1
|
Van Campenhout R, Vinken M. Hepatic cell junctions: Pulling a double-duty. Liver Int 2024; 44:2873-2889. [PMID: 39115254 DOI: 10.1111/liv.16045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 10/25/2024]
Abstract
Cell junctions, including anchoring, occluding and communicating junctions, play an indispensable role in the structural and functional organization of multicellular tissues, including in liver. Specifically, hepatic cell junctions mediate intercellular adhesion and communication between liver cells. The establishment of the hepatic cell junction network is a prerequisite for normal liver functioning. Hepatic cell junctions indeed support liver-specific features and control essential aspects of the hepatic life cycle. This review paper summarizes the role of cell junctions and their components in relation to liver physiology, thereby also discussing their involvement in hepatic dysfunctionality, including liver disease and toxicity.
Collapse
Affiliation(s)
- Raf Van Campenhout
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
2
|
Effantin G, Hograindleur MA, Fenel D, Fender P, Vassal-Stermann E. Toward the understanding of DSG2 and CD46 interaction with HAdV-11 fiber, a super-complex analysis. J Virol 2023; 97:e0091023. [PMID: 37921471 PMCID: PMC10688334 DOI: 10.1128/jvi.00910-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 11/04/2023] Open
Abstract
IMPORTANCE The main limitation of oncolytic vectors is neutralization by blood components, which prevents intratumoral administration to patients. Enadenotucirev, a chimeric HAdV-11p/HAdV-3 adenovirus identified by bio-selection, is a low seroprevalence vector active against a broad range of human carcinoma cell lines. At this stage, there's still some uncertainty about tropism and primary receptor utilization by HAdV-11. However, this information is very important, as it has a direct influence on the effectiveness of HAdV-11-based vectors. The aim of this work is to determine which of the two receptors, DSG2 and CD46, is involved in the attachment of the virus to the host, and what role they play in the early stages of infection.
Collapse
Affiliation(s)
| | | | - Daphna Fenel
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Pascal Fender
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | |
Collapse
|
3
|
Meyer KJ, Pellack D, Hedberg-Buenz A, Pomernackas N, Soukup D, Wang K, Fingert JH, Anderson MG. Recombinant adenovirus causes prolonged mobilization of macrophages in the anterior chambers of mice. Mol Vis 2021; 27:741-756. [PMID: 35136346 PMCID: PMC8763664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 12/28/2021] [Indexed: 12/04/2022] Open
Abstract
PURPOSE Ocular tissues of mice have been studied in many ways using replication-deficient species C type 5 adenovirus (Ad5) as a tool for manipulating gene expression. Whereas refinements to injection protocols and tropism have led to several advances in targeting cells of interest, there remains a relative lack of information concerning how Ad5 may influence other ocular cell types capable of confounding experimental interpretation. Here, a slit lamp is used to thoroughly photodocument the sequelae of intraocular Ad5 injections over time in mice, with attention to potentially confounding indices of inflammation. METHODS A cohort of C57BL/6J mice was randomly split into three groups (Virus, receiving unilateral intracameral injection with 5×107 plaque-forming units (pfu) of a cargo-less Ad5 construct; Saline, receiving unilateral balanced salt solution injection; and Naïve, receiving no injections). From this initial experiment, a total of 52 eyes from 26 mice were photodocumented via slit lamp at four time points (baseline and 1, 3, and 10 weeks following initiation of the experiment) by an observer masked to treatments and other parameters of the experimental design. Following the last in vivo exam, tissues were collected. Based on the slit-lamp data, tissues were studied via immunostaining with the macrophage marker F4/80. Subsequently, three iterations of the original experiment were performed with otherwise identical experimental parameters testing the effect of age, intravitreal injection, and A195 buffer, adding slit-lamp photodocumentation of an additional 32 eyes from 16 mice. RESULTS The masked investigator could use the sequential images from each mouse in the initial experiment to assign each mouse to its correct treatment group with near perfect fidelity. Virus-injected eyes were characterized by corneal damage indicative of intraocular injection and a prolonged mobilization of clump cells on the surface of the iris. Saline-injected eyes had only transient corneal opacities indicative of intraocular injections, and Naïve eyes remained normal. Immunostaining with F4/80 was consistent with ascribing the clump cells visualized via slit-lamp imaging as a type of macrophage. Experimental iterations using Ad5 indicate that all virus-injected eyes had the distinguishing feature of a prolonged presence of clump cells on the surface of the iris regardless of injection site. Mice receiving an intraocular injection of Ad5 at an advanced age displayed a protracted course of corneal cloudiness that prevented detailed visualization of the iris at the last time point. CONCLUSIONS Because the eye is often considered an "immune privileged site," we suspect that several studies have neglected to consider that the presence of Ad5 in the eye might evoke strong reactions from the innate immune system. Ad5 injection caused a sustained mobilization of clump cells-that is, macrophages. This change is likely a consequence of either direct macrophage transduction or a secondary response to cytokines produced locally by other transduced cells. Regardless of how these cells were altered, the important implication is that the adenovirus led to long-lasting changes in the environment of the anterior chamber. Thus, these findings describe a caveat of Ad5-mediated studies involving macrophage mobilization, which we encourage groups to use as a bioassay in their experiments and consider in interpretation of their ongoing experiments using adenoviruses.
