1
|
Maalouf AA, Zhu H, Zaman A, Carpino N, Hearing J, Bhatia SR, Carrico IS. Facile Conjugation Method of CpG-ODN Adjuvant to Intact Virions for Vaccine Development. Chembiochem 2025:e2400988. [PMID: 40164565 DOI: 10.1002/cbic.202400988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Vaccines are a pivotal achievement in public health, offering inexpensive, distributable, and highly effective protection against infectious diseases. Despite significant advancements in vaccine development, there are still many diseases for which vaccines are unavailable or offer limited protection. The global impact of the deficiency in vaccine-induced immunity against these diseases is profound, leading to increased rates of illness, more frequent hospitalizations, and higher mortality rates. Recent studies have demonstrated conjugation mechanisms and delivery methods to copresent adjuvants and protein epitopes to antigen-presenting cells, significantly enhancing adaptive immunity A novel approach is introduced to incorporate an adjuvant into the vaccine by covalently attaching it to whole enveloped virions. Using "clickable" azide-enabled viral particles, generated through metabolic incorporation of N-azidoacetyl glucosamine (GlcNAz), the virions with a cyclo-octyne-modified CpG-ODN is conjugated. Conjugation yielded a potent adjuvant-virus complex, eliciting higher TLR9-mediated cell activation of cultured bone marrow-derived macrophages relative to coadministered adjuvants and virions. Administration of covalent adjuvant-virion conjugates increases immune cell stimulation and may provide a generalizable and effective strategy for eliciting a heightened immune response for vaccine development.
Collapse
Affiliation(s)
- Alexandra A Maalouf
- Department of Chemistry and Institute of Chemical Biology and Drug Discovery, State University of New York Stony Brook, Stony Brook, New York, 11794-3400, USA
| | - Hengwei Zhu
- Department of Chemistry and Institute of Chemical Biology and Drug Discovery, State University of New York Stony Brook, Stony Brook, New York, 11794-3400, USA
| | - Anika Zaman
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, 11794-3400, USA
| | - Neena Carpino
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, 11794-3400, USA
| | - Janet Hearing
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, 11794-3400, USA
| | - Surita R Bhatia
- Department of Chemistry and Institute of Chemical Biology and Drug Discovery, State University of New York Stony Brook, Stony Brook, New York, 11794-3400, USA
| | - Isaac S Carrico
- Department of Chemistry and Institute of Chemical Biology and Drug Discovery, State University of New York Stony Brook, Stony Brook, New York, 11794-3400, USA
| |
Collapse
|
2
|
Tilden JAR, Doud EA, Montgomery HR, Maynard HD, Spokoyny AM. Organometallic Chemistry Tools for Building Biologically Relevant Nanoscale Systems. J Am Chem Soc 2024; 146:29989-30003. [PMID: 39468851 PMCID: PMC12009178 DOI: 10.1021/jacs.4c07110] [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] [Indexed: 10/30/2024]
Abstract
The recent emergence of organometallic chemistry for modification of biomolecular nanostructures has begun to rewrite the long-standing assumption among practitioners that small-molecule organometallics are fundamentally incompatible with biological systems. This Perspective sets out to clarify some of the existing misconceptions by focusing on the growing organometallic toolbox for biomolecular modification. Specifically, we highlight key organometallic transformations in constructing complex biologically relevant systems on the nanomolecular scale, and the organometallic synthesis of hybrid nanomaterials composed of classical nanomaterial components combined with biologically relevant species. As research progresses, many of the challenges associated with applying organometallic chemistry in this context are rapidly being reassessed. Looking to the future, the growing utility of organometallic transformations will likely make them more ubiquitous in the construction and modification of biomolecular nanostructures.
