A Novel Glycogen Synthase Kinase-3 Inhibitor Optimized for Acute

Total Page:16

File Type:pdf, Size:1020Kb

A Novel Glycogen Synthase Kinase-3 Inhibitor Optimized for Acute Published OnlineFirst May 9, 2016; DOI: 10.1158/1535-7163.MCT-15-0566 Small Molecule Therapeutics Molecular Cancer Therapeutics A Novel Glycogen Synthase Kinase-3 Inhibitor Optimized for Acute Myeloid Leukemia Differentiation Activity Sophia Hu1, Masumi Ueda2, Lindsay Stetson1, James Ignatz-Hoover1, Stephen Moreton1, Amit Chakrabarti3, Zhiqiang Xia3, Goutam Karan3, Marcos de Lima2, Mukesh K. Agrawal3,4, and David N. Wald1,3,5 Abstract Standard therapies used for the treatment of acute myeloid describe the discovery of a novel GSK3 inhibitor, GS87. GS87 leukemia (AML) are cytotoxic agents that target rapidly prolifer- was discovered in efforts to optimize GSK3 inhibition for AML ating cells. Unfortunately, this therapeutic approach has differentiation activity. Despite GS87's dramatic ability to induce limited efficacy and significant toxicity and the majority of AML AML differentiation, kinase profiling reveals its high specificity patients still die of their disease. In contrast to the poor prognosis in targeting GSK3 as compared with other kinases. GS87 demon- of most AML patients, most individuals with a rare subtype of strates high efficacy in a mouse AML model system and unlike AML, acute promyelocytic leukemia, can be cured by differenti- current AML therapeutics, exhibits little effect on normal bone ation therapy using regimens containing all-trans retinoic acid. marrow cells. GS87 induces potent differentiation by more GSK3 has been previously identified as a therapeutic target in effectively activating GSK3-dependent signaling components AML where its inhibition can lead to the differentiation and including MAPK signaling as compared with other GSK3 growth arrest of leukemic cells. Unfortunately, existing GSK3 inhibitors. GS87 is a novel GSK3 inhibitor with therapeutic inhibitors lead to suboptimal differentiation activity making potential as a differentiation agent for non-promyelocytic AML. them less useful as clinical AML differentiation agents. Here, we Mol Cancer Ther; 15(7); 1485–94. Ó2016 AACR. Introduction In efforts to find novel strategies for inducing differentiation in non-APL leukemic cells, we and others identified that inhibiting Acute myeloid leukemia (AML), the most common form of GSK3 leads to AML differentiation (3–5). GSK3 is a serine/ acute leukemia in adults, is a heterogeneous disorder defined by threonine kinase involved in multiple signaling pathways. Orig- the arrest of proliferation of immature hematopoietic cells (1). inally identified for its role in regulating glycogen synthesis, it has Standard AML chemotherapy leads to significant toxicity and is been found to play roles in many biologic processes such as frequently ineffective. Despite the fact that AML is characterized inflammation, stem cell maintenance, cell proliferation, and by a differentiation arrest, standard AML treatment targets the regulation of normal and aberrant hematopoiesis (6). In addition, rapid proliferation of leukemic cells. In contrast to the poor GSK3 plays important roles in a variety of key signaling pathways prognosis of the majority of AML patients, most individuals with such as Wnt/b-catenin, PI3K, mTOR, and MAPK signaling (6). a rare subtype of AML, acute promyelocytic leukemia (APL), can While pharmacologic inhibition of GSK3 has historically been be cured by differentiation therapy using regimens containing all- studied in a variety of diseases (7), in recent years, GSK3 has been trans retinoic acid (ATRA; ref. 2). found to play an important role in cancer. GSK3 has been demonstrated to be overexpressed and hyperactivated in a variety 1 Department of Pathology, Case Western Reserve University, Cleve- of solid cancers. In