TRIB3 Stabilizes High TWIST1 Expression to Promote Rapid APL Progression and ATRA Resistance

Total Page:16

File Type:pdf, Size:1020Kb

TRIB3 Stabilizes High TWIST1 Expression to Promote Rapid APL Progression and ATRA Resistance Author Manuscript Published OnlineFirst on June 24, 2019; DOI: 10.1158/1078-0432.CCR-19-0510 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. TRIB3 Stabilizes High TWIST1 Expression to Promote Rapid APL Progression and ATRA Resistance Jian Lin1, 3, Wu Zhang1, 3, *, Li-Ting Niu1, Yong-Mei Zhu1, Xiang-Qin Weng1, Yan Sheng1, Jiang Zhu1 and Jie Xu1, 2, * 1State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao-Tong University, 197 Ruijin Er Road, Shanghai 200025, China. 2Translational Medicine Ward, Department of Hematology, Rui-Jin Hospital, Shanghai 200025, China. 3These authors contributed equally to this work. *Correspondence: Jie Xu, State Key Laboratory for Medical Genomics and Shanghai Institute of Hematology Rui-Jin Hospital, affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, China. E-mail: [email protected]; and Wu Zhang, State Key Laboratory for Medical Genomics and Shanghai Institute of Hematology Rui-Jin Hospital, affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, China. E-mail: [email protected]; Running title: TRIB3 stabilizes TWIST1 to promote APL progression Key words: Acute promyelocytic leukemia; TWIST1; TRIB3; Early death; ATRA-resistance Conflict of interest statement: The authors declare no potential conflicts of interest. 1 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 24, 2019; DOI: 10.1158/1078-0432.CCR-19-0510 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract Purpose: The resistance to differentiation therapy and early death caused by fatal bleeding endangers the health of a significant proportion of APL patients. This study aims to investigate the molecular mechanisms of ATRA resistance and uncover new potential therapeutic strategies to block the rapid progression of early death. Experimental design: The important role of TWIST1 in APL leukemogenesis was first determined by gain- and loss-of-function assays. We then performed in vivo and in vitro experiments to explore the interaction of TWIST1 and TRIB3 and develop a potential peptide-initiated therapeutic opportunity to protect against early death and induction therapy resistance in patients with APL. Results: We found that the epithelial-mesenchymal transition (EMT)-inducing transcription factor TWIST1 is highly expressed in APL cells and is critical for leukemic cell survival. TWIST1 and TRIB3 were highly co-expressed in APL cells compared with other subtypes of acute myeloid leukemia (AML) cells. We subsequently demonstrated that TRIB3 could bind to the WR domain of TWIST1 and contribute to its stabilization by inhibiting its ubiquitination. TRIB3 depletion promoting TWIST1 degradation reverses resistance to induction therapy and improves sensitivity to ATRA. Based on a detailed functional screen of synthetic peptides, we discovered a peptide analogous to the TWIST1 WR domain that specifically represses APL cell survival by disrupting the TRIB3/TWIST1 interaction. Conclusions: Our data not only define the essential role of TWIST1 as an EMT-TF in APL patients but also suggest that disrupting the TRIB3/TWIST1 interaction reverses induction therapy resistance and blocks rapid progression of APL early death. 2 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 24, 2019; DOI: 10.1158/1078-0432.CCR-19-0510 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Translational Relevance This study was designed to improve the treatment for high-risk APL patients. This proportion of APL patients continued to suffer from early fatal bleeding and leukemic extramedullary infiltration, which remains the most important challenge and the largest obstacle to curing all APL patients. We demonstrate that the unique EMT-inducing transcription factor TWIST1 is significantly highly expressed in APL and governs the survival of APL cells. We show that TRIB3 interacts with TWIST1 and stabilizes high TWIST1 expression by repressing TWIST1 ubiquitination. Our data also suggest that a peptide similar to the WR domain disturbs the TRIB3/TWIST1 interaction, impairs rapid progression during the early death of APL and reverses resistance to ATRA therapy. These results reveal the important role of a specific oncogenic transcriptional factor, TWIST1, in APL