Collapse
Affiliation(s)
- James A. R. Tilden
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Evan A. Doud
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Hayden R. Montgomery
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Alexander M. Spokoyny
- Department of Chemistry and Biochemistry, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Ding J, Su R, Yang R, Xu J, Liu X, Yao T, Li S, Wang C, Zhang H, Yue Q, Zhan C, Li C, Gao X. Enhancing the Antitumor Efficacy of Oncolytic Adenovirus Through Sonodynamic Therapy-Augmented Virus Replication. ACS NANO 2024; 18:18282-18298. [PMID: 38953884 DOI: 10.1021/acsnano.4c01115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The therapeutic efficacy of oncolytic adenoviruses (OAs) relies on efficient viral transduction and replication. However, the limited expression of coxsackie-adenovirus receptors in many tumors, along with the intracellular antiviral signaling, poses significant obstacles to OA infection and oncolysis. Here, we present sonosensitizer-armed OAs (saOAs) that potentiate the antitumor efficacy of oncolytic virotherapy through sonodynamic therapy-augmented virus replication. The saOAs could not only efficiently infect tumor cells via transferrin receptor-mediated endocytosis but also exhibit enhanced viral replication and tumor oncolysis under ultrasound irradiation. We revealed that the sonosensitizer loaded on the viruses induced the generation of ROS within tumor cells, which triggered JNK-mediated autophagy, ultimately leading to the enhanced viral replication. In mouse models of malignant melanoma, the combination of saOAs and sonodynamic therapy elicited a robust antitumor immune response, resulting in significant inhibition of melanoma growth and improved host survival. This work highlights the potential of sonodynamic therapy in enhancing the effectiveness of OAs and provides a promising platform for fully exploiting the antitumor efficacy of oncolytic virotherapy.
Collapse
Affiliation(s)
- Junqiang Ding
- School of Pharmacy, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Runping Su
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Rong Yang
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Jinliang Xu
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Xiaoxiao Liu
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Tingting Yao
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Sha Li
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Cong Wang
- School of Pharmacy, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Hanchang Zhang
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Qi Yue
- Department of Neurosurgery, Huashan Hospital, Fudan University, 12 Urumqi Middle Road, Shanghai 200040, China
| | - Changyou Zhan
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| | - Cong Li
- School of Pharmacy, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xihui Gao
- Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences & Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, 131 Dong An Road, Shanghai 200032, China
| |
Collapse
|
4
|
Dicks MD, Rose LM, Russell RA, Bowman LA, Graham C, Jimenez-Guardeño JM, Doores KJ, Malim MH, Draper SJ, Howarth M, Biswas S. Modular capsid decoration boosts adenovirus vaccine-induced humoral immunity against SARS-CoV-2. Mol Ther 2022; 30:3639-3657. [PMID: 35949171 PMCID: PMC9364715 DOI: 10.1016/j.ymthe.2022.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/18/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022] Open
Abstract
Adenovirus vector vaccines have been widely and successfully deployed in response to coronavirus disease 2019 (COVID-19). However, despite inducing potent T cell immunity, improvement of vaccine-specific antibody responses upon homologous boosting is modest compared with other technologies. Here, we describe a system enabling modular decoration of adenovirus capsid surfaces with antigens and demonstrate potent induction of humoral immunity against these displayed antigens. Ligand attachment via a covalent bond was achieved using a protein superglue, DogTag/DogCatcher (similar to SpyTag/SpyCatcher), in a rapid and spontaneous reaction requiring only co-incubation of ligand and vector components. DogTag was inserted into surface-exposed loops in the adenovirus hexon protein to allow attachment of DogCatcher-fused ligands on virus particles. Efficient coverage of the capsid surface was achieved using various ligands, with vector infectivity retained in each case. Capsid decoration shielded particles from vector neutralizing antibodies. In prime-boost regimens, adenovirus vectors decorated with the receptor-binding domain of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike induced >10-fold higher SARS-CoV-2 neutralization titers compared with an undecorated vector encoding spike. Importantly, decorated vectors achieved equivalent or superior T cell immunogenicity against encoded antigens compared with undecorated vectors. We propose capsid decoration using protein superglues as a novel strategy to improve efficacy and boostability of adenovirus-based vaccines and therapeutics.
Collapse
Affiliation(s)
- Matthew D.J. Dicks
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK,Corresponding author: Matthew D. J. Dicks, SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK.
| | - Louisa M. Rose
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK
| | - Rebecca A. Russell
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK
| | - Lesley A.H. Bowman
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK
| | - Carl Graham
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Jose M. Jimenez-Guardeño
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Katie J. Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Michael H. Malim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Simon J. Draper
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK,Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Mark Howarth
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK,Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Sumi Biswas
- SpyBiotech Ltd, 7600 The Quorum, Oxford Business Park North, Oxford, OX4 2JZ, UK,The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
5
|
Edwardson TGW, Levasseur MD, Tetter S, Steinauer A, Hori M, Hilvert D. Protein Cages: From Fundamentals to Advanced Applications. Chem Rev 2022; 122:9145-9197. [PMID: 35394752 DOI: 10.1021/acs.chemrev.1c00877] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteins that self-assemble into polyhedral shell-like structures are useful molecular containers both in nature and in the laboratory. Here we review efforts to repurpose diverse protein cages, including viral capsids, ferritins, bacterial microcompartments, and designed capsules, as vaccines, drug delivery vehicles, targeted imaging agents, nanoreactors, templates for controlled materials synthesis, building blocks for higher-order architectures, and more. A deep understanding of the principles underlying the construction, function, and evolution of natural systems has been key to tailoring selective cargo encapsulation and interactions with both biological systems and synthetic materials through protein engineering and directed evolution. The ability to adapt and design increasingly sophisticated capsid structures and functions stands to benefit the fields of catalysis, materials science, and medicine.