addition, overexpression of GSK3 has been land, Ohio. 2Department of Hematology and Oncology, University Hospitals Case Medical Center and Case Western Reserve University, associated with resistance to therapy and poor prognosis (6, 8). Cleveland, Ohio. 3Invenio Therapeutics Inc., Cleveland, Ohio. 4MirX Prior studies have identified that inhibition of GSK3 results in 5 Pharmaceuticals, Cleveland, Ohio. University Hospitals Case Medical AML cell growth inhibition and monocytic differentiation (3–5, Center, Cleveland, Ohio. 9–12). Importantly, several studies have reported that targeting Note: Supplementary data for this article are available at Molecular Cancer GSK3 specifically impairs leukemic cell growth, but not normal Therapeutics Online (http://mct.aacrjournals.org/). bone marrow proliferation. Sophia Hu and Masumi Ueda contributed equally to this article. Because of the role of GSK3 in the molecular pathology of many Corresponding Authors: David N. Wald, Case Western Reserve University and diseases, development of specific inhibitors of this kinase with University Hospitals Case Medical Center, 2103 Cornell Road, Cleveland, OH therapeutic intentions has been pursued. While investigational 44106. Phone: 216-368-5668; Fax: 216-368-0495; E-mail: [email protected]; and GSK3 inhibitors exist, the potency and specificity of GSK3 inhibi- Mukesh K. Agarwal, Invenio Therapeutics, A262 Astecc Building, Lexington, KY tion is variable. Lithium (Li), the only FDA-approved GSK3 inhib- 40506. E-mail: [email protected] itor, has long been used clinically to treat bipolar disorder. Lithium þ doi: 10.1158/1535-7163.MCT-15-0566 directly binds GSK3 at an Mg2 binding site, leading to reversible Ó2016 American Association for Cancer Research. enzyme inhibition and inhibitory serine-9 phosphorylation www.aacrjournals.org 1485 Downloaded from mct.aacrjournals.org on October 1, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst May 9, 2016; DOI: 10.1158/1535-7163.MCT-15-0566 Hu et al. (13–15). LY2090314 and tideglusib are more specific small-mol- Proliferation and colony assay ecule GSK3 inhibitors utilized in humans in clinical trials (16, 17). Equal numbers of THP-1, HL-60, NB4, U937, and OCI cells Other small-molecule GSK3 inhibitors have been described, such were plated and treated with Li, SB or GS87 for 72 hours. Cells as SB415285 (SB), which inhibits GSK3 by competitive binding to were analyzed for proliferation using the CellTiter 96 Aqueous the ATP-binding site (18). In general, the GSK3 inhibitors One Solution Cell Proliferation Assay (Promega) as recom- described to date also inhibit many other kinases such as mended by the manufacturer using a Molecular Q3 Devices CDK2-cyclin A (CDK2A; ref. 19). Spectramax M2 plate reader. For the colony assay, after 72 hours, Although GSK3 inhibition has been demonstrated to induce the cells were washed with PBS, an equal number of viable cells evidence of AML differentiation, existing GSK3 inhibitors lead to were suspended in 0.03% noble agar (Sigma), the cells were suboptimal levels of AML differentiation that would likely pro- incubated for one week and number of colonies was counted. hibit their use as clinically useful differentiation agents. Because of the promise of differentiation therapy for AML, we aimed to Cell-cycle analysis identify a novel GSK3 inhibitor that was optimized for AML Cells were treated with Li, SB, or GS87 for 24 hours, fixed in differentiation activity. Here, we report the identification of a 90% methanol and incubated overnight at À20C. Cells were novel and highly specific GSK3 inhibitor, GS87, which induces then washed with PBS, treated with RNase (0.5 mg/mL) and extensive AML differentiation with selective growth inhibition of propidium iodide (50 mg/mL) in the dark for 60 to 90 minutes leukemic cells and exhibits promise as an AML differentiation at room