151
|
Mahfuz AMUB, Zubair-Bin-Mahfuj AM, Podder DJ. A network-biology approach for identification of key genes and pathways involved in malignant peritoneal mesothelioma. Genomics Inform 2021; 19:e16. [PMID: 34261301 PMCID: PMC8261271 DOI: 10.5808/gi.21019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/05/2021] [Accepted: 05/15/2021] [Indexed: 11/20/2022] Open
Abstract
Even in the current age of advanced medicine, the prognosis of malignant peritoneal mesothelioma (MPM) remains abysmal. Molecular mechanisms responsible for the initiation and progression of MPM are still largely not understood. Adopting an integrated bioinformatics approach, this study aims to identify the key genes and pathways responsible for MPM. Genes that are differentially expressed in MPM in comparison with the peritoneum of healthy controls have been identified by analyzing a microarray gene expression dataset. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses of these differentially expressed genes (DEG) were conducted to gain a better insight. A protein-protein interaction (PPI) network of the proteins encoded by the DEGs was constructed using STRING and hub genes were detected analyzing this network. Next, the transcription factors and miRNAs that have possible regulatory roles on the hub genes were detected. Finally, survival analyses based on the hub genes were conducted using the GEPIA2 web server. Six hundred six genes were found to be differentially expressed in MPM; 133 are upregulated and 473 are downregulated. Analyzing the STRING generated PPI network, six dense modules and 12 hub genes were identified. Fifteen transcription factors and 10 miRNAs were identified to have the most extensive regulatory functions on the DEGs. Through bioinformatics analyses, this work provides an insight into the potential genes and pathways involved in MPM.
Collapse
Affiliation(s)
- A. M. U. B. Mahfuz
- Department of Biotechnology & Genetic Engineering, Faculty of Life Science, University of Development Alternative, Dhaka 1209, Bangladesh
| | | | - Dibya Joti Podder
- Department of General Surgery, Sher-E-Bangla Medical College, Barishal 8200, Bangladesh
| |
Collapse
|
152
|
Favere K, Bosman M, Klingel K, Heymans S, Van Linthout S, Delputte PL, De Sutter J, Heidbuchel H, Guns PJ. Toll-Like Receptors: Are They Taking a Toll on the Heart in Viral Myocarditis? Viruses 2021; 13:v13061003. [PMID: 34072044 PMCID: PMC8227433 DOI: 10.3390/v13061003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
Myocarditis is an inflammatory disease of the heart with viral infections being the most common aetiology. Its complex biology remains poorly understood and its clinical management is one of the most challenging in the field of cardiology. Toll-like receptors (TLRs), a family of evolutionarily conserved pattern recognition receptors, are increasingly known to be implicated in the pathophysiology of viral myocarditis. Their central role in innate and adaptive immune responses, and in the inflammatory reaction that ensues, indeed makes them prime candidates to profoundly affect every stage of the disease process. This review describes the pathogenesis and pathophysiology of viral myocarditis, and scrutinises the role of TLRs in every phase. We conclude with directions for future research in this field.
Collapse
Affiliation(s)
- Kasper Favere
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; (M.B.); (P.-J.G.)
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium;
- Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium
- Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium;
- Correspondence:
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; (M.B.); (P.-J.G.)
| | - Karin Klingel
- Cardiopathology, Institute for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany;
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, 6229 ER Maastricht, The Netherlands;
- Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité, Universitätsmedizin Berlin, 10117 Berlin, Germany;
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Peter L. Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, 2610 Antwerp, Belgium;
| | - Johan De Sutter
- Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium;
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium;
- Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; (M.B.); (P.-J.G.)
| |
Collapse
|
153
|
Custers J, Kim D, Leyssen M, Gurwith M, Tomaka F, Robertson J, Heijnen E, Condit R, Shukarev G, Heerwegh D, van Heesbeen R, Schuitemaker H, Douoguih M, Evans E, Smith ER, Chen RT. Vaccines based on replication incompetent Ad26 viral vectors: Standardized template with key considerations for a risk/benefit assessment. Vaccine 2021; 39:3081-3101. [PMID: 33676782 PMCID: PMC7532807 DOI: 10.1016/j.vaccine.2020.09.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 11/29/2022]
Abstract
Replication-incompetent adenoviral vectors have been under investigation as a platform to carry a variety of transgenes, and express them as a basis for vaccine development. A replication-incompetent adenoviral vector based on human adenovirus type 26 (Ad26) has been evaluated in several clinical trials. The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and features of recombinant viral vector vaccines. This paper reviews features of the Ad26 vectors, including tabulation of safety and risk assessment characteristics of Ad26-based vaccines. In the Ad26 vector, deletion of E1 gene rendering the vector replication incompetent is combined with additional genetic engineering for vaccine manufacturability and transgene expression optimization. These vaccines can be manufactured in mammalian cell lines at scale providing an effective, flexible system for high-yield manufacturing. Ad26 vector vaccines have favorable thermostability profiles, compatible with vaccine supply chains. Safety data are compiled in the Ad26 vaccine safety database version 4.0, with unblinded data from 23 ongoing and completed clinical studies for 3912 participants in five different Ad26-based vaccine programs. Overall, Ad26-based vaccines have been well tolerated, with no significant safety issues identified. Evaluation of Ad26-based vaccines is continuing, with >114,000 participants vaccinated as of 4th September 2020. Extensive evaluation of immunogenicity in humans shows strong, durable humoral and cellular immune responses. Clinical trials have not revealed impact of pre-existing immunity to Ad26 on vaccine immunogenicity, even in the presence of Ad26 neutralizing antibody titers or Ad26-targeting T cell responses at baseline. The first Ad26-based vaccine, against Ebola virus, received marketing authorization from EC on 1st July 2020, as part of the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen. New developments based on Ad26 vectors are underway, including a COVID-19 vaccine, which is currently in phase 3 of clinical evaluation.
Collapse
Affiliation(s)
| | - Denny Kim
- Janssen Research & Development, Titusville, NJ, USA
| | | | - Marc Gurwith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Frank Tomaka
- Janssen Research & Development, Titusville, NJ, USA
| | | | | | - Richard Condit
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | - Eric Evans
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Emily R Smith
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T Chen
- Brighton Collaboration, A Program of the Task Force for Global Health, Decatur, GA, USA
| |
Collapse
|
154
|
Chéneau C, Eichholz K, Tran TH, Tran TTP, Paris O, Henriquet C, Bajramovic JJ, Pugniere M, Kremer EJ. Lactoferrin Retargets Human Adenoviruses to TLR4 to Induce an Abortive NLRP3-Associated Pyroptotic Response in Human Phagocytes. Front Immunol 2021; 12:685218. [PMID: 34093588 PMCID: PMC8173049 DOI: 10.3389/fimmu.2021.685218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
Despite decades of clinical and preclinical investigations, we still poorly grasp our innate immune response to human adenoviruses (HAdVs) and their vectors. In this study, we explored the impact of lactoferrin on three HAdV types that are being used as vectors for vaccines. Lactoferrin is a secreted globular glycoprotein that influences direct and indirect innate immune response against a range of pathogens following a breach in tissue homeostasis. The mechanism by which lactoferrin complexes increases HAdV uptake and induce maturation of human phagocytes is unknown. We show that lactoferrin redirects HAdV types from species B, C, and D to Toll-like receptor 4 (TLR4) cell surface complexes. TLR4-mediated internalization of the HAdV-lactoferrin complex induced an NLRP3-associated response that consisted of cytokine release and transient disruption of plasma membrane integrity, without causing cell death. These data impact our understanding of HAdV immunogenicity and may provide ways to increase the efficacy of HAdV-based vectors/vaccines.
Collapse
Affiliation(s)
- Coraline Chéneau
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Karsten Eichholz
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Tuan Hiep Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Thi Thu Phuong Tran
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Océane Paris
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| | - Corinne Henriquet
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université Montpellier, Institut Régional du Cancer, Montpellier, France
| | | | - Martine Pugniere
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université Montpellier, Institut Régional du Cancer, Montpellier, France
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
155
|
Ho CT, Wu MH, Chen MJ, Lin SP, Yen YT, Hung SC. Combination of Mesenchymal Stem Cell-Delivered Oncolytic Virus with Prodrug Activation Increases Efficacy and Safety of Colorectal Cancer Therapy. Biomedicines 2021; 9:548. [PMID: 34068264 PMCID: PMC8153168 DOI: 10.3390/biomedicines9050548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Although oncolytic viruses are currently being evaluated for cancer treatment in clinical trials, systemic administration is hindered by many factors that prevent them from reaching the tumor cells. When administered systemically, mesenchymal stem cells (MSCs) target tumors, and therefore constitute good cell carriers for oncolytic viruses. MSCs were primed with trichostatin A under hypoxia, which upregulated the expression of CXCR4, a chemokine receptor involved in tumor tropism, and coxsackievirus and adenovirus receptor that plays an important role in adenoviral infection. After priming, MSCs were loaded with conditionally replicative adenovirus that exhibits limited proliferation in cells with a functional p53 pathway and encodes Escherichia coli nitroreductase (NTR) enzymes (CRAdNTR) for targeting tumor cells. Primed MSCs increased tumor tropism and susceptibility to adenoviral infection, and successfully protected CRAdNTR from neutralization by anti-adenovirus antibodies both in vitro and in vivo, and specifically targeted p53-deficient colorectal tumors when infused intravenously. Analyses of deproteinized tissues by UPLC-MS/QTOF revealed that these MSCs converted the co-administered prodrug CB1954 into cytotoxic metabolites, such as 4-hydroxylamine and 2-amine, inducing oncolysis and tumor growth inhibition without being toxic for the host vital organs. This study shows that the combination of oncolytic viruses delivered by MSCs with the activation of prodrugs is a new cancer treatment strategy that provides a new approach for the development of oncolytic viral therapy for various cancers.
Collapse
Affiliation(s)
- Chun-Te Ho
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan; (C.-T.H.); (Y.-T.Y.)
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung 404, Taiwan
| | - Mei-Hsuan Wu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (M.-H.W.); (S.-P.L.)
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital & Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan;
| | - Shih-Pei Lin
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (M.-H.W.); (S.-P.L.)
| | - Yu-Ting Yen
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan; (C.-T.H.); (Y.-T.Y.)
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung 404, Taiwan
| | - Shih-Chieh Hung
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan; (C.-T.H.); (Y.-T.Y.)
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
156
|
Li J, Wang W, Wang J, Cao Y, Wang S, Zhao J. Viral Gene Therapy for Glioblastoma Multiforme: A Promising Hope for the Current Dilemma. Front Oncol 2021; 11:678226. [PMID: 34055646 PMCID: PMC8155537 DOI: 10.3389/fonc.2021.678226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/29/2021] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme (GBM), as one of the most common malignant brain tumors, was limited in its treatment effectiveness with current options. Its invasive and infiltrative features led to tumor recurrence and poor prognosis. Effective treatment and survival improvement have always been a challenge. With the exploration of genetic mutations and molecular pathways in neuro-oncology, gene therapy is becoming a promising therapeutic approach. Therapeutic genes are delivered into target cells with viral vectors to act specific antitumor effects, which can be used in gene delivery, play an oncolysis effect, and induce host immune response. The application of engineering technology makes the virus vector used in genetics a more prospective future. Recent advances in viral gene therapy offer hope for treating brain tumors. In this review, we discuss the types and designs of viruses as well as their study progress and potential applications in the treatment of GBM. Although still under research, viral gene therapy is promising to be a new therapeutic approach for GBM treatment in the future.