Collapse
Affiliation(s)
| | | | - Stephan Tetter
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Angela Steinauer
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Mao Hori
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
6
|
Ke X, Li C, Luo D, Wang T, Liu Y, Tan Z, Du M, He Z, Wang H, Zheng Z, Zhang Y. Metabolic labeling of enterovirus 71 with quantum dots for the study of virus receptor usage. J Nanobiotechnology 2021; 19:295. [PMID: 34583708 PMCID: PMC8477995 DOI: 10.1186/s12951-021-01046-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/16/2021] [Indexed: 11/10/2022] Open
Abstract
Fluorescent labeling and dynamic tracking is a powerful tool for exploring virus infection mechanisms. However, for small-sized viruses, virus tracking studies are usually hindered by a lack of appropriate labeling methods that do not dampen virus yield or infectivity. Here, we report a universal strategy for labeling viruses with chemical dyes and Quantum dots (QDs). Enterovirus 71 (EV71) was produced in a cell line that stably expresses a mutant methionyl-tRNA synthetase (MetRS), which can charge azidonorleucine (ANL) to the methionine sites of viral proteins during translation. Then, the ANL-containing virus was easily labeled with DBCO-AF647 and DBCO-QDs. The labeled virus shows sufficient yield and no obvious decrease in infectivity and can be used for imaging the virus entry process. Using the labeled EV71, different functions of scavenger receptor class B, member 2 (SCARB2), and heparan sulfate (HS) in EV71 infection were comparatively studied. The cell entry process of a strong HS-binding EV71 strain was investigated by real-time dynamic visualization of EV71-QDs in living cells. Taken together, our study described a universal biocompatible virus labeling method, visualized the dynamic viral entry process, and reported details of the receptor usage of EV71.
Collapse
Affiliation(s)
- Xianliang Ke
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Chunjie Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Dan Luo
- Department of Gastroenterology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430015, Wuhan, China
| | - Ting Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, 430100, China
| | - Yan Liu
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhongyuan Tan
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mingyuan Du
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, Wuhan, China
| | - Zhike He
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, Wuhan, China
| | - Hanzhong Wang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhenhua Zheng
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yuan Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
7
|
Heiss TK, Dorn RS, Prescher JA. Bioorthogonal Reactions of Triarylphosphines and Related Analogues. Chem Rev 2021; 121:6802-6849. [PMID: 34101453 PMCID: PMC10064493 DOI: 10.1021/acs.chemrev.1c00014] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bioorthogonal phosphines were introduced in the context of the Staudinger ligation over 20 years ago. Since that time, phosphine probes have been used in myriad applications to tag azide-functionalized biomolecules. The Staudinger ligation also paved the way for the development of other phosphorus-based chemistries, many of which are widely employed in biological experiments. Several reviews have highlighted early achievements in the design and application of bioorthogonal phosphines. This review summarizes more recent advances in the field. We discuss innovations in classic Staudinger-like transformations that have enabled new biological pursuits. We also highlight relative newcomers to the bioorthogonal stage, including the cyclopropenone-phosphine ligation and the phospha-Michael reaction. The review concludes with chemoselective reactions involving phosphite and phosphonite ligations. For each transformation, we describe the overall mechanism and scope. We also showcase efforts to fine-tune the reagents for specific functions. We further describe recent applications of the chemistries in biological settings. Collectively, these examples underscore the versatility and breadth of bioorthogonal phosphine reagents.
