1
|
Basis A, Sharf R, Kleinberger T. The adenoviral E4orf4 protein: A multifunctional protein serving as a guide for treating cancer, a multifactorial disease. Tumour Virus Res 2024; 19:200303. [PMID: 39681196 PMCID: PMC11928763 DOI: 10.1016/j.tvr.2024.200303] [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: 10/03/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024] Open
Abstract
Viruses exploit several cellular pathways to support their replication, and many of these virus-targeted pathways are also important for cancer growth. Consequently, studying virus-host interactions offers valuable insights into tumorigenesis and can suggest the development of novel anti-cancer therapies, with oncolytic viruses being one well-known example. The adenovirus E4orf4 protein, which disrupts several host regulatory pathways to facilitate viral infection, also functions as a potent anti-cancer agent when expressed independently. E4orf4 can selectively kill a wide range of cancer cell lines while sparing non-cancerous cells. Moreover, it effectively eliminated cancer in an in vivo Drosophila model without causing significant harm to normal tissues. In this study we provide evidence that an E4orf4-mimicking drug cocktail, comprising sublethal doses of four FDA-approved drugs targeting the pathways disrupted by E4orf4, significantly enhanced cancer cell death in many cancer cell types compared with individual drugs or less inclusive drug combinations. The quadruple drug cocktail was not toxic in non-cancerous cells. These findings provide a proof-of-principle for the potential application of virus-host interaction studies to design an effective E4orf4-based cancer therapy. Further investigation of E4orf4 interactions with the host cell will likely improve this E4orf4-based therapy by adding drugs that disrupt additional pathways. Crucially, the E4orf4-based approach offers a strategic advantage by avoiding the time-consuming development of novel drugs. Instead, it leverages existing drugs, including those that might be too toxic for use as monotherapies, by employing them at sublethal concentrations in combination. Thus, it provides a feasible and efficient method for advancing cancer therapy.
Collapse
Affiliation(s)
- Amir Basis
- Dept. of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Rakefet Sharf
- Dept. of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tamar Kleinberger
- Dept. of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
2
|
Göttig L, Schreiner S. E4orf1: The triple agent of adenovirus - Unraveling its roles in oncogenesis, infectious obesity and immune responses in virus replication and vector therapy. Tumour Virus Res 2024; 17:200277. [PMID: 38428735 PMCID: PMC10937242 DOI: 10.1016/j.tvr.2024.200277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
Human Adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous sub-types that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating cellular pathways such as PI3K-Akt-mTOR, Ras, the immune response and further HAdV replication stages than previously anticipated. In this review, we aim to explore the structure, molecular mechanisms, and biological functions of E4orf1, shedding light on its potentially multifaceted roles during HAdV infection, including metabolic diseases and oncogenesis. Furthermore, we discuss the role of functional E4orf1 in biotechnological applications such as Adenovirus (AdV) vaccine vectors and oncolytic AdV. By dissecting the intricate relationships between HAdV types and E4orf1 proteins, this review provides valuable insights into viral pathogenesis and points to promising areas of future research.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Hannover, Germany; Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Göttig L, Jummer S, Staehler L, Groitl P, Karimi M, Blanchette P, Kosulin K, Branton PE, Schreiner S. The human adenovirus PI3K-Akt activator E4orf1 is targeted by the tumor suppressor p53. J Virol 2024; 98:e0170123. [PMID: 38451084 PMCID: PMC11019960 DOI: 10.1128/jvi.01701-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/13/2024] [Indexed: 03/08/2024] Open
Abstract
Human adenoviruses (HAdV) are classified as DNA tumor viruses due to their potential to mediate oncogenic transformation in non-permissive mammalian cells and certain human stem cells. To achieve transformation, the viral early proteins of the E1 and E4 regions must block apoptosis and activate proliferation: the former predominantly through modulating the cellular tumor suppressor p53 and the latter by activating cellular pro-survival and pro-metabolism protein cascades, such as the phosphoinositide 3-kinase (PI3K-Akt) pathway, which is activated by HAdV E4orf1. Focusing on HAdV-C5, we show that E4orf1 is necessary and sufficient to stimulate Akt activation through phosphorylation in H1299 cells, which is not only hindered but repressed during HAdV-C5 infection with a loss of E4orf1 function in p53-positive A549 cells. Contrary to other research, E4orf1 localized not only in the common, cytoplasmic PI3K-Akt-containing compartment, but also in distinct nuclear aggregates. We identified a novel inhibitory mechanism, where p53 selectively targeted E4orf1 to destabilize it, also stalling E4orf1-dependent Akt phosphorylation. Co-IP and immunofluorescence studies showed that p53 and E4orf1 interact, and since p53 is bound by the HAdV-C5 E3 ubiquitin ligase complex, we also identified E4orf1 as a novel factor interacting with E1B-55K and E4orf6 during infection; overexpression of E4orf1 led to less-efficient E3 ubiquitin ligase-mediated proteasomal degradation of p53. We hypothesize that p53 specifically subverts the pro-survival function of E4orf1-mediated PI3K-Akt activation to protect the cell from metabolic hyper-activation or even transformation.IMPORTANCEHuman adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous subtypes that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. Nonetheless, E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating the cellular pathways such as phosphoinositide 3-kinase-Akt-mTOR. Our study reveals a novel and general impact of E4orf1 on host mechanisms, providing a novel basis for innovative antiviral strategies in future therapeutic settings. Ongoing investigations of the cellular pathways modulated by HAdV are of great interest, particularly since adenovirus-based vectors actually serve as vaccine or gene vectors. HAdV constitute an ideal model system to analyze the underlying molecular principles of virus-induced tumorigenesis.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Simone Jummer
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Luisa Staehler
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Peter Groitl
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Maryam Karimi
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Paola Blanchette
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Karin Kosulin
- Molecular Microbiology, Children’s Cancer Research Institute, Vienna, Austria
| | - Philip E. Branton
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Freiburg, Germany
- Institute of Virology, Medical Center—University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Javorsky A, Humbert PO, Kvansakul M. Viral manipulation of cell polarity signalling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119536. [PMID: 37437846 DOI: 10.1016/j.bbamcr.2023.119536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/14/2023]
Abstract
Cell polarity refers to the asymmetric distribution of biomacromolecules that enable the correct orientation of a cell in a particular direction. It is thus an essential component for appropriate tissue development and function. Viral infections can lead to dysregulation of polarity. This is associated with a poor prognosis due to viral interference with core cell polarity regulatory scaffolding proteins that often feature PDZ (PSD-95, DLG, and ZO-1) domains including Scrib, Dlg, Pals1, PatJ, Par3 and Par6. PDZ domains are also promiscuous, binding to several different partners through their C-terminal region which contain PDZ-binding motifs (PBM). Numerous viruses encode viral effector proteins that target cell polarity regulators for their benefit and include papillomaviruses, flaviviruses and coronaviruses. A better understanding of the mechanisms of action utilised by viral effector proteins to subvert host cell polarity sigalling will provide avenues for future therapeutic intervention, while at the same time enhance our understanding of cell polarity regulation and its role tissue homeostasis.
