1
|
Wu EY, Robertson AM, Zhu H(H, Stasiak C, Murray-Nerger LA, Romanoff E, Woon J, Bromme BA, Smith JG. CD46 Is a Protein Receptor for Human Adenovirus Type 64. Viruses 2024; 16:1827. [PMID: 39772136 PMCID: PMC11680334 DOI: 10.3390/v16121827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Certain species D human adenoviruses (HAdV-D19, -D37, and -D64) are causative agents of epidemic keratoconjunctivitis. HAdV-D37 has previously been shown to bind CD46 (membrane cofactor protein) and sialic acid as adhesion receptors. HAdV-D64 is genetically highly similar to HAdV-D37, with an identical fiber protein sequence, but differs substantially in its penton base and hexon proteins, two other major capsid components, due to genetic recombination. Here, we demonstrate that, like HAdV-D37, HAdV-D64 virions bind directly to CD46 and that CD46 and sialic acid also function as receptors for HAdV-D64 on multiple cell types. Expression of CD46 on CD46-negative cells conferred susceptibility to HAdV-D64 entry. Specifically blocking HAdV-D64 binding to CD46 on the host cell surface strongly inhibits viral entry and gene delivery into multiple cell lines that represent target tissues. We show that CD46 is expressed on human conjunctival epithelial cells and directly binds to the HAdV-D64 virion. Our results suggest that HAdV-D64 may be used to deliver genes to target conjunctival cells and that interrupting HAdV-D64 entry through its interaction with CD46 may prevent or lessen adenovirus-associated ocular disease.
Collapse
Affiliation(s)
- Eugene Y. Wu
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Alexander M. Robertson
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Hanglin (Henry) Zhu
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Corina Stasiak
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Laura A. Murray-Nerger
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Emily Romanoff
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Jesse Woon
- Department of Biology, University of Richmond, Richmond, VA 23173, USA; (A.M.R.); (H.Z.); (E.R.)
| | - Beth A. Bromme
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA (J.G.S.)
| | - Jason G. Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA (J.G.S.)
| |
Collapse
|
2
|
Gujar S, Pol JG, Kumar V, Lizarralde-Guerrero M, Konda P, Kroemer G, Bell JC. Tutorial: design, production and testing of oncolytic viruses for cancer immunotherapy. Nat Protoc 2024; 19:2540-2570. [PMID: 38769145 DOI: 10.1038/s41596-024-00985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 02/12/2024] [Indexed: 05/22/2024]
Abstract
Oncolytic viruses (OVs) represent a novel class of cancer immunotherapy agents that preferentially infect and kill cancer cells and promote protective antitumor immunity. Furthermore, OVs can be used in combination with established or upcoming immunotherapeutic agents, especially immune checkpoint inhibitors, to efficiently target a wide range of malignancies. The development of OV-based therapy involves three major steps before clinical evaluation: design, production and preclinical testing. OVs can be designed as natural or engineered strains and subsequently selected for their ability to kill a broad spectrum of cancer cells rather than normal, healthy cells. OV selection is further influenced by multiple factors, such as the availability of a specific viral platform, cancer cell permissivity, the need for genetic engineering to render the virus non-pathogenic and/or more effective and logistical considerations around the use of OVs within the laboratory or clinical setting. Selected OVs are then produced and tested for their anticancer potential by using syngeneic, xenograft or humanized preclinical models wherein immunocompromised and immunocompetent setups are used to elucidate their direct oncolytic ability as well as indirect immunotherapeutic potential in vivo. Finally, OVs demonstrating the desired anticancer potential progress toward translation in patients with cancer. This tutorial provides guidelines for the design, production and preclinical testing of OVs, emphasizing considerations specific to OV technology that determine their clinical utility as cancer immunotherapy agents.
Collapse
Affiliation(s)
- Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Jonathan G Pol
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Vishnupriyan Kumar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Manuela Lizarralde-Guerrero
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
- Ecole Normale Supérieure de Lyon, Lyon, France
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Guido Kroemer
- INSERM, U1138, Paris, France.
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
- Université Paris Cité, Paris, France.
- Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Institut Universitaire de France, Paris, France.
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - John C Bell
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada.
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
3
|
Wallace R, Bliss CM, Parker AL. The Immune System-A Double-Edged Sword for Adenovirus-Based Therapies. Viruses 2024; 16:973. [PMID: 38932265 PMCID: PMC11209478 DOI: 10.3390/v16060973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Pathogenic adenovirus (Ad) infections are widespread but typically mild and transient, except in the immunocompromised. As vectors for gene therapy, vaccine, and oncology applications, Ad-based platforms offer advantages, including ease of genetic manipulation, scale of production, and well-established safety profiles, making them attractive tools for therapeutic development. However, the immune system often poses a significant challenge that must be overcome for adenovirus-based therapies to be truly efficacious. Both pre-existing anti-Ad immunity in the population as well as the rapid development of an immune response against engineered adenoviral vectors can have detrimental effects on the downstream impact of an adenovirus-based therapeutic. This review focuses on the different challenges posed, including pre-existing natural immunity and anti-vector immunity induced by a therapeutic, in the context of innate and adaptive immune responses. We summarise different approaches developed with the aim of tackling these problems, as well as their outcomes and potential future applications.
Collapse
Affiliation(s)
- Rebecca Wallace
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
| | - Carly M. Bliss
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Alan L. Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK; (R.W.); (C.M.B.)
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
4
|
Zhao C, Porter JM, Burke PC, Arnberg N, Smith JG. Alpha-defensin binding expands human adenovirus tropism. PLoS Pathog 2024; 20:e1012317. [PMID: 38900833 PMCID: PMC11230588 DOI: 10.1371/journal.ppat.1012317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/08/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Mammalian α-defensins are a family of abundant effector peptides of the mucosal innate immune system. Although primarily considered to be antimicrobial, α-defensins can increase rather than block infection by certain prominent bacterial and viral pathogens in cell culture and in vivo. We have shown previously that exposure of mouse and human adenoviruses (HAdVs) to α-defensins is able to overcome competitive inhibitors that block cell binding, leading us to hypothesize a defensin-mediated binding mechanism that is independent of known viral receptors. To test this hypothesis, we used genetic approaches to demonstrate that none of several primary receptors nor integrin co-receptors are needed for human α-defensin-mediated binding of HAdV to cells; however, infection remains integrin dependent. Thus, our studies have revealed a novel pathway for HAdV binding to cells that bypasses viral primary receptors. We speculate that this pathway functions in parallel with receptor-mediated entry and contributes to α-defensin-enhanced infection of susceptible cells. Remarkably, we also found that in the presence of α-defensins, HAdV tropism is expanded to non-susceptible cells, even when viruses are exposed to a mixture of both susceptible and non-susceptible cells. Therefore, we propose that in the presence of sufficient concentrations of α-defensins, such as in the lung or gut, integrin expression rather than primary receptor expression will dictate HAdV tropism in vivo. In summary, α-defensins may contribute to tissue tropism not only through the neutralization of susceptible viruses but also by allowing certain defensin-resistant viruses to bind to cells independently of previously described mechanisms.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jessica M. Porter
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Phillip C. Burke
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Niklas Arnberg
- Department of Clinical Microbiology, Division of Virology and Laboratory for Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Jason G. Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
5
|
Zhao C, Porter JM, Burke PC, Arnberg N, Smith JG. Alpha-defensin binding expands human adenovirus tropism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596681. [PMID: 38854108 PMCID: PMC11160700 DOI: 10.1101/2024.05.30.596681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Mammalian α-defensins are a family of abundant effector peptides of the mucosal innate immune system. Although primarily considered to be antimicrobial, α-defensins can increase rather than block infection by certain prominent bacterial and viral pathogens in cell culture and in vivo . We have shown previously that exposure of mouse and human adenoviruses (HAdVs) to α-defensins is able to overcome competitive inhibitors that block cell binding, leading us to hypothesize a defensin-mediated binding mechanism that is independent of known viral receptors. To test this hypothesis, we used genetic approaches to demonstrate that none of several primary receptors nor integrin co-receptors are needed for human α-defensin-mediated binding of HAdV to cells; however, infection remains integrin dependent. Thus, our studies have revealed a novel pathway for HAdV binding to cells that bypasses viral primary receptors. We speculate that this pathway functions in parallel with receptor-mediated entry and contributes to α-defensin-enhanced infection of susceptible cells. Remarkably, we also found that in the presence of α-defensins, HAdV tropism is expanded to non-susceptible cells, even when viruses are exposed to a mixture of both susceptible and non-susceptible cells. Therefore, we propose that in the presence of sufficient concentrations of α-defensins, such as in the lung or gut, integrin expression rather than primary receptor expression will dictate HAdV tropism in vivo . In summary, α-defensins may contribute to tissue tropism not only through the neutralization of susceptible viruses but also by allowing certain defensin-resistant viruses to bind to cells independently of previously described mechanisms. Author Summary In this study, we demonstrate a novel mechanism for binding of human adenoviruses (HAdVs) to cells that is dependent upon interactions with α-defensin host defense peptides but is independent of known viral receptors and co-receptors. To block normal receptor-mediated HAdV infection, we made genetic changes to both host cells and HAdVs. Under these conditions, α-defensins restored cell binding; however, infection still required the function of HAdV integrin co-receptors. This was true for multiple types of HAdVs that use different primary receptors and for cells that are either naturally devoid of HAdV receptors or were engineered to be receptor deficient. These observations suggest that in the presence of concentrations of α-defensins that would be found naturally in the lung or intestine, there are two parallel pathways for HAdV binding to cells that converge on integrins for productive infection. Moreover, these binding pathways function independently, and both operate in mixed culture. Thus, we have found that viruses can co-opt host defense molecules to expand their tropism.