Collapse
|
8
|
Destito P, Vidal C, López F, Mascareñas JL. Transition Metal‐Promoted Reactions in Aqueous Media and Biological Settings. Chemistry 2021; 27:4789-4816. [DOI: 10.1002/chem.202003927] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/27/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Paolo Destito
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Cristian Vidal
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Fernando López
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
- Instituto de Química Orgánica General (CSIC) Juan de la Cierva 3 28006 Madrid Spain
| | - José L. Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais, Moleculares (CIQUS) and Departamento de Química Orgánica Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| |
Collapse
|
9
|
Barry MA, Rubin JD, Lu SC. Retargeting adenoviruses for therapeutic applications and vaccines. FEBS Lett 2020; 594:1918-1946. [PMID: 31944286 PMCID: PMC7311308 DOI: 10.1002/1873-3468.13731] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022]
Abstract
Adenoviruses (Ads) are robust vectors for therapeutic applications and vaccines, but their use can be limited by differences in their in vitro and in vivo pharmacologies. This review emphasizes that there is not just one Ad, but a whole virome of diverse viruses that can be used as therapeutics. It discusses that true vector targeting involves not only retargeting viruses, but importantly also detargeting the viruses from off-target cells.
Collapse
Affiliation(s)
- Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Department of Immunology, Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey D Rubin
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Shao-Chia Lu
- Virology and Gene Therapy Graduate Program, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Goradel NH, Negahdari B, Ghorghanlu S, Jahangiri S, Arashkia A. Strategies for enhancing intratumoral spread of oncolytic adenoviruses. Pharmacol Ther 2020; 213:107586. [PMID: 32479843 DOI: 10.1016/j.pharmthera.2020.107586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Oncolytic viruses, effectively replicate viruses within malignant cells to lyse them without affecting normal ones, have recently shown great promise in developing therapeutic options for cancer. Adenoviruses (Ads) are one of the candidates in oncolytic virotheraoy due to its easily manipulated genomic DNA and expression of wide rane of its receptors on the various cancers. Although systematic delivery of oncolytic adenoviruses can target both primary and metastatic tumors, there are some drawbacks in the effective systematic delivery of oncolytic adenoviruses, including pre-existing antibodies and liver tropism. To overcome these limitations, intratumural (IT) administration of oncolytic viruses have been proposed. However, IT injection of Ads leaves much of the tumor mass unaffected and Ads are not able to disperse more in the tumor microenvironment (TME). To this end, various strategies have been developed to enhance the IT spread of oncolytic adenoviruses, such as using extracellular matrix degradation enzymes, junction opening peptides, and fusogenic proteins. In the present paper, we reviewed different oncolytic adenoviruses, their application in the clinical trials, and strategies for enhancing their IT spread.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sajjad Ghorghanlu
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Jahangiri
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
11
|
Pied N, Wodrich H. Imaging the adenovirus infection cycle. FEBS Lett 2019; 593:3419-3448. [PMID: 31758703 DOI: 10.1002/1873-3468.13690] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022]
Abstract
Incoming adenoviruses seize control of cytosolic transport mechanisms to relocate their genome from the cell periphery to specialized sites in the nucleoplasm. The nucleus is the site for viral gene expression, genome replication, and the production of progeny for the next round of infection. By taking control of the cell, adenoviruses also suppress cell-autonomous immunity responses. To succeed in their production cycle, adenoviruses rely on well-coordinated steps, facilitated by interactions between viral proteins and cellular factors. Interactions between virus and host can impose remarkable morphological changes in the infected cell. Imaging adenoviruses has tremendously influenced how we delineate individual steps in the viral life cycle, because it allowed the development of specific optical markers to label these morphological changes in space and time. As technology advances, innovative imaging techniques and novel tools for specimen labeling keep uncovering previously unseen facets of adenovirus biology emphasizing why imaging adenoviruses is as attractive today as it was in the past. This review will summarize past achievements and present developments in adenovirus imaging centered on fluorescence microscopy approaches.
