101
|
Zhao H, Wang Y, Li B, Zheng T, Liu X, Hu BH, Che J, Zhao T, Chen J, Hatzoglou M, Zhang X, Fan Z, Zheng Q. Role of Endoplasmic Reticulum Stress in Otitis Media. Front Genet 2020; 11:495. [PMID: 32536938 PMCID: PMC7267009 DOI: 10.3389/fgene.2020.00495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 04/20/2020] [Indexed: 11/19/2022] Open
Abstract
Endoplasmic reticulum (ER) stress occurs in many inflammatory responses. Here, we investigated the role of ER stress and its associated apoptosis in otitis media (OM) to elucidate the mechanisms of OM and the signaling crosstalk between ER stress and other cell damage pathways, including inflammatory cytokines and apoptosis. We examined the expression of inflammatory cytokine- and ER stress-related genes by qRT-PCR, Western blotting, and immunohistochemistry (IHC) in the middle ear of C57BL/6J mice after challenge with peptidoglycan polysaccharide (PGPS), an agent inducing OM. We also evaluated the effect of the suppression of ER stress with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor. The study revealed the upregulation of ER stress- and apoptosis-related gene expression after the PGPS treatment, specifically ATF6, CHOP, BIP, caspase-12, and caspase-3. TUDCA treatment of PGPS-treated mice decreased OM; reduced the expression of CHOP, BIP, and caspase 3; and significantly decreased the proinflammatory gene expression of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). These results suggest that PGPS triggers ER stress and downstream proinflammatory gene expression in OM and that inhibition of ER stress alleviates OM. We propose that ER stress plays a critical role in inflammation and cell death, leading to the development of OM and points to ER stress inhibition as a potential therapeutic approach for the prevention of OM.
Collapse
Affiliation(s)
- Hongchun Zhao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Yanfei Wang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Bo Li
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Tihua Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Xiuzhen Liu
- Clinical Laboratory, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Bo Hua Hu
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Juan Che
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Tong Zhao
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Jun Chen
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Maria Hatzoglou
- Department of Genetics, Case Western Reserve University, Cleveland, OH, United States
| | - Xiaolin Zhang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | - Zhaomin Fan
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingyin Zheng
- Department of Otolaryngology-Head & Neck Surgery, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
102
|
Translational profiling of macrophages infected with Leishmania donovani identifies mTOR- and eIF4A-sensitive immune-related transcripts. PLoS Pathog 2020; 16:e1008291. [PMID: 32479529 PMCID: PMC7310862 DOI: 10.1371/journal.ppat.1008291] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/23/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022] Open
Abstract
The protozoan parasite Leishmania donovani (L. donovani) causes visceral leishmaniasis, a chronic infection which is fatal when untreated. Herein, we investigated whether in addition to altering transcription, L. donovani modulates host mRNA translation to establish a successful infection. Polysome-profiling revealed that one third of protein-coding mRNAs expressed in primary mouse macrophages are differentially translated upon infection with L. donovani promastigotes or amastigotes. Gene ontology analysis identified key biological processes enriched for translationally regulated mRNAs and were predicted to be either activated (e.g. chromatin remodeling and RNA metabolism) or inhibited (e.g. intracellular trafficking and antigen presentation) upon infection. Mechanistic in silico and biochemical analyses showed selective activation mTOR- and eIF4A-dependent mRNA translation, including transcripts encoding central regulators of mRNA turnover and inflammation (i.e. PABPC1, EIF2AK2, and TGF-β). L. donovani survival within macrophages was favored under mTOR inhibition but was dampened by pharmacological blockade of eIF4A. Overall, this study uncovers a vast yet selective reprogramming of the host cell translational landscape early during L. donovani infection, and suggests that some of these changes are involved in host defense mechanisms while others are part of parasite-driven survival strategies. Further in vitro and in vivo investigation will shed light on the contribution of mTOR- and eIF4A-dependent translational programs to the outcome of visceral leishmaniasis.
Collapse
|
103
|
Jewer M, Lee L, Leibovitch M, Zhang G, Liu J, Findlay SD, Vincent KM, Tandoc K, Dieters-Castator D, Quail DF, Dutta I, Coatham M, Xu Z, Puri A, Guan BJ, Hatzoglou M, Brumwell A, Uniacke J, Patsis C, Koromilas A, Schueler J, Siegers GM, Topisirovic I, Postovit LM. Translational control of breast cancer plasticity. Nat Commun 2020; 11:2498. [PMID: 32427827 PMCID: PMC7237473 DOI: 10.1038/s41467-020-16352-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/27/2020] [Indexed: 12/19/2022] Open
Abstract
Plasticity of neoplasia, whereby cancer cells attain stem-cell-like properties, is required for disease progression and represents a major therapeutic challenge. We report that in breast cancer cells NANOG, SNAIL and NODAL transcripts manifest multiple isoforms characterized by different 5' Untranslated Regions (5'UTRs), whereby translation of a subset of these isoforms is stimulated under hypoxia. The accumulation of the corresponding proteins induces plasticity and "fate-switching" toward stem cell-like phenotypes. Mechanistically, we observe that mTOR inhibitors and chemotherapeutics induce translational activation of a subset of NANOG, SNAIL and NODAL mRNA isoforms akin to hypoxia, engendering stem-cell-like phenotypes. These effects are overcome with drugs that antagonize translational reprogramming caused by eIF2α phosphorylation (e.g. ISRIB), suggesting that the Integrated Stress Response drives breast cancer plasticity. Collectively, our findings reveal a mechanism of induction of plasticity of breast cancer cells and provide a molecular basis for therapeutic strategies aimed at overcoming drug resistance and abrogating metastasis.
Collapse
Affiliation(s)
- Michael Jewer
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Laura Lee
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Matthew Leibovitch
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Guihua Zhang
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Jiahui Liu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Scott D Findlay
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Krista M Vincent
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Kristofferson Tandoc
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Dylan Dieters-Castator
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Daniela F Quail
- Goodman Cancer Center, McGill University, Montreal, QC, Canada
| | - Indrani Dutta
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | | | - Zhihua Xu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Aakshi Puri
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Bo-Jhih Guan
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Maria Hatzoglou
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrea Brumwell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Christos Patsis
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Antonis Koromilas
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Julia Schueler
- Charles River Discovery Research Services Germany, Freiburg, Germany
| | | | - Ivan Topisirovic
- Lady Davis Institute, Departments of Oncology and Biochemistry, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Lynne-Marie Postovit
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
104
|
Guallar D, Fuentes-Iglesias A, Souto Y, Ameneiro C, Freire-Agulleiro O, Pardavila JA, Escudero A, Garcia-Outeiral V, Moreira T, Saenz C, Xiong H, Liu D, Xiao S, Hou Y, Wu K, Torrecilla D, Hartner JC, Blanco MG, Lee LJ, López M, Walkley CR, Wang J, Fidalgo M. ADAR1-Dependent RNA Editing Promotes MET and iPSC Reprogramming by Alleviating ER Stress. Cell Stem Cell 2020; 27:300-314.e11. [PMID: 32396862 DOI: 10.1016/j.stem.2020.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 04/02/2020] [Accepted: 04/23/2020] [Indexed: 12/31/2022]
Abstract
RNA editing of adenosine to inosine (A to I) is catalyzed by ADAR1 and dramatically alters the cellular transcriptome, although its functional roles in somatic cell reprogramming are largely unexplored. Here, we show that loss of ADAR1-mediated A-to-I editing disrupts mesenchymal-to-epithelial transition (MET) during induced pluripotent stem cell (iPSC) reprogramming and impedes acquisition of induced pluripotency. Using chemical and genetic approaches, we show that absence of ADAR1-dependent RNA editing induces aberrant innate immune responses through the double-stranded RNA (dsRNA) sensor MDA5, unleashing endoplasmic reticulum (ER) stress and hindering epithelial fate acquisition. We found that A-to-I editing impedes MDA5 sensing and sequestration of dsRNAs encoding membrane proteins, which promote ER homeostasis by activating the PERK-dependent unfolded protein response pathway to consequently facilitate MET. This study therefore establishes a critical role for ADAR1 and its A-to-I editing activity during cell fate transitions and delineates a key regulatory layer underlying MET to control efficient reprogramming.