Collapse
Affiliation(s)
- Kacie J. Meyer
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | - Danielle Pellack
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | - Adam Hedberg-Buenz
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA
| | - Nicholas Pomernackas
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | - Dana Soukup
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
| | - Kai Wang
- Department of Biostatistics, University of Iowa, Iowa City, IA
| | - John H. Fingert
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA
| | - Michael G. Anderson
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA
- VA Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA
| |
Collapse
|
4
|
Wang Z, Kang B, Gao Q, Huang L, Di J, Fan Y, Yu J, Jiang B, Gao F, Wang D, Sun H, Gu Y, Li J, Su X. Quadruple-editing of the MAPK and PI3K pathways effectively blocks the progression of KRAS-mutated colorectal cancer cells. Cancer Sci 2021; 112:3895-3910. [PMID: 34185934 PMCID: PMC8409416 DOI: 10.1111/cas.15049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Mutated KRAS promotes the activation of the MAPK pathway and the progression of colorectal cancer (CRC) cells. Aberrant activation of the PI3K pathway strongly attenuates the efficacy of MAPK suppression in KRAS‐mutated CRC. The development of a novel strategy targeting a dual pathway is therefore highly essential for the therapy of KRAS‐mutated CRC. In this study, a quadruple‐depleting system for the KRAS, MEK1, PIK3CA, and MTOR genes based on CRISPR/SaCas9 was developed. Adenovirus serotype 5 (ADV5) was integrated with two engineered proteins, an adaptor and a protector, to form ADV‐protein complex (APC) for systemic delivery of the CRISPR system. Quadruple‐editing could significantly inhibit the MAPK and PI3K pathways in CRC cells with oncogenic mutations of KRAS and PIK3CA or with KRAS mutation and compensated PI3K activation. Compared with MEK and PI3K/MTOR inhibitors, quadruple‐editing induced more significant survival inhibition on primary CRC cells with oncogenic mutations of KRAS and PIK3CA. The adaptor specifically targeting EpCAM and the hexon‐shielding protector could dramatically enhance ADV5 infection efficiency to CRC cells and significantly reduce off‐targeting tropisms to many organs except the colon. Moreover, quadruple‐editing intravenously delivered by APC significantly blocked the dual pathway and tumor growth of KRAS‐mutated CRC cells, without influencing normal tissues in cell‐ and patient‐derived xenograft models. Therefore, APC‐delivered quadruple‐editing of the MAPK and PI3K pathways shows a promising therapeutic potential for KRAS‐mutated CRC.
Collapse
Affiliation(s)
- Zaozao Wang
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | | | | | | | - Jiabo Di
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yingcong Fan
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianhong Yu
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Beihai Jiang
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | | | | | | | - Ying Gu
- BGI-Shenzhen, Shenzhen, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiangqian Su
- Department of Gastrointestinal Surgery IV, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
5
|
Zhao Y, Liu Z, Li L, Wu J, Zhang H, Zhang H, Lei T, Xu B. Oncolytic Adenovirus: Prospects for Cancer Immunotherapy. Front Microbiol 2021; 12:707290. [PMID: 34367111 PMCID: PMC8334181 DOI: 10.3389/fmicb.2021.707290] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/21/2021] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy has moved to the forefront of modern oncologic treatment in the past few decades. Various forms of immunotherapy currently are emerging, including oncolytic viruses. In this therapy, viruses are engineered to selectively propagate in tumor cells and reduce toxicity for non-neoplastic tissues. Adenovirus is one of the most frequently employed oncolytic viruses because of its capacity in tumor cell lysis and immune response stimulation. Upregulation of immunostimulatory signals induced by oncolytic adenoviruses (OAds) might significantly remove local immune suppression and amplify antitumor immune responses. Existing genetic engineering technology allows us to design OAds with increasingly better tumor tropism, selectivity, and antitumor efficacy. Several promising strategies to modify the genome of OAds have been applied: capsid modifications, small deletions in the pivotal viral genes, insertion of tumor-specific promoters, and addition of immunostimulatory transgenes. OAds armed with tumor-associated antigen (TAA) transgenes as cancer vaccines provide additional therapeutic strategies to trigger tumor-specific immunity. Furthermore, the combination of OAds and immune checkpoint inhibitors (ICIs) increases clinical benefit as evidence shown in completed and ongoing clinical trials, especially in the combination of OAds with antiprogrammed death 1/programed death ligand 1 (PD-1/PD-L1) therapy. Despite remarkable antitumor potency, oncolytic adenovirus immunotherapy is confronted with tough challenges such as antiviral immune response and obstruction of tumor microenvironment (TME). In this review, we focus on genomic modification strategies of oncolytic adenoviruses and applications of OAds in cancer immunotherapy.
