide (LPS)-induced cellular infiltrates, proteases and inflammatory mediator and transcriptional profiles. Ganetespib (10-100 mg/kg, i.v.) did not directly cause myelosuppression, as assessed by video micrography and basal blood cell count, but it strongly and dose-dependently suppressed LPS-induced neutrophil mobilization into blood and neutrophil- and mononuclear cell-rich steroid-refractory lung inflammation. Ganetespib also suppressed B cell and NK cell accumulation, inflammatory cytokine and chemokine induction and MMP9 levels. These data identify non-myelosuppresssive HSP90 inhibitors as potential therapies for inflammatory diseases refractory to conventional therapy, in particular those of the lung.
  • Item
    Thumbnail Image
    Translation inhibitors induce cell death by multiple mechanisms and Mcl-1 reduction is only a minor contributor
    Lindqvist, L. M. ; Vikström, I. ; Chambers, J. M. ; McArthur, K. ; Anderson, M. Ann ; Henley, K. J. ; HAPPO, LINA ; Cluse, L. ; Johnstone, R. W. ; Roberts, A. W. ; Kile, B. T. ; Croker, B. A. ; Burns, C. J. ; Rizzacasa, M. A. ; STRASSER, ANDREAS ; Huang, D. C. S. (Nature/Macmillan, 2012)