Collapse
Affiliation(s)
- Diana Guallar
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Biochemistry and Molecular Biology, USC, Santiago de Compostela 15782, Spain.
| | - Alejandro Fuentes-Iglesias
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain
| | - Yara Souto
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain
| | - Cristina Ameneiro
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain
| | - Oscar Freire-Agulleiro
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain; NeurObesity Group & CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela 15706, Spain
| | - Jose Angel Pardavila
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain
| | - Adriana Escudero
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain
| | - Vera Garcia-Outeiral
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain
| | - Tiago Moreira
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain
| | - Carmen Saenz
- The Black Family Stem Cell Institute, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Heng Xiong
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Dongbing Liu
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Shidi Xiao
- BGI-Shenzhen, Shenzhen 518083, China; Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Yong Hou
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Kui Wu
- BGI-Shenzhen, Shenzhen 518083, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Daniel Torrecilla
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain
| | - Jochen C Hartner
- Horizon Discovery, Cambridge Research Park, Cambridge CB25 9TL, UK
| | - Miguel G Blanco
- Department of Biochemistry and Molecular Biology, USC, Santiago de Compostela 15782, Spain
| | - Leo J Lee
- BGI-Shenzhen, Shenzhen 518083, China; Department of Electrical and Computer Engineering, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Miguel López
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain; NeurObesity Group & CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela 15706, Spain
| | - Carl R Walkley
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3000, Australia
| | - Jianlong Wang
- The Black Family Stem Cell Institute, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Miguel Fidalgo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC)-Health Research Institute (IDIS), Santiago de Compostela 15782, Spain; Department of Physiology, USC, Santiago de Compostela 15782, Spain.
| |
Collapse
|
105
|
Clementi E, Inglin L, Beebe E, Gsell C, Garajova Z, Markkanen E. Persistent DNA damage triggers activation of the integrated stress response to promote cell survival under nutrient restriction. BMC Biol 2020; 18:36. [PMID: 32228693 PMCID: PMC7106853 DOI: 10.1186/s12915-020-00771-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Base-excision repair (BER) is a central DNA repair mechanism responsible for the maintenance of genome integrity. Accordingly, BER defects have been implicated in cancer, presumably by precipitating cellular transformation through an increase in the occurrence of mutations. Hence, tight adaptation of BER capacity is essential for DNA stability. However, counterintuitive to this, prolonged exposure of cells to pro-inflammatory molecules or DNA-damaging agents causes a BER deficiency by downregulating the central scaffold protein XRCC1. The rationale for this XRCC1 downregulation in response to persistent DNA damage remains enigmatic. Based on our previous findings that XRCC1 downregulation causes wide-ranging anabolic changes, we hypothesised that BER depletion could enhance cellular survival under stress, such as nutrient restriction. RESULTS Here, we demonstrate that persistent single-strand breaks (SSBs) caused by XRCC1 downregulation trigger the integrated stress response (ISR) to promote cellular survival under nutrient-restricted conditions. ISR activation depends on DNA damage signalling via ATM, which triggers PERK-mediated eIF2α phosphorylation, increasing translation of the stress-response factor ATF4. Furthermore, we demonstrate that SSBs, induced either through depletion of the transcription factor Sp1, responsible for XRCC1 levels, or through prolonged oxidative stress, trigger ISR-mediated cell survival under nutrient restriction as well. Finally, the ISR pathway can also be initiated by persistent DNA double-strand breaks. CONCLUSIONS Our results uncover a previously unappreciated connection between persistent DNA damage, caused by a decrease in BER capacity or direct induction of DNA damage, and the ISR pathway that supports cell survival in response to genotoxic stress with implications for tumour biology and beyond.
Collapse
Affiliation(s)
- Elena Clementi
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Larissa Inglin
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Corina Gsell
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Zuzana Garajova
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, 8057, Zürich, Switzerland.
| |
Collapse
|
106
|
Komar AA, Merrick WC. A Retrospective on eIF2A-and Not the Alpha Subunit of eIF2. Int J Mol Sci 2020; 21:E2054. [PMID: 32192132 PMCID: PMC7139343 DOI: 10.3390/ijms21062054] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/29/2020] [Accepted: 03/13/2020] [Indexed: 12/31/2022] Open
Abstract
Initiation of protein synthesis in eukaryotes is a complex process requiring more than 12 different initiation factors, comprising over 30 polypeptide chains. The functions of many of these factors have been established in great detail; however, the precise role of some of them and their mechanism of action is still not well understood. Eukaryotic initiation factor 2A (eIF2A) is a single chain 65 kDa protein that was initially believed to serve as the functional homologue of prokaryotic IF2, since eIF2A and IF2 catalyze biochemically similar reactions, i.e., they stimulate initiator Met-tRNAi binding to the small ribosomal subunit. However, subsequent identification of a heterotrimeric 126 kDa factor, eIF2 (α,β,γ) showed that this factor, and not eIF2A, was primarily responsible for the binding of Met-tRNAi to 40S subunit in eukaryotes. It was found however, that eIF2A can promote recruitment of Met-tRNAi to 40S/mRNA complexes under conditions of inhibition of eIF2 activity (eIF2α-phosphorylation), or its absence. eIF2A does not function in major steps in the initiation process, but is suggested to act at some minor/alternative initiation events such as re-initiation, internal initiation, or non-AUG initiation, important for translational control of specific mRNAs. This review summarizes our current understanding of the eIF2A structure and function.
Collapse
Affiliation(s)
- Anton A. Komar
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological and Environmental Sciences, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - William C. Merrick
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
| |
Collapse
|
107
|
Pharmacological induction of selective endoplasmic reticulum retention as a strategy for cancer therapy. Nat Commun 2020; 11:1304. [PMID: 32161259 PMCID: PMC7066181 DOI: 10.1038/s41467-020-15067-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
The integrated stress response (ISR) converges on eIF2α phosphorylation to regulate protein synthesis. ISR is activated by several stress conditions, including endoplasmic reticulum (ER) stress, executed by protein kinase R-like endoplasmic reticulum kinase (PERK). We report that ER stress combined with ISR inhibition causes an impaired maturation of several tyrosine kinase receptors (RTKs), consistent with a partial block of their trafficking from the ER to the Golgi. Other proteins mature or are secreted normally, indicating selective retention in the ER (sERr). sERr is relieved upon protein synthesis attenuation and is accompanied by the generation of large mixed disulfide bonded complexes, including ERp44. sERr was pharmacologically recapitulated by combining the HIV-protease inhibitor nelfinavir with ISRIB, an experimental drug that inhibits ISR. Nelfinavir/ISRIB combination is highly effective to inhibit the growth of RTK-addicted cell lines and hepatocellular (HCC) cells in vitro and in vivo. Thus, pharmacological sERr can be utilized as a modality for cancer treatment. Inhibition of PERK, an endoplasmic reticulum (ER) unfolded protein response (UPR) protein, is a potential pharmacological target for cancer treatment. Here, the authors show that inhibition of PERK under ER stress affects trafficking from the ER to the surface of several key receptor tyrosine kinases, suggesting a selective ER retention.