leukemogenesis and suggest a potential peptide-initiated therapeutic opportunity to protect against early death and induction therapy resistance in patients with APL. 3 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 24, 2019; DOI: 10.1158/1078-0432.CCR-19-0510 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Introduction Acute promyelocytic leukemia (APL), which accounts for 10-15% of AML cases, is characterized by the t(15; 17) chromosomal translocation and is now highly curable by the combination of granulocytic differentiation induction and the PML-RARα oncoprotein-targeted agents all-trans retinoic acid (ATRA) and arsenic trioxide (ATO) (1,2). Despite the striking molecular complete remission (CR) and the very few cases of relapse associated with ATRA/ATO-based regimens, mortality events typically result from early fatal bleeding, which remains the most important challenge and the largest obstacle to curing all APL patients (3). For instance, several studies have reported that the risk of early hemorrhagic death (HD) reaches an incidence of 10-20% during the first month of induction (4-6). Importantly, APL patients with a high white blood cell count face an increased risk of early HD (7). Furthermore, resistance to ATRA/ATO treatment with PML-RARα mutations still remains a therapeutic challenge for a significant proportion of APL patients. Thus, we need to better understand the molecular mechanism of APL pathogenesis and design more effective therapeutic strategies to block the rapid progression of early death and overcome resistance. Oncogenic transcription factors play an important role in the development of hematological malignancy (8,9). The dysregulation of epithelial-mesenchymal transition-inducing transcription factors (EMT-TFs), including SNAI1/SNAI2, ZEB1/ZEB2 and TWIST1/TWIST2, has also been explored within the context of the aggressive invasion, chemoresistance and poor prognosis of acute myeloid leukemia (AML) (10-12). TWIST1, a highly conserved basic helix-loop-helix (bHLH) protein, is a well-characterized EMT-TF that plays a critical role in embryonic development and cancer metastasis (13,14). Recent studies have shown that TWIST1 is overexpressed in primary AML samples (15). Our previous study reported that high expression of TWIST1 in AML contributes to extramedullary infiltration and promotes leukemic aggressiveness (16). These findings strongly suggest that 4 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 24, 2019; DOI: 10.1158/1078-0432.CCR-19-0510 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. disruption of TWIST1 is involved in leukemogenesis. However, the prognosis of AML patients with high TWIST1 expression is controversial based on several reports (15,17). These conflicting reports likely occurred because TWIST1 expression in AML is heterogeneous (18). TWIST1 expression is higher in APL, an intriguing AML subtype in which successful clinical tumor differentiation-induction therapy has been applied and for which the clinical phenotype is inconsistent with the expression level of TWIST1 (15,18). In this study, we demonstrate that the EMT-TF TWIST1 is significantly highly expressed in APL and governs the survival of APL cells. We show that TRIB3 interacts with TWIST1 and stabilizes high TWIST1 expression by repressing TWIST1 ubiquitination. Our data also suggest that a peptide similar to the WR domain disturbs the TRIB3/TWIST1 interaction, impairs rapid progression during the early death of APL and reverses resistance to ATRA therapy. These results contribute to a better understanding of the molecular mechanism of APL pathogenesis and will allow for designing more effective therapeutic strategies to block the rapid progression of early death and overcome resistance. Materials and Methods Cells and mice Leukemic cell lines were cultured in RPMI-1640 medium (Invitrogen, Grand Island, USA) supplemented with 10% FBS (Invitrogen, Grand Island, USA), and HEK293T cells were grown in DMEM (Invitrogen, Grand Island, USA) supplemented with 10% FBS. All cell lines, including NB4-R1 and PR9 cell line, were obtained from the Shanghai Institute of Hematology as previously described (19-21). NB4-R1, a de novo ATRA-resistant cell line isolated from parental NB4 cells, was obtained from Dr. Michel Lanotte (Hospital Saint Louis, Paris, France) (22). HMRP8-PML-RARα 5 Downloaded from clincancerres.aacrjournals.org on October 1, 2021.