Collapse
Affiliation(s)
- Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Wen Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Jia Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Yong Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China.,Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
157
|
ICAM-1 induced rearrangements of capsid and genome prime rhinovirus 14 for activation and uncoating. Proc Natl Acad Sci U S A 2021; 118:2024251118. [PMID: 33947819 PMCID: PMC8126848 DOI: 10.1073/pnas.2024251118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Medical visits and missed days of school and work caused by rhinoviruses cost tens of billions of US dollars annually. Currently, there are no antivirals against rhinoviruses, and the available treatments only treat the symptoms. Here, we present the molecular structure of human rhinovirus 14 in complex with its cellular receptor intercellular adhesion molecule 1. The binding of the virus to its receptor initiates the infection. Knowledge of the structure of the human rhinovirus 14–intercellular adhesion molecule 1 interface and mechanism of interaction provides the basis for the design of compounds that may block the binding of rhinoviruses to receptors and thus prevent infection. Most rhinoviruses, which are the leading cause of the common cold, utilize intercellular adhesion molecule-1 (ICAM-1) as a receptor to infect cells. To release their genomes, rhinoviruses convert to activated particles that contain pores in the capsid, lack minor capsid protein VP4, and have an altered genome organization. The binding of rhinoviruses to ICAM-1 promotes virus activation; however, the molecular details of the process remain unknown. Here, we present the structures of virion of rhinovirus 14 and its complex with ICAM-1 determined to resolutions of 2.6 and 2.4 Å, respectively. The cryo-electron microscopy reconstruction of rhinovirus 14 virions contains the resolved density of octanucleotide segments from the RNA genome that interact with VP2 subunits. We show that the binding of ICAM-1 to rhinovirus 14 is required to prime the virus for activation and genome release at acidic pH. Formation of the rhinovirus 14–ICAM-1 complex induces conformational changes to the rhinovirus 14 capsid, including translocation of the C termini of VP4 subunits, which become poised for release through pores that open in the capsids of activated particles. VP4 subunits with altered conformation block the RNA–VP2 interactions and expose patches of positively charged residues. The conformational changes to the capsid induce the redistribution of the virus genome by altering the capsid–RNA interactions. The restructuring of the rhinovirus 14 capsid and genome prepares the virions for conversion to activated particles. The high-resolution structure of rhinovirus 14 in complex with ICAM-1 explains how the binding of uncoating receptors enables enterovirus genome release.
Collapse
|
158
|
Mercier J, Ruffin M, Corvol H, Guillot L. Gene Therapy: A Possible Alternative to CFTR Modulators? Front Pharmacol 2021; 12:648203. [PMID: 33967785 PMCID: PMC8097140 DOI: 10.3389/fphar.2021.648203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a rare genetic disease that affects several organs, but lung disease is the major cause of morbidity and mortality. The gene responsible for CF, the CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) gene, has been discovered in 1989. Since then, gene therapy i.e., defective gene replacement by a functional one, remained the ultimate goal but unfortunately, it has not yet been achieved. However, patients care and symptomatic treatments considerably increased CF patients’ life expectancy ranging from 5 years old in the 1960s to 40 today. In the last decade, research works on CFTR protein structure and activity led to the development of new drugs which, by readdressing CFTR to the plasma membrane (correctors) or by enhancing its transport activity (potentiators), allow, alone or in combination, an improvement of CF patients’ lung function and quality of life. While expected, it is not yet known whether taking these drugs from an early age and for years will improve the quality of life of CF patients in the long term and further increase their life expectancy. Besides, these molecules are not available (specific variants of CFTR) or accessible (national health policies) for all patients and there is still no curative treatment. Another alternative that could benefit from new technologies, such as gene therapy, is therefore still attractive, although it is not yet offered to patients. Faced with the development of new CFTR correctors and potentiators, the question arises as to whether there is still a place for gene therapy and this is discussed in this perspective.
Collapse
Affiliation(s)
- J Mercier
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - M Ruffin
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - H Corvol
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France.,Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - L Guillot
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| |
Collapse
|
159
|
Geisler A, Hazini A, Heimann L, Kurreck J, Fechner H. Coxsackievirus B3-Its Potential as an Oncolytic Virus. Viruses 2021; 13:v13050718. [PMID: 33919076 PMCID: PMC8143167 DOI: 10.3390/v13050718] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Oncolytic virotherapy represents one of the most advanced strategies to treat otherwise untreatable types of cancer. Despite encouraging developments in recent years, the limited fraction of patients responding to therapy has demonstrated the need to search for new suitable viruses. Coxsackievirus B3 (CVB3) is a promising novel candidate with particularly valuable features. Its entry receptor, the coxsackievirus and adenovirus receptor (CAR), and heparan sulfate, which is used for cellular entry by some CVB3 variants, are highly expressed on various cancer types. Consequently, CVB3 has broad anti-tumor activity, as shown in various xenograft and syngeneic mouse tumor models. In addition to direct tumor cell killing the virus induces a strong immune response against the tumor, which contributes to a substantial increase in the efficiency of the treatment. The toxicity of oncolytic CVB3 in healthy tissues is variable and depends on the virus strain. It can be abrogated by genetic engineering the virus with target sites of microRNAs. In this review, we present an overview of the current status of the development of CVB3 as an oncolytic virus and outline which steps still need to be accomplished to develop CVB3 as a therapeutic agent for clinical use in cancer treatment.
Collapse
Affiliation(s)
- Anja Geisler
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.G.); (L.H.); (J.K.)
| | - Ahmet Hazini
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK;
| | - Lisanne Heimann
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.G.); (L.H.); (J.K.)
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.G.); (L.H.); (J.K.)
| | - Henry Fechner
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; (A.G.); (L.H.); (J.K.)
- Correspondence: ; Tel.: +49-30-31-47-21-81
| |
Collapse
|
160
|
Tessarollo NG, Domingues ACM, Antunes F, da Luz JCDS, Rodrigues OA, Cerqueira OLD, Strauss BE. Nonreplicating Adenoviral Vectors: Improving Tropism and Delivery of Cancer Gene Therapy. Cancers (Basel) 2021; 13:1863. [PMID: 33919679 PMCID: PMC8069790 DOI: 10.3390/cancers13081863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Recent preclinical and clinical studies have used viral vectors in gene therapy research, especially nonreplicating adenovirus encoding strategic therapeutic genes for cancer treatment. Adenoviruses were the first DNA viruses to go into therapeutic development, mainly due to well-known biological features: stability in vivo, ease of manufacture, and efficient gene delivery to dividing and nondividing cells. However, there are some limitations for gene therapy using adenoviral vectors, such as nonspecific transduction of normal cells and liver sequestration and neutralization by antibodies, especially when administered systemically. On the other hand, adenoviral vectors are amenable to strategies for the modification of their biological structures, including genetic manipulation of viral proteins, pseudotyping, and conjugation with polymers or biological membranes. Such modifications provide greater specificity to the target cell and better safety in systemic administration; thus, a reduction of antiviral host responses would favor the use of adenoviral vectors in cancer immunotherapy. In this review, we describe the structural and molecular features of nonreplicating adenoviral vectors, the current limitations to their use, and strategies to modify adenoviral tropism, highlighting the approaches that may allow for the systemic administration of gene therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bryan E. Strauss
- Viral Vector Laboratory, Center for Translational Investigation in Oncology, Cancer Institute of São Paulo/LIM24, University of São Paulo School of Medicine, São Paulo 01246-000, Brazil; (N.G.T.); (A.C.M.D.); (F.A.); (J.C.d.S.d.L.); (O.A.R.); (O.L.D.C.)
| |
Collapse
|
161
|
Nilchian A, Plant E, Parniewska MM, Santiago A, Rossignoli A, Skogsberg J, Hedin U, Matic L, Fuxe J. Induction of the Coxsackievirus and Adenovirus Receptor in Macrophages During the Formation of Atherosclerotic Plaques. J Infect Dis 2021; 222:2041-2051. [PMID: 32852032 PMCID: PMC7661765 DOI: 10.1093/infdis/jiaa418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/07/2020] [Indexed: 11/14/2022] Open
Abstract
Multiple viruses are implicated in atherosclerosis, but the mechanisms by which they infect cells and contribute to plaque formation in arterial walls are not well understood. Based on reports showing the presence of enterovirus in atherosclerotic plaques we hypothesized that the coxsackievirus and adenovirus receptor (CXADR/CAR), although absent in normal arteries, could be induced during plaque formation. Large-scale microarray and mass spectrometric analyses revealed significant up-regulation of CXADR messenger RNA and protein levels in plaque-invested carotid arteries compared with control arteries. Macrophages were identified as a previously unknown cellular source of CXADR in human plaques and plaques from Ldr-/-Apob100/100 mice. CXADR was specifically associated with M1-polarized macrophages and foam cells and was experimentally induced during macrophage differentiation. Furthermore, it was significantly correlated with receptors for other viruses linked to atherosclerosis. The results show that CXADR is induced in macrophages during plaque formation, suggesting a mechanism by which enterovirus infect cells in atherosclerotic plaques.
Collapse
Affiliation(s)
- Azadeh Nilchian
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Estelle Plant
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Malgorzata M Parniewska
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ana Santiago
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Aránzazu Rossignoli
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Josefin Skogsberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jonas Fuxe
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
162
|
Podshivalova ES, Semkina AS, Kravchenko DS, Frolova EI, Chumakov SP. Efficient delivery of oncolytic enterovirus by carrier cell line NK-92. MOLECULAR THERAPY-ONCOLYTICS 2021; 21:110-118. [PMID: 33981827 PMCID: PMC8065264 DOI: 10.1016/j.omto.2021.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Many members of the enterovirus family are considered as promising oncolytic agents; however, their systemic administration is largely inefficient due to the rapid neutralization of the virus in the circulation and the barrier functions of the endothelium. We aimed to evaluate natural killer cells as carriers for the delivery of oncolytic enteroviruses, which would combine the effects of cell immunotherapy with virotherapy. We tested four strains of nonpathogenic enteroviruses against the glioblastoma cell line panel and evaluated the produced infectious titers. Next, we explored whether these virus strains could be delivered to the tumor by natural killer cell line NK-92, which is being actively evaluated as a clinically acceptable therapeutic. Several strains of enteroviruses demonstrated oncolytic properties, but only coxsackievirus A7 (CVA7) could replicate in NK-92 cells efficiently. We compared the delivery efficiency of CVA7 in vivo, using NK-92 cells and direct intravenous administration, and found significant advantages of cell delivery even after a single injection. This suggests that the NK-92 cell line can be utilized as a vehicle for the delivery of the oncolytic strain of CVA7, which would improve the clinical potential of this viral oncolytic for the treatment of glioblastoma multiforme and other forms of cancer.