Collapse
|
108
|
Jaud M, Philippe C, Di Bella D, Tang W, Pyronnet S, Laurell H, Mazzolini L, Rouault-Pierre K, Touriol C. Translational Regulations in Response to Endoplasmic Reticulum Stress in Cancers. Cells 2020; 9:cells9030540. [PMID: 32111004 PMCID: PMC7140484 DOI: 10.3390/cells9030540] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
During carcinogenesis, almost all the biological processes are modified in one way or another. Among these biological processes affected, anomalies in protein synthesis are common in cancers. Indeed, cancer cells are subjected to a wide range of stresses, which include physical injuries, hypoxia, nutrient starvation, as well as mitotic, oxidative or genotoxic stresses. All of these stresses will cause the accumulation of unfolded proteins in the Endoplasmic Reticulum (ER), which is a major organelle that is involved in protein synthesis, preservation of cellular homeostasis, and adaptation to unfavourable environment. The accumulation of unfolded proteins in the endoplasmic reticulum causes stress triggering an unfolded protein response in order to promote cell survival or to induce apoptosis in case of chronic stress. Transcription and also translational reprogramming are tightly controlled during the unfolded protein response to ensure selective gene expression. The majority of stresses, including ER stress, induce firstly a decrease in global protein synthesis accompanied by the induction of alternative mechanisms for initiating the translation of mRNA, later followed by a translational recovery. After a presentation of ER stress and the UPR response, we will briefly present the different modes of translation initiation, then address the specific translational regulatory mechanisms acting during reticulum stress in cancers and highlight the importance of translational control by ER stress in tumours.
Collapse
Affiliation(s)
- Manon Jaud
- Inserm UMR1037, CRCT (Cancer Research Center of Toulouse), F-31037 Toulouse, France; (M.J.); (S.P.); (L.M.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France;
| | - Céline Philippe
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (W.T.); (K.R.-P.)
| | - Doriana Di Bella
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (W.T.); (K.R.-P.)
| | - Weiwei Tang
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (W.T.); (K.R.-P.)
| | - Stéphane Pyronnet
- Inserm UMR1037, CRCT (Cancer Research Center of Toulouse), F-31037 Toulouse, France; (M.J.); (S.P.); (L.M.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France;
| | - Henrik Laurell
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France;
- Inserm UMR1048, I2MC (Institut des Maladies Métaboliques et Cardiovasculaires), BP 84225, CEDEX 04, 31 432 Toulouse, France
| | - Laurent Mazzolini
- Inserm UMR1037, CRCT (Cancer Research Center of Toulouse), F-31037 Toulouse, France; (M.J.); (S.P.); (L.M.)
- CNRS ERL5294, CRCT, F-31037 Toulouse, France
| | - Kevin Rouault-Pierre
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (C.P.); (D.D.B.); (W.T.); (K.R.-P.)
| | - Christian Touriol
- Inserm UMR1037, CRCT (Cancer Research Center of Toulouse), F-31037 Toulouse, France; (M.J.); (S.P.); (L.M.)
- Université Toulouse III Paul-Sabatier, F-31000 Toulouse, France;
- Correspondence:
| |
Collapse
|
109
|
Oertlin C, Lorent J, Murie C, Furic L, Topisirovic I, Larsson O. Generally applicable transcriptome-wide analysis of translation using anota2seq. Nucleic Acids Res 2020; 47:e70. [PMID: 30926999 PMCID: PMC6614820 DOI: 10.1093/nar/gkz223] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 03/18/2019] [Accepted: 03/28/2019] [Indexed: 12/28/2022] Open
Abstract
mRNA translation plays an evolutionarily conserved role in homeostasis and when dysregulated contributes to various disorders including metabolic and neurological diseases and cancer. Notwithstanding that optimal and universally applicable methods are critical for understanding the complex role of translational control under physiological and pathological conditions, approaches to analyze translatomes are largely underdeveloped. To address this, we developed the anota2seq algorithm which outperforms current methods for statistical identification of changes in translation. Notably, in contrast to available analytical methods, anota2seq also allows specific identification of an underappreciated mode of gene expression regulation whereby translation acts as a buffering mechanism which maintains protein levels despite fluctuations in corresponding mRNA abundance (‘translational buffering’). Thus, the universal anota2seq algorithm allows efficient and hitherto unprecedented interrogation of translatomes which is anticipated to advance knowledge regarding the role of translation in homeostasis and disease.
Collapse
Affiliation(s)
- Christian Oertlin
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Julie Lorent
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Carl Murie
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Luc Furic
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy & Developmental Biology, Monash University, VIC, Australia.,Prostate Cancer Translational Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Ivan Topisirovic
- Lady Davis Institute, SMBD Jewish General Hospital, Gerald Bronfman Department of Oncology, and Departments of Experimental Medicine, and Biochemistry McGill University, Montreal, Canada
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
110
|
Garshott DM, Sundaramoorthy E, Leonard M, Bennett EJ. Distinct regulatory ribosomal ubiquitylation events are reversible and hierarchically organized. eLife 2020; 9:54023. [PMID: 32011234 PMCID: PMC7064338 DOI: 10.7554/elife.54023] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/01/2020] [Indexed: 11/13/2022] Open
Abstract
Activation of the integrated stress response (ISR) or the ribosome-associated quality control (RQC) pathway stimulates regulatory ribosomal ubiquitylation (RRub) on distinct 40S ribosomal proteins, yet the cellular role and fate of ubiquitylated proteins remain unclear. We demonstrate that uS10 and uS5 ubiquitylation are dependent upon eS10 or uS3 ubiquitylation, respectively, suggesting that a hierarchical relationship exists among RRub events establishing a ubiquitin code on ribosomes. We show that stress dependent RRub events diminish after initial stimuli and that demodification by deubiquitylating enzymes contributes to reduced RRub levels during stress recovery. Utilizing an optical RQC reporter we identify OTUD3 and USP21 as deubiquitylating enzymes that antagonize ZNF598-mediated 40S ubiquitylation and can limit RQC activation. Critically, cells lacking USP21 or OTUD3 have altered RQC activity and delayed eS10 deubiquitylation indicating a functional role for deubiquitylating enzymes within the RQC pathway. Ribosomes are cellular machines that build proteins by latching on and then reading template molecules known as mRNAs. Several ribosomes may be moving along the same piece of mRNA at the same time, each making their own copy of the same protein. Damage to an mRNA or other problems may cause a ribosome to stall, leading to subsequent collisions. A quality control pathway exists to identify stalled ribosomes and fix the ‘traffic jams’. It relies on enzymes that tag halted ribosomes with molecules known as ubiquitin. The cell then removes these ribosomes from the mRNA and destroys the proteins they were making. Afterwards, additional enzymes take off the ubiquitin tags so the cell can recycle the ribosomes. These enzymes are key to signaling the end of the quality control event, yet their identity was still unclear. Garshott et al. used genetic approaches to study traffic jams of ribosomes in mammalian cells. The experiments showed that cells added sets of ubiquitin tags to stalled ribosomes in a specific order. Two enzymes, known as USP21 and OTUD3, could stop this process; this allowed ribosomes to carry on reading mRNA. Further work revealed that the ribosomes in cells that produce higher levels of USP21 and OTUD3 were less likely to stall on mRNA. On the other hand, ribosomes in cells lacking USP1 and OTUD3 retained their ubiquitin tags for longer and were more likely to stall. The findings of Garshott et al. reveal that USP21 and OTUD3 are involved in the quality control pathway which fixes ribosome traffic jams. In mice, problems in this pathway have been linked with neurons dying or being damaged because toxic protein products start to accumulate in cells; this is similar to what happens in human conditions such as Alzheimer's and Parkinson's diseases. Using ubiquitin to target and potentially fix the pathway could therefore open the door to new therapies.