Recommended publications
  • Supplemental Information to Mammadova-Bach Et Al., “Laminin Α1 Orchestrates VEGFA Functions in the Ecosystem of Colorectal Carcinogenesis”
    Supplemental information to Mammadova-Bach et al., “Laminin α1 orchestrates VEGFA functions in the ecosystem of colorectal carcinogenesis” Supplemental material and methods Cloning of the villin-LMα1 vector The plasmid pBS-villin-promoter containing the 3.5 Kb of the murine villin promoter, the first non coding exon, 5.5 kb of the first intron and 15 nucleotides of the second villin exon, was generated by S. Robine (Institut Curie, Paris, France). The EcoRI site in the multi cloning site was destroyed by fill in ligation with T4 polymerase according to the manufacturer`s instructions (New England Biolabs, Ozyme, Saint Quentin en Yvelines, France). Site directed mutagenesis (GeneEditor in vitro Site-Directed Mutagenesis system, Promega, Charbonnières-les-Bains, France) was then used to introduce a BsiWI site before the start codon of the villin coding sequence using the 5’ phosphorylated primer: 5’CCTTCTCCTCTAGGCTCGCGTACGATGACGTCGGACTTGCGG3’. A double strand annealed oligonucleotide, 5’GGCCGGACGCGTGAATTCGTCGACGC3’ and 5’GGCCGCGTCGACGAATTCACGC GTCC3’ containing restriction site for MluI, EcoRI and SalI were inserted in the NotI site (present in the multi cloning site), generating the plasmid pBS-villin-promoter-MES. The SV40 polyA region of the pEGFP plasmid (Clontech, Ozyme, Saint Quentin Yvelines, France) was amplified by PCR using primers 5’GGCGCCTCTAGATCATAATCAGCCATA3’ and 5’GGCGCCCTTAAGATACATTGATGAGTT3’ before subcloning into the pGEMTeasy vector (Promega, Charbonnières-les-Bains, France). After EcoRI digestion, the SV40 polyA fragment was purified with the NucleoSpin Extract II kit (Machery-Nagel, Hoerdt, France) and then subcloned into the EcoRI site of the plasmid pBS-villin-promoter-MES. Site directed mutagenesis was used to introduce a BsiWI site (5’ phosphorylated AGCGCAGGGAGCGGCGGCCGTACGATGCGCGGCAGCGGCACG3’) before the initiation codon and a MluI site (5’ phosphorylated 1 CCCGGGCCTGAGCCCTAAACGCGTGCCAGCCTCTGCCCTTGG3’) after the stop codon in the full length cDNA coding for the mouse LMα1 in the pCIS vector (kindly provided by P.
    [Show full text]
  • TRIB2 Functions As Novel Oncogene in Colorectal Cancer by Blocking
    Hou et al. Molecular Cancer (2018) 17:172 https://doi.org/10.1186/s12943-018-0922-x RESEARCH Open Access TRIB2 functions as novel oncogene in colorectal cancer by blocking cellular senescence through AP4/p21 signaling Zhenlin Hou1, Kaixuan Guo1, Xuling Sun1, Fuqing Hu1, Qianzhi Chen1, Xuelai Luo1, Guihua Wang1, Junbo Hu1 and Li Sun2* Abstract Background: Cellular senescence is a state of irreversible cell growth arrest and senescence cells permanently lose proliferation potential. Induction of cellular senescence might be a novel therapy for cancer cells. TRIB2 has been reported to participate in regulating proliferation and drug resistance of various cancer cells. However, the role of TRIB2 in cellular senescence of colorectal cancer (CRC) and its molecular mechanism remains unclear. Methods: The expression of TRIB2 in colorectal cancer tissues and adjacent tissues was detected by immunohistochemistry and RT-PCR. The growth, cell cycle distribution and cellular senescence of colorectal cancer cells were evaluated by Cell Counting Kit-8 (CCK8) assay, flow cytometry detection and senescence- associated β-galactosidase staining, respectively. Western blot, RT-PCR and luciferase assay were performed to determine how TRIB2 regulates p21. Immunoprecipitation (IP) and chromatin-immunoprecipitation (ChIP) were used to investigate the molecular mechanisms. Results: We found that TRIB2 expression was elevated in CRC tissues compared to normal adjacent tissues and high TRIB2 expression indicated poor prognosis of CRC patients. Functionally, depletion of TRIB2 inhibited cancer cells proliferation, induced cell cycle arrest and promoted cellular senescence, whereas overexpression of TRIB2 accelerated cell growth, cell cycle progression and blocked cellular senescence. Further studies showed that TRIB2 physically interacted with AP4 and inhibited p21 expression through enhancing transcription activities of AP4.