temperature and analyzed by flow cytometry. therapeutic. Western blot analysis Materials and Methods Cells treated as indicated were washed with PBS, centrifuged, Reagents and cells and lysed with a triton-containing lysis buffer for whole cell The 2,000 compound small-molecule library was custom extracts. Protein lysates (50 mg/lane) were resolved on appropriate designed based upon similarities to known chemical and struc- SDS–PAGE gel and transferred to polyvinylidene difluoride mem- tural features of previously reported GSK3 inhibitors by Enamine brane (Millipore) using Bio-Rad transfer apparatus (Bio-Rad). The (Ukraine). Additional quantities of GS87 (T5729983) were membranes were blocked, incubated with the indicated primary obtained from Enamine. SB415286 (SB) was obtained from antibodies at 4 C overnight, and incubated with the appropriate Tocris Biosciences. Lithium (Li) was obtained from Sigma. horseradish peroxidase–conjugated secondary antibodies. Immu- SB203580 (SB203), PD08959 (PD), and SP600125 (SP) were noreactive protein bands were detected by enhanced chemilumi- obtained from Selleck Chem. MTT reagent was obtained from the nescence (Pierce) using XAR-5 film. CellTiter 96 Aqueous One Solution Cell Proliferation Assay (Promega). Phospho-Ser9 GSK3b (5558), GAPDH (5174), phos- RNA microarray pho-p38 (4511), phospho-ERK (4370), and phospho-JNK HL-60 cells were treated with Li, SB, or GS87 for 48 hours and (4668) was obtained from Cell Signaling Technology. b-Catenin, total RNA was isolated using TRIzol (Invitrogen). The cRNA was p21 (397), c-myc (40), c-MYB (7874), and MAFB (74521) anti- prepared using the TargetAmp-Nano Kit (Epicenter) and hybrid- bodies were obtained from Santa Cruz Biotechnology. OCI-AML3 ized to the human HT-12 Expression BeadChip (Illumina). cells were obtained from DSMZ, and HL-60, NB4, THP-1, U937, Microarray results were evaluated using Genomestudio (Illu- and HeLa cells were obtained from ATCC. All cell lines were
Recommended publications
  • Deregulated Gene Expression Pathways in Myelodysplastic Syndrome Hematopoietic Stem Cells
    Leukemia (2010) 24, 756–764 & 2010 Macmillan Publishers Limited All rights reserved 0887-6924/10 $32.00 www.nature.com/leu ORIGINAL ARTICLE Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells A Pellagatti1, M Cazzola2, A Giagounidis3, J Perry1, L Malcovati2, MG Della Porta2,MJa¨dersten4, S Killick5, A Verma6, CJ Norbury7, E Hellstro¨m-Lindberg4, JS Wainscoat1 and J Boultwood1 1LRF Molecular Haematology Unit, NDCLS, John Radcliffe Hospital, Oxford, UK; 2Department of Hematology Oncology, University of Pavia Medical School, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 3Medizinische Klinik II, St Johannes Hospital, Duisburg, Germany; 4Division of Hematology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; 5Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK; 6Albert Einstein College of Medicine, Bronx, NY, USA and 7Sir William Dunn School of Pathology, University of Oxford, Oxford, UK To gain insight into the molecular pathogenesis of the the World Health Organization.6,7 Patients with refractory myelodysplastic syndromes (MDS), we performed global gene anemia (RA) with or without ringed sideroblasts, according to expression profiling and pathway analysis on the hemato- poietic stem cells (HSC) of 183 MDS patients as compared with the the French–American–British classification, were subdivided HSC of 17 healthy controls. The most significantly deregulated based on the presence or absence of multilineage dysplasia. In pathways in MDS include interferon signaling, thrombopoietin addition, patients with RA with excess blasts (RAEB) were signaling and the Wnt pathways. Among the most signifi- subdivided into two categories, RAEB1 and RAEB2, based on the cantly deregulated gene pathways in early MDS are immuno- percentage of bone marrow blasts.