Collapse
Affiliation(s)
- Airah Javorsky
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Patrick O Humbert
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia; Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Victoria 3086, Australia; Department of Biochemistry & Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Marc Kvansakul
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia; Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
5
|
Tessier TM, Dodge MJ, MacNeil KM, Evans AM, Prusinkiewicz MA, Mymryk JS. Almost famous: Human adenoviruses (and what they have taught us about cancer). Tumour Virus Res 2021; 12:200225. [PMID: 34500123 PMCID: PMC8449131 DOI: 10.1016/j.tvr.2021.200225] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/25/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
Papillomaviruses, polyomaviruses and adenoviruses are collectively categorized as the small DNA tumour viruses. Notably, human adenoviruses were the first human viruses demonstrated to be able to cause cancer, albeit in non-human animal models. Despite their long history, no human adenovirus is a known causative agent of human cancers, unlike a subset of their more famous cousins, including human papillomaviruses and human Merkel cell polyomavirus. Nevertheless, seminal research using human adenoviruses has been highly informative in understanding the basics of cell cycle control, gene expression, apoptosis and cell differentiation. This review highlights the contributions of human adenovirus research in advancing our knowledge of the molecular basis of cancer.
Collapse
Affiliation(s)
- Tanner M Tessier
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Mackenzie J Dodge
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Katelyn M MacNeil
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Andris M Evans
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Martin A Prusinkiewicz
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Joe S Mymryk
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada; Department of Otolaryngology, Head & Neck Surgery, The University of Western Ontario, London, ON, Canada; Department of Oncology, The University of Western Ontario, London, ON, Canada; London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
6
|
Garcia G, Sharma A, Ramaiah A, Sen C, Purkayastha A, Kohn DB, Parcells MS, Beck S, Kim H, Bakowski MA, Kirkpatrick MG, Riva L, Wolff KC, Han B, Yuen C, Ulmert D, Purbey PK, Scumpia P, Beutler N, Rogers TF, Chatterjee AK, Gabriel G, Bartenschlager R, Gomperts B, Svendsen CN, Betz UAK, Damoiseaux RD, Arumugaswami V. Antiviral drug screen identifies DNA-damage response inhibitor as potent blocker of SARS-CoV-2 replication. Cell Rep 2021; 35:108940. [PMID: 33784499 PMCID: PMC7969873 DOI: 10.1016/j.celrep.2021.108940] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 01/26/2021] [Accepted: 03/12/2021] [Indexed: 12/16/2022] Open
Abstract
SARS-CoV-2 has currently precipitated the COVID-19 global health crisis. We developed a medium-throughput drug-screening system and identified a small-molecule library of 34 of 430 protein kinase inhibitors that were capable of inhibiting the SARS-CoV-2 cytopathic effect in human epithelial cells. These drug inhibitors are in various stages of clinical trials. We detected key proteins involved in cellular signaling pathways mTOR-PI3K-AKT, ABL-BCR/MAPK, and DNA-damage response that are critical for SARS-CoV-2 infection. A drug-protein interaction-based secondary screen confirmed compounds, such as the ATR kinase inhibitor berzosertib and torin2 with anti-SARS-CoV-2 activity. Berzosertib exhibited potent antiviral activity against SARS-CoV-2 in multiple cell types and blocked replication at the post-entry step. Berzosertib inhibited replication of SARS-CoV-1 and the Middle East respiratory syndrome coronavirus (MERS-CoV) as well. Our study highlights key promising kinase inhibitors to constrain coronavirus replication as a host-directed therapy in the treatment of COVID-19 and beyond as well as provides an important mechanism of host-pathogen interactions.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arunachalam Ramaiah
- Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, CA 92697, USA; Section of Cell and Developmental Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Chandani Sen
- UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Arunima Purkayastha
- UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Donald B Kohn
- UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA
| | - Mark S Parcells
- Department of Animal and Food Sciences, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Sebastian Beck
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Heeyoung Kim
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - Malina A Bakowski
- Calibr, a division of Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Melanie G Kirkpatrick
- Calibr, a division of Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Laura Riva
- Calibr, a division of Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Karen C Wolff
- Calibr, a division of Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Brandon Han
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Constance Yuen
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David Ulmert
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Prabhat K Purbey
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Philip Scumpia
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Thomas F Rogers
- Department of Immunology and Microbiology, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; UC San Diego Division of Infectious Diseases and Global Public Health, UC San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Arnab K Chatterjee
- Calibr, a division of Scripps Research Institute, 11119 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Gülsah Gabriel
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany; German Center for Infection Research, Heidelberg partner site, Heidelberg, Germany; Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Brigitte Gomperts
- UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Robert D Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
7
|
Laevskaya A, Borovjagin A, Timashev PS, Lesniak MS, Ulasov I. Metabolome-Driven Regulation of Adenovirus-Induced Cell Death. Int J Mol Sci 2021; 22:ijms22010464. [PMID: 33466472 PMCID: PMC7796492 DOI: 10.3390/ijms22010464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 02/06/2023] Open
Abstract
A viral infection that involves virus invasion, protein synthesis, and virion assembly is typically accompanied by sharp fluctuations in the intracellular levels of metabolites. Under certain conditions, dramatic metabolic shifts can result in various types of cell death. Here, we review different types of adenovirus-induced cell death associated with changes in metabolic profiles of the infected cells. As evidenced by experimental data, in most cases changes in the metabolome precede cell death rather than represent its consequence. In our previous study, the induction of autophagic cell death was observed following adenovirus-mediated lactate production, acetyl-CoA accumulation, and ATP release, while apoptosis was demonstrated to be modulated by alterations in acetate and asparagine metabolism. On the other hand, adenovirus-induced ROS production and ATP depletion were demonstrated to play a significant role in the process of necrotic cell death. Interestingly, the accumulation of ceramide compounds was found to contribute to the induction of all the three types of cell death mentioned above. Eventually, the characterization of metabolite analysis could help in uncovering the molecular mechanism of adenovirus-mediated cell death induction and contribute to the development of efficacious oncolytic adenoviral vectors.