Collapse
Affiliation(s)
- Danielle M Garshott
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Elayanambi Sundaramoorthy
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Marilyn Leonard
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Eric J Bennett
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| |
Collapse
|
111
|
Wang LY, Cui JJ, Guo CX, Yin JY. A New Way to Discover IRESs in Pathology or Stress Conditions? Harnessing Latest High-Throughput Technologies. Bioessays 2020; 42:e1900180. [PMID: 31909834 DOI: 10.1002/bies.201900180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/10/2019] [Indexed: 11/12/2022]
Abstract
The cellular internal ribosomal entry site (IRES) is one of the most important elements to mediate cap-independent translational initiation, especially under conditions of stress and pathology. However, a high-throughput method to discover IRESs in these conditions is still lacking. Here, a possible way IRES long-read sequencing based on the latest high-throughput technologies is proposed to solve this problem. Based on this design, diversity and integrity of the transcriptome from original samples can be kept. The micro-environment that stimulates or inhibits IRES activity can also be mimicked. By using long read-length sequencing technology, additional experiments that are essential for ruling out the cryptic promoters or splicing events in routine IRES identification processes can be circumvented. It is hoped that this proposed methodology may be adopted for IRES element discovery, hence uncovering the full extent of the role of IRESs in disease, development, and stress. Also see the video abstract here https://youtu.be/JuWBbMzWXS8.
Collapse
Affiliation(s)
- Lei-Yun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410078, P. R. China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Jia-Jia Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410078, P. R. China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, P. R. China
| | - Cheng-Xian Guo
- Central of Clinic Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, 410013, P. R. China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410078, P. R. China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China.,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, P. R. China.,Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Changsha, 410078, P. R. China.,Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research Center, Changsha, 410078, P. R. China
| |
Collapse
|
112
|
The integrated stress response promotes B7H6 expression. J Mol Med (Berl) 2019; 98:135-148. [PMID: 31838577 PMCID: PMC6952340 DOI: 10.1007/s00109-019-01859-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
The B7 family member, B7H6, is a ligand for the natural killer cell receptor NKp30. B7H6 is hardly expressed on normal tissues, but undergoes upregulation on different types of tumors, implicating it as an attractive target for cancer immunotherapy. The molecular mechanisms that control B7H6 expression are poorly understood. We report that in contrast to other NK cell ligands, endoplasmic reticulum (ER) stress upregulates B7H6 mRNA levels and surface expression. B7H6 induction by ER stress requires protein kinase R-like ER kinase (PERK), one of the three canonical sensors of the unfolded protein response. PERK phosphorylates eIF2α, which regulates protein synthesis and gene expression. Because eIF2α is phosphorylated by several kinases following different stress conditions, the program downstream to eIF2α phosphorylation is called the integrated stress response (ISR). Several drugs were reported to promote the ISR. Nelfinavir and lopinavir, two clinically approved HIV protease inhibitors, promote eIF2α phosphorylation by different mechanisms. We show that nelfinavir and lopinavir sustainably instigate B7H6 expression at their pharmacologically relevant concentrations. As such, ER stress and ISR conditions sensitize melanoma targets to CAR-T cells directed against B7H6. Our study highlights a novel mechanism to induce B7H6 expression and suggests a pharmacological approach to improve B7H6-directed immunotherapy. KEY MESSAGES: B7H6 is induced by ER stress in a PERK-dependent mechanism. Induction of B7H6 is obtained pharmacologically by HIV protease inhibitors. Exposure of tumor cells to the HIV protease inhibitor nelfinavir improves the recognition by B7H6-directed CAR-T.
Collapse
|
113
|
Young-Baird SK, Lourenço MB, Elder MK, Klann E, Liebau S, Dever TE. Suppression of MEHMO Syndrome Mutation in eIF2 by Small Molecule ISRIB. Mol Cell 2019; 77:875-886.e7. [PMID: 31836389 DOI: 10.1016/j.molcel.2019.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/07/2019] [Accepted: 11/06/2019] [Indexed: 12/27/2022]
Abstract
Dysregulation of cellular protein synthesis is linked to a variety of diseases. Mutations in EIF2S3, encoding the γ subunit of the heterotrimeric eukaryotic translation initiation factor eIF2, cause MEHMO syndrome, an X-linked intellectual disability disorder. Here, using patient-derived induced pluripotent stem cells, we show that a mutation at the C terminus of eIF2γ impairs CDC123 promotion of eIF2 complex formation and decreases the level of eIF2-GTP-Met-tRNAiMet ternary complexes. This reduction in eIF2 activity results in dysregulation of global and gene-specific protein synthesis and enhances cell death upon stress induction. Addition of the drug ISRIB, an activator of the eIF2 guanine nucleotide exchange factor, rescues the cell growth, translation, and neuronal differentiation defects associated with the EIF2S3 mutation, offering the possibility of therapeutic intervention for MEHMO syndrome.
Collapse
Affiliation(s)
- Sara K Young-Baird
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA; National Institute of General Medical Sciences, NIH, Bethesda, MD 20892, USA.
| | - Maíra Bertolessi Lourenço
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Megan K Elder
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Thomas E Dever
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
114
|
Lorent J, Kusnadi EP, van Hoef V, Rebello RJ, Leibovitch M, Ristau J, Chen S, Lawrence MG, Szkop KJ, Samreen B, Balanathan P, Rapino F, Close P, Bukczynska P, Scharmann K, Takizawa I, Risbridger GP, Selth LA, Leidel SA, Lin Q, Topisirovic I, Larsson O, Furic L. Translational offsetting as a mode of estrogen receptor α-dependent regulation of gene expression. EMBO J 2019; 38:e101323. [PMID: 31556460 PMCID: PMC6885737 DOI: 10.15252/embj.2018101323] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor alpha (ERα) activity is associated with increased cancer cell proliferation. Studies aiming to understand the impact of ERα on cancer-associated phenotypes have largely been limited to its transcriptional activity. Herein, we demonstrate that ERα coordinates its transcriptional output with selective modulation of mRNA translation. Importantly, translational perturbations caused by depletion of ERα largely manifest as "translational offsetting" of the transcriptome, whereby amounts of translated mRNAs and corresponding protein levels are maintained constant despite changes in mRNA abundance. Transcripts whose levels, but not polysome association, are reduced following ERα depletion lack features which limit translation efficiency including structured 5'UTRs and miRNA target sites. In contrast, mRNAs induced upon ERα depletion whose polysome association remains unaltered are enriched in codons requiring U34-modified tRNAs for efficient decoding. Consistently, ERα regulates levels of U34-modifying enzymes and thereby controls levels of U34-modified tRNAs. These findings unravel a hitherto unprecedented mechanism of ERα-dependent orchestration of transcriptional and translational programs that may be a pervasive mechanism of proteome maintenance in hormone-dependent cancers.