Collapse
|
6
|
King CR, Dodge MJ, MacNeil KM, Tessier TM, Mymryk JS, Mehle A. Expanding the adenovirus toolbox: reporter viruses for studying the dynamics of human adenovirus replication. J Virol 2024; 98:e0020724. [PMID: 38639487 PMCID: PMC11092356 DOI: 10.1128/jvi.00207-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
To streamline standard virological assays, we developed a suite of nine fluorescent or bioluminescent replication competent human species C5 adenovirus reporter viruses that mimic their parental wild-type counterpart. These reporter viruses provide a rapid and quantitative readout of various aspects of viral infection and replication based on EGFP, mCherry, or NanoLuc measurement. Moreover, they permit real-time non-invasive measures of viral load, replication dynamics, and infection kinetics over the entire course of infection, allowing measurements that were not previously possible. This suite of replication competent reporter viruses increases the ease, speed, and adaptability of standard assays and has the potential to accelerate multiple areas of human adenovirus research.IMPORTANCEIn this work, we developed a versatile toolbox of nine HAdV-C5 reporter viruses and validated their functions in cell culture. These reporter viruses provide a rapid and quantitative readout of various aspects of viral infection and replication based on EGFP, mCherry, or NanoLuc measurement. The utility of these reporter viruses could also be extended for use in 3D cell culture, organoids, live cell imaging, or animal models, and provides a conceptual framework for the development of new reporter viruses representing other clinically relevant HAdV species.
Collapse
Affiliation(s)
- Cason R. King
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mackenzie J. Dodge
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Katelyn M. MacNeil
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Tanner M. Tessier
- Division of Protective Immunity, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joe S. Mymryk
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
- Department of Otolaryngology, University of Western Ontario, London, Ontario, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Andrew Mehle
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
7
|
Pinski AN, Gan T, Lin SC, Droit L, Diamond M, Barouch DH, Wang D. Isolation of a recombinant simian adenovirus encoding the human adenovirus G52 hexon suggests a simian origin for human adenovirus G52. J Virol 2024; 98:e0004324. [PMID: 38497664 PMCID: PMC11019922 DOI: 10.1128/jvi.00043-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/25/2024] [Indexed: 03/19/2024] Open
Abstract
Human adenoviruses (HAdVs) are causative agents of morbidity and mortality throughout the world. These double-stranded DNA viruses are phylogenetically classified into seven different species (A-G). HAdV-G52, originally isolated in 2008 from a patient presenting with gastroenteritis, is the sole human-derived member of species G. Phylogenetic analysis previously suggested that HAdV-G52 may have a simian origin, indicating a potential zoonotic spillover into humans. However, evidence of HAdV-G52 in either human or simian populations has not been reported since. Here, we describe the isolation and in vitro characterization of rhesus (rh)AdV-69, a novel simian AdV with clear evidence of recombination with HAdV-G52, from the stool of a rhesus macaque. Specifically, the rhAdV-69 hexon capsid protein is 100% identical to that of HAdV-G52, whereas the remainder of the genome is most similar to rhAdV-55, sharing 95.36% nucleic acid identity. A second recombination event with an unknown adenovirus (AdV) is evident at the short fiber gene. From the same sample, we also isolated a second, highly related recombinant AdV (rhAdV-68) that harbors a distinct hexon gene but nearly identical backbone compared to rhAdV-69. In vitro, rhAdV-68 and rhAdV-69 demonstrate comparable growth kinetics and tropisms in human cell lines, nonhuman cell lines, and human enteroids. Furthermore, we show that coinfection of highly related AdVs is not unique to this sample since we also isolated coinfecting rhAdVs from two additional rhesus macaque stool samples. Our data collectively contribute to elucidating the origins of HAdV-G52 and provide insights into the frequency of coinfections and subsequent recombination in AdV evolution.IMPORTANCEUnderstanding the host origins of adenoviruses (AdVs) is critical for public health as transmission of viruses from animals to humans can lead to emergent viruses. Recombination between animal and human AdVs can also produce emergent viruses. HAdV-G52 is the only human-derived member of the HAdV G species. It has been suggested that HAdV-G52 has a simian origin. Here, we isolated from a rhesus macaque, a novel rhAdV, rhAdV-69, that encodes a hexon protein that is 100% identical to that of HAdV-G52. This observation suggests that HAdV-G52 may indeed have a simian origin. We also isolated a highly related rhAdV, differing only in the hexon gene, from the same rhesus macaque stool sample as rhAdV-69, illustrating the potential for co-infection of closely related AdVs and recombination at the hexon gene. Furthermore, our study highlights the critical role of whole-genome sequencing in understanding AdV evolution and monitoring the emergence of pathogenic AdVs.
Collapse
Affiliation(s)
- Amanda N. Pinski
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Tianyu Gan
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Shih-Ching Lin
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lindsay Droit
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael Diamond
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Medicine, Division of Infectious Diseases, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David Wang
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Gheitasi H, Sabbaghian M, Fadaee M, Mohammadzadeh N, Shekarchi AA, Poortahmasebi V. The relationship between autophagy and respiratory viruses. Arch Microbiol 2024; 206:136. [PMID: 38436746 DOI: 10.1007/s00203-024-03838-3] [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: 11/26/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 03/05/2024]
Abstract
Respiratory viruses have caused severe global health problems and posed essential challenges to the medical community. In recent years, the role of autophagy as a critical process in cells in viral respiratory diseases has been noticed. One of the vital catabolic biological processes in the body is autophagy. Autophagy contributes to energy recovery by targeting and selectively directing foreign microorganisms, organelles, and senescent intracellular proteins to the lysosome for degradation and phagocytosis. Activation or suppression of autophagy is often initiated when foreign pathogenic organisms such as viruses infect cells. Because of its antiviral properties, several viruses may escape or resist this process by encoding viral proteins. Viruses can also use autophagy to enhance their replication or prolong the persistence of latent infections. Here, we provide an overview of autophagy and respiratory viruses such as coronavirus, rhinovirus, parainfluenza, influenza, adenovirus, and respiratory syncytial virus, and examine the interactions between them and the role of autophagy in the virus-host interaction process and the resulting virus replication strategy.
Collapse
Affiliation(s)
- Hamidreza Gheitasi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sabbaghian
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader Mohammadzadeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Scarsella L, Ehrke-Schulz E, Paulussen M, Thal SC, Ehrhardt A, Aydin M. Advances of Recombinant Adenoviral Vectors in Preclinical and Clinical Applications. Viruses 2024; 16:377. [PMID: 38543743 PMCID: PMC10974029 DOI: 10.3390/v16030377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 05/23/2024] Open
Abstract
Adenoviruses (Ad) have the potential to induce severe infections in vulnerable patient groups. Therefore, understanding Ad biology and antiviral processes is important to comprehend the signaling cascades during an infection and to initiate appropriate diagnostic and therapeutic interventions. In addition, Ad vector-based vaccines have revealed significant potential in generating robust immune protection and recombinant Ad vectors facilitate efficient gene transfer to treat genetic diseases and are used as oncolytic viruses to treat cancer. Continuous improvements in gene delivery capacity, coupled with advancements in production methods, have enabled widespread application in cancer therapy, vaccine development, and gene therapy on a large scale. This review provides a comprehensive overview of the virus biology, and several aspects of recombinant Ad vectors, as well as the development of Ad vector, are discussed. Moreover, we focus on those Ads that were used in preclinical and clinical applications including regenerative medicine, vaccine development, genome engineering, treatment of genetic diseases, and virotherapy in tumor treatment.
Collapse
Affiliation(s)
- Luca Scarsella
- Department of Anesthesiology, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Science (ZBAF), Department of Human Medicine, Faculty of Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Eric Ehrke-Schulz
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
| | - Michael Paulussen
- Chair of Pediatrics, University Children’s Hospital, Vestische Kinder- und Jugendklinik Datteln, Witten/Herdecke University, 45711 Datteln, Germany;
| | - Serge C. Thal
- Department of Anesthesiology, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
| | - Malik Aydin
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department Human Medicine, Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany; (E.E.-S.); (A.E.)
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Science (ZBAF), Department of Human Medicine, Faculty of Medicine, Witten/Herdecke University, 58453 Witten, Germany
- Chair of Pediatrics, University Children’s Hospital, Vestische Kinder- und Jugendklinik Datteln, Witten/Herdecke University, 45711 Datteln, Germany;
- Institute for Medical Laboratory Diagnostics, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany
| |
Collapse
|
10
|
Casasnovas JM. Virus-Receptor Interactions and Receptor-Mediated Virus Entry into Host Cells. Subcell Biochem 2024; 105:533-566. [PMID: 39738957 DOI: 10.1007/978-3-031-65187-8_15] [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] [Indexed: 01/02/2025]
Abstract
The virus particles described in the previous chapters of this book are vehicles that transmit the viral genome and the infection from cell to cell. To initiate the infective cycle, the viral genome must therefore translocate from the viral particle to the cell cytoplasm. Via distinct proteins or motifs in their outermost shell, the particles of animal viruses or bacteriophages attach initially to specific receptors on the host cell surface. These viral receptors thus mediate penetration of the viral genome inside the cell, where the intracellular infective cycle starts. The presence of these receptors on the cell surface is a principal determinant of virus-host tropism. Viruses can use diverse types of molecules to attach to and enter into cells. In addition, virus-receptor recognition can evolve over the course of an infection, and viral variants with distinct receptor-binding specificities and tropism can appear. The identification of viral receptors and the characterization of virus-receptor interactions have been major research goals in virology. In this chapter, we will describe, from a structural perspective, several virus-receptor interactions and the active role of receptor molecules in virus cell entry.