Collapse
Affiliation(s)
- Noémie Pied
- CNRS UMR 5234, Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, France
| | - Harald Wodrich
- CNRS UMR 5234, Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, France
| |
Collapse
|
12
|
Serwa RA, Sekine E, Brown J, Teo SHC, Tate EW, O’Hare P. Analysis of a fully infectious bio-orthogonally modified human virus reveals novel features of virus cell entry. PLoS Pathog 2019; 15:e1007956. [PMID: 31589653 PMCID: PMC6797222 DOI: 10.1371/journal.ppat.1007956] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/17/2019] [Accepted: 09/12/2019] [Indexed: 12/28/2022] Open
Abstract
We report the analysis of a complex enveloped human virus, herpes simplex virus (HSV), assembled after in vivo incorporation of bio-orthogonal methionine analogues homopropargylglycine (HPG) or azidohomoalanine (AHA). We optimised protocols for the production of virions incorporating AHA (termed HSVAHA), identifying conditions which resulted in normal yields of HSV and normal particle/pfu ratios. Moreover we show that essentially every single HSVAHA capsid-containing particle was detectable at the individual particle level by chemical ligation of azide-linked fluorochromes to AHA-containing structural proteins. This was a completely specific chemical ligation, with no capsids assembled under normal methionine-containing conditions detected in parallel. We demonstrate by quantitative mass spectrometric analysis that HSVAHA virions exhibit no qualitative or quantitative differences in the repertoires of structural proteins compared to virions assembled under normal conditions. Individual proteins and AHA incorporation sites were identified in capsid, tegument and envelope compartments, including major essential structural proteins. Finally we reveal novel aspects of entry pathways using HSVAHA and chemical fluorochrome ligation that were not apparent from conventional immunofluorescence. Since ligation targets total AHA-containing protein and peptides, our results demonstrate the presence of abundant AHA-labelled products in cytoplasmic macrodomains and tubules which no longer contain intact particles detectable by immunofluorescence. Although these do not co-localise with lysosomal markers, we propose they may represent sites of proteolytic virion processing. Analysis of HSVAHA also enabled the discrimination from primary entering from secondary assembling virions, demonstrating assembly and second round infection within 6 hrs of initial infection and dual infections of primary and secondary virus in spatially restricted cytoplasmic areas of the same cell. Together with other demonstrated applications e.g., in genome biology, lipid and protein trafficking, this work further exemplifies the utility and potential of bio-orthogonal chemistry for studies in many aspects of virus-host interactions.
Collapse
Affiliation(s)
- Remigiusz A. Serwa
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, London, United Kingdom
| | - Eiki Sekine
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jonathan Brown
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Su Hui Catherine Teo
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Edward W. Tate
- Department of Chemistry, Molecular Sciences Research Hub, White City Campus, London, United Kingdom
| | - Peter O’Hare
- Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Chen MY, Butler SS, Chen W, Suh J. Physical, chemical, and synthetic virology: Reprogramming viruses as controllable nanodevices. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1545. [PMID: 30411529 PMCID: PMC6461522 DOI: 10.1002/wnan.1545] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/03/2018] [Accepted: 10/04/2018] [Indexed: 01/24/2023]
Abstract
The fields of physical, chemical, and synthetic virology work in partnership to reprogram viruses as controllable nanodevices. Physical virology provides the fundamental biophysical understanding of how virus capsids assemble, disassemble, display metastability, and assume various configurations. Chemical virology considers the virus capsid as a chemically addressable structure, providing chemical pathways to modify the capsid exterior, interior, and subunit interfaces. Synthetic virology takes an engineering approach, modifying the virus capsid through rational, combinatorial, and bioinformatics-driven design strategies. Advances in these three subfields of virology aim to develop virus-based materials and tools that can be applied to solve critical problems in biomedicine and biotechnology, including applications in gene therapy and drug delivery, diagnostics, and immunotherapy. Examples discussed include mammalian viruses, such as adeno-associated virus (AAV), plant viruses, such as cowpea mosaic virus (CPMV), and bacterial viruses, such as Qβ bacteriophage. Importantly, research efforts in physical, chemical, and synthetic virology have further unraveled the design principles foundational to the form and function of viruses. This article is categorized under: Diagnostic Tools > Diagnostic Nanodevices Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
| | - Susan S Butler
- Department of Bioengineering, Rice University, Houston, Texas
| | - Weitong Chen
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, Texas
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, Texas
| |
Collapse
|
14
|
Chen G, Katrekar D, Mali P. RNA-Guided Adenosine Deaminases: Advances and Challenges for Therapeutic RNA Editing. Biochemistry 2019; 58:1947-1957. [PMID: 30943016 DOI: 10.1021/acs.biochem.9b00046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Targeted transcriptome engineering, in contrast to genome engineering, offers a complementary and potentially tunable and reversible strategy for cellular engineering. In this regard, adenosine to inosine (A-to-I) RNA base editing was recently engineered to make programmable base conversions on target RNAs. Similar to the DNA base editing technology, A-to-I RNA editing may offer an attractive alternative in a therapeutic setting, especially for the correction of point mutations. This Perspective introduces five currently characterized RNA editing systems and serves as a reader's guide for implementing an appropriate RNA editing strategy for applications in research or therapeutics.