    [Show full text]
  • Gene Symbol Gene Description ACVR1B Activin a Receptor, Type IB
    Table S1. Kinase clones included in human kinase cDNA library for yeast two-hybrid screening Gene Symbol Gene Description ACVR1B activin A receptor, type IB ADCK2 aarF domain containing kinase 2 ADCK4 aarF domain containing kinase 4 AGK multiple substrate lipid kinase;MULK AK1 adenylate kinase 1 AK3 adenylate kinase 3 like 1 AK3L1 adenylate kinase 3 ALDH18A1 aldehyde dehydrogenase 18 family, member A1;ALDH18A1 ALK anaplastic lymphoma kinase (Ki-1) ALPK1 alpha-kinase 1 ALPK2 alpha-kinase 2 AMHR2 anti-Mullerian hormone receptor, type II ARAF v-raf murine sarcoma 3611 viral oncogene homolog 1 ARSG arylsulfatase G;ARSG AURKB aurora kinase B AURKC aurora kinase C BCKDK branched chain alpha-ketoacid dehydrogenase kinase BMPR1A bone morphogenetic protein receptor, type IA BMPR2 bone morphogenetic protein receptor, type II (serine/threonine kinase) BRAF v-raf murine sarcoma viral oncogene homolog B1 BRD3 bromodomain containing 3 BRD4 bromodomain containing 4 BTK Bruton agammaglobulinemia tyrosine kinase BUB1 BUB1 budding uninhibited by benzimidazoles 1 homolog (yeast) BUB1B BUB1 budding uninhibited by benzimidazoles 1 homolog beta (yeast) C9orf98 chromosome 9 open reading frame 98;C9orf98 CABC1 chaperone, ABC1 activity of bc1 complex like (S. pombe) CALM1 calmodulin 1 (phosphorylase kinase, delta) CALM2 calmodulin 2 (phosphorylase kinase, delta) CALM3 calmodulin 3 (phosphorylase kinase, delta) CAMK1 calcium/calmodulin-dependent protein kinase I CAMK2A calcium/calmodulin-dependent protein kinase (CaM kinase) II alpha CAMK2B calcium/calmodulin-dependent
    [Show full text]
  • MAP2K3 (Human) Recombinant Protein (Q01)
    MAP2K3 (Human) Recombinant phosphorylates and thus activates MAPK14/p38-MAPK. Protein (Q01) This kinase can be activated by insulin, and is necessary for the expression of glucose transporter. Expression of Catalog Number: H00005606-Q01 RAS oncogene is found to result in the accumulation of the active form of this kinase, which thus leads to the Regulation Status: For research use only (RUO) constitutive activation of MAPK14, and confers oncogenic transformation of primary cells. The inhibition Product Description: Human MAP2K3 partial ORF ( of this kinase is involved in the pathogenesis of Yersina AAH32478, 1 a.a. - 100 a.a.) recombinant protein with pseudotuberculosis. Multiple alternatively spliced GST-tag at N-terminal. transcript variants that encode distinct isoforms have been reported for this gene. [provided by RefSeq] Sequence: MESPASSQPASMPQSKGKSKRKKDLRISCMSKPPAP NPTPPRNLDSRTFITIGDRNFEVEADDLVTISELGRGAY GVVEKVRHAQSGTIMAVKRIRATVN Host: Wheat Germ (in vitro) Theoretical MW (kDa): 36.63 Applications: AP, Array, ELISA, WB-Re (See our web site product page for detailed applications information) Protocols: See our web site at http://www.abnova.com/support/protocols.asp or product page for detailed protocols Preparation Method: in vitro wheat germ expression system Purification: Glutathione Sepharose 4 Fast Flow Storage Buffer: 50 mM Tris-HCI, 10 mM reduced Glutathione, pH=8.0 in the elution buffer. Storage Instruction: Store at -80°C. Aliquot to avoid repeated freezing and thawing. Entrez GeneID: 5606 Gene Symbol: MAP2K3 Gene Alias: MAPKK3, MEK3, MKK3, PRKMK3 Gene Summary: The protein encoded by this gene is a dual specificity protein kinase that belongs to the MAP kinase kinase family. This kinase is activated by mitogenic and environmental stress, and participates in the MAP kinase-mediated signaling cascade.