Collapse
Affiliation(s)
- José M Casasnovas
- Department of Macromolecular Structure, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
11
|
Mundy RM, Baker AT, Bates EA, Cunliffe TG, Teijeira-Crespo A, Moses E, Rizkallah PJ, Parker AL. Broad sialic acid usage amongst species D human adenovirus. NPJ VIRUSES 2023; 1:1. [PMID: 38665237 PMCID: PMC11041768 DOI: 10.1038/s44298-023-00001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/27/2023] [Indexed: 04/28/2024]
Abstract
Human adenoviruses (HAdV) are widespread pathogens causing usually mild infections. The Species D (HAdV-D) cause gastrointestinal tract infections and epidemic keratoconjunctivitis (EKC). Despite being significant pathogens, knowledge around HAdV-D mechanism of cell infection is lacking. Sialic acid (SA) usage has been proposed as a cell infection mechanism for EKC causing HAdV-D. Here we highlight an important role for SA engagement by many HAdV-D. We provide apo state crystal structures of 7 previously undetermined HAdV-D fiber-knob proteins, and structures of HAdV-D25, D29, D30 and D53 fiber-knob proteins in complex with SA. Biologically, we demonstrate that removal of cell surface SA reduced infectivity of HAdV-C5 vectors pseudotyped with HAdV-D fiber-knob proteins, whilst engagement of the classical HAdV receptor CAR was variable. Our data indicates variable usage of SA and CAR across HAdV-D. Better defining these interactions will enable improved development of antivirals and engineering of the viruses into refined therapeutic vectors.
Collapse
Affiliation(s)
- Rosie M. Mundy
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Alexander T. Baker
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Emily A. Bates
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Tabitha G. Cunliffe
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Alicia Teijeira-Crespo
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Elise Moses
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Pierre J. Rizkallah
- Division of Infection & Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| | - Alan L. Parker
- Division of Cancer & Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
- Systems Immunity University Research Institute, Cardiff University School of Medicine, Cardiff, CF14 4XN UK
| |
Collapse
|
12
|
Parsons AJ, Stein KR, Atanasoff KE, Ophir SI, Casado JP, Tortorella D. The CD46 ectodomain participates in human cytomegalovirus infection of epithelial cells. J Gen Virol 2023; 104:001892. [PMID: 37668349 PMCID: PMC10484303 DOI: 10.1099/jgv.0.001892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) primary infections are typically asymptomatic in healthy individuals yet can cause increased morbidity and mortality in organ transplant recipients, AIDS patients, neonates, and the elderly. The successful, widespread dissemination of this virus among the population can be attributed in part to its wide cellular tropism and ability to establish life-long latency. HCMV infection is a multi-step process that requires numerous cellular and viral factors. The viral envelope consists of envelope protein complexes that interact with cellular factors; such interactions dictate virus recognition and attachment to different cell types, followed by fusion either at the cell membrane or within an endocytic vesicle. Several HCMV entry factors, including neuropilin-2 (Nrp2), THBD, CD147, OR14I1, and CD46, are characterized as participating in HCMV pentamer-specific entry of non-fibroblast cells such as epithelial, trophoblast, and endothelial cells, respectively. This study focuses on characterizing the structural elements of CD46 that impact HCMV infection. Infectivity studies of wild-type and CD46 knockout epithelial cells demonstrated that levels of CD46 expressed on the cell surface were directly related to HCMV infectivity. Overexpression of CD46 isomers BC1, BC2, and C2 enhanced infection. Further, CD46 knockout epithelial cells expressing CD46 deletion and chimeric molecules identified that the intact ectodomain was critical for rescue of HCMV infection in CD46 knockout cells. Collectively, these data support a model that the extracellular domain of CD46 participates in HCMV infection due to its surface expression.
Collapse
Affiliation(s)
- Andrea J. Parsons
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kathryn R. Stein
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristina E. Atanasoff
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sabrina I. Ophir
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jailene Paredes Casado
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
13
|
Kosoltanapiwat N, van der Hoek L, Kinsella CM, Tongshoob J, Prasittichai L, Klein M, Jebbink MF, Deijs M, Reamtong O, Boonnak K, Khongsiri W, Phadungsombat J, Tongthainan D, Tulayakul P, Yindee M. A Novel Simian Adenovirus Associating with Human Adeno-virus Species G Isolated from Long-Tailed Macaque Feces. Viruses 2023; 15:1371. [PMID: 37376670 PMCID: PMC10303043 DOI: 10.3390/v15061371] [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: 04/27/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Metagenomics has demonstrated its capability in outbreak investigations and pathogen surveillance and discovery. With high-throughput and effective bioinformatics, many disease-causing agents, as well as novel viruses of humans and animals, have been identified using metagenomic analysis. In this study, a VIDISCA metagenomics workflow was used to identify potential unknown viruses in 33 fecal samples from asymptomatic long-tailed macaques (Macaca fascicularis) in Ratchaburi Province, Thailand. Putatively novel astroviruses, enteroviruses, and adenoviruses were detected and confirmed by PCR analysis of long-tailed macaque fecal samples collected from areas in four provinces, Ratchaburi, Kanchanaburi, Lopburi, and Prachuap Khiri Khan, where humans and monkeys live in proximity (total n = 187). Astroviruses, enteroviruses, and adenoviruses were present in 3.2%, 7.5%, and 4.8% of macaque fecal samples, respectively. One adenovirus, named AdV-RBR-6-3, was successfully isolated in human cell culture. Whole-genome analysis suggested that it is a new member of the species Human adenovirus G, closely related to Rhesus adenovirus 53, with evidence of genetic recombination and variation in the hexon, fiber, and CR1 genes. Sero-surveillance showed neutralizing antibodies against AdV-RBR-6-3 in 2.9% and 11.2% of monkeys and humans, respectively, suggesting cross-species infection of monkeys and humans. Overall, we reported the use of metagenomics to screen for possible new viruses, as well as the isolation and molecular and serological characterization of the new adenovirus with cross-species transmission potential. The findings emphasize that zoonotic surveillance is important and should be continued, especially in areas where humans and animals interact, to predict and prevent the threat of emerging zoonotic pathogens.
Collapse
Affiliation(s)
- Nathamon Kosoltanapiwat
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (J.T.); (W.K.)
| | - Lia van der Hoek
- Amsterdam UMC, Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.v.d.H.); (C.M.K.); (M.K.); (M.F.J.); (M.D.)
| | - Cormac M. Kinsella
- Amsterdam UMC, Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.v.d.H.); (C.M.K.); (M.K.); (M.F.J.); (M.D.)
| | - Jarinee Tongshoob
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (J.T.); (W.K.)
| | - Luxsana Prasittichai
- Wildlife Conservation Division, Protected Areas Regional Office 3 (Ban Pong), Department of National Parks, Wildlife and Plant Conservation, Ministry of Natural Resources and Environment, Ratchaburi 70110, Thailand;
| | - Michelle Klein
- Amsterdam UMC, Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.v.d.H.); (C.M.K.); (M.K.); (M.F.J.); (M.D.)
| | - Maarten F. Jebbink
- Amsterdam UMC, Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.v.d.H.); (C.M.K.); (M.K.); (M.F.J.); (M.D.)
| | - Martin Deijs
- Amsterdam UMC, Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.v.d.H.); (C.M.K.); (M.K.); (M.F.J.); (M.D.)
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Kobporn Boonnak
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Wathusiri Khongsiri
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (J.T.); (W.K.)
| | - Juthamas Phadungsombat
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan;
| | - Daraka Tongthainan
- Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-ok, Chonburi 20110, Thailand;
| | - Phitsanu Tulayakul
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Kasetsart University, Nakhon Pathom 73140, Thailand;
| | - Marnoch Yindee
- Akkhraratchakumari Veterinary College, Walailak University, Nakhonsithammarat 80161, Thailand;
| |
Collapse
|
14
|
Sutherland DM, Strebl M, Koehler M, Welsh OL, Yu X, Hu L, dos Santos Natividade R, Knowlton JJ, Taylor GM, Moreno RA, Wörz P, Lonergan ZR, Aravamudhan P, Guzman-Cardozo C, Kour S, Pandey UB, Alsteens D, Wang Z, Prasad BVV, Stehle T, Dermody TS. NgR1 binding to reovirus reveals an unusual bivalent interaction and a new viral attachment protein. Proc Natl Acad Sci U S A 2023; 120:e2219404120. [PMID: 37276413 PMCID: PMC10268256 DOI: 10.1073/pnas.2219404120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/19/2023] [Indexed: 06/07/2023] Open
Abstract
Nogo-66 receptor 1 (NgR1) binds a variety of structurally dissimilar ligands in the adult central nervous system to inhibit axon extension. Disruption of ligand binding to NgR1 and subsequent signaling can improve neuron outgrowth, making NgR1 an important therapeutic target for diverse neurological conditions such as spinal crush injuries and Alzheimer's disease. Human NgR1 serves as a receptor for mammalian orthoreovirus (reovirus), but the mechanism of virus-receptor engagement is unknown. To elucidate how NgR1 mediates cell binding and entry of reovirus, we defined the affinity of interaction between virus and receptor, determined the structure of the virus-receptor complex, and identified residues in the receptor required for virus binding and infection. These studies revealed that central NgR1 surfaces form a bridge between two copies of viral capsid protein σ3, establishing that σ3 serves as a receptor ligand for reovirus. This unusual binding interface produces high-avidity interactions between virus and receptor to prime early entry steps. These studies refine models of reovirus cell-attachment and highlight the evolution of viruses to engage multiple receptors using distinct capsid components.
Collapse
Affiliation(s)
- Danica M. Sutherland
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Michael Strebl
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Olivia L. Welsh
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Xinzhe Yu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Rita dos Santos Natividade
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Jonathan J. Knowlton
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Cryo-Electron Microscopy and Tomography Core, Baylor College of Medicine, Houston, TX77030
| | - Gwen M. Taylor
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Rodolfo A. Moreno
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Patrick Wörz
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Zachery R. Lonergan
- Cryo-Electron Microscopy and Tomography Core, Baylor College of Medicine, Houston, TX77030
| | - Pavithra Aravamudhan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Camila Guzman-Cardozo
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Sukhleen Kour
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
| | - Udai Bhan Pandey
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN37232
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA15261
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
- Children’s Neuroscience Institute, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Zhao Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
- Walloon Excellence in Life Sciences and Biotechnology, 1300Wavre, Belgium
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - B. V. Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15219
| |
Collapse
|
15
|
Gryciuk A, Rogalska M, Baran J, Kuryk L, Staniszewska M. Oncolytic Adenoviruses Armed with Co-Stimulatory Molecules for Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071947. [PMID: 37046608 PMCID: PMC10093006 DOI: 10.3390/cancers15071947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
In clinical trials, adenovirus vectors (AdVs) are commonly used platforms for human gene delivery therapy. High genome capacity and flexibility in gene organization make HAdVs suitable for cloning. Recent advancements in molecular techniques have influenced the development of genetically engineered adenovirus vectors showing therapeutic potential. Increased molecular understanding of the benefits and limitations of HAdVs in preclinical research and clinical studies is a crucial point in the engineering of refined oncolytic vectors. This review presents HAdV species (A-G) used in oncotherapy. We describe the adenovirus genome organizations and modifications, the possibilities oncolytic viruses offer, and their current limitations. Ongoing and ended clinical trials based on oncolytic adenoviruses are presented. This review provides a broad overview of the current knowledge of oncolytic therapy. HAdV-based strategies targeting tumors by employing variable immune modifiers or delivering immune stimulatory factors are of great promise in the field of immune oncologyy This approach can change the face of the fight against cancer, supplying the medical tools to defeat tumors more selectively and safely.