Collapse
Affiliation(s)
- Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Anton Borovjagin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Peter S. Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Department of Polymers and Composites, N.N.Semenov Institute of Chemical Physics, 4 Kosygin St., 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, 119991 Moscow, Russia
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60601, USA;
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Correspondence:
| |
Collapse
|
8
|
Baugh R, Khalique H, Seymour LW. Convergent Evolution by Cancer and Viruses in Evading the NKG2D Immune Response. Cancers (Basel) 2020; 12:E3827. [PMID: 33352921 PMCID: PMC7766243 DOI: 10.3390/cancers12123827] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The natural killer group 2 member D (NKG2D) receptor and its family of NKG2D ligands (NKG2DLs) are key components in the innate immune system, triggering NK, γδ and CD8+ T cell-mediated immune responses. While surface NKG2DL are rarely found on healthy cells, expression is significantly increased in response to various types of cellular stress, viral infection, and tumour cell transformation. In order to evade immune-mediated cytotoxicity, both pathogenic viruses and cancer cells have evolved various mechanisms of subverting immune defences and preventing NKG2DL expression. Comparisons of the mechanisms employed following virus infection or malignant transformation reveal a pattern of converging evolution at many of the key regulatory steps involved in NKG2DL expression and subsequent immune responses. Exploring ways to target these shared steps in virus- and cancer-mediated immune evasion may provide new mechanistic insights and therapeutic opportunities, for example, using oncolytic virotherapy to re-engage the innate immune system towards cancer cells.
Collapse
Affiliation(s)
| | | | - Leonard W. Seymour
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.B.); (H.K.)
| |
Collapse
|
9
|
Soh TK, Davies CTR, Muenzner J, Hunter LM, Barrow HG, Connor V, Bouton CR, Smith C, Emmott E, Antrobus R, Graham SC, Weekes MP, Crump CM. Temporal Proteomic Analysis of Herpes Simplex Virus 1 Infection Reveals Cell-Surface Remodeling via pUL56-Mediated GOPC Degradation. Cell Rep 2020; 33:108235. [PMID: 33027661 PMCID: PMC7539533 DOI: 10.1016/j.celrep.2020.108235] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 07/15/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Herpesviruses are ubiquitous in the human population and they extensively remodel the cellular environment during infection. Multiplexed quantitative proteomic analysis over the time course of herpes simplex virus 1 (HSV-1) infection was used to characterize changes in the host-cell proteome and the kinetics of viral protein production. Several host-cell proteins are targeted for rapid degradation by HSV-1, including the cellular trafficking factor Golgi-associated PDZ and coiled-coil motif-containing protein (GOPC). We show that the poorly characterized HSV-1 pUL56 directly binds GOPC, stimulating its ubiquitination and proteasomal degradation. Plasma membrane profiling reveals that pUL56 mediates specific changes to the cell-surface proteome of infected cells, including loss of interleukin-18 (IL18) receptor and Toll-like receptor 2 (TLR2), and that cell-surface expression of TLR2 is GOPC dependent. Our study provides significant resources for future investigation of HSV-host interactions and highlights an efficient mechanism whereby a single virus protein targets a cellular trafficking factor to modify the surface of infected cells. Multiplexed proteomic screens reveal regulation of host protein abundance by HSV-1 HSV-1 pUL56 targets host proteins such as GOPC for proteasomal degradation HSV-1-mediated degradation of GOPC remodels the plasma membrane of infected cells GOPC is important for cell-surface expression of immune receptor TLR2 in keratinocytes
Collapse
Affiliation(s)
- Timothy K Soh
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Colin T R Davies
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Julia Muenzner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Leah M Hunter
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Henry G Barrow
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Viv Connor
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Clément R Bouton
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Cameron Smith
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Edward Emmott
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Stephen C Graham
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Colin M Crump
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.
| |
Collapse
|
10
|
Characterization of the adipogenic protein E4orf1 from adenovirus 36 through an in silico approach. J Mol Model 2020; 26:285. [PMID: 32978703 DOI: 10.1007/s00894-020-04531-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 09/03/2020] [Indexed: 10/23/2022]
Abstract
Adenovirus 36 (Ad-36) is related to human obesity due to its adipogenic activity mediated by the early 4 open reading frame 1 (E4orf1) protein. Mechanisms underlying the adipogenic effect of E4orf1 are not completely understood; however, the proliferation and differentiation of fat cells are increased through the activation of the phosphatidyl inositol 3 kinase pathway by binding proteins containing PDZ domain. This study characterized E4orf1 tridimensional structure and analyzed its interactions with PDZ domain-containing proteins in order to provide new information about the behavior of this viral protein and its targets, which could provide an interesting druggable target for obesity-related cardiometabolic alterations. In silico strategies such as homology modeling, docking, and molecular dynamics (MD) were used to study the interaction of E4orf1 with five PDZ domains of disk large homolog 1 (PDZ-1 and PDZ-2), membrane-associated guanylate kinase 1 (PDZ-3), and multi-PDZ domain protein 1 (PDZ-7 and PDZ-10). Mutagenesis analysis of selected residues was performed to evaluate their effects on the stabilization of E4orf1:PDZ complexes. MD simulations showed that the E4orf1:PDZ10 complex was more stable than the others ones. The highly hydrophobic residues at the C-terminal region (114-125) of the E4orf1 are essential in the initial phase stabilization of the complexes. Moreover, the residues 80-85 in the core region contribute to longer stabilization of the E4orf1:PDZ10 complex, a result that was confirmed by in silico mutagenesis. In conclusion, E4orf1 forms a stable complex with PDZ10 domain, and the residues 80-85 are of particular importance. The characterization of E4orf1 interactions with PDZ domains provides an initial approach to discover druggable targets for Ad-36-induced obesity.
Collapse
|
11
|
Marziali F, Dizanzo MP, Cavatorta AL, Gardiol D. Differential expression of DLG1 as a common trait in different human diseases: an encouraging issue in molecular pathology. Biol Chem 2020; 400:699-710. [PMID: 30517074 DOI: 10.1515/hsz-2018-0350] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022]
Abstract
Human disc large (DLG1) is a scaffolding protein that through the interaction with diverse cell partners participates in the control of key cellular processes such as polarity, proliferation and migration. Experimental data have mainly identified DLG1 as a tumor suppressor. An outstanding point for DLG1 protein is that altered DLG1 expression and DLG1 gene mutations were observed in different pathologies, including cancer and neurological and immunological disorders. Evident changes in DLG1 abundance and/or cell localization were identified in a number of studies suggesting its participation in molecular mechanisms responsible for the development of such illnesses. In this review, we focus on some of the latest findings regarding DLG1 alterations in different diseases as well as its potential use as a biomarker for pathological progression. We further address the current knowledge on the molecular mechanisms regulating DLG1 expression and the posttranslational modifications that may affect DLG1 cell localization and functions. Despite the advances in this field, there are still open questions about the precise molecular link between alterations in DLG1 expression and the development of each specific pathology. The complete understanding of this concern will give us new scenarios for the design of promising diagnosis and therapeutic tools.