    [Show full text]
  • Human MAPK10 Peptide (DAG-P1762) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Human MAPK10 peptide (DAG-P1762) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Antigen Description The protein encoded by this gene is a member of the MAP kinase family. MAP kinases act as an integration point for multiple biochemical signals, and are involved in a wide variety of cellular processes such as proliferation, differentiation, transcription regulation and development. This protein is a neuronal-specific form of c-Jun N-terminal kinases (JNKs). Through its phosphorylation and nuclear localization, this kinase plays regulatory roles in the signaling pathways during neuronal apoptosis. Beta-arrestin 2, a receptor-regulated MAP kinase scaffold protein, is found to interact with, and stimulate the phosphorylation of this kinase by MAP kinase kinase 4 (MKK4). Cyclin-dependent kianse 5 can phosphorylate, and inhibit the activity of this kinase, which may be important in preventing neuronal apoptosis. Four alternatively spliced transcript variants encoding distinct isoforms have been reported. [provided by RefSeq, Jul 2008] Specificity Specific to a subset of neurons in the nervous system. Present in the hippocampus and areas, cerebellum, striatum, brain stem, and weakly in the spinal cord. Very weak expression in testis and kidney. Nature Synthetic Expression System N/A Purity 70 - 90% by HPLC. Conjugate Unconjugated Sequence Similarities Belongs to the protein kinase superfamily. CMGC Ser/Thr protein kinase family. MAP kinase subfamily.Contains 1 protein kinase domain. Cellular Localization Cytoplasm. Procedure None Format Liquid Preservative None Storage Shipped at 4°C. Upon delivery aliquot and store at -20°C or -80°C. Avoid repeated freeze / thaw cycles.
    [Show full text]
  • A Central Role of IKK2 and TPL2 in JNK Activation and Viral B-Cell Transformation
    ARTICLE https://doi.org/10.1038/s41467-020-14502-x OPEN A central role of IKK2 and TPL2 in JNK activation and viral B-cell transformation Stefanie Voigt1, Kai R. Sterz 1, Fabian Giehler1,2, Anne-Wiebe Mohr 1,2, Joanna B. Wilson 3, Andreas Moosmann 1,2,4 & Arnd Kieser 1,2* IκB kinase 2 (IKK2) is well known for its pivotal role as a mediator of the canonical NF-κB pathway, which has important functions in inflammation and immunity, but also in cancer. 1234567890():,; Here we identify a novel and critical function of IKK2 and its co-factor NEMO in the activation of oncogenic c-Jun N-terminal kinase (JNK) signaling, induced by the latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV). Independent of its kinase activity, the TGFβ- activated kinase 1 (TAK1) mediates LMP1 signaling complex formation, NEMO ubiquitination and subsequent IKK2 activation. The tumor progression locus 2 (TPL2) kinase is induced by LMP1 via IKK2 and transmits JNK activation signals downstream of IKK2. The IKK2-TPL2-JNK axis is specific for LMP1 and differs from TNFα, Interleukin−1 and CD40 signaling. This pathway mediates essential LMP1 survival signals in EBV-transformed human B cells and post-transplant lymphoma, and thus qualifies as a target for treatment of EBV-induced cancer. 1 Helmholtz Centre Munich - German Research Centre for Environmental Health, Research Unit Gene Vectors, Marchioninistrasse 25, 81377 Munich, Germany. 2 German Centre for Infection Research (DZIF), Partner Site Munich, Munich, Germany. 3 School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
    [Show full text]
  • Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC)
    cancers Review Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC) Chiara Diquigiovanni * and Elena Bonora Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; [email protected] * Correspondence: [email protected]; Tel.: +39-051-208-8418 Simple Summary: Non-medullary thyroid carcinoma (NMTC) originates from thyroid follicular epithelial cells and is considered familial when occurs in two or more first-degree relatives of the patient, in the absence of predisposing environmental factors. Familial NMTC (FNMTC) cases show a high genetic heterogeneity, thus impairing the identification of pivotal molecular changes. In the past years, linkage-based approaches identified several susceptibility loci and variants associated with NMTC risk, however only few genes have been identified. The advent of next-generation sequencing technologies has improved the discovery of new predisposing genes. In this review we report the most significant genes where variants predispose to FNMTC, with the perspective that the integration of these new molecular findings in the clinical data of patients might allow an early detection and tailored therapy of the disease, optimizing patient management. Abstract: Non-medullary thyroid carcinoma (NMTC) is the most frequent endocrine tumor and originates from the follicular epithelial cells of the thyroid. Familial NMTC (FNMTC) has been defined in pedigrees where two or more first-degree relatives of the patient present the disease in absence of other predisposing environmental factors. Compared to sporadic cases, FNMTCs are often multifocal, recurring more frequently and showing an early age at onset with a worse outcome. FNMTC cases Citation: Diquigiovanni, C.; Bonora, E.