Collapse
Affiliation(s)
| | - Alevtina Sergeevna Semkina
- Department of Medical Nanobiotechnologies, Pirogov Russian National Research Medical University, Ostrovityanova 1, Moscow 117997, Russia.,Department of Basic and Applied Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Kropotkinskiy 23, Moscow 119991, Russia
| | - Dmitry Sergeevich Kravchenko
- Department of Peptide and Protein Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Elena Ivanovna Frolova
- Department of Peptide and Protein Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Stepan Petrovich Chumakov
- Department of Peptide and Protein Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| |
Collapse
|
163
|
Modeling the complete kinetics of coxsackievirus B3 reveals human determinants of host-cell feedback. Cell Syst 2021; 12:304-323.e13. [PMID: 33740397 DOI: 10.1016/j.cels.2021.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/13/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Complete kinetic models are pervasive in chemistry but lacking in biological systems. We encoded the complete kinetics of infection for coxsackievirus B3 (CVB3), a compact and fast-acting RNA virus. The model consists of separable, detailed modules describing viral binding-delivery, translation-replication, and encapsidation. Specific module activities are dampened by the type I interferon response to viral double-stranded RNAs (dsRNAs), which is itself disrupted by viral proteinases. The experimentally validated kinetics uncovered that cleavability of the dsRNA transducer mitochondrial antiviral signaling protein (MAVS) becomes a stronger determinant of viral outcomes when cells receive supplemental interferon after infection. Cleavability is naturally altered in humans by a common MAVS polymorphism, which removes a proteinase-targeted site but paradoxically elevates CVB3 infectivity. These observations are reconciled with a simple nonlinear model of MAVS regulation. Modeling complete kinetics is an attainable goal for small, rapidly infecting viruses and perhaps viral pathogens more broadly. A record of this paper's transparent peer review process is included in the Supplemental information.
Collapse
|
164
|
Garcia-Moure M, Gonzalez-Huarriz M, Labiano S, Guruceaga E, Bandres E, Zalacain M, Marrodan L, de Andrea C, Villalba M, Martinez-Velez N, Laspidea V, Puigdelloses M, Gallego Perez-Larraya J, Iñigo-Marco I, Stripecke R, Chan JA, Raabe EH, Kool M, Gomez-Manzano C, Fueyo J, Patiño-García A, Alonso MM. Delta-24-RGD, an Oncolytic Adenovirus, Increases Survival and Promotes Proinflammatory Immune Landscape Remodeling in Models of AT/RT and CNS-PNET. Clin Cancer Res 2021; 27:1807-1820. [PMID: 33376098 PMCID: PMC7617079 DOI: 10.1158/1078-0432.ccr-20-3313] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/14/2020] [Accepted: 12/22/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Atypical teratoid/rhabdoid tumors (AT/RT) and central nervous system primitive neuroectodermal tumors (CNS-PNET) are pediatric brain tumors with poor survival and life-long negative side effects. Here, the aim was to characterize the efficacy and safety of the oncolytic adenovirus, Delta-24-RGD, which selectively replicates in and kills tumor cells. EXPERIMENTAL DESIGN Delta-24-RGD determinants for infection and replication were evaluated in patient expression datasets. Viral replication and cytotoxicity were assessed in vitro in a battery of CNS-PNET and AT/RT cell lines. In vivo, efficacy was determined in different orthotopic mouse models, including early and established tumor models, a disseminated AT/RT lesion model, and immunocompetent humanized mouse models (hCD34+-NSG-SGM3). RESULTS Delta-24-RGD infected and replicated efficiently in all the cell lines tested. In addition, the virus induced dose-dependent cytotoxicity [IC50 value below 1 plaque-forming unit (PFU)/cell] and the release of immunogenic markers. In vivo, a single intratumoral Delta-24-RGD injection (107 or 108 PFU) significantly increased survival and led to long-term survival in AT/RT and PNET models. Delta-24-RGD hindered the dissemination of AT/RTs and increased survival, leading to 70% of long-term survivors. Of relevance, viral administration to established tumor masses (30 days after engraftment) showed therapeutic benefit. In humanized immunocompetent models, Delta-24-RGD significantly extended the survival of mice bearing AT/RTs or PNETs (ranging from 11 to 27 days) and did not display any toxicity associated with inflammation. Immunophenotyping of Delta-24-RGD-treated tumors revealed increased CD8+ T-cell infiltration. CONCLUSIONS Delta-24-RGD is a feasible therapeutic option for AT/RTs and CNS-PNETs. This work constitutes the basis for potential translation to the clinical setting.
Collapse
Affiliation(s)
- Marc Garcia-Moure
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain.
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Elizabeth Guruceaga
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Bioinformatics Platform, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Navarra, Spain
| | - Eva Bandres
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Immunology Unit, Department of Hematology, Complejo Hospitalario de Navarra, Pamplona, Navarra, Spain
| | - Marta Zalacain
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lucia Marrodan
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Carlos de Andrea
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Naiara Martinez-Velez
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Virginia Laspidea
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Montse Puigdelloses
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Jaime Gallego Perez-Larraya
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Iñigo-Marco
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Renata Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Laboratory of Regenerative Immune Therapies Applied of the Research Network REBIRTH, German Centre for Infection Research (DZIF), partner site Hannover, Hannover, Germany
| | - Jennifer A Chan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eric H Raabe
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Division of Pediatric Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center (KITZ), Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Candelaria Gomez-Manzano
- Department of NeuroOncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juan Fueyo
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana Patiño-García
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Marta M Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain.
- Program in Solid Tumors, Foundation for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| |
Collapse
|
165
|
Holcomb D, Alexaki A, Hernandez N, Hunt R, Laurie K, Kames J, Hamasaki-Katagiri N, Komar AA, DiCuccio M, Kimchi-Sarfaty C. Gene variants of coagulation related proteins that interact with SARS-CoV-2. PLoS Comput Biol 2021; 17:e1008805. [PMID: 33730015 PMCID: PMC8007013 DOI: 10.1371/journal.pcbi.1008805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/29/2021] [Accepted: 02/15/2021] [Indexed: 12/30/2022] Open
Abstract
Thrombosis is a recognized complication of Coronavirus disease of 2019 (COVID-19) and is often associated with poor prognosis. There is a well-recognized link between coagulation and inflammation, however, the extent of thrombotic events associated with COVID-19 warrants further investigation. Poly(A) Binding Protein Cytoplasmic 4 (PABPC4), Serine/Cysteine Proteinase Inhibitor Clade G Member 1 (SERPING1) and Vitamin K epOxide Reductase Complex subunit 1 (VKORC1), which are all proteins linked to coagulation, have been shown to interact with SARS proteins. We computationally examined the interaction of these with SARS-CoV-2 proteins and, in the case of VKORC1, we describe its binding to ORF7a in detail. We examined the occurrence of variants of each of these proteins across populations and interrogated their potential contribution to COVID-19 severity. Potential mechanisms, by which some of these variants may contribute to disease, are proposed. Some of these variants are prevalent in minority groups that are disproportionally affected by severe COVID-19. Therefore, we are proposing that further investigation around these variants may lead to better understanding of disease pathogenesis in minority groups and more informed therapeutic approaches.
Collapse
Affiliation(s)
- David Holcomb
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Aikaterini Alexaki
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Nancy Hernandez
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Ryan Hunt
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Kyle Laurie
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jacob Kames
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Nobuko Hamasaki-Katagiri
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Anton A. Komar
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio, United States of America
| | - Michael DiCuccio
- National Center of Biotechnology Information, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chava Kimchi-Sarfaty
- Center for Biologics Evaluation and Research, Office of Tissues and Advanced Therapies, Division of Plasma Protein Therapeutics, Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
166
|
Heat Shock Protein 90 Chaperones E1A Early Protein of Adenovirus 5 and Is Essential for Replication of the Virus. Int J Mol Sci 2021; 22:ijms22042020. [PMID: 33670684 PMCID: PMC7921956 DOI: 10.3390/ijms22042020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/04/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Adenovirus infections tend to be mild, but they may pose a serious threat for young and immunocompromised individuals. The treatment is complicated because there are no approved safe and specific drugs for adenovirus infections. Here, we present evidence that 17-(Allylamino)-17-demethoxygeldanamycin (17-AAG), an inhibitor of Hsp90 chaperone, decreases the rate of human adenovirus 5 (HAdV-5) replication in cell cultures by 95%. 17-AAG inhibited the transcription of early and late genes of HAdV-5, replication of viral DNA, and expression of viral proteins. 6 h after infection, Hsp90 inhibition results in a 6.3-fold reduction of the newly synthesized E1A protein level without a decrease in the E1A mRNA level. However, the Hsp90 inhibition does not increase the decay rate of the E1A protein that was constitutively expressed in the cell before exposure to the inhibitor. The co-immunoprecipitation proved that E1A protein interacted with Hsp90. Altogether, the presented results show, for the first time. that Hsp90 chaperones newly synthesized, but not mature, E1A protein. Because E1A serves as a transcriptional co-activator of adenovirus early genes, the anti-adenoviral activity of the Hsp90 inhibitor might be explained by the decreased E1A level.
Collapse
|
167
|
Heparan Sulfate Is a Cellular Receptor for Enteric Human Adenoviruses. Viruses 2021; 13:v13020298. [PMID: 33672966 PMCID: PMC7918131 DOI: 10.3390/v13020298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/16/2022] Open
Abstract
Human adenovirus (HAdV)-F40 and -F41 are leading causes of diarrhea and diarrhea-associated mortality in children under the age of five, but the mechanisms by which they infect host cells are poorly understood. HAdVs initiate infection through interactions between the knob domain of the fiber capsid protein and host cell receptors. Unlike most other HAdVs, HAdV-F40 and -F41 possess two different fiber proteins-a long fiber and a short fiber. Whereas the long fiber binds to the Coxsackievirus and adenovirus receptor (CAR), no binding partners have been identified for the short fiber. In this study, we identified heparan sulfate (HS) as an interaction partner for the short fiber of enteric HAdVs. We demonstrate that exposure to acidic pH, which mimics the environment of the stomach, inactivates the interaction of enteric adenovirus with CAR. However, the short fiber:HS interaction is resistant to and even enhanced by acidic pH, which allows attachment to host cells. Our results suggest a switch in receptor usage of enteric HAdVs after exposure to acidic pH and add to the understanding of the function of the short fibers. These results may also be useful for antiviral drug development and the utilization of enteric HAdVs for clinical applications such as vaccine development.
Collapse
|
168
|
Adenovirus and the Cornea: More Than Meets the Eye. Viruses 2021; 13:v13020293. [PMID: 33668417 PMCID: PMC7917768 DOI: 10.3390/v13020293] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/07/2021] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
Human adenoviruses cause disease at multiple mucosal sites, including the respiratory, gastrointestinal, and genitourinary tracts, and are common agents of conjunctivitis. One site of infection that has received sparse attention is the cornea, a transparent tissue and the window of the eye. While most adenovirus infections are self-limited, corneal inflammation (keratitis) due to adenovirus can persist or recur for months to years after infection, leading to reduced vision, discomfort, and light sensitivity. Topical corticosteroids effectively suppress late adenovirus keratitis but are associated with vision-threatening side effects. In this short review, we summarize current knowledge on infection of the cornea by adenoviruses, including corneal epithelial cell receptors and determinants of corneal tropism. We briefly discuss mechanisms of stromal keratitis due to adenovirus infection, and review an emerging therapy to mitigate adenovirus corneal infections based on evolving knowledge of corneal epithelial receptor usage.
Collapse
|
169
|
Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduct Target Ther 2021; 6:53. [PMID: 33558455 PMCID: PMC7868676 DOI: 10.1038/s41392-021-00487-6] [Citation(s) in RCA: 718] [Impact Index Per Article: 179.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/05/2020] [Accepted: 10/23/2020] [Indexed: 01/30/2023] Open
Abstract
Throughout its 40-year history, the field of gene therapy has been marked by many transitions. It has seen great strides in combating human disease, has given hope to patients and families with limited treatment options, but has also been subject to many setbacks. Treatment of patients with this class of investigational drugs has resulted in severe adverse effects and, even in rare cases, death. At the heart of this dichotomous field are the viral-based vectors, the delivery vehicles that have allowed researchers and clinicians to develop powerful drug platforms, and have radically changed the face of medicine. Within the past 5 years, the gene therapy field has seen a wave of drugs based on viral vectors that have gained regulatory approval that come in a variety of designs and purposes. These modalities range from vector-based cancer therapies, to treating monogenic diseases with life-altering outcomes. At present, the three key vector strategies are based on adenoviruses, adeno-associated viruses, and lentiviruses. They have led the way in preclinical and clinical successes in the past two decades. However, despite these successes, many challenges still limit these approaches from attaining their full potential. To review the viral vector-based gene therapy landscape, we focus on these three highly regarded vector platforms and describe mechanisms of action and their roles in treating human disease.