Collapse
Affiliation(s)
- Julie Lorent
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Eric P Kusnadi
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| | - Vincent van Hoef
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Richard J Rebello
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
| | - Matthew Leibovitch
- Gerald Bronfman Department of Oncology and Departments of Biochemistry and Experimental MedicineLady Davis InstituteMcGill UniversityMontrealQCCanada
| | - Johannes Ristau
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Shan Chen
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Mitchell G Lawrence
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| | - Krzysztof J Szkop
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Baila Samreen
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Preetika Balanathan
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
| | - Francesca Rapino
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | - Pierre Close
- Laboratory of Cancer SignalingGIGA‐InstituteUniversity of LiègeLiègeBelgium
| | - Patricia Bukczynska
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
| | - Karin Scharmann
- Max Planck Institute for Molecular BiomedicineMünsterGermany
- Cells‐in‐Motion Cluster of ExcellenceUniversity of MünsterMünsterGermany
| | - Itsuhiro Takizawa
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
| | - Gail P Risbridger
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's HealthAdelaide Medical SchoolFaculty of Health and Medical SciencesThe University of AdelaideAdelaideSAAustralia
| | - Sebastian A Leidel
- Max Planck Institute for Molecular BiomedicineMünsterGermany
- Cells‐in‐Motion Cluster of ExcellenceUniversity of MünsterMünsterGermany
- Department of Chemistry and BiochemistryUniversity of BernBernSwitzerland
| | - Qishan Lin
- RNA Epitranscriptomics & Proteomics ResourceDepartment of ChemistryUniversity at AlbanyAlbanyNYUSA
| | - Ivan Topisirovic
- Gerald Bronfman Department of Oncology and Departments of Biochemistry and Experimental MedicineLady Davis InstituteMcGill UniversityMontrealQCCanada
| | - Ola Larsson
- Science for Life LaboratoryDepartment of Oncology‐PathologyKarolinska InstitutetSolnaSweden
| | - Luc Furic
- Prostate Cancer Translational Research LaboratoryPeter MacCallum Cancer CentreMelbourneVic.Australia
- Cancer ProgramBiomedicine Discovery Institute and Department of Anatomy and Developmental BiologyMonash UniversityClaytonVic.Australia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVic.Australia
| |
Collapse
|
115
|
Pizzinga M, Harvey RF, Garland GD, Mordue R, Dezi V, Ramakrishna M, Sfakianos A, Monti M, Mulroney TE, Poyry T, Willis AE. The cell stress response: extreme times call for post‐transcriptional measures. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 11:e1578. [DOI: 10.1002/wrna.1578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/09/2019] [Accepted: 10/16/2019] [Indexed: 12/26/2022]
Affiliation(s)
| | | | | | - Ryan Mordue
- MRC Toxicology Unit University of Cambridge Leicester UK
| | - Veronica Dezi
- MRC Toxicology Unit University of Cambridge Leicester UK
| | | | | | - Mie Monti
- MRC Toxicology Unit University of Cambridge Leicester UK
| | | | - Tuija Poyry
- MRC Toxicology Unit University of Cambridge Leicester UK
| | - Anne E. Willis
- MRC Toxicology Unit University of Cambridge Leicester UK
| |
Collapse
|
116
|
Pathria G, Lee JS, Hasnis E, Tandoc K, Scott DA, Verma S, Feng Y, Larue L, Sahu AD, Topisirovic I, Ruppin E, Ronai ZA. Translational reprogramming marks adaptation to asparagine restriction in cancer. Nat Cell Biol 2019; 21:1590-1603. [PMID: 31740775 PMCID: PMC7307327 DOI: 10.1038/s41556-019-0415-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 09/25/2019] [Indexed: 01/24/2023]
Abstract
While amino acid restriction remains an attractive strategy for cancer therapy, metabolic adaptations limit its effectiveness. Here we demonstrate a role of translational reprogramming in the survival of asparagine-restricted cancer cells. Asparagine limitation in melanoma and pancreatic cancer cells activates RTK-MAPK as part of a feedforward mechanism involving mTORC1-dependent increase in MNK1 and eIF4E, resulting in enhanced translation of ATF4 mRNA. MAPK inhibition attenuates translational induction of ATF4 and the expression of its target asparagine biosynthesis enzyme ASNS, sensitizing melanoma and pancreatic tumors to asparagine restriction, reflected in their growth inhibition. Correspondingly, low ASNS expression is among the top predictors of response to MAPK signaling inhibitors in melanoma patients and is associated with favorable prognosis, when combined with low MAPK signaling activity. While unveiling a previously unknown axis of adaptation to asparagine deprivation, these studies offer the rationale for clinical evaluation of MAPK inhibitors in combination with asparagine restriction approaches.
Collapse
Affiliation(s)
- Gaurav Pathria
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Joo Sang Lee
- Cancer Data Science Lab (CDSL), National Cancer Institute, National Institute of Health, Bethesda, MD, USA.,Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Erez Hasnis
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Kristofferson Tandoc
- Gerald Bronfman Department of Oncology, Lady Davis Institute, SMBD Jewish General Hospital, and Departments of Experimental Medicine and Biochemistry, McGill University, Montreal, Quebec, Canada
| | - David A Scott
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sachin Verma
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Yongmei Feng
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lionel Larue
- Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, INSERM U1021, Orsay, France.,Universitê Paris-Sud and Université Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipe Labellisée Ligue Contre le Cancer, Orsay, France
| | - Avinash D Sahu
- Harvard School of Public Health and Massachusetts General Hospital, Boston, MA, USA
| | - Ivan Topisirovic
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Eytan Ruppin
- Cancer Data Science Lab (CDSL), National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
117
|
RITA requires eIF2α-dependent modulation of mRNA translation for its anti-cancer activity. Cell Death Dis 2019; 10:845. [PMID: 31699971 PMCID: PMC6838152 DOI: 10.1038/s41419-019-2074-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/30/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022]
Abstract
Tumor protein 53 (p53, encoded by the TP53 gene) is a key tumor suppressor regulating cell fates in response to internal and external stresses. As TP53 is mutated or silenced in a majority of tumors, reactivation of p53 by small molecules represents a promising strategy in cancer therapeutics. One such agent is RITA (reactivation of p53 and induction of tumor cell apoptosis), which restores p53 expression in cells with hyperactive HDM2 and induces apoptosis. Yet, mechanisms underlying the anticancer activity of RITA are incompletely understood. Here we show that RITA suppresses mRNA translation independently of p53 by inducing eIF2α phosphorylation. Surprisingly, reactivation of p53 following RITA treatment is critically dependent on eIF2α phosphorylation. Moreover, inhibition of eIF2α phosphorylation attenuates pro-apoptotic and anti-neoplastic effects of RITA, while inducing phosphorylation of eIF2α enhances the anticancer activity of RITA. Collectively, these findings demonstrate that the translational machinery plays a major role in determining the antineoplastic activity of RITA, and suggest that combining p53 activators and translation modulators may be beneficial.