Collapse
Affiliation(s)
- Federico Marziali
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - María Paula Dizanzo
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Ana Laura Cavatorta
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Daniela Gardiol
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| |
Collapse
|
12
|
Ip WH, Dobner T. Cell transformation by the adenovirus oncogenes E1 and E4. FEBS Lett 2019; 594:1848-1860. [PMID: 31821536 DOI: 10.1002/1873-3468.13717] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/21/2022]
Abstract
Extensive studies on viral-mediated oncogenic transformation by human adenoviruses have revealed much of our current understanding on the molecular mechanisms that are involved in the process. To date, these studies have shown that cell transformation is a multistep process regulated by the cooperation of several adenoviral gene products encoded in the early regions 1 (E1) and 4 (E4). Early region 1A immortalizes primary rodent cells, whereas co-expression of early region protein 1B induces full manifestation of the transformed phenotype. Beside E1 proteins, also some E4 proteins have partial transforming activities through regulating many cellular pathways. Here, we summarize recent data of how adenoviral oncoproteins may contribute to viral transformation and discuss the challenge of pinpointing the underlying mechanisms.
Collapse
Affiliation(s)
- Wing Hang Ip
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Thomas Dobner
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| |
Collapse
|
13
|
Jiao Y, Liang X, Hou J, Aisa Y, Wu H, Zhang Z, Nuermaimaiti N, Zhao Y, Jiang S, Guan Y. Adenovirus type 36 regulates adipose stem cell differentiation and glucolipid metabolism through the PI3K/Akt/FoxO1/PPARγ signaling pathway. Lipids Health Dis 2019; 18:70. [PMID: 30902099 PMCID: PMC6429705 DOI: 10.1186/s12944-019-1004-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background This study aims to investigate the molecular mechanism of Adenovirus type 36 (Ad36) in adipocyte differentiation and glucolipid metabolism. Methods Rat obesity model was established by Ad36 infection and high-fat diet, respectively. Comparison of the body weight, clinical biochemical indicators, insulin sensitivity and lipid heterotopic deposition between these two models was performed. Ad36-induced adipocyte in vitro model was also established. The binding rate of FoxO1, PPARγ and its target gene promoter was detected using ChIP. The mRNA and protein expression levels of PPARγ and downstream target genes were detected by RT-PCR and Western blot, respectively. Oil red O staining was used to measure differentiation into adipocyte. Wortmannin (WM), inhibitor of PI3K, was used to act on Ad36-induced hADSCs. Results Ad36-induced obese rats did not exhibit disorders in blood glucose and blood TG, insulin resistance and lipid ectopic deposition. The expression of Adipoq, Lpin1 and Glut4 in the adipose tissue increased. Oil red O staining showed that Ad36 induced the differentiation of hAMSCs into human adipocytes in vitro. During this process, the binding rate of FoxO1 and PPARγ promoter regions was weakened. However, the binding rate of the transcription factor PPARγ to its target genes Acc, Adipoq, Lpin1 and Glut4 was enhanced, and thus increased the protein expression of P-FoxO1, PPARγ2, ACC, LPIN1, GLUT4 and ADIPOQ. The PI3K inhibitor Wortmannin reduced the expression of P-Akt, P-FoxO1 and PPARγ2, thereby inhibiting adipogenesis of hADSC. Conclusion Ad36 may promote fatty acid and triglyceride synthesis, and improve insulin sensitivity by affecting the PI3K/Akt/FoxO1/PPARγ signaling pathway.
Collapse
Affiliation(s)
- Yi Jiao
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Xiaodi Liang
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Jianfei Hou
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Yiliyasi Aisa
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Han Wu
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Zhilu Zhang
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Nuerbiye Nuermaimaiti
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China
| | - Yang Zhao
- Department of Burn and Plastic Surgery, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Sheng Jiang
- Department of Endocrinology, the First Affiliated Hospital of Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China.
| | - Yaqun Guan
- Department of Biochemistry, Preclinical Medicine College, Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
14
|
Shastri AA, Hegde V, Peddibhotla S, Feizy Z, Dhurandhar NV. E4orf1: A protein for enhancing glucose uptake despite impaired proximal insulin signaling. PLoS One 2018; 13:e0208427. [PMID: 30521580 PMCID: PMC6283569 DOI: 10.1371/journal.pone.0208427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 11/17/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Type 2 diabetes is often linked with impaired proximal insulin signaling. Hence, a therapeutic agent that enhances cellular glucose uptake without requiring proximal insulin signaling would be desirable for improving glycemic control. The E4orf1 peptide (E4) derived from human adenovirus 36 (Ad36) promotes cellular glucose uptake in vitro and in vivo, independent of insulin. E4 bypasses a part of insulin signaling to upregulate cellular glucose uptake. We tested the hypothesis that E4 requires the distal but not proximal insulin signaling to enhance cellular glucose disposal. METHODS 3T3-L1 preadipocytes inducibly expressing E4 or a null vector (NV) were treated with inhibitor of insulin receptor (S961), inhibitor of insulin like growth factor-1receptor (IGF-1R) (Picropodophyllin, PPP), PPP+S961, or phosphatidyl inositol-3 kinase (PI3K) inhibitor (Wortmannin, WM). We used PPP and S961 to block the proximal insulin signaling, or WM to block the distal insulin signaling. Cells were exposed to 0 or 100nM insulin. RESULTS As expected, when the proximal or distal insulin signaling was blocked in NV cells, insulin could not enhance pAKT protein abundance, Glut4 translocation, or glucose uptake. Whereas, E4 cells significantly increased pAKT abundance, Glut4 translocation and glucose uptake independent of the presence of insulin or proximal insulin signaling. Enhanced glucose disposal in E4 cells was completely abrogated when the distal insulin signaling was blocked. CONCLUSIONS E4 bypasses the proximal insulin signaling but uses the distal insulin signaling to activate pAkt and in turn Glut4 translocation to improve cellular glucose uptake. E4 offers a promising template to improve glycemic control when the proximal insulin signaling is impaired.
Collapse
Affiliation(s)
- Anuradha A. Shastri
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - Vijay Hegde
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - Swetha Peddibhotla
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - Zahra Feizy
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, United States of America
| | - Nikhil V. Dhurandhar
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Thomas M, Banks L. Upsetting the Balance: When Viruses Manipulate Cell Polarity Control. J Mol Biol 2018; 430:3481-3503. [PMID: 29680664 PMCID: PMC7094317 DOI: 10.1016/j.jmb.2018.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/12/2018] [Accepted: 04/14/2018] [Indexed: 12/20/2022]
Abstract
The central importance of cell polarity control is emphasized by the frequency with which it is targeted by many diverse viruses. It is clear that in targeting key polarity control proteins, viruses affect not only host cell polarity, but also influence many cellular processes, including transcription, replication, and innate and acquired immunity. Examination of the interactions of different virus proteins with the cell and its polarity controls during the virus life cycles, and in virally-induced cell transformation shows ever more clearly how intimately all cellular processes are linked to the control of cell polarity.