Collapse
Affiliation(s)
- Jote T Bulcha
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic medical sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hong Ma
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
170
|
Xu L, Zheng Q, Zhu R, Yin Z, Yu H, Lin Y, Wu Y, He M, Huang Y, Jiang Y, Sun H, Zha Z, Yang H, Huang Q, Zhang D, Chen Z, Ye X, Han J, Yang L, Liu C, Que Y, Fang M, Gu Y, Zhang J, Luo W, Zhou ZH, Li S, Cheng T, Xia N. Cryo-EM structures reveal the molecular basis of receptor-initiated coxsackievirus uncoating. Cell Host Microbe 2021; 29:448-462.e5. [PMID: 33539764 DOI: 10.1016/j.chom.2021.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/16/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
Enterovirus uncoating receptors bind at the surface depression ("canyon") that encircles each capsid vertex causing the release of a host-derived lipid called "pocket factor" that is buried in a hydrophobic pocket formed by the major viral capsid protein, VP1. Coxsackievirus and adenovirus receptor (CAR) is a universal uncoating receptor of group B coxsackieviruses (CVB). Here, we present five high-resolution cryoEM structures of CVB representing different stages of virus infection. Structural comparisons show that the CAR penetrates deeper into the canyon than other uncoating receptors, leading to a cascade of events: collapse of the VP1 hydrophobic pocket, high-efficiency release of the pocket factor and viral uncoating and genome release under neutral pH, as compared with low pH. Furthermore, we identified a potent therapeutic antibody that can neutralize viral infection by interfering with virion-CAR interactions, destabilizing the capsid and inducing virion disruption. Together, these results define the structural basis of CVB cell entry and antibody neutralization.
Collapse
Affiliation(s)
- Longfa Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Rui Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhichao Yin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yu Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yuanyuan Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Maozhou He
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yang Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yichao Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Sun
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhenghui Zha
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongwei Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qiongzi Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Dongqing Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhenqin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiangzhong Ye
- Beijing Wantai Enterprise Community Partners, Beijing 102206, China
| | - Jinle Han
- Beijing Wantai Enterprise Community Partners, Beijing 102206, China
| | - Lisheng Yang
- Beijing Wantai Enterprise Community Partners, Beijing 102206, China
| | - Che Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yuqiong Que
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Mujin Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wenxin Luo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Z Hong Zhou
- California NanoSystems Institute (CNSI), UCLA, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen 361102, China; Research Unit of Frontier Technology of Structural Vaccinology, Chinese Academy of Medical Sciences, Xiamen, Fujian 361102, China.
| |
Collapse
|
171
|
Ene CI, Fueyo J, Lang FF. Delta-24 adenoviral therapy for glioblastoma: evolution from the bench to bedside and future considerations. Neurosurg Focus 2021; 50:E6. [PMID: 33524949 DOI: 10.3171/2020.11.focus20853] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/16/2020] [Indexed: 11/06/2022]
Abstract
Delta-24-based oncolytic viruses are conditional replication adenoviruses developed to selectively infect and replicate in retinoblastoma 1 (Rb)-deficient cancer cells but not normal cell with intact Rb1 pathways. Over the years, there has been a significant evolution in the design of Delta-24 based on a better understanding of the underlying basis for infection, replication, and spread within cancer. One example is the development of Delta-24-RGD (DNX-2401), where the arginine-glycine-aspartate (RGD) domain enhances the infectivity of Delta-24 for cancer cells. DNX-2401 demonstrated objective biological and clinical responses during a phase I window of opportunity clinical trial for recurrent human glioblastoma. In long-term responders (> 3 years), there was evidence of immune infiltration (T cells and macrophages) into the tumor microenvironment with minimal toxicity. Although more in-depth analysis and phase III studies are pending, these results indicate that Delta-24-based adenovirus therapy may induce an antitumor response in glioblastoma, resulting in long-term antitumor immune response. In this review, the authors discuss the preclinical and clinical development of Delta-24 oncolytic adenoviral therapy for glioblastoma and describe structural improvements to Delta-24 that have enhanced its efficacy in vivo. They also highlight ongoing research that attempts to address the remaining obstacles limiting efficacy of Delta-24 adenovirus therapy for glioblastoma.
Collapse
Affiliation(s)
| | - Juan Fueyo
- Departments of1Neurosurgery and.,2Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
172
|
Vupputuri S, Tayebi L, Hikkaduwa Koralege RS, Nigatu A, Mozafari M, Mishra A, Liu L, Ramsey JD. Polyethylene glycol–modified DOTAP:cholesterol/adenovirus hybrid vectors have improved transduction efficiency and reduced immunogenicity. JOURNAL OF NANOPARTICLE RESEARCH 2021; 23:37. [DOI: 10.1007/s11051-020-05134-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 12/26/2020] [Indexed: 03/07/2025]
|
173
|
Böhnke J, Pinkert S, Schmidt M, Binder H, Bilz NC, Jung M, Reibetanz U, Beling A, Rujescu D, Claus C. Coxsackievirus B3 Infection of Human iPSC Lines and Derived Primary Germ-Layer Cells Regarding Receptor Expression. Int J Mol Sci 2021; 22:1220. [PMID: 33513663 PMCID: PMC7865966 DOI: 10.3390/ijms22031220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
The association of members of the enterovirus family with pregnancy complications up to miscarriages is under discussion. Here, infection of two different human induced pluripotent stem cell (iPSC) lines and iPSC-derived primary germ-layer cells with coxsackievirus B3 (CVB3) was characterized as an in vitro cell culture model for very early human development. Transcriptomic analysis of iPSC lines infected with recombinant CVB3 expressing enhanced green fluorescent protein (EGFP) revealed a reduction in the expression of pluripotency genes besides an enhancement of genes involved in RNA metabolism. The initial distribution of CVB3-EGFP-positive cells within iPSC colonies correlated with the distribution of its receptor coxsackie- and adenovirus receptor (CAR). Application of anti-CAR blocking antibodies supported the requirement of CAR, but not of the co-receptor decay-accelerating factor (DAF) for infection of iPSC lines. Among iPSC-derived germ-layer cells, mesodermal cells were especially vulnerable to CVB3-EGFP infection. Our data implicate further consideration of members of the enterovirus family in the screening program of human pregnancies. Furthermore, iPSCs with their differentiation capacity into cell populations of relevant viral target organs could offer a reliable screening approach for therapeutic intervention and for assessment of organ-specific enterovirus virulence.
Collapse
Affiliation(s)
- Janik Böhnke
- Institute of Medical Microbiology and Virology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (J.B.); (N.C.B.)
| | - Sandra Pinkert
- Institute of Biochemistry, Berlin Institute of Health (BIH) and Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (A.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Side, 10115 Berlin, Germany
| | - Maria Schmidt
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany; (M.S.); (H.B.)
| | - Hans Binder
- Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107 Leipzig, Germany; (M.S.); (H.B.)
| | - Nicole Christin Bilz
- Institute of Medical Microbiology and Virology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (J.B.); (N.C.B.)
| | - Matthias Jung
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Martin Luther University Halle Wittenberg, Julius-Kuehn-Strasse 7, 06112 Halle (Saale), Germany; (M.J.); (D.R.)
| | - Uta Reibetanz
- Institute for Medical Physics and Biophysics, Medical Faculty, University of Leipzig, Härtelstrasse 16-18, 04107 Leipzig, Germany;
| | - Antje Beling
- Institute of Biochemistry, Berlin Institute of Health (BIH) and Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (S.P.); (A.B.)
- DZHK (German Centre for Cardiovascular Research), Partner Side, 10115 Berlin, Germany
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Martin Luther University Halle Wittenberg, Julius-Kuehn-Strasse 7, 06112 Halle (Saale), Germany; (M.J.); (D.R.)
| | - Claudia Claus
- Institute of Medical Microbiology and Virology, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (J.B.); (N.C.B.)
| |
Collapse
|
174
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
175
|
Persson BD, John L, Rafie K, Strebl M, Frängsmyr L, Ballmann MZ, Mindler K, Havenga M, Lemckert A, Stehle T, Carlson LA, Arnberg N. Human species D adenovirus hexon capsid protein mediates cell entry through a direct interaction with CD46. Proc Natl Acad Sci U S A 2021; 118:e2020732118. [PMID: 33384338 PMCID: PMC7826407 DOI: 10.1073/pnas.2020732118] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Human adenovirus species D (HAdV-D) types are currently being explored as vaccine vectors for coronavirus disease 2019 (COVID-19) and other severe infectious diseases. The efficacy of such vector-based vaccines depends on functional interactions with receptors on host cells. Adenoviruses of different species are assumed to enter host cells mainly by interactions between the knob domain of the protruding fiber capsid protein and cellular receptors. Using a cell-based receptor-screening assay, we identified CD46 as a receptor for HAdV-D56. The function of CD46 was validated in infection experiments using cells lacking and overexpressing CD46, and by competition infection experiments using soluble CD46. Remarkably, unlike HAdV-B types that engage CD46 through interactions with the knob domain of the fiber protein, HAdV-D types infect host cells through a direct interaction between CD46 and the hexon protein. Soluble hexon proteins (but not fiber knob) inhibited HAdV-D56 infection, and surface plasmon analyses demonstrated that CD46 binds to HAdV-D hexon (but not fiber knob) proteins. Cryoelectron microscopy analysis of the HAdV-D56 virion-CD46 complex confirmed the interaction and showed that CD46 binds to the central cavity of hexon trimers. Finally, soluble CD46 inhibited infection by 16 out of 17 investigated HAdV-D types, suggesting that CD46 is an important receptor for a large group of adenoviruses. In conclusion, this study identifies a noncanonical entry mechanism used by human adenoviruses, which adds to the knowledge of adenovirus biology and can also be useful for development of adenovirus-based vaccine vectors.