Collapse
Affiliation(s)
- Aleksander Gryciuk
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Marta Rogalska
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Joanna Baran
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH-NRI, 00-791 Warsaw, Poland
- Valo Therapeutics, 00790 Helsinki, Finland
| | - Monika Staniszewska
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| |
Collapse
|
16
|
Podgorski II, Harrach B, Benkő M, Papp T. Characterization of monkey adenoviruses with three fiber genes. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 108:105403. [PMID: 36610683 DOI: 10.1016/j.meegid.2023.105403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Although the occurrence of three fiber genes in monkey adenoviruses had already been described, the relatedness of the "extra" fibers have not yet been discussed. Here we report the genome analysis of two simian adenovirus (SAdV) serotypes from Old World monkeys and the phylogenetic analysis of the multiple fiber genes found in these and related AdVs. One of the newly sequenced serotypes (SAdV-2), isolated from a rhesus macaque (Macaca mulatta), was classified into species Human mastadenovirus G (HAdV-G), while the other serotype (SAdV-17), originating from a grivet (Chlorocebus aethiops), classified to Simian mastadenovirus F (SAdV-F). We identified unique features in the gene content of these SAdVs compared to those typical for other members of the genus Mastadenovirus. Namely, in the E1B region of SAdV-2, the 19K gene was replaced by an ITR repetition and a copy of the E4 ORF1 gene. Among the 37 genes in both SAdVs, three genes of different lengths, predicted to code for the cellular attachment proteins (the fibers), were found. These proteins exhibit high diversity. Yet, phylogenetic calculations of their conserved parts could reveal the probable evolutionary steps leading to the multiple-fibered contemporary HAdV and SAdV species. Seemingly, there existed (a) common ancestor(s) with two fiber genes for the lineages of the AdVs in species SAdV-B, -E, -F and HAdV-F, alongside a double-fibered ancestor for today's SAdV-C and HAdV-G, which later diverged into descendants forming today's species. Additionally, some HAdV-G members picked up a third fiber gene either to the left-hand or to the in-between position from the existing two. A SAdV-F progenitor also obtained a third copy to the middle, as observed in SAdV-17. The existence of three fiber genes in these contemporary AdVs brings novel possibilities for the design of optimised AdV-based vectors with potential multiple target binding abilities.
Collapse
Affiliation(s)
- Iva I Podgorski
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary.
| | - Balázs Harrach
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary.
| | - Mária Benkő
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary.
| | - Tibor Papp
- Veterinary Medical Research Institute, H-1143 Budapest, Hungary.
| |
Collapse
|
17
|
Mardi N, Haiaty S, Rahbarghazi R, Mobarak H, Milani M, Zarebkohan A, Nouri M. Exosomal transmission of viruses, a two-edged biological sword. Cell Commun Signal 2023; 21:19. [PMID: 36691072 PMCID: PMC9868521 DOI: 10.1186/s12964-022-01037-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
As a common belief, most viruses can egress from the host cells as single particles and transmit to uninfected cells. Emerging data have revealed en bloc viral transmission as lipid bilayer-cloaked particles via extracellular vesicles especially exosomes (Exo). The supporting membrane can be originated from multivesicular bodies during intra-luminal vesicle formation and autophagic response. Exo are nano-sized particles, ranging from 40-200 nm, with the ability to harbor several types of signaling molecules from donor to acceptor cells in a paracrine manner, resulting in the modulation of specific signaling reactions in target cells. The phenomenon of Exo biogenesis consists of multiple and complex biological steps with the participation of diverse constituents and molecular pathways. Due to similarities between Exo biogenesis and virus replication and the existence of shared pathways, it is thought that viruses can hijack the Exo biogenesis machinery to spread and evade immune cells. To this end, Exo can transmit complete virions (as single units or aggregates), separate viral components, and naked genetic materials. The current review article aims to scrutinize challenges and opportunities related to the exosomal delivery of viruses in terms of viral infections and public health. Video Abstract.
Collapse
Affiliation(s)
- Narges Mardi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
| | - Morteza Milani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
MacNeil KM, Dodge MJ, Evans AM, Tessier TM, Weinberg JB, Mymryk JS. Adenoviruses in medicine: innocuous pathogen, predator, or partner. Trends Mol Med 2023; 29:4-19. [PMID: 36336610 PMCID: PMC9742145 DOI: 10.1016/j.molmed.2022.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/09/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The consequences of human adenovirus (HAdV) infections are generally mild. However, despite the perception that HAdVs are harmless, infections can cause severe disease in certain individuals, including newborns, the immunocompromised, and those with pre-existing conditions, including respiratory or cardiac disease. In addition, HAdV outbreaks remain relatively common events and the recent emergence of more pathogenic genomic variants of various genotypes has been well documented. Coupled with evidence of zoonotic transmission, interspecies recombination, and the lack of approved AdV antivirals or widely available vaccines, HAdVs remain a threat to public health. At the same time, the detailed understanding of AdV biology garnered over nearly 7 decades of study has made this group of viruses a molecular workhorse for vaccine and gene therapy applications.
Collapse
Affiliation(s)
- Katelyn M MacNeil
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Mackenzie J Dodge
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Andris M Evans
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Tanner M Tessier
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Jason B Weinberg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA.
| | - Joe S Mymryk
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada; Department of Otolaryngology, Head & Neck Surgery, The University of Western Ontario, London, ON, Canada; Department of Oncology, The University of Western Ontario, London, ON, Canada; London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
19
|
Wang SY, Liu WQ, Li YQ, Li JX, Zhu FC. A China-developed adenovirus vector-based COVID-19 vaccine: review of the development and application of Ad5-nCov. Expert Rev Vaccines 2023; 22:704-713. [PMID: 37501516 DOI: 10.1080/14760584.2023.2242528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION The global spread of COVID-19 has prompted the development of vaccines. A recombinant adenovirus type-5 vectored COVID-19 vaccine (Ad5-nCoV) developed by Chinese scientists has been authorized for use as a prime and booster dose in China and several other countries. AREAS COVERED We searched published articles as of 4 May 2023, on PubMed using keywords related to Adenovirus vector, vaccine, and SARS-CoV-2. We reported the progress and outcomes of Ad5-nCov, including vaccine efficacy, safety, immunogenicity based on pre-clinical trials, clinical trials, and real-world studies for primary and booster doses. EXPERT OPINION Ad5-nCoV is a significant advancement in Chinese vaccine development technology. Evidence from clinical trials and real-world studies has demonstrated well-tolerated, highly immunogenic, and efficacy of Ad5-nCoV in preventing severe/critical COVID-19. Aerosolized Ad5-nCoV, given via a novel route, could elicit mucosal immunity and improve the vaccine efficacy, enhance the production capacity and availability, and reduce the potential negative impact of preexisting antibodies. However, additional research is necessary to evaluate the long-term safety and immunogenicity of Ad5-nCoV, its efficacy against emerging variants, its effectiveness in a real-world context of hybrid immunity, and its cost-effectiveness, particularly with respect to aerosolized Ad5-nCoV.
Collapse
Affiliation(s)
- Shen-Yu Wang
- Department of Immunization Programe, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wen-Qing Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu-Qing Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing-Xin Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng-Cai Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- NHC Key Laboratory of Enteric Pathogenic Microbiology (Jiangsu Provincial Center for Disease Control and Prevention), Nanjing, China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
20
|
Schröer K, Alshawabkeh M, Schellhorn S, Bronder K, Zhang W, Ehrhardt A. Influence of Heparan Sulfate Proteoglycans and Factor X on species D Human Adenovirus Uptake and Transduction. Viruses 2022; 15:55. [PMID: 36680095 PMCID: PMC9866072 DOI: 10.3390/v15010055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
More than 100 human adenovirus (Ad) types were identified, of which species D comprises the largest group. Heparan sulfate proteoglycans (HSPGs) were shown to function as cell surface receptors for cell binding and uptake of some Ads, but a systematic analysis of species D Ads is lacking. Previous research focused on Ad5 and blood coagulation factor X (FX) complexes, which revealed that Ad5 can transduce cells with low expression levels of its main coxsackievirus-adenovirus receptor in the presence of high HSPG expression levels in a FX dependent manner. Based on our reporter gene-tagged Ad-library, we explored for the first time a broad spectrum of species D Ads to study the role of HSPG on their cellular uptake. This study was performed on three Chinese Hamster Ovary (CHO) cell lines with different forms of HSPG (only proteoglycan (745), non-sulfated HSPG (606) or sulfated HSPG (K1)). The effect of Ad:FX complexes on Ad uptake was explored in the presence of physiological levels of FX in blood (6-10 µg/mL). We found that sulfation of HSPG plays an important role in cellular uptake and transduction of FX-bound Ad5 but neither HSPG nor FX influenced uptake of all tested species D Ads. Because FX has no influence on transduction efficiencies of species D Ads and therefore may not bind to them, these Ads may not be protected from attack by neutralizing IgM antibodies or the complement pathway, which may have implications for species D Ads used as vaccine and gene therapy vectors.