    [Show full text]
  • Spitz Melanoma Is a Distinct Subset of Spitzoid Melanoma
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Mod Pathol Manuscript Author . Author manuscript; Manuscript Author available in PMC 2020 July 03. Published in final edited form as: Mod Pathol. 2020 June ; 33(6): 1122–1134. doi:10.1038/s41379-019-0445-z. Spitz Melanoma is a Distinct Subset of Spitzoid Melanoma Shyam Raghavan, MD1, Sandra Peternel, MD2,3, Thaddeus W. Mully, MD2, Jeffrey P. North, MD2, Laura B. Pincus, MD2, Philip E. LeBoit, MD2, Timothy H. McCalmont, MD2, Boris C. Bastian, MD, PhD2, Iwei Yeh, MD, PhD2 1Department of Pathology, Stanford University, Stanford, CA 2Departments of Pathology and Dermatology, University of California San Francisco, San Francisco, CA 3Department of Dermatovenerology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia Abstract Melanomas that have histopathologic features that overlap with those of Spitz nevus are referred to as spitzoid melanomas. However, the diagnostic concept is used inconsistently and genomic analyses suggest it is a heterogeneous category. Spitz tumors, the spectrum of melanocytic neoplasms extending from Spitz nevi to their malignant counterpart Spitz melanoma, are defined in the 2018 WHO classification of skin tumors by the presence of specific genetic alterations such a kinase fusions or HRAS mutations. It is unclear what fraction of ‘spitzoid melanomas’ defined solely by their histopathologic features belong to the category of Spitz melanoma or to other melanoma subtypes. We assembled a cohort of 25 spitzoid melanomas diagnosed at a single institution over an eight-year period and performed high coverage DNA sequencing of 480 cancer related genes. Transcriptome wide RNA sequencing was performed for select cases.
    [Show full text]
  • Application of a MYC Degradation
    SCIENCE SIGNALING | RESEARCH ARTICLE CANCER Copyright © 2019 The Authors, some rights reserved; Application of a MYC degradation screen identifies exclusive licensee American Association sensitivity to CDK9 inhibitors in KRAS-mutant for the Advancement of Science. No claim pancreatic cancer to original U.S. Devon R. Blake1, Angelina V. Vaseva2, Richard G. Hodge2, McKenzie P. Kline3, Thomas S. K. Gilbert1,4, Government Works Vikas Tyagi5, Daowei Huang5, Gabrielle C. Whiten5, Jacob E. Larson5, Xiaodong Wang2,5, Kenneth H. Pearce5, Laura E. Herring1,4, Lee M. Graves1,2,4, Stephen V. Frye2,5, Michael J. Emanuele1,2, Adrienne D. Cox1,2,6, Channing J. Der1,2* Stabilization of the MYC oncoprotein by KRAS signaling critically promotes the growth of pancreatic ductal adeno- carcinoma (PDAC). Thus, understanding how MYC protein stability is regulated may lead to effective therapies. Here, we used a previously developed, flow cytometry–based assay that screened a library of >800 protein kinase inhibitors and identified compounds that promoted either the stability or degradation of MYC in a KRAS-mutant PDAC cell line. We validated compounds that stabilized or destabilized MYC and then focused on one compound, Downloaded from UNC10112785, that induced the substantial loss of MYC protein in both two-dimensional (2D) and 3D cell cultures. We determined that this compound is a potent CDK9 inhibitor with a previously uncharacterized scaffold, caused MYC loss through both transcriptional and posttranslational mechanisms, and suppresses PDAC anchorage- dependent and anchorage-independent growth. We discovered that CDK9 enhanced MYC protein stability 62 through a previously unknown, KRAS-independent mechanism involving direct phosphorylation of MYC at Ser .