Collapse
Affiliation(s)
- B David Persson
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| | - Lijo John
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| | - Karim Rafie
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-90187 Umeå, Sweden
- Department of Medical Biochemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Michael Strebl
- Interfaculty Institute of Biochemistry, The University of Tübingen, D-72076 Tübingen, Germany
| | - Lars Frängsmyr
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| | | | - Katja Mindler
- Interfaculty Institute of Biochemistry, The University of Tübingen, D-72076 Tübingen, Germany
| | - Menzo Havenga
- Batavia Biosciences, 2333 CL Leiden, The Netherlands
| | | | - Thilo Stehle
- Interfaculty Institute of Biochemistry, The University of Tübingen, D-72076 Tübingen, Germany
| | - Lars-Anders Carlson
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-90187 Umeå, Sweden
- Department of Medical Biochemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Niklas Arnberg
- Department of Clinical Microbiology, Division of Virology, Umeå University, SE-90185 Umeå, Sweden;
- Laboratory for Molecular Infection Medicine Sweden, Umeå University, SE-90185 Umeå, Sweden
| |
Collapse
|
176
|
Tsang JOL, Zhou J, Zhao X, Li C, Zou Z, Yin F, Yuan S, Yeung ML, Chu H, Chan JFW. Development of Three-Dimensional Human Intestinal Organoids as a Physiologically Relevant Model for Characterizing the Viral Replication Kinetics and Antiviral Susceptibility of Enteroviruses. Biomedicines 2021; 9:88. [PMID: 33477611 PMCID: PMC7831294 DOI: 10.3390/biomedicines9010088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/16/2022] Open
Abstract
Enteroviruses are important causes of hand, foot, and mouth disease, respiratory infections, and neurological infections in human. A major hurdle for the development of anti-enterovirus agents is the lack of physiologically relevant evaluation platforms that closely correlate with the in vivo state. We established the human small intestinal organoids as a novel platform for characterizing the viral replication kinetics and evaluating candidate antivirals for enteroviruses. The organoids supported productive replication of enterovirus (EV)-A71, coxsackievirus B2, and poliovirus type 3, as evidenced by increasing viral loads, infectious virus titers, and the presence of cytopathic effects. In contrast, EV-D68, which mainly causes respiratory tract infection in humans, did not replicate significantly in the organoids. The differential expression profiles of the receptors for these enteroviruses correlated with their replication kinetics. Using itraconazole as control, we showed that the results of various antiviral assays, including viral load reduction, plaque reduction, and cytopathic effect inhibition assays, were highly reproducible in the organoids. Moreover, itraconazole attenuated virus-induced inflammatory response in the organoids, which helped to explain its antiviral effects and mechanism. Collectively, these data showed that the human small intestinal organoids may serve as a robust platform for investigating the pathogenesis and evaluating antivirals for enteroviruses.
Collapse
Affiliation(s)
- Jessica Oi-Ling Tsang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Xiaoyu Zhao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Cun Li
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Zijiao Zou
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Feifei Yin
- Hainan-Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China;
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Man-Lung Yeung
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (J.O.-L.T.); (J.Z.); (X.Z.); (C.L.); (Z.Z.); (S.Y.); (M.-L.Y.); (H.C.)
- Hainan-Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China;
| |
Collapse
|
177
|
Lin CH, Chang YC, Chang TK, Huang CH, Lu YC, Huang CH, Chen MJ. Enhanced expression of coxsackievirus and adenovirus receptor in lipopolysaccharide-induced inflammatory macrophages is through TRIF-dependent innate immunity pathway. Life Sci 2021; 265:118832. [PMID: 33259866 DOI: 10.1016/j.lfs.2020.118832] [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: 07/09/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
AIMS Inflammatory macrophages have been proposed as a therapeutic target for joint disorders caused by inflammation. This study aimed to investigate the expression and regulation of coxsackievirus-adenovirus receptor (CAR) in lipopolysaccharide (LPS)-stimulated inflammatory macrophages whereby to evaluate the feasibility of virus-directed enzyme prodrug therapy (VDEPT). MAIN METHODS Macrophage cell lines (RAW264.7 and J774A.1) and primary macrophage cells derived from rat spleen were used to evaluate the expression of CAR protein or CAR mRNA. Specific inhibitors for TLR4 pathway were used to investigate the regulation of CAR expression. CAR expression in rat joints was documented by immunohistochemistry. Conditionally replicating adenovirus, CRAd-EGFP(PS1217L) or CRAd-NTR(PS1217H6), and non-replicating adenovirus CTL102 were used to transduce genes for enhanced green fluorescent protein (EGFP) or nitroreductase (NTR), respectively. The expression of EGFP, NTR, and the toxicity induced by CB1954 activation were evaluated. KEY FINDINGS The in vitro experiments revealed that CAR upregulation was mediated through the TLR4/TRIF/IRF3 pathway in LPS-stimulated inflammatory macrophage RAW264.7 and J774A.1 cells. The inflammatory RAW264.7 cells upregulated CAR expression following LPS stimulation, leading to higher infectability, increased NTR expression, and enhanced sensitization to CB1954. In animal experiments, the induction of CAR expression was observed in the CD68-expressing primary macrophages and in the CD68-expressing macrophages within joints following LPS stimulation. SIGNIFICANCE In conclusion, we report an enhanced CAR expression in inflammatory macrophages in vitro and in vivo through the immune response elicited by LPS. Thus, the TLR4/TRIF/IRF3 pathway of macrophages, when activated, could facilitate the therapeutic application of adenovirus-mediated VDEPT.
Collapse
Affiliation(s)
- Chi-Hsin Lin
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan; Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City, Taiwan
| | - Yuan-Ching Chang
- Department of Surgery, MacKay Memorial Hospital, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Ting-Kuo Chang
- Department of Surgery, MacKay Memorial Hospital, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Department of Orthopedics, MacKay Memorial Hospital, New Taipei City, Taiwan
| | - Chang-Hung Huang
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan; Department of Surgery, MacKay Memorial Hospital, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Yung-Chang Lu
- Department of Surgery, MacKay Memorial Hospital, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Department of Orthopedics, MacKay Memorial Hospital, New Taipei City, Taiwan
| | - Chun-Hsiung Huang
- Department of Surgery, MacKay Memorial Hospital, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Department of Orthopedics, MacKay Memorial Hospital, New Taipei City, Taiwan
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.
| |
Collapse
|
178
|
Dumitrescu M, Vacaru AM, Trusca VG, Fenyo IM, Ionita R, Gafencu AV. K2 Transfection System Boosts the Adenoviral Transduction of Murine Mesenchymal Stromal Cells. Int J Mol Sci 2021; 22:E598. [PMID: 33435318 PMCID: PMC7826527 DOI: 10.3390/ijms22020598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Adenoviral vectors are important vehicles for delivering therapeutic genes into mammalian cells. However, the yield of the adenoviral transduction of murine mesenchymal stromal cells (MSC) is low. Here, we aimed to improve the adenoviral transduction efficiency of bone marrow-derived MSC. Our data showed that among all the potential transduction boosters that we tested, the K2 Transfection System (K2TS) greatly increased the transduction efficiency. After optimization of both K2TS components, the yield of the adenoviral transduction increased from 18% to 96% for non-obese diabetic (NOD)-derived MSC, from 30% to 86% for C57BL/6-derived MSC, and from 0.6% to 63% for BALB/c-derived MSC, when 250 transduction units/cell were used. We found that MSC derived from these mouse strains expressed different levels of the coxsackievirus and adenovirus receptors (MSC from C57BL/6≥NOD>>>BALB/c). K2TS did not increase the level of the receptor expression, but desensitized the cells to foreign DNA and facilitated the virus entry into the cell. The expression of Stem cells antigen-1 (Sca-1) and 5'-nucleotidase (CD73) MSC markers, the adipogenic and osteogenic differentiation potential, and the immunosuppressive capacity were preserved after the adenoviral transduction of MSC in the presence of the K2TS. In conclusion, K2TS significantly enhanced the adenoviral transduction of MSC, without interfering with their main characteristics and properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Anca Violeta Gafencu
- Institute of Cellular Biology and Pathology “N. Simionescu”, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (M.D.); (A.M.V.); (V.G.T.); (I.M.F.); (R.I.)
| |
Collapse
|
179
|
Ono J, Okada Y, Kanri Y, Sano H, Hasegawa H. Immunohistochemical Study of Differential Expressions of CAR, E-Cadherin, CK-13, -17, p53 and Ki-67 in Oral Lichen Planus, Lichenoid Lesion and Lichenoid Epithelial Dysplasia. J HARD TISSUE BIOL 2021. [DOI: 10.2485/jhtb.30.355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Junya Ono
- Department of Pathology, The Nippon Dental University School of Life Dentistry at Niigata
| | - Yasuo Okada
- Department of Pathology, The Nippon Dental University School of Life Dentistry at Niigata
| | - Yoriaki Kanri
- Department of Pathology, The Nippon Dental University School of Life Dentistry at Niigata
| | - Hiroto Sano
- Department of Pathology, The Nippon Dental University School of Life Dentistry at Niigata
| | - Hitoshi Hasegawa
- Department of Pathology, The Nippon Dental University School of Life Dentistry at Niigata
| |
Collapse
|
180
|
Sharma V, Perry DJ, Eghtesady P. Role of coxsackie-adenovirus receptor in cardiac development and pathogenesis of congenital heart disease. Birth Defects Res 2020; 113:535-545. [PMID: 33369284 DOI: 10.1002/bdr2.1860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/30/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
The coxsackie-adenovirus receptor (CAR) is a cell surface transmembrane protein originally recognized for its role as a binding site for coxsackie- and adeno-viruses. As such, it is believed to play an important role in pathogenesis of myocarditis. Other studies have suggested that CAR also plays an important role in embryonic development, which is not surprising given the strong expression of the receptor in heart, brain, liver, pancreas, kidney, small intestine, and various epithelia during development. A number of studies have looked at downregulation and upregulation of CAR and have confirmed the central role of CAR during critical periods of development. These studies all demonstrated embryonic lethality with variable phenotypes: electrophysiological abnormalities, cardiac structural deformations, and extracardiac abnormalities, such as lymphatic malformations. The purpose of this review is to summarize the existing literature about CAR and formulate some questions for future studies, with an emphasis on the role of CAR during embryonic heart development.
Collapse
Affiliation(s)
- Vipul Sharma
- Division of Pediatric Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel J Perry
- Division of Pediatric Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pirooz Eghtesady
- Division of Pediatric Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
181
|
E3 ubiquitin ligase Mindbomb 1 facilitates nuclear delivery of adenovirus genomes. Proc Natl Acad Sci U S A 2020; 118:2015794118. [PMID: 33443154 DOI: 10.1073/pnas.2015794118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The journey from plasma membrane to nuclear pore is a critical step in the lifecycle of DNA viruses, many of which must successfully deposit their genomes into the nucleus for replication. Viral capsids navigate this vast distance through the coordinated hijacking of a number of cellular host factors, many of which remain unknown. We performed a gene-trap screen in haploid cells to identify host factors for adenovirus (AdV), a DNA virus that can cause severe respiratory illness in immune-compromised individuals. This work identified Mindbomb 1 (MIB1), an E3 ubiquitin ligase involved in neurodevelopment, as critical for AdV infectivity. In the absence of MIB1, we observed that viral capsids successfully traffic to the proximity of the nucleus but ultimately fail to deposit their genomes within. The capacity of MIB1 to promote AdV infection was dependent on its ubiquitination activity, suggesting that MIB1 may mediate proteasomal degradation of one or more negative regulators of AdV infection. Employing complementary proteomic approaches to characterize proteins proximal to MIB1 upon AdV infection and differentially ubiquitinated in the presence or absence of MIB1, we observed an intersection between MIB1 and ribonucleoproteins (RNPs) largely unexplored in mammalian cells. This work uncovers yet another way that viruses utilize host cell machinery for their own replication, highlighting a potential target for therapeutic interventions that counter AdV infection.
Collapse
|
182
|
Zhou YC, Zhang YN, Yang X, Wang SB, Hu PY. Delivery systems for enhancing oncolytic adenoviruses efficacy. Int J Pharm 2020; 591:119971. [PMID: 33059014 DOI: 10.1016/j.ijpharm.2020.119971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/24/2022]
Abstract
Oncolytic adenovirus (OAds) has long been considered a promising biotherapeutic agent against various types of cancer owing to selectively replicate in and lyse cancer cells, while remaining dormant in healthy cells. In the last years, multiple (pre)clinical studies using genetic engineering technologies enhanced OAds anti-tumor effects in a broad range of cancers. However, poor targeting delivery, tropism toward healthy tissues, low-level expression of Ad receptors on tumor cells, and pre-existing neutralizing antibodies are major hurdles for systemic administration of OAds. Different vehicles have been developed for addressing these obstacles, such as stem cells, nanoparticles (NPs) and shielding polymers, extracellular vesicles (EVs), hydrogels, and microparticles (MPs). These carriers can enhance the therapeutic efficacy of OVs through enhancing transfection, circulatory longevity, cellular interactions, specific targeting, and immune responses against cancer. In this paper, we reviewed adenovirus structure and biology, different types of OAds, and the efficacy of different carriers in systemic administration of OAds.