Collapse
Affiliation(s)
- Yaqi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheming Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huibo Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haohan Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianyu Lei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Adenoviral Vector DNA- and SARS-CoV-2 mRNA-Based Covid-19 Vaccines: Possible Integration into the Human Genome - Are Adenoviral Genes Expressed in Vector-based Vaccines? Virus Res 2021; 302:198466. [PMID: 34087261 PMCID: PMC8168329 DOI: 10.1016/j.virusres.2021.198466] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 11/22/2022]
Abstract
Vigorous vaccination programs against SARS-CoV-2-causing Covid-19 are the major chance to fight this dreadful pandemic. The currently administered vaccines depend on adenovirus DNA vectors or on SARS-CoV-2 mRNA that might become reverse transcribed into DNA, however infrequently. In some societies, people have become sensitized against the potential short- or long-term side effects of foreign DNA being injected into humans. In my laboratory, the fate of foreign DNA in mammalian (human) cells and organisms has been investigated for many years. In this review, a summary of the results obtained will be presented. This synopsis has been put in the evolutionary context of retrotransposon insertions into pre-human genomes millions of years ago. In addition, studies on adenovirus vector-based DNA, on the fate of food-ingested DNA as well as the long-term persistence of SARS-CoV-2 RNA/DNA will be described. Actual integration of viral DNA molecules and of adenovirus vector DNA will likely be chance events whose frequency and epigenetic consequences cannot with certainty be assessed. The review also addresses problems of remaining adenoviral gene expression in adenoviral-based vectors and their role in side effects of vaccines. Eventually, it will come down to weighing the possible risks of genomic insertions of vaccine-associated foreign DNA and unknown levels of vector-carried adenoviral gene expression versus protection against the dangers of Covid-19. A decision in favor of vaccination against life-threatening disease appears prudent. Informing the public about the complexities of biology will be a reliable guide when having to reach personal decisions about vaccinations.
Collapse
|
7
|
Gao J, Zhang W, Mese K, Bunz O, Lu F, Ehrhardt A. Transient Chimeric Ad5/37 Fiber Enhances NK-92 Carrier Cell-Mediated Delivery of Oncolytic Adenovirus Type 5 to Tumor Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:376-389. [PMID: 32695840 PMCID: PMC7358217 DOI: 10.1016/j.omtm.2020.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023]
Abstract
Methods for customizing and improving virus vector tropism are limited. In this study, we introduce a microRNA (miRNA)-regulated molecular method to enhance vector transduction without genome alteration. Based on the importance of adenovirus (Ad) vectors for cancer and gene treatment, we exemplified this technology for an Ad type 5 (Ad5) vector temporally carrying a knob from Ad37. We constructed a producer cell line stably expressing a fused Ad5/37 chimeric fiber comprising the Ad5 shaft-tail and the Ad37 knob and a miRNA inhibiting Ad5 knob expression (HEK293-Ad5/37-miRNA). The chimeric Ad5/37 vector resulted in enhanced transduction rates in Ad37 adequately and Ad5 poorly transduced cells. Particularly, encapsidation of the oncolytic Ad5-human telomerase reverse transcriptase (hTERT) vector genome into the chimeric Ad5/37 capsid showed efficient transduction of NK-92 carrier cells. These infected carrier cells then delivered the oncolytic vector to tumor cells, which resulted in enhanced Ad5-hTERT-mediated tumor cell killing. We show that this transiently capsid-modified chimeric vector carrying an Ad5 genome displayed higher transduction efficiencies of natural killer cell-derived NK-92 cells utilized as carriers in cancer immune therapy. In summary, transiently modified adenoviral vectors will have important implications for cancer and gene therapy.