    [Show full text]
  • Characterization of TRIB2-Mediated Resistance to Pharmacological Inhibition of MEK
    VANESSA MENDES HENRIQUES Characterization of TRIB2-mediated resistance to pharmacological inhibition of MEK Oncobiology Master Thesis Faro, 2017 VANESSA MENDES HENRIQUES Characterization of TRIB2-mediated resistance to pharmacological inhibition of MEK Supervisors: Dr. Wolfgang Link Dr. Bibiana Ferreira Oncobiology Master Thesis Faro, 2017 Título: “Characterization of TRIB2-mediated resistance to pharmacological inhibition of MEK” Declaração de autoria do trabalho Declaro ser a autora deste trabalho, que é original e inédito. Autores e trabalhos consultados estão devidamente citados no texto e constam da listagem de referências incluída. Copyright Vanessa Mendes Henriques _____________________________ A Universidade do Algarve tem o direito, perpétuo e sem limites geográficos, de arquivar e publicitar este trabalho através de exemplares impressos reproduzidos em papel ou de forma digital, ou por qualquer outro meio conhecido ou que venha a ser inventado, de o divulgar através de repositórios científicos e de admitir a sua cópia e distribuição com objetivos educacionais ou de investigação, não comerciais, desde que seja dado crédito ao autor e editor. i Acknowledgements First, I would like to thank the greatest opportunity given by professor doctor Wolfgang Link to accepting me into his team, contributing to my scientific and personal progress. Thank You for all Your knowledge and help across the year. A special thanks to Bibiana Ferreira who stayed by me all year and trained me. Thank You for all you taught me, thank you for all your patience and time and all the support. Thank you for all the great times that You provided me. It was an amazing experience to learn and work with You, which made me grow as a scientist and also as a person.
    [Show full text]
  • The New Antitumor Drug ABTL0812 Inhibits the Akt/Mtorc1 Axis By
    Published OnlineFirst December 15, 2015; DOI: 10.1158/1078-0432.CCR-15-1808 Cancer Therapy: Preclinical Clinical Cancer Research The New Antitumor Drug ABTL0812 Inhibits the Akt/mTORC1 Axis by Upregulating Tribbles-3 Pseudokinase Tatiana Erazo1, Mar Lorente2,3, Anna Lopez-Plana 4, Pau Munoz-Guardiola~ 1,5, Patricia Fernandez-Nogueira 4, Jose A. García-Martínez5, Paloma Bragado4, Gemma Fuster4, María Salazar2, Jordi Espadaler5, Javier Hernandez-Losa 6, Jose Ramon Bayascas7, Marc Cortal5, Laura Vidal8, Pedro Gascon 4,8, Mariana Gomez-Ferreria 5, Jose Alfon 5, Guillermo Velasco2,3, Carles Domenech 5, and Jose M. Lizcano1 Abstract Purpose: ABTL0812 is a novel first-in-class, small molecule tion of Tribbles-3 pseudokinase (TRIB3) gene expression. Upre- which showed antiproliferative effect on tumor cells in pheno- gulated TRIB3 binds cellular Akt, preventing its activation by typic assays. Here we describe the mechanism of action of this upstream kinases, resulting in Akt inhibition and suppression of antitumor drug, which is currently in clinical development. the Akt/mTORC1 axis. Pharmacologic inhibition of PPARa/g or Experimental Design: We investigated the effect of ABTL0812 TRIB3 silencing prevented ABTL0812-induced cell death. on cancer cell death, proliferation, and modulation of intracel- ABTL0812 treatment induced Akt inhibition in cancer cells, tumor lular signaling pathways, using human lung (A549) and pancre- xenografts, and peripheral blood mononuclear cells from patients atic (MiaPaCa-2) cancer cells and tumor xenografts. To identify enrolled in phase I/Ib first-in-human clinical trial. cellular targets, we performed in silico high-throughput screening Conclusions: ABTL0812 has a unique and novel mechanism of comparing ABTL0812 chemical structure against ChEMBL15 action, that defines a new and drugable cellular route that links database.