Collapse
Affiliation(s)
- Yu-Cheng Zhou
- Gastroenterological & Pancreatic Surgery Department, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China; Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - You-Ni Zhang
- Clinical Laboratory, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Taizhou 317200, Zhejiang Province, China
| | - Xue Yang
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China.
| | - Pei-Yang Hu
- Department of Traumatology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Taizhou 317200, Zhejiang Province, China.
| |
Collapse
|
183
|
Khosa S, Bravo Araya M, Griebel P, Arsic N, Tikoo SK. Bovine Adenovirus-3 Tropism for Bovine Leukocyte Sub-Populations. Viruses 2020; 12:E1431. [PMID: 33322850 PMCID: PMC7763465 DOI: 10.3390/v12121431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 11/20/2022] Open
Abstract
A number of characteristics including lack of virulence and the ability to grow to high titers, have made bovine adenovirus-3 (BAdV-3) a vector of choice for further development as a vaccine-delivery vehicle for cattle. Despite the importance of blood leukocytes, including dendritic cells (DC), in the induction of protective immune responses, little is known about the interaction between BAdV-3 and bovine blood leukocytes. Here, we demonstrate that compared to other leukocytes, bovine blood monocytes and neutrophils are significantly transduced by BAdV404a (BAdV-3, expressing enhanced yellow green fluorescent protein [EYFP]) at a MOI of 1-5 without a significant difference in the mean fluorescence of EYFP expression. Moreover, though expression of some BAdV-3-specific proteins was observed, no progeny virions were detected in the transduced monocytes or neutrophils. Interestingly, addition of the "RGD" motif at the C-terminus of BAdV-3 minor capsid protein pIX (BAV888) enhanced the ability of the virus to enter the monocytes without altering the tropism of BAdV-3. The increased uptake of BAV888 by monocytes was associated with a significant increase in viral genome copies and the abundance of EYFP and BAdV-3 19K transcripts compared to BAdV404a-transduced monocytes. Our results suggest that BAdV-3 efficiently transduces monocytes and neutrophils in the absence of viral replication. Moreover, RGD-modified capsid significantly increases vector uptake without affecting the initial interaction with monocytes.
Collapse
Affiliation(s)
- Sugandhika Khosa
- VIDO-InterVac., 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (S.K.); (M.B.A.); (P.G.); (N.A.)
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Maria Bravo Araya
- VIDO-InterVac., 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (S.K.); (M.B.A.); (P.G.); (N.A.)
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Philip Griebel
- VIDO-InterVac., 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (S.K.); (M.B.A.); (P.G.); (N.A.)
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Natasa Arsic
- VIDO-InterVac., 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (S.K.); (M.B.A.); (P.G.); (N.A.)
| | - Suresh K. Tikoo
- VIDO-InterVac., 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; (S.K.); (M.B.A.); (P.G.); (N.A.)
- Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| |
Collapse
|
184
|
Kim J, Beidler P, Wang H, Li C, Quassab A, Coles C, Drescher C, Carter D, Lieber A. Desmoglein-2 as a prognostic and biomarker in ovarian cancer. Cancer Biol Ther 2020; 21:1154-1162. [PMID: 33218274 PMCID: PMC7722792 DOI: 10.1080/15384047.2020.1843323] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/31/2023] Open
Abstract
Greater than 80% of all cancer cases are carcinomas, formed by the malignant transformation of epithelial cells. One of the key features of epithelial tumors is the presence of intercellular junctions, which link cells to one another and act as barriers to the penetration of molecules. This study assessed the expression of desmoglein-2, an epithelial junction protein, as a prognostic and diagnostic biomarker for ovarian cancer. Ovarian cancer sections were stained for DSG2 and signal intensity was correlated to cancer type and grade. DSG2 immunohistochemistry signals and mRNA levels were analyzed in chemo-resistant and chemo-sensitive cases. Ovarian cancer patient serum levels of shed DSG2 were correlated to disease-free and overall survival. Primary ovarian cancer cells were used to study DSG2 levels as they changed in response to cisplatin treatment. DSG2 expression was found to be positively correlated with cancer grade. Ovarian cancer patients with high serum levels of shed DSG2 fared significantly worse in both progression-free survival (median survival of 16 months vs. 26 months, p = .0023) and general survival (median survival of 37 months vs. undefined, p < .0001). A subgroup of primary chemotherapy-resistant cases had stronger DSG2 IHC/Western signals and higher DSG2 mRNA levels. Furthermore, our in vitro studies indicate that non-cytotoxic doses of cisplatin can enhance DSG2 expression, which, in turn, can contribute to chemo-resistance. We suggest that DSG2 can be used in stratifying patients, deciding on where to use aggressive treatment strategies, predicting chemoresistance, and as a companion diagnostic for treatments targeting DSG2.
Collapse
Affiliation(s)
- Jiho Kim
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
- R&D Department, PAI Life Sciences Inc, Seattle, Washington, USA
| | - Peter Beidler
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Hongjie Wang
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Chang Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Abdullah Quassab
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Cari Coles
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Charles Drescher
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Darrick Carter
- R&D Department, PAI Life Sciences Inc, Seattle, Washington, USA
- R&D Department, Onc Bio, Seattle, Washington, USA
| | - André Lieber
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
185
|
He L, Yu A, Deng L, Zhang H. Eradicating the Roots: Advanced Therapeutic Approaches Targeting Breast Cancer Stem Cells. Curr Pharm Des 2020; 26:2009-2021. [PMID: 32183663 DOI: 10.2174/1381612826666200317132949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/08/2020] [Indexed: 12/30/2022]
Abstract
Accumulating evidences have demonstrated that the existence of breast cancer-initiating cells, which drives the original tumorigenicity, local invasion and migration propensity of breast cancer. These cells, termed as breast cancer stem cells (BCSCs), possess properties including self-renewal, multidirectional differentiation and proliferative potential, and are believed to play important roles in the intrinsic drug resistance of breast cancer. One of the reasons why BCBCs cause difficulties in breast cancer treating is that BCBCs can control both genetic and non-genetic elements to keep their niches safe and sound, which allows BCSCs for constant self-renewal and differentiation. Therapeutic strategies designed to target BCSCs may ultimately result in effective interventions for the treatment of breast cancer. Novel strategies including nanomedicine, oncolytic virus therapy, immunotherapy and induced differentiation therapy are emerging and proved to be efficient in anti-BCSCs therapy. In this review, we summarized breast tumor biology and the current challenges of breast cancer therapies, focused on breast cancer stem cells, and introduced promising therapeutic strategies targeting BCSCs.
Collapse
Affiliation(s)
- Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Anran Yu
- The State University of New York, Buffalo, NY 12246, United States
| | - Li Deng
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hongwei Zhang
- School of Pharmacy, MCPHS University, Boston, MA 02115, United States
| |
Collapse
|
186
|
Honkimaa A, Kimura B, Sioofy-Khojine AB, Lin J, Laiho J, Oikarinen S, Hyöty H. Genetic Adaptation of Coxsackievirus B1 during Persistent Infection in Pancreatic Cells. Microorganisms 2020; 8:microorganisms8111790. [PMID: 33203081 PMCID: PMC7697981 DOI: 10.3390/microorganisms8111790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022] Open
Abstract
Coxsackie B (CVB) viruses have been associated with type 1 diabetes. We have recently observed that CVB1 was linked to the initiation of the autoimmune process leading to type 1 diabetes in Finnish children. Viral persistency in the pancreas is currently considered as one possible mechanism. In the current study persistent infection was established in pancreatic ductal and beta cell lines (PANC-1 and 1.1B4) using four different CVB1 strains, including the prototype strain and three clinical isolates. We sequenced 5′ untranslated region (UTR) and regions coding for structural and non-structural proteins and the second single open reading frame (ORF) protein of all persisting CVB1 strains using next generation sequencing to identify mutations that are common for all of these strains. One mutation, K257R in VP1, was found from all persisting CVB1 strains. The mutations were mainly accumulated in viral structural proteins, especially at BC, DE, EF loops and C-terminus of viral capsid protein 1 (VP1), the puff region of VP2, the knob region of VP3 and infection-enhancing epitope of VP4. This showed that the capsid region of the viruses sustains various changes during persistency some of which could be hallmark(s) of persistency.
Collapse
Affiliation(s)
- Anni Honkimaa
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
- Correspondence:
| | - Bryn Kimura
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Amir-Babak Sioofy-Khojine
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Jake Lin
- Finnish Institute of Molecular Medicine (FIMM), University of Helsinki, 00290 Helsinki, Finland;
| | - Jutta Laiho
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
- Fimlab Laboratories, Pirkanmaa Hospital District, 33520 Tampere, Finland
| |
Collapse
|
187
|
Cunliffe TG, Bates EA, Parker AL. Hitting the Target but Missing the Point: Recent Progress towards Adenovirus-Based Precision Virotherapies. Cancers (Basel) 2020; 12:E3327. [PMID: 33187160 PMCID: PMC7696810 DOI: 10.3390/cancers12113327] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 10/31/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
More people are surviving longer with cancer. Whilst this can be partially attributed to advances in early detection of cancers, there is little doubt that the improvement in survival statistics is also due to the expansion in the spectrum of treatments available for efficacious treatment. Transformative amongst those are immunotherapies, which have proven effective agents for treating immunogenic forms of cancer, although immunologically "cold" tumour types remain refractive. Oncolytic viruses, such as those based on adenovirus, have great potential as anti-cancer agents and have seen a resurgence of interest in recent years. Amongst their many advantages is their ability to induce immunogenic cell death (ICD) of infected tumour cells, thus providing the alluring potential to synergise with immunotherapies by turning immunologically "cold" tumours "hot". Additionally, enhanced immune mediated cell killing can be promoted through the local overexpression of immunological transgenes, encoded from within the engineered viral genome. To achieve this full potential requires the development of refined, tumour selective "precision virotherapies" that are extensively engineered to prevent off-target up take via native routes of infection and targeted to infect and replicate uniquely within malignantly transformed cells. Here, we review the latest advances towards this holy grail within the adenoviral field.