Collapse
|
118
|
Gonen N, Meller A, Sabath N, Shalgi R. Amino Acid Biosynthesis Regulation during Endoplasmic Reticulum Stress Is Coupled to Protein Expression Demands. iScience 2019; 19:204-213. [PMID: 31377665 PMCID: PMC6698312 DOI: 10.1016/j.isci.2019.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/11/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) stress response, also known as the unfolded protein response (UPR), is a complex cellular response to ER protein misfolding that involves transcriptional regulatory branches and a PERK-mediated translational regulatory branch. Here we revealed that amino acid biosynthesis regulation is coupled to protein synthesis demands during ER stress. Specifically, we demonstrated that the UPR leads to PERK-dependent induction in the biosynthesis of specific amino acids, and to upregulation of their corresponding tRNA synthetases. Furthermore, we found that sequences of UPR-upregulated proteins are significantly enriched with these UPR-induced amino acids. Interestingly, whereas the UPR leads to repression of ER target proteins, we showed that secreted proteins tended to escape this repression and were highly enriched for the UPR-induced amino acids. Our results unravel coordination between amino acid supply, namely, biosynthesis and tRNA loading, and demand from UPR-induced proteins under ER stress, thus revealing an additional regulatory layer of protein synthesis. Coordination of amino acid supply and protein synthesis demand during ER stress Specific amino acid biosynthesis and cognate tRNA synthetases induction by the UPR UPR-induced amino acids support amino acid demand of UPR-upregulated proteins UPR-induced amino acids are highly enriched within secreted proteins
Collapse
Affiliation(s)
- Nir Gonen
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Anatoly Meller
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Niv Sabath
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Reut Shalgi
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
119
|
Hetz C, Axten JM, Patterson JB. Pharmacological targeting of the unfolded protein response for disease intervention. Nat Chem Biol 2019; 15:764-775. [PMID: 31320759 DOI: 10.1038/s41589-019-0326-2] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
Accumulation of unfolded proteins at the endoplasmic reticulum (ER) is a salient attribute of many human diseases including obesity, liver disorders, cancer, diabetes and neurodegeneration. To restore ER proteostasis, cells activate the unfolded protein response (UPR), a signaling pathway that imposes adaptive programs or triggers apoptosis of damaged cells. The UPR is critical to sustain the normal function of specialized secretory cells (i.e., pancreatic β cells and B lymphocytes) and to control the production of lipids and cholesterol in the liver. In the context of disease, adaptive UPR responses have been linked to the growth of solid tumors, whereas chronic ER stress contributes to cell dysfunction in brain diseases, metabolic syndromes, among other conditions. Here we discuss recent developments in the design and optimization of novel compounds to manipulate UPR signaling and their efficacy in various disease models.
Collapse
Affiliation(s)
- Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile. .,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, USA. .,Department of Immunology and Infectious diseases, Harvard School of Public Health, Boston, MA, USA.
| | - Jeffrey M Axten
- Medicinal Chemistry, Medicine Design, GlaxoSmithKline, Collegeville, PA, USA.
| | | |
Collapse
|
120
|
Papadopoli D, Boulay K, Kazak L, Pollak M, Mallette FA, Topisirovic I, Hulea L. mTOR as a central regulator of lifespan and aging. F1000Res 2019; 8:F1000 Faculty Rev-998. [PMID: 31316753 PMCID: PMC6611156 DOI: 10.12688/f1000research.17196.1] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
The mammalian/mechanistic target of rapamycin (mTOR) is a key component of cellular metabolism that integrates nutrient sensing with cellular processes that fuel cell growth and proliferation. Although the involvement of the mTOR pathway in regulating life span and aging has been studied extensively in the last decade, the underpinning mechanisms remain elusive. In this review, we highlight the emerging insights that link mTOR to various processes related to aging, such as nutrient sensing, maintenance of proteostasis, autophagy, mitochondrial dysfunction, cellular senescence, and decline in stem cell function.
Collapse
Affiliation(s)
- David Papadopoli
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Karine Boulay
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Lawrence Kazak
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada
- Goodman Cancer Research Centre, 1160 Pine Avenue West, Montréal, QC, H3A 1A3, Canada
| | - Michael Pollak
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Goodman Cancer Research Centre, 1160 Pine Avenue West, Montréal, QC, H3A 1A3, Canada
- Department of Experimental Medicine, McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Frédérick A. Mallette
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
- Département de Médecine, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Ivan Topisirovic
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada
- Department of Experimental Medicine, McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Laura Hulea
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
- Département de Médecine, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| |
Collapse
|
121
|
Widespread PERK-dependent repression of ER targets in response to ER stress. Sci Rep 2019; 9:4330. [PMID: 30867432 PMCID: PMC6416471 DOI: 10.1038/s41598-019-38705-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/04/2019] [Indexed: 12/11/2022] Open
Abstract
The UPR (Unfolded Protein Response) is a well-orchestrated response to ER protein folding and processing overload, integrating both transcriptional and translational outputs. Its three arms in mammalian cells, the PERK translational response arm, together with the ATF6 and IRE1-XBP1-mediated transcriptional arms, have been thoroughly investigated. Using ribosome footprint profiling, we performed a deep characterization of gene expression programs involved in the early and late ER stress responses, within WT or PERK -/- Mouse Embryonic Fibroblasts (MEFs). We found that both repression and activation gene expression programs, affecting hundreds of genes, are significantly hampered in the absence of PERK. Specifically, PERK -/- cells do not show global translational inhibition, nor do they specifically activate early gene expression programs upon short exposure to ER stress. Furthermore, while PERK -/- cells do activate/repress late ER-stress response genes, the response is substantially weaker. Importantly, we highlight a widespread PERK-dependent repression program, consisting of ER targeted proteins, including transmembrane proteins, glycoproteins, and proteins with disulfide bonds. This phenomenon occurs in various different cell types, and has a major translational regulatory component. Moreover, we revealed a novel interplay between PERK and the XBP1-ATF6 arms of the UPR, whereby PERK attenuates the expression of a specific subset of XBP1-ATF6 targets, further illuminating the complexity of the integrated ER stress response.
Collapse
|
122
|
Uchenunu O, Pollak M, Topisirovic I, Hulea L. Oncogenic kinases and perturbations in protein synthesis machinery and energetics in neoplasia. J Mol Endocrinol 2019; 62:R83-R103. [PMID: 30072418 PMCID: PMC6347283 DOI: 10.1530/jme-18-0058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 08/01/2018] [Indexed: 12/17/2022]
Abstract
Notwithstanding that metabolic perturbations and dysregulated protein synthesis are salient features of cancer, the mechanism underlying coordination of cellular energy balance with mRNA translation (which is the most energy consuming process in the cell) is poorly understood. In this review, we focus on recently emerging insights in the molecular underpinnings of the cross-talk between oncogenic kinases, translational apparatus and cellular energy metabolism. In particular, we focus on the central signaling nodes that regulate these processes (e.g. the mechanistic/mammalian target of rapamycin MTOR) and the potential implications of these findings on improving the anti-neoplastic efficacy of oncogenic kinase inhibitors.
Collapse
Affiliation(s)
- Oro Uchenunu
- Lady Davis Institute, SMBD JGH, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, Montreal, Quebec, Canada
| | - Michael Pollak
- Lady Davis Institute, SMBD JGH, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, Montreal, Quebec, Canada
| | - Ivan Topisirovic
- Lady Davis Institute, SMBD JGH, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, Montreal, Quebec, Canada
- Biochemistry Department, McGill University, Montreal, Quebec, Canada
| | - Laura Hulea
- Lady Davis Institute, SMBD JGH, McGill University, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, Montreal, Quebec, Canada
- Correspondence should be addressed to L Hulea:
| |
Collapse
|
123
|
High-Throughput Screening Identified Compounds Sensitizing Tumor Cells to Glucose Starvation in Culture and VEGF Inhibitors In Vivo. Cancers (Basel) 2019; 11:cancers11020156. [PMID: 30704052 PMCID: PMC6406438 DOI: 10.3390/cancers11020156] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/17/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor cells utilize glucose to fuel their anabolic needs, including rapid proliferation. However, due to defective vasculature and increased glucose uptake, tumor cells must overcome glucose deprivation. Accordingly, tumor cells depend on cellular pathways promoting survival under such conditions. Targeting these survival mechanisms can thus serve as a new therapeutic strategy in oncology. As such, we sought to identify small-molecule inhibitors which sensitize tumor cells to glucose starvation by high-throughput drug screening in vitro. Specifically, we searched for inhibitors that selectively killed tumor cells growing in glucose-free but not in normal medium. This phenotypic drug screen of 7000 agents with MCF7 cells led to the identification of 67 potential candidates, 31 of which were validated individually. Among the identified compounds, we found a high number of compounds known to target mitochondria. The efficacies of two of the identified compounds, QNZ (EVP4593) and papaverine, were validated in four different tumor cell lines. We found that these agents inhibited the mTOR(Mechamistic\Mammilian Target of Rapamycin) pathway in tumor cells growing under glucose starvation, but not under normal conditions. The results were validated and confirmed in vivo, with QNZ and papaverine exhibiting superior antitumor activity in a tumor xenograft model when combined with the VEGF inhibitor bevacizumab (avastin). Administering these drug combinations (i.e., avastin and papaverine, and avastin and QNZ) led to significant reductions in proliferation and mTOR activity of the aggressive DLD1 colon cell line in mice. Given our findings, we propose that compounds targeting metabolically challenged tumors, such as inhibitors of mitochondrial activity, be considered as a therapeutic strategy in cancer.