Collapse
Affiliation(s)
- Katrin Schröer
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Montaha Alshawabkeh
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Sebastian Schellhorn
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Katrin Bronder
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Witten/Herdecke University, 58453 Witten, Germany
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Witten/Herdecke University, 58453 Witten, Germany
| |
Collapse
|
21
|
杨 中, 付 扬, 任 洛, 陈 诗, 刘 恩, 臧 娜. [Silencing CD46 and DSG2 in host A549 cells inhibits entry of human adenovirus type 3 and type 7 and reduces interleukin-8 release]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1344-1350. [PMID: 36210707 PMCID: PMC9550555 DOI: 10.12122/j.issn.1673-4254.2022.09.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the effect of silencing CD46 and desmoglein 2 (DSG2) in host A549 cells on the entry of human adenovirus type 3 (HAdV-3) and type 7 (HAdV-7) and host cell secretion of inflammatory cytokines. METHODS RNA interference technique was use to silence the expression of CD46 or DSG2 in human epithelial alveolar A549 cells as the host cells of HAdV-3 or HAdV-7. The binding of the viruses with CD46 and DSG2 were observed with immunofluorescence staining at 0.5 and 1 h after viral infection. The viral load in the host cells was determined with qRT-PCR, and IL-8 secretion level was measured using ELISA. RESULTS In infected A549 cells, immunofluorescent staining revealed colocalization of HAdV-3 and HAdV-37 with their receptors CD46 and DSG2 at 0.5 h and 2 h after infection, and the copy number of the viruses increased progressively after the infection in a time-dependent manner. In A549 cells with CD46 silencing, the virus titers were significantly lower at 2, 6, 12 and 24 h postinfection in comparison with the cells without gene silencing; the virus titers were also significantly decreased in the cells with DSG2 silencing. The secretion level of IL-8 increased significantly in A549 cells without siRNA transfection following infection with HAdV-3 and HAdV-7 (P < 0.0001), but decreased significantly in cells with CD46 and DSG2 silencing (P < 0.0001). CONCLUSION HAdV-3 and HAdV-7 enter host cells by binding to their receptors CD46 and DSG2, and virus titer and cytokines release increase with infection time. Silencing CD46 and DSG2 can inhibit virus entry and cytokine IL-8 production in host cells.
Collapse
Affiliation(s)
- 中英 杨
- />重庆医科大学附属儿童医院呼吸科//国家儿童健康与疾病临床医学研究中心//儿童发育与疾病教育部重点实验室//儿科学重庆市重点实验室//儿童感染免疫重庆市重点实验室,重庆 400014Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - 扬喜 付
- />重庆医科大学附属儿童医院呼吸科//国家儿童健康与疾病临床医学研究中心//儿童发育与疾病教育部重点实验室//儿科学重庆市重点实验室//儿童感染免疫重庆市重点实验室,重庆 400014Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - 洛 任
- />重庆医科大学附属儿童医院呼吸科//国家儿童健康与疾病临床医学研究中心//儿童发育与疾病教育部重点实验室//儿科学重庆市重点实验室//儿童感染免疫重庆市重点实验室,重庆 400014Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - 诗懿 陈
- />重庆医科大学附属儿童医院呼吸科//国家儿童健康与疾病临床医学研究中心//儿童发育与疾病教育部重点实验室//儿科学重庆市重点实验室//儿童感染免疫重庆市重点实验室,重庆 400014Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - 恩梅 刘
- />重庆医科大学附属儿童医院呼吸科//国家儿童健康与疾病临床医学研究中心//儿童发育与疾病教育部重点实验室//儿科学重庆市重点实验室//儿童感染免疫重庆市重点实验室,重庆 400014Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| | - 娜 臧
- />重庆医科大学附属儿童医院呼吸科//国家儿童健康与疾病临床医学研究中心//儿童发育与疾病教育部重点实验室//儿科学重庆市重点实验室//儿童感染免疫重庆市重点实验室,重庆 400014Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing 400014, China
| |
Collapse
|
22
|
Human Adenovirus Type 26 Infection Mediated by αvβ3 Integrin Is Caveolin-1-Dependent. Microbiol Spectr 2022; 10:e0109722. [PMID: 35924932 PMCID: PMC9430667 DOI: 10.1128/spectrum.01097-22] [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] [Indexed: 12/02/2022] Open
Abstract
Human adenovirus type 26 (HAdV26) has been recognized as a promising platform for vaccine vector development, and very recently vaccine against COVID-19 based on HAdV26 was authorized for emergency use. Nevertheless, basic biology of this virus, namely, pathway which HAdV26 uses to enter the cell, is still insufficiently known. We have shown here that HAdV26 infection of human epithelial cells expressing low amount of αvβ3 integrin involves clathrin and is caveolin-1-independent, while HAdV26 infection of cells with high amount of αvβ3 integrin does not involve clathrin but is caveolin-1-dependent. Thus, this study demonstrates that caveolin-1 is limiting factor in αvβ3 integrin-mediated HAdV26 infection. Regardless of αvβ3 integrin expression, HAdV26 infection involves dynamin-2. Our data provide for the first-time description of HAdV26 cell entry pathway, hence increase our knowledge of HAdV26 infection. Knowing that functionality of adenovirus vector is influenced by its cell entry pathway and intracellular trafficking, our results will contribute to better understanding of HAdV26 immunogenicity and antigen presentation when used as vaccine vector. IMPORTANCE In order to fulfill its role as a vector, adenovirus needs to successfully deliver its DNA genome to the host nucleus, a process highly influenced by adenovirus intracellular translocation. Thus, cell entry pathway and intracellular trafficking determine functionality of human adenovirus-based vectors. Endocytosis of HAdV26, currently extensively studied as a vaccine vector, has not been described so far. We present here that HAdV26 infection of human epithelial cells with high expression of αvβ3 integrin, one of the putative HAdV26 receptors, is caveolin-1- and partially dynamin-2-dependent. Since caveolin containing domains provide a unique environment for specific signaling events and participate in inflammatory signaling one can imagine that directing HAdV26 cell entry toward caveolin-1-mediate pathway might play role in immunogenicity of this virus. Therefore, our results contribute to better understanding of HAdV26 infection pathway, hence, can be helpful in explaining induction of immune response and antigen presentation by HAdV26-based vaccine vector.
Collapse
|
23
|
Dienst EGT, Kremer EJ. Adenovirus receptors on antigen-presenting cells of the skin. Biol Cell 2022; 114:297-308. [PMID: 35906865 DOI: 10.1111/boc.202200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 12/01/2022]
Abstract
Skin, the largest human organ, is part of the first line of physical and immunological defense against many pathogens. Understanding how skin antigen-presenting cells (APCs) respond to viruses or virus-based vaccines is crucial to develop antiviral pharmaceutics, and efficient and safe vaccines. Here, we discuss the way resident and recruited skin APCs engage adenoviruses and the impact on innate immune responses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
24
|
Bates EA, Davies JA, Váňová J, Nestić D, Meniel VS, Koushyar S, Cunliffe TG, Mundy RM, Moses E, Uusi-Kerttula HK, Baker AT, Cole DK, Majhen D, Rizkallah PJ, Phesse T, Chester JD, Parker AL. Development of a low-seroprevalence, αvβ6 integrin-selective virotherapy based on human adenovirus type 10. Mol Ther Oncolytics 2022; 25:43-56. [PMID: 35399606 PMCID: PMC8971729 DOI: 10.1016/j.omto.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/13/2022] [Indexed: 11/29/2022] Open
Abstract
Oncolytic virotherapies (OV) hold immense clinical potential. OV based on human adenoviruses (HAdV) derived from HAdV with naturally low rates of pre-existing immunity will be beneficial for future clinical translation. We generated a low-seroprevalence HAdV-D10 serotype vector incorporating an αvβ6 integrin-selective peptide, A20, to target αvβ6-positive tumor cell types. HAdV-D10 has limited natural tropism. Structural and biological studies of HAdV-D10 knob protein highlighted low-affinity engagement with native adenoviral receptors CAR and sialic acid. HAdV-D10 fails to engage blood coagulation factor X, potentially eliminating "off-target" hepatic sequestration in vivo. We engineered an A20 peptide that selectively binds αvβ6 integrin into the DG loop of HAdV-D10 fiber knob. Assays in αvβ6+ cancer cell lines demonstrated significantly increased transduction mediated by αvβ6-targeted variants compared with controls, confirmed microscopically. HAdV-D10.A20 resisted neutralization by neutralizing HAdV-C5 sera. Systemic delivery of HAdV-D10.A20 resulted in significantly increased GFP expression in BT20 tumors. Replication-competent HAdV-D10.A20 demonstrated αvβ6 integrin-selective cell killing in vitro and in vivo. HAdV-D10 possesses characteristics of a promising virotherapy, combining low seroprevalence, weak receptor interactions, and reduced off-target uptake. Incorporation of an αvβ6 integrin-selective peptide resulted in HAdV-D10.A20, with significant potential for clinical translation.
Collapse
Affiliation(s)
- Emily A. Bates
- 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
| | - Jana Váňová
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic
| | - Davor Nestić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Valerie S. Meniel
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - Sarah Koushyar
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - Tabitha G. Cunliffe
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Rosie M. Mundy
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Elise Moses
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Hanni K. Uusi-Kerttula
- 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
| | - David K. Cole
- Division of Infection and Immunity, 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
| | - Pierre J. Rizkallah
- Division of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Toby Phesse
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - 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
|
25
|
Majhen D. Human adenovirus type 26 basic biology and its usage as vaccine vector. Rev Med Virol 2022; 32:e2338. [PMID: 35278248 DOI: 10.1002/rmv.2338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/10/2022]
Abstract
Due to their nature, adenoviruses have been recognised as promising candidates for vaccine vector development. Since they mimic natural infection, recombinant adenovirus vectors have been proven as ideal shuttles to deliver foreign transgenes aiming at inducing both humoral and cellular immune response. In addition, a potent adjuvant effect can be exerted due to the adenovirus inherent stimulation of various elements of innate and adaptive immunity. Due to its low seroprevalence in humans as well as induction of favourable immune response to inserted transgene, human adenovirus type 26 (HAdV-D26) has been recognised as a promising platform for vaccine vector development and is studied in number of completed or ongoing clinical studies. Very recently HAdV-D26 based Ebola and Covid-19 vaccines were approved for medical use. In this review, current state of the art regarding HAdV-D26 basic biology and its usage as vaccine vector will be discussed.