Collapse
Affiliation(s)
| | | | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK; (T.G.C.); (E.A.B.)
| |
Collapse
|
188
|
Rathjen FG. The CAR group of Ig cell adhesion proteins–Regulators of gap junctions? Bioessays 2020; 42:e2000031. [DOI: 10.1002/bies.202000031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/01/2020] [Indexed: 12/29/2022]
|
189
|
Generation of Genetically RGD σ1-Modified Oncolytic Reovirus That Enhances JAM-A-Independent Infection of Tumor Cells. J Virol 2020; 94:JVI.01703-20. [PMID: 32907973 DOI: 10.1128/jvi.01703-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/14/2022] Open
Abstract
Mammalian reovirus (MRV) strain type 3 Dearing (T3D) is a naturally occurring oncolytic virus that has been developed as a potential cancer therapeutic. However, MRV treatment cannot be applied to cancer cells expressing low levels of junctional adhesion molecule A (JAM-A), which is the entry receptor of MRV. In this study, we developed a reverse genetics system for MRV strain T3D-L, which showed high oncolytic potency. To modify the cell tropism of MRV, an arginine-glycine-aspartic acid (RGD) peptide with an affinity to integrin was inserted at the C terminus or loop structures of the viral cell attachment protein σ1. The recombinant RGD σ1-modified viruses induced remarkable cell lysis in human cancer cell lines with marginal JAM-A expression and in JAM-A knockout cancer cell lines generated by a CRISPR/Cas9 system. Pretreatment of cells with anti-integrin antibody decreased cell death caused by the RGD σ1-modified virus, suggesting the infection to the cells was via a specific interaction with integrin αV. By using mouse models, we assessed virulence of the RGD σ1-modified viruses in vivo This system will open new avenues for the use of genetically modified oncolytic MRV for use as a cancer therapy.IMPORTANCE Oncolytic viruses kill tumors without affecting normal cells. A variety of oncolytic viruses are used as cancer therapeutics. Mammalian reovirus (MRV), which belongs to the genus Orthoreovirus, family Reoviridae, is one such natural oncolytic virus. The anticancer effects of MRV are being evaluated in clinical trials. Unlike other oncolytic viruses, MRV has not been genetically modified for use as a cancer therapeutic in clinical trials. Here, we used a reverse genetic approach to introduce an integrin-affinity peptide sequence into the MRV cell attachment protein σ1 to alter the natural tropism of the virus. The recombinant viruses were able to infect cancer cell lines expressing very low levels of the MRV entry receptor, junctional adhesion molecule A (JAM-A), and cause tumor cell death while maintaining its original tropism via JAM-A. This is a novel report of a genetically modified oncolytic MRV by introducing a peptide sequence into σ1.
Collapse
|
190
|
Koike S. The risk of unintentional propagation of poliovirus can be minimized by using human cell lines lacking the functional CD155 gene. Microbiol Immunol 2020; 64:835-839. [PMID: 32902876 DOI: 10.1111/1348-0421.12843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/30/2020] [Accepted: 09/06/2020] [Indexed: 11/29/2022]
Abstract
After eradication and containment of wild poliovirus (PV) and cessation of oral polio vaccinations, it is critical to minimize the risk of reintroducing PV into PV-free communities via facilities that handle the virus. The potential risk of unintentional PV propagation through unidentified contaminated materials is a serious issue. This study reports the generation of HeLa and RD-A cells deficient in functional CD155 gene (∆PVR cells); these cells are not susceptible to PV but remain susceptible to other picornaviruses. These ∆PVR cells will minimize the risk of unintentional transmission of PV and support performing the experiments more safely.
Collapse
Affiliation(s)
- Satoshi Koike
- Neurovirology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
191
|
Mochizuki Y, Tazawa H, Demiya K, Kure M, Kondo H, Komatsubara T, Sugiu K, Hasei J, Yoshida A, Kunisada T, Urata Y, Kagawa S, Ozaki T, Fujiwara T. Telomerase-specific oncolytic immunotherapy for promoting efficacy of PD-1 blockade in osteosarcoma. Cancer Immunol Immunother 2020; 70:1405-1417. [PMID: 33151368 DOI: 10.1007/s00262-020-02774-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/19/2020] [Indexed: 12/20/2022]
Abstract
Immune checkpoint inhibitors including anti-programmed cell death 1 (PD-1) antibody have recently improved clinical outcome in certain cancer patients; however, osteosarcoma (OS) patients are refractory to PD-1 blockade. Oncolytic virotherapy has emerged as novel immunogenic therapy to augment antitumor immune response. We developed a telomerase-specific replication-competent oncolytic adenovirus OBP-502 that induces lytic cell death via binding to integrins. In this study, we assessed the combined effect of PD-1 blockade and OBP-502 in OS cells. The expression of coxsackie and adenovirus receptor (CAR), integrins αvβ3 and αvβ5, and programmed cell death ligand 1 (PD-L1) was analyzed in two murine OS cells (K7M2, NHOS). The cytopathic activity of OBP-502 in both cells was analyzed using the XTT assay. OBP-502-induced immunogenic cell death was assessed by analyzing the level of extracellular ATP and high-mobility group box protein B1 (HMGB1). Subcutaneous tumor models for K7M2 and NHOS cells were used to evaluate the antitumor effect and number of tumor-infiltrating CD8+ cells in combination therapy. K7M2 and NHOS cells showed high expression of integrins αvβ3 and αvβ5, but not CAR. OBP-502 significantly suppressed the viability of both cells, in which PD-L1 expression and the release of ATP and HMGB1 were significantly increased. Intratumoral injection of OBP-502 significantly augmented the efficacy of PD-1 blockade on subcutaneous K2M2 and NHOS tumor models via enhancement of tumor-infiltrating CD8+ T cells. Our results suggest that telomerase-specific oncolytic virotherapy is a promising antitumor strategy to promote the efficacy of PD-1 blockade in OS.
Collapse
Affiliation(s)
- Yusuke Mochizuki
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Hiroshi Tazawa
- Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan. .,Center for Innovative Clinical Medicine, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Koji Demiya
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Miho Kure
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Hiroya Kondo
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tadashi Komatsubara
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Kazuhisa Sugiu
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Joe Hasei
- Sports Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, 700-8558, Japan
| | - Aki Yoshida
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshiyuki Kunisada
- Medical Materials for Musculoskeletal Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc, Tokyo, 105-0001, Japan
| | - Shunsuke Kagawa
- Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.,Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, 700-8558, Japan
| | - Toshifumi Ozaki
- Departments of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| |
Collapse
|
192
|
Clinical and Preclinical Systematic Review of Astragalus Membranaceus for Viral Myocarditis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1560353. [PMID: 33204391 PMCID: PMC7652609 DOI: 10.1155/2020/1560353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/20/2020] [Indexed: 12/09/2022]
Abstract
Astragalus membranaceus (AM) is a traditional Chinese medicine, which possesses a variety of biological activities in the cardiovascular systems. We conducted a clinical and preclinical systematic review of 28 randomized clinical control studies with 2522 participants and 16 animal studies with 634 animals to evaluate the efficacy, safety, and possible mechanisms of AM for viral myocarditis (VM). The search strategies were performed in 7 databases from inception to January 2020. Application of the Cochrane Collaboration's tool 7-item checklist, SYRCLE's tool 10-item checklist, and Rev-Man 5.3 software to analyze the risk of bias of studies and data. The results show the score of clinical study quality ranged from 3 to 7 points with an average of 3.32, and the score of animal study quality ranged from 2 to 5 points with an average of 3. In clinical study, AM significantly reduced serum myocardial enzymes and cardiac troponin I levels and improved the clinical treatment efficiency in VM patients compared with the control group (P < 0.05). There was no significant difference in the incidence of adverse reactions (P > 0.05). Significant increase of the survival rate and decrease of the cardiac cardiology score, cardiac enzymes, and cardiac troponin I were compared with the placebo group in animal studies (P < 0.05). The possible mechanisms of AM are largely through antivirus and antivirus receptors, anti-inflammatory, antioxidation, antiapoptotic, antifibrosis, and reducing cardiac calcium load. In conclusion, the findings suggested that AM is a cardioprotection candidate drug for VM.
Collapse
|
193
|
Hulin-Curtis SL, Davies JA, Nestić D, Bates EA, Baker AT, Cunliffe TG, Majhen D, Chester JD, Parker AL. Identification of folate receptor α (FRα) binding oligopeptides and their evaluation for targeted virotherapy applications. Cancer Gene Ther 2020; 27:785-798. [PMID: 31902944 PMCID: PMC7661341 DOI: 10.1038/s41417-019-0156-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
Oncolytic virotherapies (OV) based on human adenoviral (HAdV) vectors hold significant promise for the treatment of advanced ovarian cancers where local, intraperitoneal delivery to tumour metastases is feasible, bypassing many complexities associated with intravascular delivery. The efficacy of HAdV-C5-based OV is hampered by a lack of tumour selectivity, where the primary receptor, hCAR, is commonly downregulated during malignant transformation. Conversely, folate receptor alpha (FRα) is highly expressed on ovarian cancer cells, providing a compelling target for tumour selective delivery of virotherapies. Here, we identify high-affinity FRα-binding oligopeptides for genetic incorporation into HAdV-C5 vectors. Biopanning identified a 12-mer linear peptide, DWSSWVYRDPQT, and two 7-mer cysteine-constrained peptides, CIGNSNTLC and CTVRTSAEC that bound FRα in the context of the phage particle. Synthesised lead peptide, CTVRTSAEC, bound specifically to FRα and could be competitively inhibited with folic acid. To assess the capacity of the elucidated FRα-binding oligopeptides to target OV to FRα, we genetically incorporated the peptides into the HAdV-C5 fiber-knob HI loop including in vectors genetically ablated for hCAR interactions. Unfortunately, the recombinant vectors failed to efficiently target transduction via FRα due to defective intracellular trafficking following entry via FRα, indicating that whilst the peptides identified may have potential for applications for targeted drug delivery, they require additional refinement for targeted virotherapy applications.
Collapse
Affiliation(s)
- Sarah L Hulin-Curtis
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - James A Davies
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Davor Nestić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Emily A Bates
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Alexander T Baker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Tabitha G Cunliffe
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - John D Chester
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
- Velindre Cancer Centre, Whitchurch, Cardiff, CF14 2TL, UK
| | - Alan L Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
194
|
Huang N, Perez P, Kato T, Mikami Y, Okuda K, Gilmore RC, Domínguez Conde C, Gasmi B, Stein S, Beach M, Pelayo E, Maldonado J, LaFont B, Padilla R, Murrah V, Maile R, Lovell W, Wallet S, Bowman NM, Meinig SL, Wolfgang MC, Choudhury SN, Novotny M, Aevermann BD, Scheuermann R, Cannon G, Anderson C, Marchesan J, Bush M, Freire M, Kimple A, Herr DL, Rabin J, Grazioli A, French BN, Pranzatelli T, Chiorini JA, Kleiner DE, Pittaluga S, Hewitt S, Burbelo PD, Chertow D, Frank K, Lee J, Boucher RC, Teichmann SA, Warner BM, Byrd KM. Integrated Single-Cell Atlases Reveal an Oral SARS-CoV-2 Infection and Transmission Axis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 33140061 DOI: 10.1101/2020.10.26.20219089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite signs of infection, the involvement of the oral cavity in COVID-19 is poorly understood. To address this, single-cell RNA sequencing data-sets were integrated from human minor salivary glands and gingiva to identify 11 epithelial, 7 mesenchymal, and 15 immune cell clusters. Analysis of SARS-CoV-2 viral entry factor expression showed enrichment in epithelia including the ducts and acini of the salivary glands and the suprabasal cells of the mucosae. COVID-19 autopsy tissues confirmed in vivo SARS-CoV-2 infection in the salivary glands and mucosa. Saliva from SARS-CoV-2-infected individuals harbored epithelial cells exhibiting ACE2 expression and SARS-CoV-2 RNA. Matched nasopharyngeal and saliva samples found distinct viral shedding dynamics and viral burden in saliva correlated with COVID-19 symptoms including taste loss. Upon recovery, this cohort exhibited salivary antibodies against SARS-CoV-2 proteins. Collectively, the oral cavity represents a robust site for COVID-19 infection and implicates saliva in viral transmission.