Collapse
|
124
|
M(en)TORship lessons on life and death by the integrated stress response. Biochim Biophys Acta Gen Subj 2018; 1863:644-649. [PMID: 30572003 DOI: 10.1016/j.bbagen.2018.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022]
Abstract
Cells employ pro-survival and pro-adaptive pathways to cope with different forms of environmental stress. When stress is excessive, and the damage caused by it is unsustainable, cells engage pro-death pathways, which are in place to protect the host from the deleterious effects of harmed cells. Two important pathways that determine the balance between survival and death of stressed cells are the integrated stress response (ISR) and the mammalian target of rapamycin (mTOR), both of which converge at the level of mRNA translation. The two pathways have established avenues of communication to control their activity and determine the fate of stressed cells in a context-dependent manner. The functional interplay between the ISR and mTOR may have significant ramifications in the development and treatment of human diseases such as diabetes, neurodegeneration and cancer.
Collapse
|
125
|
Shen P, Reineke LC, Knutsen E, Chen M, Pichler M, Ling H, Calin GA. Metformin blocks MYC protein synthesis in colorectal cancer via mTOR-4EBP-eIF4E and MNK1-eIF4G-eIF4E signaling. Mol Oncol 2018; 12:1856-1870. [PMID: 30221473 PMCID: PMC6210051 DOI: 10.1002/1878-0261.12384] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
The antidiabetic drug metformin has been associated with reduced colorectal cancer (CRC) risk and improved prognosis of CRC patients. However, the detailed mechanisms underlying such beneficial effects remain unknown. In this study, we aimed to evaluate metformin activity in CRC models and unveil the underlying molecular mechanisms. We showed that metformin inhibits CRC cell proliferation by arresting cells in the G1 phase of the cell cycle and dramatically reduces colony formation of CRC cells. We discovered that metformin causes a robust reduction of MYC protein level. Through the use of luciferase assay and coincubation with either protein synthesis or proteasome inhibitors, we demonstrated that regulation of MYC by metformin is independent of the proteasome and 3' UTR-mediated regulation, but depends on protein synthesis. Data from polysome profiling and ribopuromycylation assays showed that metformin induced widespread inhibition of protein synthesis. Repression of protein synthesis by metformin preferentially affects cell cycle-associated proteins, by altering signaling through the mTOR-4EBP-eIF4E and MNK1-eIF4G-eIF4E axes. The inhibition of MYC protein synthesis may underlie metformin's beneficial effects on CRC risk and prognosis.
Collapse
Affiliation(s)
- Peng Shen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lucas C Reineke
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Erik Knutsen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Meng Chen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martin Pichler
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Division of Oncology, Research Unit of Non-Coding RNA and Genome Editing in Cancer, Medical University of Graz, Austria
| | - Hui Ling
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
126
|
Rendleman J, Cheng Z, Maity S, Kastelic N, Munschauer M, Allgoewer K, Teo G, Zhang YBM, Lei A, Parker B, Landthaler M, Freeberg L, Kuersten S, Choi H, Vogel C. New insights into the cellular temporal response to proteostatic stress. eLife 2018; 7:39054. [PMID: 30272558 PMCID: PMC6185107 DOI: 10.7554/elife.39054] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/28/2018] [Indexed: 12/13/2022] Open
Abstract
Maintaining a healthy proteome involves all layers of gene expression regulation. By quantifying temporal changes of the transcriptome, translatome, proteome, and RNA-protein interactome in cervical cancer cells, we systematically characterize the molecular landscape in response to proteostatic challenges. We identify shared and specific responses to misfolded proteins and to oxidative stress, two conditions that are tightly linked. We reveal new aspects of the unfolded protein response, including many genes that escape global translation shutdown. A subset of these genes supports rerouting of energy production in the mitochondria. We also find that many genes change at multiple levels, in either the same or opposing directions, and at different time points. We highlight a variety of putative regulatory pathways, including the stress-dependent alternative splicing of aminoacyl-tRNA synthetases, and protein-RNA binding within the 3’ untranslated region of molecular chaperones. These results illustrate the potential of this information-rich resource.
Collapse
Affiliation(s)
- Justin Rendleman
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Zhe Cheng
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Shuvadeep Maity
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Nicolai Kastelic
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mathias Munschauer
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Kristina Allgoewer
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Guoshou Teo
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Yun Bin Matteo Zhang
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Amy Lei
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Brian Parker
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Integrative Research Institute for the Life Sciences, Institute of Biology, Humboldt University, Berlin, Germany
| | | | | | - Hyungwon Choi
- National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Christine Vogel
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, United States
| |
Collapse
|
127
|
|
128
|
Reineke LC, Cheema SA, Dubrulle J, Neilson JR. Chronic starvation induces noncanonical pro-death stress granules. J Cell Sci 2018; 131:jcs220244. [PMID: 30185525 PMCID: PMC6198455 DOI: 10.1242/jcs.220244] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023] Open
Abstract
Stress granules (SGs) assemble under stress-induced conditions that inhibit protein synthesis, including phosphorylation of eIF2α, inhibition of the RNA helicase eIF4a proteins or inactivation of mTORC1. Classically defined SGs are composed of translation initiation factors, 40S ribosomes, RNA-binding proteins and poly(A)+ mRNAs. As such, they represent an important compartment for storage of mRNAs and regulation of their translation. Emerging work on SGs indicates that these structures might promote cellular survival in diverse disease states. Yet, much work on SG formation and function employs acute stress conditions, which might not accurately reflect the chronic stresses that manifest in human disease. Here, we used prolonged nutrient starvation to model and investigate SG formation and function during chronic stress in a human cell line and mouse embryonic fibroblasts. Surprisingly, we found that SGs that form under chronic nutrient starvation lack 40S ribosomes, do not actively exchange their constituent components with cytoplasmic pools and promote cell death. We named these SGs starvation-induced SGs (stSGs). Our results on stSGs imply that SG assembly and function in the context of prolonged nutrient starvation stress differ significantly from what has been described for acute stress conditions.
Collapse
Affiliation(s)
- Lucas C Reineke
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shebna A Cheema
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julien Dubrulle
- Integrated Microscopy Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joel R Neilson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
129
|
James CC, Smyth JW. Alternative mechanisms of translation initiation: An emerging dynamic regulator of the proteome in health and disease. Life Sci 2018; 212:138-144. [PMID: 30290184 DOI: 10.1016/j.lfs.2018.09.054] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/18/2018] [Accepted: 09/27/2018] [Indexed: 01/06/2023]
Abstract
Eukaryotic mRNAs were historically thought to rely exclusively on recognition and binding of their 5' cap by initiation factors to effect protein translation. While internal ribosome entry sites (IRESs) are well accepted as necessary for the cap-independent translation of many viral genomes, there is now recognition that eukaryotic mRNAs also undergo non-canonical modes of translation initiation. Recently, high-throughput assays have identified thousands of mammalian transcripts with translation initiation occurring at non-canonical start codons, upstream of and within protein coding regions. In addition to IRES-mediated events, regulatory mechanisms of translation initiation have been described involving alternate 5' cap recognition, mRNA sequence elements, and ribosome selection. These mechanisms ensure translation of specific mRNAs under conditions where cap-dependent translation is shut down and contribute to pathological states including cardiac hypertrophy and cancer. Such global and gene-specific dynamic regulation of translation presents us with an increasing number of novel therapeutic targets. While these newly discovered modes of translation initiation have been largely studied in isolation, it is likely that several act on the same mRNA and exquisite coordination is necessary to maintain 'normal' translation. In this short review, we summarize the current state of knowledge of these alternative mechanisms of eukaryotic protein translation, their contribution to normal and pathological cell biology, and the potential of targeting translation initiation therapeutically in human disease.