Collapse
Affiliation(s)
- Dragomira Majhen
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
26
|
Reddy VS, Yu X, Barry MA. Refined Capsid Structure of Human Adenovirus D26 at 3.4 Å Resolution. Viruses 2022; 14:v14020414. [PMID: 35216007 PMCID: PMC8878492 DOI: 10.3390/v14020414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Various adenoviruses are being used as viral vectors for the generation of vaccines against chronic and emerging diseases (e.g., AIDS, COVID-19). Here, we report the improved capsid structure for one of these vectors, human adenovirus D26 (HAdV-D26), at 3.4 Å resolution, by reprocessing the previous cryo-electron microscopy dataset and obtaining a refined model. In addition to overall improvements in the model, the highlights of the structure include (1) locating a segment of the processed peptide of VIII that was previously believed to be released from the mature virions, (2) reorientation of the helical appendage domain (APD) of IIIa situated underneath the vertex region relative to its counterpart observed in the cleavage defective (ts1) mutant of HAdV-C5 that resulted in the loss of interactions between the APD and hexon bases, and (3) the revised conformation of the cleaved N-terminal segments of pre-protein VI (pVIn), located in the hexon cavities, is highly conserved, with notable stacking interactions between the conserved His13 and Phe18 residues. Taken together, the improved model of HAdV-D26 capsid provides a better understanding of protein–protein interactions in HAdV capsids and facilitates the efforts to modify and/or design adenoviral vectors with altered properties. Last but not least, we provide some insights into clotting factors (e.g., FX and PF4) binding to AdV vectors.
Collapse
Affiliation(s)
- Vijay S Reddy
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xiaodi Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael A Barry
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
27
|
Ex Vivo and In Vivo CD46 Receptor Utilization by Species D Human Adenovirus Serotype 26 (HAdV26). J Virol 2022; 96:e0082621. [PMID: 34787457 PMCID: PMC8826919 DOI: 10.1128/jvi.00826-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human adenovirus serotype 26 (Ad26) is used as a gene-based vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and HIV-1. However, its primary receptor portfolio remains controversial, potentially including sialic acid, coxsackie and adenovirus receptor (CAR), integrins, and CD46. We and others have shown that Ad26 can use CD46, but these observations were questioned on the basis of the inability to cocrystallize Ad26 fiber with CD46. Recent work demonstrated that Ad26 binds CD46 with its hexon protein rather than its fiber. We examined the functional consequences of Ad26 for infection in vitro and in vivo. Ectopic expression of human CD46 on Chinese hamster ovary cells increased Ad26 infection significantly. Deletion of the complement control protein domain CCP1 or CCP2 or the serine-threonine-proline (STP) region of CD46 reduced infection. Comparing wild-type and sialic acid-deficient CHO cells, we show that the usage of CD46 is independent of its sialylation status. Ad26 transduction was increased in CD46 transgenic mice after intramuscular (i.m.) injection but not after intranasal (i.n.) administration. Ad26 transduction was 10-fold lower than Ad5 transduction after intratumoral (i.t.) injection of CD46-expressing tumors. Ad26 transduction of liver was 1,000-fold lower than that ofAd5 after intravenous (i.v.) injection. These data demonstrate the use of CD46 by Ad26 in certain situations but also show that the receptor has little consequence by other routes of administration. Finally, i.v. injection of high doses of Ad26 into CD46 mice induced release of liver enzymes into the bloodstream and reduced white blood cell counts but did not induce thrombocytopenia. This suggests that Ad26 virions do not induce direct clotting side effects seen during coronavirus disease 2019 (COVID-19) vaccination with this serotype of adenovirus. IMPORTANCE The human species D Ad26 is being investigated as a low-seroprevalence vector for oncolytic virotherapy and gene-based vaccination against HIV-1 and SARS-CoV-2. However, there is debate in the literature about its tropism and receptor utilization, which directly influence its efficiency for certain applications. This work was aimed at determining which receptor(s) this virus uses for infection and its role in virus biology, vaccine efficacy, and, importantly, vaccine safety.
Collapse
|
28
|
Adenovirus-α-defensin complexes induce NLRP3-associated maturation of human phagocytes via TLR4 engagement. J Virol 2022; 96:e0185021. [PMID: 35080426 DOI: 10.1128/jvi.01850-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intramuscular delivery of human adenovirus (HAdV)-based vaccines leads to rapid recruitment of neutrophils, which then release antimicrobial peptides/proteins (AMPs). How these AMPs influence vaccine efficacy over the subsequent 24 h is poorly understood. In this study, we asked if human neutrophil protein 1 (HNP-1), an α-defensin that influences the direct and indirect innate immune responses to a range of pathogens, impacts the response of human phagocytes to three HAdV species/types (HAdV-C5, -D26, -B35). We show that HNP-1 binds to the capsids, redirects HAdV-C5, -D26, -B35 to Toll-like receptor 4 (TLR4), which leads to internalization, an NLRP3-mediated inflammasome response, and IL-1β release. Surprisingly, IL-1β release was not associated with notable disruption of plasma membrane integrity. These data further our understanding of HAdV vaccine immunogenicity and may provide pathways to extend the efficacy. Importance This study examines the interactions between danger-associated molecular patterns and human adenoviruses and its impact on vaccines. HAdVs and HNP-1 can interact, these interactions will modify the response of antigen-presenting cells., which will influence vaccine efficacy.
Collapse
|
29
|
Baker AT, Boyd RJ, Sarkar D, Teijeira-Crespo A, Chan CK, Bates E, Waraich K, Vant J, Wilson E, Truong CD, Lipka-Lloyd M, Fromme P, Vermaas J, Williams D, Machiesky L, Heurich M, Nagalo BM, Coughlan L, Umlauf S, Chiu PL, Rizkallah PJ, Cohen TS, Parker AL, Singharoy A, Borad MJ. ChAdOx1 interacts with CAR and PF4 with implications for thrombosis with thrombocytopenia syndrome. SCIENCE ADVANCES 2021; 7:eabl8213. [PMID: 34851659 PMCID: PMC8635433 DOI: 10.1126/sciadv.abl8213] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/19/2021] [Indexed: 05/09/2023]
Abstract
Vaccines derived from chimpanzee adenovirus Y25 (ChAdOx1), human adenovirus type 26 (HAdV-D26), and human adenovirus type 5 (HAdV-C5) are critical in combatting the severe acute respiratory coronavirus 2 (SARS-CoV-2) pandemic. As part of the largest vaccination campaign in history, ultrarare side effects not seen in phase 3 trials, including thrombosis with thrombocytopenia syndrome (TTS), a rare condition resembling heparin-induced thrombocytopenia (HIT), have been observed. This study demonstrates that all three adenoviruses deployed as vaccination vectors versus SARS-CoV-2 bind to platelet factor 4 (PF4), a protein implicated in the pathogenesis of HIT. We have determined the structure of the ChAdOx1 viral vector and used it in state-of-the-art computational simulations to demonstrate an electrostatic interaction mechanism with PF4, which was confirmed experimentally by surface plasmon resonance. These data confirm that PF4 is capable of forming stable complexes with clinically relevant adenoviruses, an important step in unraveling the mechanisms underlying TTS.
Collapse
Affiliation(s)
- Alexander T. Baker
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ 85054, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Ryan J. Boyd
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Daipayan Sarkar
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
- MSU-DOE Plant Research Laboratory, Michigan State University, East Lansing, MI 48824
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Alicia Teijeira-Crespo
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Chun Kit Chan
- Computational Structural Biology and Molecular Biophysics, Beckman institute, University of Illinois, IL 61801, USA
| | - Emily Bates
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Kasim Waraich
- Institute of Infection Immunity and Inflammation, MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - John Vant
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Eric Wilson
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Chloe D. Truong
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Magdalena Lipka-Lloyd
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK
| | - Petra Fromme
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Josh Vermaas
- MSU-DOE Plant Research Laboratory, Michigan State University, East Lansing, MI 48824
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Dewight Williams
- Eyring Materials Center, Arizona State University, Tempe, AZ 85281, USA
| | - LeeAnn Machiesky
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Meike Heurich
- School of Pharmacy and Pharmaceutical Science, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF10 3NB, UK
| | - Bolni M. Nagalo
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ 85054, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, MD 21201, USA
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 W. Baltimore Street, MD 21201, USA
| | - Scott Umlauf
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Po-Lin Chiu
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Pierre J. Rizkallah
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Taylor S. Cohen
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Alan L. Parker
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Abhishek Singharoy
- Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85281, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85251, USA
| | - Mitesh J. Borad
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ 85054, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA
| |
Collapse
|
30
|
Bieri M, Hendrickx R, Bauer M, Yu B, Jetzer T, Dreier B, Mittl PRE, Sobek J, Plückthun A, Greber UF, Hemmi S. The RGD-binding integrins αvβ6 and αvβ8 are receptors for mouse adenovirus-1 and -3 infection. PLoS Pathog 2021; 17:e1010083. [PMID: 34910784 PMCID: PMC8673666 DOI: 10.1371/journal.ppat.1010083] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Mammalian adenoviruses (AdVs) comprise more than ~350 types including over 100 human (HAdVs) and just three mouse AdVs (MAdVs). While most HAdVs initiate infection by high affinity/avidity binding of their fiber knob (FK) protein to either coxsackievirus AdV receptor (CAR), CD46 or desmoglein (DSG)-2, MAdV-1 (M1) infection requires arginine-glycine-aspartate (RGD) binding integrins. To identify the receptors mediating MAdV infection we generated five novel reporter viruses for MAdV-1/-2/-3 (M1, M2, M3) transducing permissive murine (m) CMT-93 cells, but not B16 mouse melanoma cells expressing mCAR, human (h) CD46 or hDSG-2. Recombinant M1 or M3 FKs cross-blocked M1 and M3 but not M2 infections. Profiling of murine and human cells expressing RGD-binding integrins suggested that αvβ6 and αvβ8 heterodimers are associated with M1 and M3 infections. Ectopic expression of mβ6 in B16 cells strongly enhanced M1 and M3 binding, infection, and progeny production comparable with mαvβ6-positive CMT-93 cells, whereas mβ8 expressing cells were more permissive to M1 than M3. Anti-integrin antibodies potently blocked M1 and M3 binding and infection of CMT-93 cells and hαvβ8-positive M000216 cells. Soluble integrin αvβ6, and synthetic peptides containing the RGDLXXL sequence derived from FK-M1, FK-M3 and foot and mouth disease virus coat protein strongly interfered with M1/M3 infections, in agreement with high affinity interactions of FK-M1/FK-M3 with αvβ6/αvβ8, determined by surface plasmon resonance measurements. Molecular docking simulations of ternary complexes revealed a bent conformation of RGDLXXL-containing FK-M3 peptides on the subunit interface of αvβ6/β8, where the distal leucine residue dips into a hydrophobic pocket of β6/8, the arginine residue ionically engages αv aspartate215, and the aspartate residue coordinates a divalent cation in αvβ6/β8. Together, the RGDLXXL-bearing FKs are part of an essential mechanism for M1/M3 infection engaging murine and human αvβ6/8 integrins. These integrins are highly conserved in other mammals, and may favour cross-species virus transmission.