    [Show full text]
  • TRIB2 Confers Resistance to Anti-Cancer Therapy by Activating the Serine/Threonine Protein Kinase AKT
    ARTICLE Received 23 Nov 2015 | Accepted 23 Jan 2017 | Published 9 Mar 2017 DOI: 10.1038/ncomms14687 OPEN TRIB2 confers resistance to anti-cancer therapy by activating the serine/threonine protein kinase AKT Richard Hill1,2,3, Patricia A. Madureira2, Bibiana Ferreira2, Ineˆs Baptista2, Susana Machado2, Laura Colac¸o2, Marta dos Santos2, Ningshu Liu4, Ana Dopazo5, Selma Ugurel6, Angyal Adrienn7, Endre Kiss-Toth7, Murat Isbilen8, Ali O. Gure8 & Wolfgang Link1,2,9 Intrinsic and acquired resistance to chemotherapy is the fundamental reason for treatment failure for many cancer patients. The identification of molecular mechanisms involved in drug resistance or sensitization is imperative. Here we report that tribbles homologue 2 (TRIB2) ablates forkhead box O activation and disrupts the p53/MDM2 regulatory axis, conferring resistance to various chemotherapeutics. TRIB2 suppression is exerted via direct interaction with AKT a key signalling protein in cell proliferation, survival and metabolism pathways. Ectopic or intrinsic high expression of TRIB2 induces drug resistance by promoting phospho- AKT (at Ser473) via its COP1 domain. TRIB2 expression is significantly increased in tumour tissues from patients correlating with an increased phosphorylation of AKT, FOXO3a, MDM2 and an impaired therapeutic response. This culminates in an extremely poor clinical outcome. Our study reveals a novel regulatory mechanism underlying drug resistance and suggests that TRIB2 functions as a regulatory component of the PI3K network, activating AKT in cancer cells. 1 Department of Biomedical Sciences and Medicine (DCBM), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal. 2 Centre for Biomedical Research (CBMR), University of Algarve, Campus de Gambelas, Faro 8005-139, Portugal.
    [Show full text]
  • The Cell Biology Behind the Oncogenic PIP3 Lipids Ana C
    © 2019. Published by The Company of Biologists Ltd | Journal of Cell Science (2019) 132, jcs228395. doi:10.1242/jcs.228395 MEETING REPORT The cell biology behind the oncogenic PIP3 lipids Ana C. Carrera1,* and Richard Anderson2 ABSTRACT favors the interaction of PI3K with the plasma membrane. Thus, α The different mechanisms of phosphoinositide 3-kinase (PI3K) blocking the plasma membrane recruitment of PI3K could be an activation in cancer as well as the events that result in PI3K effective therapeutic approach. pathway reactivation after patient treatment with PI3K inhibitors was Activation of PI3K is triggered by ligand binding to receptor discussed on October 15–17th, 2018, in the medieval town of Baeza tyrosine kinases (RTKs; EGF receptor etc.) and is enhanced by Ras (Universidad Internacional de Andalucıa,́ Spain) at the workshop (Jiménez et al., 2002). The work of Jonathan M. Backer (Albert entitled ‘The cell biology behind the oncogenic PIP3 lipids’. These Einstein College of Medicine, NY) showed that, in addition to topics and the data presented regarding cellular functions altered by RTK-mediated activation, agonist binding to G protein-coupled β PI3K deregulation, the cooperation of PI3K/PTEN mutations with receptors (GPCRs) also activates PI3K ; simultaneous switch on β other tumor drivers, and the lessons learned for PI3K-targeted of RTK and GPCRs induces optimal PI3K activation (Houslay β therapy, are discussed below. et al., 2016) (Fig. 1). PI3K is not frequently mutated in cancer; however, its overexpression is found in several cancer types and can trigger cell transformation (Liu et al., 2009; Denley et al., 2007). Jonathan’s studies on the contribution of PI3Kβ to breast INTRODUCTION cancer metastasis illustrate that GPCR signaling to PI3Kβ is The phosphoinositide 3-kinase (PI3K) family comprises eight kinases required for the formation of invadopodia (Khalil et al., 2016).