Collapse
|
16
|
Marziali F, Bugnon Valdano M, Brunet Avalos C, Moriena L, Cavatorta AL, Gardiol D. Interference of HTLV-1 Tax Protein with Cell Polarity Regulators: Defining the Subcellular Localization of the Tax-DLG1 Interaction. Viruses 2017; 9:E355. [PMID: 29168728 PMCID: PMC5744130 DOI: 10.3390/v9120355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 12/17/2022] Open
Abstract
Human T cell leukemia virus (HTLV)-1 Tax (Tax) protein is very important in viral replication and cell transformation. Tax localizes in the nucleus and cytoplasm in association with organelles. Some activities of Tax depend on interactions with PDZ (PSD-95/Discs Large/Z0-1) domain-containing proteins such as Discs large protein 1 (DLG1) which is involved in cell polarity and proliferation. The DLG1 interaction results in a cytoplasmic co-localization pattern resembling vesicular aggregates, the nature of which is still unknown. To further explore the role of PDZ proteins in HTLV-1 cell transformation, we deeply investigated the Tax-DLG1 association. By fluorescence resonance energy transfer (FRET), we detected, for the first time, the direct binding of Tax to DLG1 within the cell. We showed that the interaction specifically affects the cellular distribution of not only DLG1, but also Tax. After studying different cell structures, we demonstrated that the aggregates distribute into the Golgi apparatus in spatial association with the microtubule-organizing center (MTOC). This study contributes to understand the biological significance of Tax-PDZ interactions.
Collapse
Affiliation(s)
- Federico Marziali
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| | - Marina Bugnon Valdano
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| | - Clarisse Brunet Avalos
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| | - Lucía Moriena
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| | - Ana Laura Cavatorta
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| | - Daniela Gardiol
- Instituto de Biología Molecular y Celular de Rosario-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina.
| |
Collapse
|
17
|
Chaudhary N, Gonzalez E, Chang SH, Geng F, Rafii S, Altorki NK, McGraw TE. Adenovirus Protein E4-ORF1 Activation of PI3 Kinase Reveals Differential Regulation of Downstream Effector Pathways in Adipocytes. Cell Rep 2017; 17:3305-3318. [PMID: 28009298 DOI: 10.1016/j.celrep.2016.11.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 10/05/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
Insulin activation of phosphatidylinositol 3-kinase (PI3K) regulates metabolism, including the translocation of the Glut4 glucose transporter to the plasma membrane and inactivation of the FoxO1 transcription factor. Adenoviral protein E4-ORF1 stimulates cellular glucose metabolism by mimicking growth-factor activation of PI3K. We have used E4-ORF1 as a tool to dissect PI3K-mediated signaling in adipocytes. E4-ORF1 activation of PI3K in adipocytes recapitulates insulin regulation of FoxO1 but not regulation of Glut4. This uncoupling of PI3K effects occurs despite E4-ORF1 activating PI3K and downstream signaling to levels achieved by insulin. Although E4-ORF1 does not fully recapitulate insulin's effects on Glut4, it enhances insulin-stimulated insertion of Glut4-containing vesicles to the plasma membrane independent of Rab10, a key regulator of Glut4 trafficking. E4-ORF1 also stimulates plasma membrane translocation of ubiquitously expressed Glut1 glucose transporter, an effect that is likely essential for E4-ORF1 to promote an anabolic metabolism in a broad range of cell types.
Collapse
Affiliation(s)
- Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Eva Gonzalez
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sung-Hee Chang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Fuqiang Geng
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shahin Rafii
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nasser K Altorki
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10065, USA; Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10065, USA; Lung Cancer Program, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
18
|
Dyer A, Di Y, Calderon H, Illingworth S, Kueberuwa G, Tedcastle A, Jakeman P, Chia SL, Brown A, Silva MA, Barlow D, Beadle J, Hermiston T, Ferguson DJ, Champion B, Fisher KD, Seymour LW. Oncolytic Group B Adenovirus Enadenotucirev Mediates Non-apoptotic Cell Death with Membrane Disruption and Release of Inflammatory Mediators. Mol Ther Oncolytics 2017; 4:18-30. [PMID: 28345021 PMCID: PMC5363721 DOI: 10.1016/j.omto.2016.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/28/2016] [Indexed: 11/22/2022] Open
Abstract
Enadenotucirev (EnAd) is a chimeric group B adenovirus isolated by bioselection from a library of adenovirus serotypes. It replicates selectively in and kills a diverse range of carcinoma cells, shows effective anticancer activity in preclinical systems, and is currently undergoing phase I/II clinical trials. EnAd kills cells more quickly than type 5 adenovirus, and speed of cytotoxicity is dose dependent. The EnAd death pathway does not involve p53, is predominantly caspase independent, and appears to involve a rapid fall in cellular ATP. Infected cells show early loss of membrane integrity; increased exposure of calreticulin; extracellular release of ATP, HSP70, and HMGB1; and influx of calcium. The virus also causes an obvious single membrane blister reminiscent of ischemic cell death by oncosis. In human tumor biopsies maintained in ex vivo culture, EnAd mediated release of pro-inflammatory mediators such as TNF-α, IL-6, and HMGB1. In accordance with this, EnAd-infected tumor cells showed potent stimulation of dendritic cells and CD4+ T cells in a mixed tumor-leukocyte reaction in vitro. Whereas many viruses have evolved for efficient propagation with minimal inflammation, bioselection of EnAd for rapid killing has yielded a virus with a short life cycle that combines potent cytotoxicity with a proinflammatory mechanism of cell death.
Collapse
Affiliation(s)
- Arthur Dyer
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Ying Di
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Hugo Calderon
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 4SD, UK
| | - Sam Illingworth
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 4SD, UK
| | - Gray Kueberuwa
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alison Tedcastle
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Phil Jakeman
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Suet Lin Chia
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alice Brown
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 4SD, UK
| | - Michael A. Silva
- Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - David Barlow
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - John Beadle
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 4SD, UK
| | - Terry Hermiston
- Bayer HealthCare, 455 Mission Bay Blvd. S., San Francisco, CA 94158, USA
| | - David J.P. Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford, Oxford OX3 9DU, UK
| | - Brian Champion
- PsiOxus Therapeutics, Ltd., Milton Park, Abingdon OX14 4SD, UK
| | - Kerry D. Fisher
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | | |
Collapse
|
19
|
McMurphy TB, Huang W, Xiao R, Liu X, Dhurandhar NV, Cao L. Hepatic Expression of Adenovirus 36 E4ORF1 Improves Glycemic Control and Promotes Glucose Metabolism Through AKT Activation. Diabetes 2017; 66:358-371. [PMID: 27903748 PMCID: PMC5248996 DOI: 10.2337/db16-0876] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
Considering that impaired proximal insulin signaling is linked with diabetes, approaches that enhance glucose disposal independent of insulin signaling are attractive. In vitro data indicate that the E4ORF1 peptide derived from human adenovirus 36 (Ad36) interacts with cells from adipose tissue, skeletal muscle, and liver to enhance glucose disposal, independent of proximal insulin signaling. Adipocyte-specific expression of Ad36E4ORF1 improves hyperglycemia in mice. To determine the hepatic interaction of Ad36E4ORF1 in enhancing glycemic control, we expressed E4ORF1 of Ad36 or Ad5 or fluorescent tag alone by using recombinant adeno-associated viral vector in the liver of three mouse models. In db/db or diet-induced obesity (DIO) mice, hepatic expression of Ad36E4ORF1 but not Ad5E4ORF1 robustly improved glycemic control. In normoglycemic wild-type mice, hepatic expression of Ad36E4ORF1 lowered nonfasting blood glucose at a high dose of expression. Of note, Ad36E4ORF1 significantly reduced insulin levels in db/db and DIO mice. The improvement in glycemic control was observed without stimulation of the proximal insulin signaling pathway. Collectively, these data indicate that Ad36E4ORF1 is not a typical sensitizer, mimetic, or secretagogue of insulin. Instead, it may have insulin-sparing action, which seems to reduce the need for insulin and, hence, to reduce insulin levels.