Collapse
Affiliation(s)
- Jian Gao
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Kemal Mese
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Oskar Bunz
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Fengmin Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Department of Human Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| |
Collapse
|
8
|
Pan Q, Wang J, Gao Y, Wang Q, Cui H, Liu C, Qi X, Zhang Y, Wang Y, Li K, Gao L, Liu A, Wang X. Identification of chicken CAR homology as a cellular receptor for the emerging highly pathogenic fowl adenovirus 4 via unique binding mechanism. Emerg Microbes Infect 2020; 9:586-596. [PMID: 32174269 PMCID: PMC7144210 DOI: 10.1080/22221751.2020.1736954] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Since 2015, the prevalence of severe hepatitis-hydropericardium syndrome, which is caused by the novel genotype fowl adenovirus serotype 4 (FAdV-4), has increased in China and led to considerable economic losses. The replication cycle of FAdV-4, especially the emerging highly pathogenic novel genotype FAdV-4, remains largely unknown. The adenovirus fibre interacts with the cellular receptor as the initial step in adenovirus (AdV) infection. In our previous studies, the complete genome sequence showed that the fibre patterns of FAdV-4 were distinct from all other AdVs. Here, protein-blockage and antibody-neutralization assays were performed to confirm that the novel FAdV-4 short fibre was critical for binding to susceptible leghorn male hepatocellular (LMH) cells. Subsequently, fibre 1 was used as bait to investigate the receptor on LMH cells via mass spectrometry. The chicken coxsackie and adenovirus receptor (CAR) protein was confirmed as the novel FAdV-4 receptor in competition assays. We further identified the D2 domain of CAR (D2-CAR) as the active domain responsible for binding to the short fibre of the novel FAdV-4. Taken together, these findings demonstrate for the first time that the chicken CAR homolog is a cellular receptor for the novel FAdV-4, which facilitates viral entry by interacting with the viral short fibre through the D2 domain. Collectively, these findings provide an in-depth understanding of the mechanisms of the emerging novel genotype FAdV-4 invasion and pathogenesis.
Collapse
Affiliation(s)
- Qing Pan
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Jing Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yulong Gao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Qi Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Hongyu Cui
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Changjun Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xiaole Qi
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yanping Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yongqiang Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Kai Li
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Li Gao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Aijing Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Xiaomei Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
9
|
Human Adenovirus Serotype 5 Is Sensitive to IgM-Independent Neutralization In Vitro and In Vivo. Viruses 2019; 11:v11070616. [PMID: 31284434 PMCID: PMC6669743 DOI: 10.3390/v11070616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/23/2019] [Accepted: 07/03/2019] [Indexed: 01/03/2023] Open
Abstract
Human adenovirus 5 (HAdV-5) is used as a vector in gene therapy clinical trials, hence its interactions with the host immune system have been widely studied. Previous studies have demonstrated that HAdV-5 binds specifically to murine coagulation factor X (mFX), inhibiting IgM and complement-mediated neutralization. Here, we examined the physical binding of immune components to HAdV-5 by nanoparticle tracking analysis, neutralization assays, mass spectrometry analysis and in vivo experiments. We observed that purified mouse Immunoglobulin M (IgM) antibodies bound to HAdV-5 only in the presence of complement components. Active serum components were demonstrated to bind to HAdV-5 in the presence or absence of mFX, indicating that immune molecules and mFX might bind to different sites. Since binding of mFX to HAdV-5 blocks the neutralization cascade, these findings suggested that not all complement-binding sites may be involved in virion neutralization. Furthermore, the data obtained from serum neutralization experiments suggested that immune molecules other than IgM and IgG may trigger activation of the complement cascade in vitro. In vivo experiments were conducted in immunocompetent C57BL/6 or immuno-deficient Rag2-/- mice. HAdV-5T* (a mutant HAdV-5 unable to bind to human or mFX) was neutralized to some extent in both mouse models, suggesting that murine immunoglobulins were not required for neutralization of HAdV-5 in vivo. Liquid Chromatography-Mass Spectrometry (LC-MS/MS) analysis of HAdV-5 and HAdV-5T* after exposure to murine sera showed stable binding of C3 and C4b in the absence of mFX. In summary, these results suggest that HAdV-5 neutralization can be mediated by both the classical and alternative pathways and that, in the absence of immunoglobulins, the complement cascade can be activated by direct binding of C3 to the virion.