    [Show full text]
  • Tpl2 (Map3k8) Regulates the Migration, Differentiation, and Function
    TPL2 (MAP3K8) REGULATES THE MIGRATION, DIFFERENTIATION, AND FUNCTION OF CRITICAL INNATE IMMUNE CELLS DURING THE INFLAMMATORY RESPONSE by SEAN MICHAEL ROWLEY (Under the Direction of Wendy Watford) ABSTRACT The protein kinase Tpl2 (MAP3K8) regulates innate inflammatory responses and is being actively pursued for therapeutic inhibition during chronic autoimmunity. Herein, we addressed the contribution of Tpl2 to pro-inflammatory responses of NK cells and macrophages. Despite the Tpl2-dependent regulation of IFN secretion by CD4+ T-cells, NK cell IFNγ production, STAT4 expression, and expression of cytotoxic machinery occurred independently of Tpl2. In contrast, tpl2-/- macrophages were functionally defective, as they displayed impaired chemokine and chemokine receptor expression following LPS stimulation and were defective in migrating in vivo to inflamed tissues. Tpl2-/- macrophages were also impaired in their differentiation towards a pro-inflammatory phenotype (M1) while conversely displaying an enhanced anti-inflammatory phenotype (M2). Overall, this work provides additional support for targeting Tpl2, through its effects on macrophage recruitment and differentiation, for the treatment of autoimmunity. INDEX WORDS: autoimmunity, inflammation, cytokines, chemokines, cellular trafficking, NK cells, macrophage classical activation, macrophage alternative activation TPL2 (MAP3K8) REGULATES THE MIGRATION, DIFFERENTIATION, AND FUNCTION OF CRITICAL INNATE IMMUNE CELLS DURING THE INFLAMMATORY RESPONSE by SEAN MICHAEL ROWLEY B.S., University
    [Show full text]
  • MAPK Kinase 3 Is a Tumor Suppressor with Reduced Copy Number in Breast Cancer
    Author Manuscript Published OnlineFirst on November 14, 2013; DOI: 10.1158/0008-5472.CAN-13-1310 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. MAPK kinase 3 is a tumor suppressor with reduced copy number in breast cancer Adam J. MacNeil1,2,3, Shun-Chang Jiao4, Lori A. McEachern5, Yong Jun Yang6, Amanda Dennis1,2, Haiming Yu4, Zhaolin Xu3,7, Jean S. Marshall1,3, and Tong-Jun Lin1,2,3,7 1 Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, B3K 6R8, Canada. 2 Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, B3K 6R8, Canada. 3 Beatrice Hunter Cancer Research Institute, Suite 2L-A2, Tupper Link, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada. 4 Department of Medical Oncology, General Hospital of the People's Liberation Army, Beijing, 100853, China. 5 Department of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, B3K 6R8, Canada. 6 Institute of Zoonosis, College of Animal Sciences and Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, China. 7 Department of Pathology, Dalhousie University, Halifax, Nova Scotia, B3K 6R8, Canada. Correspondence: Shun-Chang Jiao, email:[email protected]; Department of Medical Oncology, General Hospital of the People's Liberation Army, Beijing, 100853, China. Tong-Jun Lin, email: [email protected]; phone 902-470-8834; IWK Health Centre, Department of Pediatrics, Dalhousie University, 5850/5980 University Ave, Halifax, Nova Scotia, B3K 6R8, Canada. The authors have declared that no conflict of interest exists. Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2013 American Association for Cancer Research.