Collapse
Affiliation(s)
- Manuela Bieri
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Molecular Life Sciences Graduate School, ETH and University Of Zurich, Switzerland
| | - Rodinde Hendrickx
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
- Molecular Life Sciences Graduate School, ETH and University Of Zurich, Switzerland
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Tania Jetzer
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Peer R. E. Mittl
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Jens Sobek
- Functional Genomics Center Zurich, Eidgenössische Technische Hochschule (ETH) Zurich and University of Zurich, Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Urs F. Greber
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Silvio Hemmi
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Bertzbach LD, Ip WH, Dobner T. Animal Models in Human Adenovirus Research. BIOLOGY 2021; 10:biology10121253. [PMID: 34943168 PMCID: PMC8698265 DOI: 10.3390/biology10121253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary Animal models are widely used to study various aspects of human diseases and disorders. Likewise, they are indispensable for preclinical testing of medicals and vaccines. Human adenovirus infections are usually self-limiting, and can cause mild respiratory symptoms with fever, eye infection or gastrointestinal symptoms, but occasional local outbreaks with severe disease courses have been reported. In addition, adenovirus infections pose a serious risk for children and patients with a weakened immune system. Human adenovirus research in animal models to study adenovirus-induced disease and tumor development started in the 1950s. Various animal species have been tested for their susceptibility to human adenovirus infection since then, and some have been shown to mimic key characteristics of the infection in humans, including persistent infection. Furthermore, some rodent species have been found to develop tumors upon human adenovirus infection. Our review summarizes the current knowledge on animal models in human adenovirus research, describing the pros and cons along with important findings and future perspectives. Abstract Human adenovirus (HAdV) infections cause a wide variety of clinical symptoms, ranging from mild upper respiratory tract disease to lethal outcomes, particularly in immunocompromised individuals. To date, neither widely available vaccines nor approved antiadenoviral compounds are available to efficiently deal with HAdV infections. Thus, there is a need to thoroughly understand HAdV-induced disease, and for the development and preclinical evaluation of HAdV therapeutics and/or vaccines, and consequently for suitable standardizable in vitro systems and animal models. Current animal models to study HAdV pathogenesis, persistence, and tumorigenesis include rodents such as Syrian hamsters, mice, and cotton rats, as well as rabbits. In addition, a few recent studies on other species, such as pigs and tree shrews, reported promising data. These models mimic (aspects of) HAdV-induced pathological changes in humans and, although they are relevant, an ideal HAdV animal model has yet to be developed. This review summarizes the available animal models of HAdV infection with comprehensive descriptions of virus-induced pathogenesis in different animal species. We also elaborate on rodent HAdV animal models and how they contributed to insights into adenovirus-induced cell transformation and cancer.
Collapse
|
32
|
Othman M, Baker AT, Gupalo E, Elsebaie A, Bliss CM, Rondina MT, Lillicrap D, Parker AL. To clot or not to clot? Ad is the question-Insights on mechanisms related to vaccine-induced thrombotic thrombocytopenia. J Thromb Haemost 2021; 19:2845-2856. [PMID: 34351057 PMCID: PMC8420166 DOI: 10.1111/jth.15485] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 02/05/2023]
Abstract
Vaccine-induced immune thrombotic thrombocytopenia (VITT) has caused global concern. VITT is characterized by thrombosis and thrombocytopenia following COVID-19 vaccinations with the AstraZeneca ChAdOx1 nCov-19 and the Janssen Ad26.COV2.S vaccines. Patients present with thrombosis, severe thrombocytopenia developing 5-24 days following first dose of vaccine, with elevated D-dimer, and PF4 antibodies, signifying platelet activation. As of June 1, 2021, more than 1.93 billion COVID-19 vaccine doses had been administered worldwide. Currently, 467 VITT cases (0.000024%) have been reported across the UK, Europe, Canada, and Australia. Guidance on diagnosis and management of VITT has been reported but the pathogenic mechanism is yet to be fully elucidated. Here, we propose and discuss potential mechanisms in relation to adenovirus induction of VITT. We provide insights and clues into areas warranting investigation into the mechanistic basis of VITT, highlighting the unanswered questions. Further research is required to help solidify a pathogenic model for this condition.
Collapse
Affiliation(s)
- Maha Othman
- Department of Biomedical and Molecular SciencesSchool of MedicineQueen's UniversityKingstonOntarioCanada
- School of Baccalaureate NursingSt. Lawrence CollegeKingstonOntarioCanada
| | - Alexander T. Baker
- Center for Individualized MedicineMayo ClinicScottsdaleArizonaUSA
- Division of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| | - Elena Gupalo
- National Medical Research Center for CardiologyMoscowRussia
| | - Abdelrahman Elsebaie
- Department of Biomedical and Molecular SciencesSchool of MedicineQueen's UniversityKingstonOntarioCanada
| | - Carly M. Bliss
- Division of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| | - Matthew T. Rondina
- Departments of Internal Medicine and Pathology, and the Molecular Medicine ProgramUniversity of Utah HealthSalt Lake CityUtahUSA
- Department of Internal Medicine and GRECCGeorge E. Wahlen VAMCSalt Lake CityUtahUSA
| | - David Lillicrap
- Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - Alan L. Parker
- Division of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| |
Collapse
|
33
|
Ballmann MZ, Raus S, Engelhart R, Kaján GL, Beqqali A, Hadoke PWF, van der Zalm C, Papp T, John L, Khan S, Boedhoe S, Danskog K, Frängsmyr L, Custers J, Bakker WAM, van der Schaar HM, Arnberg N, Lemckert AAC, Havenga M, Baker AH. Human AdV-20-42-42, a Promising Novel Adenoviral Vector for Gene Therapy and Vaccine Product Development. J Virol 2021; 95:e0038721. [PMID: 34469243 PMCID: PMC8549523 DOI: 10.1128/jvi.00387-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 08/29/2021] [Indexed: 02/07/2023] Open
Abstract
Preexisting immune responses toward adenoviral vectors limit the use of a vector based on particular serotypes and its clinical applicability for gene therapy and/or vaccination. Therefore, there is a significant interest in vectorizing novel adenoviral types that have low seroprevalence in the human population. Here, we describe the discovery and vectorization of a chimeric human adenovirus, which we call HAdV-20-42-42. Full-genome sequencing revealed that this virus is closely related to human serotype 42, except for the penton base, which is derived from serotype 20. The HAdV-20-42-42 vector could be propagated stably to high titers on existing E1-complementing packaging cell lines. Receptor-binding studies revealed that the vector utilized both CAR and CD46 as receptors for cell entry. Furthermore, the HAdV-20-42-42 vector was potent in transducing human and murine cardiovascular cells and tissues, irrespective of the presence of blood coagulation factor X. In vivo characterizations demonstrate that when delivered intravenously (i.v.) in mice, HAdV-20-42-42 mainly targeted the lungs, liver, and spleen and triggered robust inflammatory immune responses. Finally, we demonstrate that potent T-cell responses against vector-delivered antigens could be induced upon intramuscular vaccination in mice. In summary, from the data obtained we conclude that HAdV-20-42-42 provides a valuable addition to the portfolio of adenoviral vectors available to develop efficacious products in the fields of gene therapy and vaccination. IMPORTANCE Adenoviral vectors are under investigation for a broad range of therapeutic indications in diverse fields, such as oncology and gene therapy, as well as for vaccination both for human and veterinary use. A wealth of data shows that preexisting immune responses may limit the use of a vector. Particularly in the current climate of global pandemic, there is a need to expand the toolbox with novel adenoviral vectors for vaccine development. Our data demonstrate that we have successfully vectorized a novel adenovirus type candidate with low seroprevalence. The cell transduction data and antigen-specific immune responses induced in vivo demonstrate that this vector is highly promising for the development of gene therapy and vaccine products.