Collapse
|
10
|
Stepanenko AA, Chekhonin VP. Tropism and transduction of oncolytic adenovirus 5 vectors in cancer therapy: Focus on fiber chimerism and mosaicism, hexon and pIX. Virus Res 2018; 257:40-51. [PMID: 30125593 DOI: 10.1016/j.virusres.2018.08.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 02/09/2023]
Abstract
The cellular internalization (infection of cells) of adenovirus 5 (Ad5) is mediated by the initial attachment of the globular knob domain of the capsid fiber protein to the cell surface coxsackievirus and adenovirus receptor (CAR), then followed by the interaction of the virus penton base proteins with cellular integrins. In tumors, there is a substantial intra- and intertumoral variability in CAR expression. The CAR-negative cells generally exhibit very low infectability. Since the fiber knob is a primary mediator of Ad5 binding to the cell surface, improved infectivity of Ad5-based vectors as oncolytic agents may be achieved via genetic modifications of this domain. The strategies to modify or broaden tropism and increase transduction efficiency of Ad5-based vectors include: 1) an incorporation of a targeting peptide into the fiber knob domain (the HI loop and/or C-terminus); 2) fiber knob serotype switching, or pseudotyping, by constructing chimeric fibers consisting of the knob domain derived from an alternate serotype (e.g., Ad5/3 or Ad5/35 chimeras), which binds to receptor(s) other than CAR (e.g., desmoglein 2/DSG2 and/or CD46); 3) "fiber complex mosaicism", an approach of combining serotype chimerism with peptide ligand(s) incorporation (e.g., Ad5/3-RGD); 4) "dual fiber mosaicism" by expressing two separate fibers with distinct receptor-binding capabilities on the same viral particle (e.g., Ad5-5/3 or Ad5-5/σ1); 5) fiber xenotyping by replacing the knob and shaft domains of wild-type Ad5 fiber protein with fibritin trimerization domain of T4 bacteriophage or σ1 attachment protein of reovirus. Other genetic approaches to increase the CAR-independent transduction efficiency include insertion of a targeting peptide into the hypervariable region of the capsid protein hexon or fusion to the C-terminus of pIX. Finally, we consider a yet unsolved molecular mechanism of liver targeting by Ad5-based vectors (CAR-, integrin-, fiber shaft KKTK motif-, and hepatic heparan sulfate glycosaminoglycans-independent, but fiber-, hexon- and blood factor X-dependent).
Collapse
Affiliation(s)
- Aleksei A Stepanenko
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky lane 23, 119034 Moscow, Russia.
| | - Vladimir P Chekhonin
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky lane 23, 119034 Moscow, Russia; Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov str. 1, 117997 Moscow, Russia.
| |
Collapse
|
11
|
Schmid M, Ernst P, Honegger A, Suomalainen M, Zimmermann M, Braun L, Stauffer S, Thom C, Dreier B, Eibauer M, Kipar A, Vogel V, Greber UF, Medalia O, Plückthun A. Adenoviral vector with shield and adapter increases tumor specificity and escapes liver and immune control. Nat Commun 2018; 9:450. [PMID: 29386504 PMCID: PMC5792622 DOI: 10.1038/s41467-017-02707-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/20/2017] [Indexed: 01/16/2023] Open
Abstract
Most systemic viral gene therapies have been limited by sequestration and degradation of virions, innate and adaptive immunity, and silencing of therapeutic genes within the target cells. Here we engineer a high-affinity protein coat, shielding the most commonly used vector in clinical gene therapy, human adenovirus type 5. Using electron microscopy and crystallography we demonstrate a massive coverage of the virion surface through the hexon-shielding scFv fragment, trimerized to exploit the hexon symmetry and gain avidity. The shield reduces virion clearance in the liver. When the shielded particles are equipped with adaptor proteins, the virions deliver their payload genes into human cancer cells expressing HER2 or EGFR. The combination of shield and adapter also increases viral gene delivery to xenografted tumors in vivo, reduces liver off-targeting and immune neutralization. Our study highlights the power of protein engineering for viral vectors overcoming the challenges of local and systemic viral gene therapies. Viral gene therapy can be limited by the efficacy of virion sequestration, immune responses and the silencing of genetic payloads. Here the authors engineer an advenovirus protein coat which shields the virion from the immune system while targeting cancer cells.
Collapse
Affiliation(s)
- Markus Schmid
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Patrick Ernst
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Annemarie Honegger
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Maarit Suomalainen
- Department of Molecular Life Science, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Martina Zimmermann
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Lukas Braun
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, 8093, Zurich, Switzerland
| | - Sarah Stauffer
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Cristian Thom
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, 8057, Zurich, Switzerland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, 8093, Zurich, Switzerland
| | - Urs F Greber
- Department of Molecular Life Science, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland.,Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, 84105, Israel
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland.
| |
Collapse
|