    [Show full text]
  • Identification of Oncogenic Driver Mutations by Genome-Wide CRISPR-Cas9 Dropout Screening Michael K
    Kiessling et al. BMC Genomics (2016) 17:723 DOI 10.1186/s12864-016-3042-2 METHODOLOGY ARTICLE Open Access Identification of oncogenic driver mutations by genome-wide CRISPR-Cas9 dropout screening Michael K. Kiessling1, Sven Schuierer2, Silke Stertz3, Martin Beibel2, Sebastian Bergling2, Judith Knehr2, Walter Carbone2, Cheryl de Vallière1, Joelle Tchinda4, Tewis Bouwmeester2, Klaus Seuwen2, Gerhard Rogler1*† and Guglielmo Roma2*† Abstract Background: Genome-wide CRISPR-Cas9 dropout screens can identify genes whose knockout affects cell viability. Recent CRISPR screens detected thousands of essential genes required for cellular survival and key cellular processes; however discovering novel lineage-specific genetic dependencies from the many hits still remains a challenge. Results: To assess whether CRISPR-Cas9 dropout screens can help identify cancer dependencies, we screened two human cancer cell lines carrying known and distinct oncogenic mutations using a genome-wide sgRNA library. We found that the gRNA targeting the driver mutation EGFR was one of the highest-ranking candidates in the EGFR- mutant HCC-827 lung adenocarcinoma cell line. Likewise, sgRNAs for NRAS and MAP2K1 (MEK1), a downstream kinase of mutant NRAS, were identified among the top hits in the NRAS-mutant neuroblastoma cell line CHP-212. Depletion of these genes targeted by the sgRNAs strongly correlated with the sensitivity to specific kinase inhibitors of the EGFR or RAS pathway in cell viability assays. In addition, we describe other dependencies such as TBK1 in HCC-827 cells and TRIB2 in CHP-212 cells which merit further investigation. Conclusions: We show that genome-wide CRISPR dropout screens are suitable for the identification of oncogenic drivers and other essential genes.
    [Show full text]
  • Highlights of the 2Nd International Symposium on Tribbles and Diseases: Tribbles Tremble in Therapeutics for Immunity, Metabolism, Fundamental Cell Biology and Cancer
    Acta Pharmaceutica Sinica B 2019;9(2):443–454 Chinese Pharmaceutical Association Institute of Materia Medica, Chinese Academy of Medical Sciences Acta Pharmaceutica Sinica B www.elsevier.com/locate/apsb www.sciencedirect.com MEETING REPORT Highlights of the 2nd International Symposium on Tribbles and Diseases: tribbles tremble in therapeutics for immunity, metabolism, fundamental cell biology and cancer Bing Cuia, Patrick A. Eyersb, Leonard L. Dobensc, Nguan Soon Tand,e,f,g, Peter D. Maceh, Wolfgang A. Linki,j,k, Endre Kiss-Tothl, Karen Keeshanm, Takuro Nakamuran, Warren S. Pearo, Yodit Fesehap,q, Jessica Johnstonl, Arkatiz Carracedor,s,t,u, Marcel Scheidelerv,w,x,y, Zabran llyasl, Robert C. Bauerz, Jorge D. Erusalimskyaa, Dominika Grzesikab, Juan Salamanca-Viloriaac,ad, Xiaoxi Lva, Yishi Jinae,KeLia,af, Guillermo Velascoag, Shuang Shanga, Jose M. Lizcanoah, Xiaowei Zhanga, Jichao Zhoua, Jiaojiao Yua, Fang Huaa, Feng Wanga, Shanshan Liua, Jinmei Yua, Zhuowei Hua,n aState Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China bDepartment of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK cDivision of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA dSchool of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore eLee Kong Chian School of Medicine,
    [Show full text]
  • TRIB2 Modulates Proteasome Function to Reduce Ubiquitin Stability And