Collapse
Affiliation(s)
- Carissa C James
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA, USA; Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA; Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, VA, USA
| | - James W Smyth
- Virginia Tech Carilion Research Institute and School of Medicine, Roanoke, VA, USA; Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, VA, USA.
| |
Collapse
|
130
|
McGrath EP, Logue SE, Mnich K, Deegan S, Jäger R, Gorman AM, Samali A. The Unfolded Protein Response in Breast Cancer. Cancers (Basel) 2018; 10:cancers10100344. [PMID: 30248920 PMCID: PMC6211039 DOI: 10.3390/cancers10100344] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/12/2018] [Accepted: 09/18/2018] [Indexed: 01/18/2023] Open
Abstract
In 2018, in the US alone, it is estimated that 268,670 people will be diagnosed with breast cancer, and that 41,400 will die from it. Since breast cancers often become resistant to therapies, and certain breast cancers lack therapeutic targets, new approaches are urgently required. A cell-stress response pathway, the unfolded protein response (UPR), has emerged as a promising target for the development of novel breast cancer treatments. This pathway is activated in response to a disturbance in endoplasmic reticulum (ER) homeostasis but has diverse physiological and disease-specific functions. In breast cancer, UPR signalling promotes a malignant phenotype and can confer tumours with resistance to widely used therapies. Here, we review several roles for UPR signalling in breast cancer, highlighting UPR-mediated therapy resistance and the potential for targeting the UPR alone or in combination with existing therapies.
Collapse
Affiliation(s)
- Eoghan P McGrath
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Susan E Logue
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Katarzyna Mnich
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Shane Deegan
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Richard Jäger
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, 53359 Rheinbach, Germany.
| | - Adrienne M Gorman
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Afshin Samali
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| |
Collapse
|
131
|
Translation Links Nutrient Availability with Inflammation. Trends Biochem Sci 2018; 43:849-852. [PMID: 30224182 DOI: 10.1016/j.tibs.2018.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 08/29/2018] [Indexed: 11/22/2022]
Abstract
Translation plays a crucial role in shaping the proteome during adaptation to various types of stress. A recent study by Gameiro and Struhl identified an inflammatory response which comprises coordination of transcriptional and translational programs, and which appears to be required for recovery from nutrient deprivation.
Collapse
|
132
|
Pavitt GD. Regulation of translation initiation factor eIF2B at the hub of the integrated stress response. WILEY INTERDISCIPLINARY REVIEWS-RNA 2018; 9:e1491. [PMID: 29989343 DOI: 10.1002/wrna.1491] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/08/2018] [Accepted: 05/22/2018] [Indexed: 12/29/2022]
Abstract
Phosphorylation of the translation initiation factor eIF2 is one of the most widely used and well-studied mechanisms cells use to respond to diverse cellular stresses. Known as the integrated stress response (ISR), the control pathway uses modulation of protein synthesis to reprogram gene expression and restore homeostasis. Here the current knowledge of the molecular mechanisms of eIF2 activation and its control by phosphorylation at a single-conserved phosphorylation site, serine 51 are discussed with a major focus on the regulatory roles of eIF2B and eIF5 where a current molecular view of ISR control of eIF2B activity is presented. How genetic disorders affect eIF2 or eIF2B is discussed, as are syndromes where excess signaling through the ISR is a component. Finally, studies into the action of recently identified compounds that modulate the ISR in experimental systems are discussed; these suggest that eIF2B is a potential therapeutic target for a wide range of conditions. This article is categorized under: Translation > Translation Regulation.
Collapse
Affiliation(s)
- Graham D Pavitt
- Division Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
133
|
Moon SL, Parker R. EIF2B2 mutations in vanishing white matter disease hypersuppress translation and delay recovery during the integrated stress response. RNA (NEW YORK, N.Y.) 2018; 24:841-852. [PMID: 29632131 PMCID: PMC5959252 DOI: 10.1261/rna.066563.118] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 04/07/2018] [Indexed: 05/29/2023]
Abstract
Mutations in eIF2B genes cause vanishing white matter disease (VWMD), a fatal leukodystrophy that can manifest following physical trauma or illness, conditions that activate the integrated stress response (ISR). EIF2B is the guanine exchange factor for eIF2, facilitating ternary complex formation and translation initiation. During the ISR, eIF2α is phosphorylated and inhibits eIF2B, causing global translation suppression and stress-induced gene translation, allowing stress adaptation and recovery. We demonstrate that VWMD patient cells hypersuppress translation during the ISR caused by acute ER stress, delaying stress-induced gene expression and interrupting a negative feedback loop that allows translational recovery by GADD34-mediated dephosphorylation of phospho-eIF2α. Thus, cells from VWMD patients undergo a prolonged state of translational hyperrepression and fail to recover from stress. We demonstrate that small molecules targeting eIF2B or the eIF2α kinase PERK rescue translation defects in patient cells. Therefore, defects in the ISR could contribute to white matter loss in VWMD.
Collapse
Affiliation(s)
- Stephanie L Moon
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80303, USA
| | - Roy Parker
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80303, USA
- Howard Hughes Medical Institute, University of Colorado, Boulder, Colorado 80303, USA
| |
Collapse
|
134
|
Shatsky IN, Terenin IM, Smirnova VV, Andreev DE. Cap-Independent Translation: What's in a Name? Trends Biochem Sci 2018; 43:882-895. [PMID: 29789219 DOI: 10.1016/j.tibs.2018.04.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/15/2018] [Accepted: 04/22/2018] [Indexed: 02/05/2023]
Abstract
Eukaryotic translation initiation relies on the m7G cap present at the 5' end of all mRNAs. Some viral mRNAs employ alternative mechanisms of initiation based on internal ribosome entry. The 'IRES ideology' was adopted by researchers to explain the differential translation of cellular mRNAs when the cap recognition is suppressed. However, some cellular IRESs have already been challenged and others are awaiting their validation. As an alternative cap-independent mechanism, we propose adopting the concept of cap-independent translation enhancers (CITEs) for mammalian mRNAs. Unlike IRESs, CITEs can be located both within 5' and 3' UTRs and bind mRNA-recruiting translational components. The respective 5' UTRs are then inspected by the scanning machinery essentially in the same way as under cap-dependent translation.
Collapse
Affiliation(s)
- Ivan N Shatsky
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Leninskie Gory 1, Bldg. 40, Moscow 119992, Russia.
| | - Ilya M Terenin
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Leninskie Gory 1, Bldg. 40, Moscow 119992, Russia; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Trubetskaya Str. 8-2, 119991, Moscow, Russia
| | - Victoria V Smirnova
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Leninskie Gory 1, Bldg. 40, Moscow 119992, Russia
| | - Dmitri E Andreev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Leninskie Gory 1, Bldg. 40, Moscow 119992, Russia
| |
Collapse
|