Collapse
|
195
|
Hepatocytes trap and silence coxsackieviruses, protecting against systemic disease in mice. Commun Biol 2020; 3:580. [PMID: 33067530 PMCID: PMC7568585 DOI: 10.1038/s42003-020-01303-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Previous research suggests that hepatocytes catabolize chemical toxins but do not remove microbial agents, which are filtered out by other liver cells (Kupffer cells and endothelial cells). Here we show that, contrary to current understanding, hepatocytes trap and rapidly silence type B coxsackieviruses (CVBs). In genetically wildtype mice, this activity causes hepatocyte damage, which is alleviated in mice carrying a hepatocyte-specific deletion of the coxsackievirus-adenovirus receptor. However, in these mutant mice, there is a dramatic early rise in blood-borne virus, followed by accelerated systemic disease and increased mortality. Thus, wild type hepatocytes act similarly to a sponge for CVBs, protecting against systemic illness at the expense of their own survival. We speculate that hepatocytes may play a similar role in other viral infections as well, thereby explaining why hepatocytes have evolved their remarkable regenerative capacity. Our data also suggest that, in addition to their many other functions, hepatocytes might be considered an integral part of the innate immune system. Kimura, Flynn and Whitton find that hepatocytes act as a sponge to trap viruses, but that doing so damages the liver cells. They show that, when mouse hepatocytes are altered to prevent trapping of circulating virus, the mice are more likely to develop systemic disease and die, providing strong evidence for an important overlooked function of hepatocytes.
Collapse
|
196
|
Combination of rAd-p53 in situ gene therapy and anti-PD-1 antibody immunotherapy induced anti-tumor activity in mouse syngeneic urogenital cancer models. Sci Rep 2020; 10:17464. [PMID: 33060772 PMCID: PMC7562933 DOI: 10.1038/s41598-020-74660-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022] Open
Abstract
In this study we undertook a novel combination therapy using rAd-p53 in situ gene therapy and immunotherapy with immune checkpoint inhibitor (ICI) anti-PD-1 antibody for urogenital cancers. Three mouse syngeneic tumor cell lines, TRAMP-C2 (prostate cancer derived from C57BL/6 mice), MBT-2 (bladder cancer derived from C3H mice) and Renca (kidney cancer derived from BALB/c mice) were used in this study. The highest coxsackie and adenovirus receptor (CAR) mRNA expression was observed in TRAMP-C2 cells, followed by Renca and then MBT-2 cells. Consistent with the CAR expressions, rAd-p53 at 160 multiplicity of infection (MOI) significantly inhibited the cell proliferation of TRAMP-C2 and Renca cells, but not MBT-2 cells. In in vivo experiments, the combination of intratumoral injections of rAd-p53 (1 × 109 plaque-forming units) every other day and intraperitoneal injections of anti-mouse PD-1 antibody (200 μg) twice a week suppressed tumor growth and prolonged survival compared to rAd-p53 or anti-PD-1 antibody monotherapy in both the TRAMP-C2 and Renca models. Our results encourage the clinical development of combination therapy comprised of in situ gene therapy with rAd-p53 and immunotherapy with an ICI anti-PD-1 antibody for urogenital cancers.
Collapse
|
197
|
Orsay Virus CP-δ Adopts a Novel β-Bracelet Structural Fold and Incorporates into Virions as a Head Fiber. J Virol 2020; 94:JVI.01560-20. [PMID: 32817218 PMCID: PMC7565637 DOI: 10.1128/jvi.01560-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/08/2020] [Indexed: 11/20/2022] Open
Abstract
Viruses often have extended fibers to mediate host cell recognition and entry, serving as promising targets for antiviral drug development. Unlike other known viral fibers, the δ proteins from the three recently discovered nematode viruses are incorporated into infectious particles as protruding fibers covalently linked to the capsid. Crystal structures of δ revealed novel pentameric folding repeats, which we term β-bracelets, in the intermediate shaft region. Based on sequence analysis, the β-bracelet motif of δ is conserved in all three nematode viruses and could account for ∼60% of the total length of the fiber. Our study indicated that δ plays important roles in cell attachment for this group of nematode viruses. In addition, the tightly knitted β-bracelet fold, which presumably allows δ to survive harsh environments in the worm gut, could be applicable to bioengineering applications given its potentially high stability. Fiber proteins are commonly found in eukaryotic and prokaryotic viruses, where they play important roles in mediating viral attachment and host cell entry. They typically form trimeric structures and are incorporated into virions via noncovalent interactions. Orsay virus, a small RNA virus which specifically infects the laboratory model nematode Caenorhabditis elegans, encodes a fibrous protein δ that can be expressed as a free protein and as a capsid protein-δ (CP-δ) fusion protein. Free δ has previously been demonstrated to facilitate viral exit following intracellular expression; however, the biological significance and prevalence of CP-δ remained relatively unknown. Here, we demonstrate that Orsay CP-δ is covalently incorporated into infectious particles, the first example of any attached viral fibers known to date. The crystal structure of δ(1–101) (a deletion mutant containing the first 101 amino acid [aa] residues of δ) reveals a pentameric, 145-Å long fiber with an N-terminal coiled coil followed by multiple β-bracelet repeats. Electron micrographs of infectious virions depict particle-associated CP-δ fibers with dimensions similar to free δ. The δ proteins from two other nematode viruses, Le Blanc and Santeuil, which both specifically infect Caenorhabditis briggsae, were also found to form fibrous molecules. Recombinant Le Blanc δ was able to block Orsay virus infection in worm culture and vice versa, suggesting these two viruses likely compete for the same cell receptor(s). Thus, we propose that while CP-δ likely mediates host cell attachment for all three nematode viruses, additional downstream factor(s) ultimately determine the host specificity and range of each virus. IMPORTANCE Viruses often have extended fibers to mediate host cell recognition and entry, serving as promising targets for antiviral drug development. Unlike other known viral fibers, the δ proteins from the three recently discovered nematode viruses are incorporated into infectious particles as protruding fibers covalently linked to the capsid. Crystal structures of δ revealed novel pentameric folding repeats, which we term β-bracelets, in the intermediate shaft region. Based on sequence analysis, the β-bracelet motif of δ is conserved in all three nematode viruses and could account for ∼60% of the total length of the fiber. Our study indicated that δ plays important roles in cell attachment for this group of nematode viruses. In addition, the tightly knitted β-bracelet fold, which presumably allows δ to survive harsh environments in the worm gut, could be applicable to bioengineering applications given its potentially high stability.
Collapse
|
198
|
Cao QT, Aguiar JA, Tremblay BJM, Abbas N, Tiessen N, Revill S, Makhdami N, Ayoub A, Cox G, Ask K, Doxey AC, Hirota JA. ABCF1 Regulates dsDNA-induced Immune Responses in Human Airway Epithelial Cells. Front Cell Infect Microbiol 2020; 10:487. [PMID: 33042865 PMCID: PMC7525020 DOI: 10.3389/fcimb.2020.00487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 08/05/2020] [Indexed: 11/24/2022] Open
Abstract
Background: The airway epithelium represents a critical component of the human lung that helps orchestrate defenses against respiratory tract viral infections, which are responsible for more than 2.5 million deaths/year globally. Innate immune activities of the airway epithelium rely on Toll-like receptors (TLRs), nucleotide binding and leucine-rich-repeat pyrin domain containing (NLRP) receptors, and cytosolic nucleic acid sensors. ATP Binding Cassette (ABC) transporters are ubiquitous across all three domains of life—Archaea, Bacteria, and Eukarya—and expressed in the human airway epithelium. ABCF1, a unique ABC family member that lacks a transmembrane domain, has been defined as a cytosolic nucleic acid sensor that regulates CXCL10, interferon-β expression, and downstream type I interferon responses. We tested the hypothesis that ABCF1 functions as a dsDNA nucleic acid sensor in human airway epithelial cells important in regulating antiviral responses. Methods: Expression and localization experiments were performed using in situ hybridization and immunohistochemistry in human lung tissue, while confirmatory transcript and protein expression was performed in human airway epithelial cells. Functional experiments were performed with siRNA methods in a human airway epithelial cell line. Complementary transcriptomic analyses were performed to explore the contributions of ABCF1 to gene expression patterns. Results: Using archived human lung and human airway epithelial cells, we confirm expression of ABCF1 gene and protein expression in these tissue samples, with a role for mediating CXCL10 production in response to dsDNA viral mimic challenge. Although, ABCF1 knockdown was associated with an attenuation of select genes involved in the antiviral responses, Gene Ontology analyses revealed a greater interaction of ABCF1 with TLR signaling suggesting a multifactorial role for ABCF1 in innate immunity in human airway epithelial cells. Conclusion: ABCF1 is a candidate cytosolic nucleic acid sensor and modulator of TLR signaling that is expressed at gene and protein levels in human airway epithelial cells. The precise level where ABCF1 protein functions to modulate immune responses to pathogens remains to be determined but is anticipated to involve IRF-3 and CXCL10 production.
Collapse
Affiliation(s)
- Quynh T Cao
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | | | | | - Nadin Abbas
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Nicholas Tiessen
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Spencer Revill
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Nima Makhdami
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Anmar Ayoub
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Gerard Cox
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Andrew C Doxey
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada.,Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Jeremy A Hirota
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada.,Department of Biology, University of Waterloo, Waterloo, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
199
|
Surface Modification of Adenovirus Vector to Improve Immunogenicity and Tropism. Methods Mol Biol 2020. [PMID: 32959253 DOI: 10.1007/978-1-0716-0795-4_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Although adenovirus is a popular vector for delivering genes, there are several drawbacks that limit its effectiveness, including tropism and both the innate and adaptive immune responses. One approach that has been used to ameliorate these drawbacks is PEGylation of the virus with subsequent modification to add functional moieties for the purpose of cell targeting or enhancing infection. Here, we describe a general approach for PEGylating adenovirus and conjugating cell-penetrating peptides to the surface of the virus to impart the ability to transduce CAR-negative cells.
Collapse
|
200
|
Holcomb D, Alexaki A, Hernandez N, Laurie K, Kames J, Hamasaki-Katagiri N, Komar AA, DiCuccio M, Kimchi-Sarfaty C. Potential impact on coagulopathy of gene variants of coagulation related proteins that interact with SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32935103 DOI: 10.1101/2020.09.08.272328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Thrombosis has been one of the complications of the Coronavirus disease of 2019 (COVID-19), often associated with poor prognosis. There is a well-recognized link between coagulation and inflammation, however, the extent of thrombotic events associated with COVID-19 warrants further investigation. Poly(A) Binding Protein Cytoplasmic 4 (PABPC4), Serine/Cysteine Proteinase Inhibitor Clade G Member 1 (SERPING1) and Vitamin K epOxide Reductase Complex subunit 1 (VKORC1), which are all proteins linked to coagulation, have been shown to interact with SARS proteins. We computationally examined the interaction of these with SARS-CoV-2 proteins and, in the case of VKORC1, we describe its binding to ORF7a in detail. We examined the occurrence of variants of each of these proteins across populations and interrogated their potential contribution to COVID-19 severity. Potential mechanisms by which some of these variants may contribute to disease are proposed. Some of these variants are prevalent in minority groups that are disproportionally affected by severe COVID-19. Therefore, we are proposing that further investigation around these variants may lead to better understanding of disease pathogenesis in minority groups and more informed therapeutic approaches. Author summary Increased blood clotting, especially in the lungs, is a common complication of COVID-19. Infectious diseases cause inflammation which in turn can contribute to increased blood clotting. However, the extent of clot formation that is seen in the lungs of COVID-19 patients suggests that there may be a more direct link. We identified three human proteins that are involved indirectly in the blood clotting cascade and have been shown to interact with proteins of SARS virus, which is closely related to the novel coronavirus. We examined computationally the interaction of these human proteins with the viral proteins. We looked for genetic variants of these proteins and examined how these variants are distributed across populations. We investigated whether variants of these genes could impact severity of COVID-19. Further investigation around these variants may provide clues for the pathogenesis of COVID-19 particularly in minority groups.
Collapse
|