Collapse
Affiliation(s)
| | - Svjetlana Raus
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ruben Engelhart
- Batavia Biosciences B.V., Leiden, The Netherlands
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Győző L. Kaján
- Department of Clinical Microbiology, Division of Virology, Umeå University, Umeå, Sweden
| | - Abdelaziz Beqqali
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Tibor Papp
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Lijo John
- Department of Clinical Microbiology, Division of Virology, Umeå University, Umeå, Sweden
| | - Selina Khan
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Satish Boedhoe
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Katarina Danskog
- Department of Clinical Microbiology, Division of Virology, Umeå University, Umeå, Sweden
| | - Lars Frängsmyr
- Department of Clinical Microbiology, Division of Virology, Umeå University, Umeå, Sweden
| | - Jerome Custers
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | | | | | - Niklas Arnberg
- Department of Clinical Microbiology, Division of Virology, Umeå University, Umeå, Sweden
| | | | | | - Andrew H. Baker
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Liszewski MK, Atkinson JP. Membrane cofactor protein (MCP; CD46): deficiency states and pathogen connections. Curr Opin Immunol 2021; 72:126-134. [PMID: 34004375 PMCID: PMC8123722 DOI: 10.1016/j.coi.2021.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
Membrane cofactor protein (MCP; CD46), a ubiquitously expressed complement regulatory protein, serves as a cofactor for serine protease factor I to cleave and inactivate C3b and C4b deposited on host cells. However, CD46 also plays roles in human reproduction, autophagy, modulating T cell activation and effector functions and is a member of the newly identified intracellular complement system (complosome). CD46 also is a receptor for 11 pathogens ('pathogen magnet'). While CD46 deficiencies contribute to inflammatory disorders, its overexpression in cancers and role as a receptor for some adenoviruses has led to its targeting by oncolytic agents and adenoviral-based therapeutic vectors, including coronavirus disease of 2019 (COVID-19) vaccines. This review focuses on recent advances in identifying disease-causing CD46 variants and its pathogen connections.
Collapse
Affiliation(s)
- M Kathryn Liszewski
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
35
|
Sahu SK, Kulkarni DH, Ozanturk AN, Ma L, Kulkarni HS. Emerging roles of the complement system in host-pathogen interactions. Trends Microbiol 2021; 30:390-402. [PMID: 34600784 DOI: 10.1016/j.tim.2021.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022]
Abstract
The complement system has historically been entertained as a fluid-phase, hepatically derived system which protects the intravascular space from encapsulated bacteria. However, there has been an increasing appreciation for its role in protection against non-encapsulated pathogens. Specifically, we have an improved understanding of how pathogens are recognized by specific complement proteins, as well as how they trigger and evade them. Additionally, we have an improved understanding of locally derived complement proteins, many of which promote host defense. Moreover, intracellular complement proteins have been identified that facilitate local protection and barrier function despite pathogen invasion. Our review aims to summarize these advances in the field as well as provide an insight into the pathophysiological changes occurring when the system is dysregulated in infection.
Collapse
Affiliation(s)
- Sanjaya K Sahu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Devesha H Kulkarni
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ayse N Ozanturk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lina Ma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
36
|
Wang Z, Zhang X. Adenovirus vector-attributed hepatotoxicity blocks clinical application in gene therapy. Cytotherapy 2021; 23:1045-1052. [PMID: 34548241 DOI: 10.1016/j.jcyt.2021.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023]
Abstract
Adenoviruses (Ads), common self-limiting pathogens in humans and animals, usually cause conjunctivitis, mild upper respiratory tract infection or gastroenteritis in humans and hepatotoxicity syndrome in chickens and dogs, posing great threats to public health and livestock husbandry. Artificially modified Ads, which wipe out virulence-determining genes, are the most frequently used viral vectors in gene therapy, and some Ad vector (AdV)-related medicines and vaccines have been licensed and applied. Inherent liver tropism enables AdVs to specifically deliver drugs/genes to the liver; however, AdVs are closely associated with acute hepatotoxicity in immunocompromised individuals, and the side effects of AdVs, which stimulate a strong inflammatory reaction in the liver and cause acute hepatotoxicity, have largely limited clinical application. Therefore, this review systematically elucidates the intimate relationship between AdVs and hepatotoxicity in terms of virus and host and precisely illustrates the accumulated understanding in this field over the past decades. This review demonstrates the liver tropism of AdVs and molecular mechanism of AdV-induced hepatotoxicity and looks at the studies on AdV-mediated animal hepatotoxicity, which will undoubtedly deepen the understanding of AdV-caused liver injury and be of benefit in the further safe development of AdVs.
Collapse
Affiliation(s)
- Zeng Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.
| | - Xiaozhan Zhang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| |
Collapse
|
37
|
Shieh WJ. Human adenovirus infections in pediatric population - an update on clinico-pathologic correlation. Biomed J 2021; 45:38-49. [PMID: 34506970 PMCID: PMC9133246 DOI: 10.1016/j.bj.2021.08.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 01/23/2023] Open
Abstract
Human adenoviruses can cause infections at any age but most commonly in pediatric population, especially in young children and infants. By the time of 10 years old, most children have had at least one episode of adenovirus infection. Adenoviruses can cause many symptoms similar to common cold, including rhinorrhea, fever, cough, and sore throat. Lower respiratory infections such as bronchitis, bronchiolitis, and pneumonia can be severe and even fatal. Other diseases such as conjunctivitis, gastroenteritis, cystitis, myocarditis, cardiomyopathy, and meningoencephalitis can also be associated with adenovirus infections. A variety of recent advancement of structural and molecular biology methods have revamped the taxonomy of adenoviruses and furthered our understanding of the diversity of related clinical diseases. Because of the wide spectrum and complexity of diseases associated with human adenovirus infections, the scope of this review is limited to basic virology and epidemiology of adenoviruses with a main focus on the clinico–pathologic correlation. Clinical manifestations and pathology of any infectious disease are always related; therefore, it is logical to review clinico–pathologic correlation within the specific disease entity caused by adenoviruses to better understand this common viral infection in pediatric population.
Collapse
Affiliation(s)
- Wun-Ju Shieh
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan.
| |
Collapse
|
38
|
Aydin M, Schellhorn S, Wirth S, Zhang W, Ehrhardt A. Human Species D Adenoviruses Isolated from Diarrheal Feces Show Low Infection Rates in Primary Nasal Epithelial Cells. CHILDREN (BASEL, SWITZERLAND) 2021; 8:563. [PMID: 34208817 PMCID: PMC8307086 DOI: 10.3390/children8070563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 01/20/2023]
Abstract
The importance of adenovirus (Ad) research is significantly increasing with respect to virotherapy for vaccine development, tumor, and gene therapy. Due to the different species and subtypes of this virus, the characterization of the biological significance of especially rare Ad is necessary. Previously, rare Ad types 70, 73, and 74 were originally isolated from fecal samples of immunocompromised patients and they represent recombinants of other Ad types. Here we investigated transduction experiments of these reporter gene tagged Ad types in primary cells exemplified by subject-derived primary nasal epithelial cells (NAEPCs). To analyze the transduction rates, we performed flow cytometry, quantitative polymerase chain reaction (PCR), and cytokine analyses 25 h post-infection. We found that, in contrast to Ad type 5 (as a positive control), the transduction rates of NAEPCs with Ad types 70, 73, and 74 were interestingly low. The major Ad receptor (coxsackievirus-adenovirus receptor and CD46) expression levels showed no significant change after infection with Ad types 70, 73 and 74. Moreover, Interleukin 6 (IL-6) was not released after in vitro Ad transduction. Due to the high risk of developing life-threatening complications in immunocompromised patients by these human species D Ads, even more attention needs to be investigated into the development of diagnostic and therapeutic concepts to prevent and treat those opportunistic infections in susceptible patients.
Collapse
Affiliation(s)
- Malik Aydin
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
- Center for Child and Adolescent Medicine, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
| | - Sebastian Schellhorn
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany; (S.S.); (W.Z.); (A.E.)
| | - Stefan Wirth
- Center for Child and Adolescent Medicine, Helios University Hospital Wuppertal, Witten/Herdecke University, 42283 Wuppertal, Germany;
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany; (S.S.); (W.Z.); (A.E.)
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany; (S.S.); (W.Z.); (A.E.)
| |
Collapse
|
39
|
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
|
40
|
Adenovirus Structure: What Is New? Int J Mol Sci 2021; 22:ijms22105240. [PMID: 34063479 PMCID: PMC8156859 DOI: 10.3390/ijms22105240] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Adenoviruses are large (~950 Å) and complex non-enveloped, dsDNA icosahedral viruses. They have a pseudo-T = 25 triangulation number with at least 12 different proteins composing the virion. These include the major and minor capsid proteins, core proteins, maturation protease, terminal protein, and packaging machinery. Although adenoviruses have been studied for more than 60 years, deciphering their architecture has presented a challenge for structural biology techniques. An outstanding event was the first near-atomic resolution structure of human adenovirus type 5 (HAdV-C5), solved by cryo-electron microscopy (cryo-EM) in 2010. Discovery of new adenovirus types, together with methodological advances in structural biology techniques, in particular cryo-EM, has lately produced a considerable amount of new, high-resolution data on the organization of adenoviruses belonging to different species. In spite of these advances, the organization of the non-icosahedral core is still a great unknown. Nevertheless, alternative techniques such as atomic force microscopy (AFM) are providing interesting glimpses on the role of the core proteins in genome condensation and virion stability. Here we summarize the current knowledge on adenovirus structure, with an emphasis on high-resolution structures obtained since 2010.
Collapse
|
41
|
Adenovirus - a blueprint for gene delivery. Curr Opin Virol 2021; 48:49-56. [PMID: 33892224 DOI: 10.1016/j.coviro.2021.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 11/23/2022]
Abstract
A central quest in gene therapy and vaccination is to achieve effective and long-lasting gene expression at minimal dosage. Adenovirus vectors are widely used therapeutics and safely deliver genes into many cell types. Adenoviruses evolved to use elaborate trafficking and particle deconstruction processes, and efficient gene expression and progeny formation. Here, we discuss recent insights into how human adenoviruses deliver their double-stranded DNA genome into cell nuclei, and effect lytic cell killing, non-lytic persistent infection or vector gene expression. The mechanisms underlying adenovirus entry, uncoating, nuclear transport and gene expression provide a blueprint for the emerging field of synthetic virology, where artificial virus-like particles are evolved to deliver therapeutic payload into human cells without viral proteins and genomes.
Collapse
|
42
|
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
|