    [Show full text]
  • The MKKK62-MKK3-MAPK7/14 Module Negatively Regulates Seed
    Mao et al. Rice (2019) 12:2 https://doi.org/10.1186/s12284-018-0260-z ORIGINAL ARTICLE Open Access The MKKK62-MKK3-MAPK7/14 module negatively regulates seed dormancy in rice Xingxue Mao1,2†, Jianjun Zhang3†, Wuge Liu1,2†, Shijuan Yan4, Qing Liu1,2, Hua Fu1,2, Junliang Zhao1,2, Wenjie Huang4, Jingfang Dong1,2, Shaohong Zhang1,2, Tifeng Yang1,2, Wu Yang1,2, Bin Liu1,2* and Feng Wang1,2* Abstract Background: Seed dormancy directly affects the phenotype of pre-harvest sprouting, and ultimately affects the quality and yield of rice seeds. Although many genes controlling seed dormancy have been cloned from cereals, the regulatory mechanisms controlling this process are complex, and much remains unknown. The MAPK cascade is involved in many signal transduction pathways. Recently, MKK3 has been reported to be involved in the regulation of seed dormancy, but its mechanism of action is unclear. Results: We found that MKKK62-overexpressing rice lines (OE) lost seed dormancy. Further analyses showed that the abscisic acid (ABA) sensitivity of OE lines was decreased. In yeast two-hybrid experiments, MKKK62 interacted with MKK3, and MKK3 interacted with MAPK7 and MAPK14. Knock-out experiments confirmed that MKK3, MAPK7, and MAPK14 were involved in the regulation of seed dormancy. The OE lines showed decreased transcript levels of OsMFT, a homolog of a gene that controls seed dormancy in wheat. The up-regulation of OsMFT in MKK3-knockout lines (OE/mkk3) and MAPK7/14-knockout lines (OE/mapk7/mapk14) indicated that the MKKK62-MKK3-MAPK7/ MAPK14 system controlled seed dormancy by regulating the transcription of OsMFT.
    [Show full text]
  • Integrated Molecular Characterisation of the MAPK Pathways in Human
    ARTICLE https://doi.org/10.1038/s42003-020-01552-6 OPEN Integrated molecular characterisation of the MAPK pathways in human cancers reveals pharmacologically vulnerable mutations and gene dependencies 1234567890():,; ✉ Musalula Sinkala 1 , Panji Nkhoma 2, Nicola Mulder 1 & Darren Patrick Martin1 The mitogen-activated protein kinase (MAPK) pathways are crucial regulators of the cellular processes that fuel the malignant transformation of normal cells. The molecular aberrations which lead to cancer involve mutations in, and transcription variations of, various MAPK pathway genes. Here, we examine the genome sequences of 40,848 patient-derived tumours representing 101 distinct human cancers to identify cancer-associated mutations in MAPK signalling pathway genes. We show that patients with tumours that have mutations within genes of the ERK-1/2 pathway, the p38 pathways, or multiple MAPK pathway modules, tend to have worse disease outcomes than patients with tumours that have no mutations within the MAPK pathways genes. Furthermore, by integrating information extracted from various large-scale molecular datasets, we expose the relationship between the fitness of cancer cells after CRISPR mediated gene knockout of MAPK pathway genes, and their dose-responses to MAPK pathway inhibitors. Besides providing new insights into MAPK pathways, we unearth vulnerabilities in specific pathway genes that are reflected in the re sponses of cancer cells to MAPK targeting drugs: a revelation with great potential for guiding the development of innovative therapies.
    [Show full text]