Collapse
Affiliation(s)
- Travis B McMurphy
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH
| | - Wei Huang
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH
| | - Run Xiao
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH
| | - Xianglan Liu
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH
| | | | - Lei Cao
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH
- The Comprehensive Cancer Center, Ohio State University, Columbus, OH
| |
Collapse
|
20
|
Patwary NIA, Islam MS, Sohel M, Ara I, Sikder MOF, Shahik SM. In silico structure analysis and epitope prediction of E3 CR1-beta protein of Human Adenovirus E for vaccine design. Biomed J 2016; 39:382-390. [PMID: 28043417 PMCID: PMC6138513 DOI: 10.1016/j.bj.2016.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 07/12/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Human Adenoviruses are divided into 7 species of Human Adenovirus A to G based on DNA genome homology. The Human Adenovirus E (HAdVs-E) genome is a linear, double-stranded DNA containing 38 protein-coding genes. Wild-type adenoviruses type E, are linked to a number of slight illnesses. The most important part of HAdVs-E is E3 CR1-beta protein which controls the host immune response and viral attachment. METHOD We use numerous bio-informatics and immuno-informatics implements comprising sequence and construction tools for construction of 3D model and epitope prediction for HAdVs-E. RESULTS The 3D structure of E3 CR1-beta protein was generated and total of ten antigenic B cell epitopes, 6 MHC class I and 11 MHC class II binding peptides were predicted. CONCLUSION The study was carried out to predict antigenic determinants/epitopes of the E3 CR1-beta protein of Human Adenovirus E along with the 3D protein modeling. The study revealed potential T-cell and B-cell epitopes that can raise the desired immune response against E3 CR1-beta protein and useful in developing effective vaccines against HAdVs-E.
Collapse
Affiliation(s)
- Noman Ibna Amin Patwary
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Bangladesh
| | - Md Saiful Islam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Bangladesh
| | - Md Sohel
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Bangladesh
| | - Ismot Ara
- Department of Computer Science and Engineering, Faculty of Science and Technology, Atish Dipankar University of Science and Technology, Bangladesh; Department of Computer Science and Engineering, American International University-Bangladesh, Bangladesh
| | - Mohd Omar Faruk Sikder
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Bangladesh
| | - Shah Md Shahik
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Bangladesh.
| |
Collapse
|
21
|
Le Sage V, Cinti A, Amorim R, Mouland AJ. Adapting the Stress Response: Viral Subversion of the mTOR Signaling Pathway. Viruses 2016; 8:v8060152. [PMID: 27231932 PMCID: PMC4926172 DOI: 10.3390/v8060152] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a central regulator of gene expression, translation and various metabolic processes. Multiple extracellular (growth factors) and intracellular (energy status) molecular signals as well as a variety of stressors are integrated into the mTOR pathway. Viral infection is a significant stress that can activate, reduce or even suppress the mTOR signaling pathway. Consequently, viruses have evolved a plethora of different mechanisms to attack and co-opt the mTOR pathway in order to make the host cell a hospitable environment for replication. A more comprehensive knowledge of different viral interactions may provide fruitful targets for new antiviral drugs.
Collapse
Affiliation(s)
- Valerie Le Sage
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
| | - Alessandro Cinti
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
- Department of Medicine, McGill University, Montréal, QC H3A 0G4, Canada.
| | - Raquel Amorim
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
- Department of Medicine, McGill University, Montréal, QC H3A 0G4, Canada.
| | - Andrew J Mouland
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
- Department of Medicine, McGill University, Montréal, QC H3A 0G4, Canada.
| |
Collapse
|
22
|
The high-risk HPV E6 target scribble (hScrib) is required for HPV E6 expression in cervical tumour-derived cell lines. PAPILLOMAVIRUS RESEARCH 2016; 2:70-77. [PMID: 29074188 PMCID: PMC5886876 DOI: 10.1016/j.pvr.2016.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/08/2016] [Accepted: 04/05/2016] [Indexed: 11/21/2022]
Abstract
The ability of high-risk HPV E6 oncoproteins to target cellular proteins which harbor PDZ domains is believed to play an important role in the virus life cycle and to influence the ability of these viruses to bring about malignant transformation. Whilst many of these PDZ proteins are potential tumour suppressors, involved in the control of cell polarity and cell-contact, recent studies suggest that mislocalisation or overexpression might result in the emergence of oncogenic functions. This has been shown most clearly for two E6 targets, hDlg and hScrib. In this study we show that hScrib plays such a role in HeLa cells, where its expression is required for maintaining high levels of HPV-18 E6 protein. Loss of hScrib has no effect on E6 stability but results in lower levels of E6 transcription and a reduced rate of E6 translation. We further show that, in the context of cervical tumour-derived cell lines, both hScrib and E6 cooperate in the activation of the S6 kinase signaling pathway, and thereby contribute towards maintaining high rates of protein translation. These results indicate that the residual hScrib that is present within HPV transformed cells is pro-oncogenic, and highlights the dual functions of E6 cell polarity targets.