    Guo et al. Cell Death and Disease (2021) 12:42 https://doi.org/10.1038/s41419-020-03299-8 Cell Death & Disease ARTICLE Open Access TRIB2 modulates proteasome function to reduce ubiquitin stability and protect liver cancer cells against oxidative stress Susu Guo1,YuxinChen2,YueyueYang1,XiaoZhang3,LifangMa3,XiangfeiXue1, Yongxia Qiao4 and Jiayi Wang 1,3 Abstract The regulation of homeostasis in the Ubiquitin (Ub) proteasome system (UPS) is likely to be important for the development of liver cancer. Tribbles homolog 2 (TRIB2) is known to affect Ub E3 ligases (E3s) in liver cancer. However, whether TRIB2 regulates the UPS in other ways and the relevant mechanisms are still unknown. Here, we reveal that TRIB2 decreased Ub levels largely by stimulating proteasome degradation of Ub. In the proteasome, proteasome 20S subunit beta 5 (PSMB5) was critical for the function of TRIB2, although it did not directly interact with TRIB2. However, poly (rC) binding protein 2 (PCBP2), which was identified by mass spectrometry, directly interacted with both TRIB2 and PSMB5. PCBP2 was a prerequisite for the TRIB2 induction of PSMB5 activity and decreased Ub levels. A significant correlation between TRIB2 and PCBP2 was revealed in liver cancer specimens. Interestingly, TRIB2 suppressed the K48- ubiquitination of PCBP2 to increase its level. Therefore, a model showing that TRIB2 cooperates and stimulates PCBP2 to reduce Ub levels was established. Additionally, the reduction in Ub levels induced by TRIB2 and PCBP2 was dependent on K48-ubiquitination. PCBP2 was one of the possible downstream factors of TRIB2 and their interaction relied on the DQLVPD element of TRIB2 and the KH3 domain of PCBP2.
    [Show full text]
  • Tribbles Homolog 2 (Trib2), a Pseudo Serine/Threonine Kinase In
    Fang et al. Cell Commun Signal (2021) 19:41 https://doi.org/10.1186/s12964-021-00725-y REVIEW Open Access Tribbles homolog 2 (Trib2), a pseudo serine/ threonine kinase in tumorigenesis and stem cell fate decisions Yu Fang1,2*, Angelina Olegovna Zekiy3, Farhoodeh Ghaedrahmati4, Anton Timoshin5, Maryam Farzaneh6 , Amir Anbiyaiee7 and Seyed Esmaeil Khoshnam8 Abstract The family of Tribbles proteins play many critical nonenzymatic roles and regulate a wide range of key signaling pathways. Tribbles homolog 2 (Trib2) is a pseudo serine/threonine kinase that functions as a scafold or adaptor in various physiological and pathological processes. Trib2 can interact with E3 ubiquitin ligases and control protein sta- bility of downstream efectors. This protein is induced by mitogens and enhances the propagation of several cancer cells, including myeloid leukemia, liver, lung, skin, bone, brain, and pancreatic. Thus, Trib2 can be a predictive and valuable biomarker for the diagnosis and treatment of cancer. Recent studies have illustrated that Trib2 plays a major role in cell fate determination of stem cells. Stem cells have the capacity to self-renew and diferentiate into specifc cell types. Stem cells are important sources for cell-based regenerative medicine and drug screening. Trib2 has been found to increase the self-renewal ability of embryonic stem cells, the reprogramming efciency of somatic cells, and chondrogenesis. In this review, we will focus on the recent advances of Trib2 function in tumorigenesis and stem cell fate decisions. Keywords: Pluripotent stem cells, Tribbles homolog 2, Tumorigenesis, Pluripotency, Reprogramming, Stem cell fate, Regenerative medicine Background E3 ubiquitin ligases and control protein stability of down- Tribbles homolog 2 (Trib2) is a pseudo serine/threo- stream efectors [7].