Collapse
|
23
|
Hegde V, Na HN, Dubuisson O, Burke SJ, Collier JJ, Burk D, Mendoza T, Dhurandhar NV. An adenovirus-derived protein: A novel candidate for anti-diabetic drug development. Biochimie 2016; 121:140-50. [PMID: 26700141 PMCID: PMC5125614 DOI: 10.1016/j.biochi.2015.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/08/2015] [Indexed: 01/04/2023]
Abstract
AIMS Exposure to human adenovirus Ad36 is causatively and correlatively linked with better glycemic control in animals and humans, respectively. Although the anti-hyperglycemic property of Ad36 may offer some therapeutic potential, it is impractical to use an infectious agent for therapeutic benefit. Cell-based studies identified that Ad36 enhances cellular glucose disposal via its E4orf1 protein. Ability to improve glycemic control in vivo is a critical prerequisite for further investigating the therapeutic potential of E4orf1. Therefore, the aim of this study was to determine the ability of E4orf1 to improve glycemic control independent of insulin despite high fat diet. MATERIALS & METHODS 8-9wk old male C57BL/6J mice fed a high-fat diet (60% kcal) were injected with a retrovirus plasmid expressing E4orf1, or a null vector (Control). Glycemic control was determined by glucose and insulin tolerance test. Islet cell size, amount of insulin and glucagon were determined in formalin-fixed pancreas. Rat insulinoma cell line (832/13) was infected with E4orf1 or control to determine changes in glucose stimulated insulin secretion. Protein from flash frozen adipose tissue depots, liver and muscle was used to determine molecular signaling by western blotting. RESULTS In multiple experiments, retrovirus-mediated E4orf1 expression in C57BL/6J mice significantly and reproducibly improved glucose excursion following a glucose load despite a high fat diet (60% energy). Importantly, E4orf1 improved glucose clearance without increasing insulin sensitivity, production or secretion, underscoring its insulin-independent effect. E4orf1 modulated molecular signaling in mice tissue, which included greater protein abundance of adiponectin, p-AKT and Glucose transporter Glu4. CONCLUSIONS This study provides the proof of concept for translational development of E4orf1 as a potential anti-diabetic agent. High fat intake and impaired insulin signaling are often associated with obesity, diabetes and insulin resistance. Hence, the ability of E4orf1 to improve glycemic control despite high fat diet and independent of insulin, is particularly attractive.
Collapse
Affiliation(s)
- Vijay Hegde
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Ha-Na Na
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Olga Dubuisson
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Susan J Burke
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - J Jason Collier
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - David Burk
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Tamra Mendoza
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | | |
Collapse
|
24
|
James CD, Roberts S. Viral Interactions with PDZ Domain-Containing Proteins-An Oncogenic Trait? Pathogens 2016; 5:pathogens5010008. [PMID: 26797638 PMCID: PMC4810129 DOI: 10.3390/pathogens5010008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 02/06/2023] Open
Abstract
Many of the human viruses with oncogenic capabilities, either in their natural host or in experimental systems (hepatitis B and C, human T cell leukaemia virus type 1, Kaposi sarcoma herpesvirus, human immunodeficiency virus, high-risk human papillomaviruses and adenovirus type 9), encode in their limited genome the ability to target cellular proteins containing PSD95/ DLG/ZO-1 (PDZ) interaction modules. In many cases (but not always), the viruses have evolved to bind the PDZ domains using the same short linear peptide motifs found in host protein-PDZ interactions, and in some cases regulate the interactions in a similar fashion by phosphorylation. What is striking is that the diverse viruses target a common subset of PDZ proteins that are intimately involved in controlling cell polarity and the structure and function of intercellular junctions, including tight junctions. Cell polarity is fundamental to the control of cell proliferation and cell survival and disruption of polarity and the signal transduction pathways involved is a key event in tumourigenesis. This review focuses on the oncogenic viruses and the role of targeting PDZ proteins in the virus life cycle and the contribution of virus-PDZ protein interactions to virus-mediated oncogenesis. We highlight how many of the viral associations with PDZ proteins lead to deregulation of PI3K/AKT signalling, benefitting virus replication but as a consequence also contributing to oncogenesis.
Collapse
Affiliation(s)
- Claire D James
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK.
- Present address; Virginia Commonwealth University, School of Dentistry, W. Baxter Perkinson Jr. Building, 521 North 11th Street, P.O. Box 980566, Richmond, VA 23298-0566, USA.
| | - Sally Roberts
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK.
| |
Collapse
|
25
|
Lévy P, Bartosch B. Metabolic reprogramming: a hallmark of viral oncogenesis. Oncogene 2015; 35:4155-64. [DOI: 10.1038/onc.2015.479] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 02/07/2023]
|
26
|
Adenovirus E4-ORF1 Dysregulates Epidermal Growth Factor and Insulin/Insulin-Like Growth Factor Receptors To Mediate Constitutive Myc Expression. J Virol 2015; 89:10774-85. [PMID: 26269183 DOI: 10.1128/jvi.01463-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/05/2015] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED The E4-ORF1 protein encoded by human adenovirus stimulates viral replication in human epithelial cells by binding and activating cellular phosphatidylinositol 3-kinase (PI3K) at the plasma membrane and cellular Myc in the nucleus. In this study, we showed that E4-ORF1 hijacks the tyrosine kinase activities of cellular epidermal growth factor receptor (EGFR) and insulin receptor (InsR)/insulin-like growth factor receptor 1 (IGF1R), as well as the lipid kinase activity of PI3K, to mediate constitutive Myc protein expression. We additionally demonstrated that EGFR contributes to constitutive Myc expression through the capacity of E4-ORF1 to induce ligand-independent EGFR activation and stimulation of the Ras/Mek/Erk pathway, the latter activity of which was conserved by human adenoviruses. Results further suggested that EGFR normally forms a complex with the cellular PDZ protein Discs Large 1 (Dlg1), a component of the Dlg1:E4-ORF1:PI3K ternary complex that mediates E4-ORF1-induced PI3K activation, and that E4-ORF1 binds the Dlg1:EGFR complex and promotes the association of EGFR with InsR and IGF1R. In addition to its role in constitutive Myc expression, InsR/IGF1R also negatively regulates EGFR autophosphorylation and EGFR-mediated Ras/Mek/Erk signaling, and data suggested that E4-ORF1 binding to Dlg1 antagonizes these activities. Collectively, our findings suggest that in human epithelial cells, E4-ORF1 targets EGFR, InsR/IGF1R, and PI3K at the plasma membrane to activate cytosolic signaling pathways that sustain Myc protein levels in the nucleus. We postulate that E4-ORF1-induced constitutive Myc expression functions to ensure the formation of nuclear E4-ORF1:Myc complexes, which have been shown to activate Myc and to enhance adenovirus replication. IMPORTANCE While human adenoviruses primarily produce self-limited acute infections in humans, these agents are associated with life-threatening diseases in immunocompromised patients and in otherwise healthy individuals infected with certain virulent serotypes. The adenovirus E4-ORF1 protein enhances viral replication by activating the cellular lipid kinase PI3K and the cellular transcription factor Myc. Here we report that E4-ORF1 usurps the functions of the cellular tyrosine kinase receptors EGFR and InsR /: IGF1R, as well as PI3K, to sustain Myc protein expression in cells. Furthermore, sustained Myc expression depended on E4-ORF1-induced ligand-independent EGFR activation that stimulated Ras/Mek/Erk signaling, a function found to be conserved by human adenoviruses. Given the established roles of PI3K, the Ras/Mek/Erk pathway, and Myc in the adenovirus life cycle, our findings may aid in the development of safer, more effective therapeutic strategies to treat severe adenovirus infections as well as improved adenovirus vectors for use in vaccination and gene and cancer therapy.