    [Show full text]
  • 1 Title: the BRAF and MEK Inhibitors Dabrafenib and Trametinib
    Author Manuscript Published OnlineFirst on January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2339 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Title: The BRAF and MEK Inhibitors Dabrafenib and Trametinib: Effects on Immune Function and in Combination with Immunomodulatory Antibodies Targeting PD1, PD-L1 and CTLA-4 Authors: Li Liu1*, Patrick A Mayes1*, Stephen Eastman1, Hong Shi1, Sapna Yadavilli1, Tianqian Zhang1, Jingsong Yang1, Laura Seestaller-Wehr1, Shu-Yun Zhang1, Chris Hopson1, Lyuben Tsvetkov1, Junping Jing2, Shu Zhang3 , James Smothers1, and Axel Hoos1 Affiliations: GlaxoSmithKline, 1Immuno-Oncology & Combination DPU, 2Molecular Medicine Unit, Oncology R&D, 3Statistical Science, Collegeville, PA19426, Pennsylvania, USA, * These authors contributed equally Running title: Dabrafenib and trametinib effect on immune function Financial support: This study was financially supported by GlaxoSmithKline. Corresponding author: Axel Hoos, GlaxoSmithKline, Immuno-Oncology & Combination DPU, 1250 S. Collegeville Rd., Collegeville, PA, USA, 19426. Tel: +1 610 427 3733. Email: [email protected]. Conflict of interest disclosure: L. Liu, P. Mayes, S. Eastman, H. Shi, S. Yadavilli, J. Yang, L. Seestaller-Wehr, C. Hopson, J. Jing, S. Zhang and J. Smothers are employees and stockholders of GlaxoSmithKline. T. Zhang, S-Y. Zhang, L. Tsvetkov and A. Hoos are employees of GlaxoSmithKline. Word count: 4985 Total number of figures and tables: 6 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2015 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2339 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]
  • Kinome Expression Profiling to Target New Therapeutic Avenues in Multiple Myeloma
    Plasma Cell DIsorders SUPPLEMENTARY APPENDIX Kinome expression profiling to target new therapeutic avenues in multiple myeloma Hugues de Boussac, 1 Angélique Bruyer, 1 Michel Jourdan, 1 Anke Maes, 2 Nicolas Robert, 3 Claire Gourzones, 1 Laure Vincent, 4 Anja Seckinger, 5,6 Guillaume Cartron, 4,7,8 Dirk Hose, 5,6 Elke De Bruyne, 2 Alboukadel Kassambara, 1 Philippe Pasero 1 and Jérôme Moreaux 1,3,8 1IGH, CNRS, Université de Montpellier, Montpellier, France; 2Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium; 3CHU Montpellier, Laboratory for Monitoring Innovative Therapies, Department of Biologi - cal Hematology, Montpellier, France; 4CHU Montpellier, Department of Clinical Hematology, Montpellier, France; 5Medizinische Klinik und Poliklinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany; 6Nationales Centrum für Tumorerkrankungen, Heidelberg , Ger - many; 7Université de Montpellier, UMR CNRS 5235, Montpellier, France and 8 Université de Montpellier, UFR de Médecine, Montpel - lier, France ©2020 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol. 2018.208306 Received: October 5, 2018. Accepted: July 5, 2019. Pre-published: July 9, 2019. Correspondence: JEROME MOREAUX - [email protected] Supplementary experiment procedures Kinome Index A list of 661 genes of kinases or kinases related have been extracted from literature9, and challenged in the HM cohort for OS prognostic values The prognostic value of each of the genes was computed using maximally selected rank test from R package MaxStat. After Benjamini Hochberg multiple testing correction a list of 104 significant prognostic genes has been extracted. This second list has then been challenged for similar prognosis value in the UAMS-TT2 validation cohort.
    [Show full text]