Collapse
|
27
|
Ganti K, Broniarczyk J, Manoubi W, Massimi P, Mittal S, Pim D, Szalmas A, Thatte J, Thomas M, Tomaić V, Banks L. The Human Papillomavirus E6 PDZ Binding Motif: From Life Cycle to Malignancy. Viruses 2015; 7:3530-51. [PMID: 26147797 PMCID: PMC4517114 DOI: 10.3390/v7072785] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 01/01/2023] Open
Abstract
Cancer-causing HPV E6 oncoproteins are characterized by the presence of a PDZ binding motif (PBM) at their extreme carboxy terminus. It was long thought that this region of E6 had a sole function to confer interaction with a defined set of cellular substrates. However, more recent studies have shown that the E6 PBM has a complex pattern of regulation, whereby phosphorylation within the PBM can regulate interaction with two classes of cellular proteins: those containing PDZ domains and the members of the 14-3-3 family of proteins. In this review, we explore the roles that the PBM and its ligands play in the virus life cycle, and subsequently how these can inadvertently contribute towards the development of malignancy. We also explore how subtle alterations in cellular signal transduction pathways might result in aberrant E6 phosphorylation, which in turn might contribute towards disease progression.
Collapse
Affiliation(s)
- Ketaki Ganti
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Justyna Broniarczyk
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Wiem Manoubi
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Paola Massimi
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Suruchi Mittal
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - David Pim
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Anita Szalmas
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Jayashree Thatte
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Miranda Thomas
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Vjekoslav Tomaić
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| | - Lawrence Banks
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy.
| |
Collapse
|
28
|
Ikonomov OC, Sbrissa D, Venkatareddy M, Tisdale E, Garg P, Shisheva A. Class III PI 3-kinase is the main source of PtdIns3P substrate and membrane recruitment signal for PIKfyve constitutive function in podocyte endomembrane homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1240-50. [PMID: 25619930 DOI: 10.1016/j.bbamcr.2015.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/22/2014] [Accepted: 01/16/2015] [Indexed: 11/19/2022]
Abstract
The evolutionarily conserved PIKfyve, which synthesizes PtdIns5P from PtdIns, and PtdIns(3,5)P2 from PtdIns3P, requires PtdIns3P as both an enzyme substrate and a membrane recruitment signal. Whereas the PtdIns3P source is undetermined, class III PI3K (Vps34), the only evolutionarily conserved of the eight mammalian PI3Ks, is presumed as a main candidate. A hallmark of PIKfyve deficiency is formation of multiple translucent cytoplasmic vacuoles seen by light microscopy in cells cultured in complete media. Such an aberrant phenotype is often observed in cells from conditional Vps34 knockout (KO) mice. To clarify the mechanism of Vps34 KO-triggered vacuolation and the PtdIns3P source for PIKfyve functionality, here we have characterized a podocyte cell type derived from Vps34fl/fl mice, which, upon Cre-mediated gene KO, robustly formed cytoplasmic vacuoles resembling those in PikfyveKO MEFs. Vps34wt, expressed in Vps34KO podocytes restored the normal morphology, but only if the endogenous PIKfyve activity was intact. Conversely, expressed PIKfyvewt rescued completely the vacuolation only in PikfyveKO MEFs but not in Vps34KO podocytes. Analyses of phosphoinositide profiles by HPLC and localization patterns by a PtdIns3P biosensor revealed that Vps34 is the main supplier of localized PtdIns3P not only for PIKfyve activity but also for membrane recruitment. Concordantly, Vps34KO podocytes had severely reduced steady-state levels of both PtdIns(3,5)P2 and PtdIns5P, along with PtdIns3P. We further revealed a plausible physiologically-relevant Vps34-independent PtdIns3P supply for PIKfyve, operating through activated class I PI3Ks. Our data provide the first evidence that the vacuolation phenotype in Vps34KO podocytes is due to PIKfyve dysfunction and that Vps34 is a main PtdIns3P source for constitutive PIKfyve functionality.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Diego Sbrissa
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | - Ellen Tisdale
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Puneet Garg
- Division of Nephrology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
29
|
Hijacking Dlg1 for oncogenic phosphatidylinositol 3-kinase activation in human epithelial cells is a conserved mechanism of human adenovirus E4-ORF1 proteins. J Virol 2014; 88:14268-77. [PMID: 25253337 DOI: 10.1128/jvi.02324-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED The E4-ORF1 gene of human adenoviruses encodes a 14-kDa protein that promotes viral replication as well as cellular metabolic reprogramming, survival, and transformation by constitutively activating cellular phosphatidylinositol 3-kinase (PI3K). We recently reported that the E4-ORF1 protein from subgroup D human adenovirus type 9 upregulates and oncogenically activates PI3K by a novel mechanism involving separate interactions of E4-ORF1 with cellular discs large 1 (Dlg1) and PI3K to form a ternary complex that translocates to the plasma membrane (K. Kong, M. Kumar, M. Taruishi, and R. T. Javier, PLoS Pathog. 10:e1004102, 2014, doi:10.1371/journal.ppat.1004102). The current study was carried out to investigate whether other human adenovirus E4-ORF1 proteins share this mechanism of action. The results showed that in human MCF10A epithelial cells, stable expression of E4-ORF1 proteins encoded by representative human adenovirus serotypes from subgroups A to D induce ternary complex formation, Dlg1-dependent PI3K activation, PI3K protein elevation, Dlg1 and PI3K membrane recruitment, and PI3K-dependent cellular transformation. The first three of these E4-ORF1 activities were also observed in MCF10A cells infected with each wild-type human adenovirus from subgroups A to D. Our findings indicate that most, if not all, human adenovirus E4-ORF1 proteins share a conserved molecular mechanism of PI3K activation, which confers a common capacity to promote oncogenic transformation in human epithelial cells. IMPORTANCE PI3K activation by the adenovirus E4-ORF1 protein mediates oncogenic cellular transformation by human adenovirus type 9, augments viral protein expression and replication by human adenovirus type 5, and dysregulates cellular glucose and lipid metabolism by human adenovirus type 36. For the first time, we report that E4-ORF1 proteins from human adenoviruses in subgroups A to D evolved a conserved molecular mechanism to mediate constitutive PI3K activation that can provoke oncogenic transformation in human epithelial cells. The results raise potential safety concerns about the use of vectors encoding the E4-ORF1 gene in human gene therapy and vaccination. Our findings further suggest that the conserved mechanism revealed here may be targeted for development of therapeutic drugs to treat and prevent adenovirus-associated human diseases.
Collapse
|