Collapse
Affiliation(s)
- Genghao Chen
- Department of Bioengineering , University of California, San Diego , La Jolla , California 92093-0412 , United States
| | - Dhruva Katrekar
- Department of Bioengineering , University of California, San Diego , La Jolla , California 92093-0412 , United States
| | - Prashant Mali
- Department of Bioengineering , University of California, San Diego , La Jolla , California 92093-0412 , United States
| |
Collapse
|
15
|
Müller TG, Sakin V, Müller B. A Spotlight on Viruses-Application of Click Chemistry to Visualize Virus-Cell Interactions. Molecules 2019; 24:molecules24030481. [PMID: 30700005 PMCID: PMC6385038 DOI: 10.3390/molecules24030481] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/18/2019] [Accepted: 01/19/2019] [Indexed: 01/03/2023] Open
Abstract
The replication of a virus within its host cell involves numerous interactions between viral and cellular factors, which have to be tightly controlled in space and time. The intricate interplay between viral exploitation of cellular pathways and the intrinsic host defense mechanisms is difficult to unravel by traditional bulk approaches. In recent years, novel fluorescence microscopy techniques and single virus tracking have transformed the investigation of dynamic virus-host interactions. A prerequisite for the application of these imaging-based methods is the attachment of a fluorescent label to the structure of interest. However, their small size, limited coding capacity and multifunctional proteins render viruses particularly challenging targets for fluorescent labeling approaches. Click chemistry in conjunction with genetic code expansion provides virologists with a novel toolbox for site-specific, minimally invasive labeling of virion components, whose potential has just recently begun to be exploited. Here, we summarize recent achievements, current developments and future challenges for the labeling of viral nucleic acids, proteins, glycoproteins or lipids using click chemistry in order to study dynamic processes in virus-cell interactions.
Collapse
Affiliation(s)
- Thorsten G Müller
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Volkan Sakin
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Barbara Müller
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
16
|
Salem ESB, Murakami K, Takahashi T, Bernhard E, Borra V, Bethi M, Nakamura T. Isolation of Primary Mouse Hepatocytes for Nascent Protein Synthesis Analysis by Non-radioactive L-azidohomoalanine Labeling Method. J Vis Exp 2018. [PMID: 30417869 DOI: 10.3791/58323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatocytes are parenchymal cells of the liver and engage multiple metabolic functions, including synthesis and secretion of proteins essential for systemic energy homeostasis. Primary hepatocytes isolated from the murine liver constitute a valuable biological tool to understand the functional properties or alterations occurring in the liver. Herein we describe a method for the isolation and culture of primary mouse hepatocytes by performing a two-step collagenase perfusion technique and discuss their utilization for investigating protein metabolism. The liver of an adult mouse is sequentially perfused with ethylene glycol-bis tetraacetic acid (EGTA) and collagenase, followed by the isolation of hepatocytes with the density gradient buffer. These isolated hepatocytes are viable on culture plates and maintain the majority of endowed characteristics of hepatocytes. These hepatocytes can be used for assessments of protein metabolism including nascent protein synthesis with non-radioactive reagents. We show that the isolated hepatocytes are readily controlled and comprise a higher quality and volume stability of protein synthesis linked to energy metabolism by utilizing the chemo-selective ligation reaction with a Tetramethylrhodamine (TAMRA) protein detection method and western blotting analyses. Therefore, this method is valuable for investigating hepatic nascent protein synthesis linked to energy homeostasis. The following protocol outlines the materials and methods for the isolation of high-quality primary mouse hepatocytes and detection of nascent protein synthesis.
Collapse
Affiliation(s)
- Esam S B Salem
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati; Division of Endocrinology, Cincinnati Children's Hospital Medical Center
| | - Kazutoshi Murakami
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center
| | | | - Elise Bernhard
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center
| | - Vishnupriya Borra
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center
| | - Mridula Bethi
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center
| | - Takahisa Nakamura
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, College of Medicine, University of Cincinnati;
| |
Collapse
|
17
|
Kelemen RE, Erickson SB, Chatterjee A. Synthesis at the interface of virology and genetic code expansion. Curr Opin Chem Biol 2018; 46:164-171. [PMID: 30086446 DOI: 10.1016/j.cbpa.2018.07.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/18/2018] [Accepted: 07/13/2018] [Indexed: 01/24/2023]
Abstract
How a virus efficiently invades its host cell and masterfully engineers its properties provides valuable lessons and resources for the emerging discipline of synthetic biology, which seeks to create engineered biological systems with novel functions. Recently, the toolbox of synthetic biology has also been enriched by the genetic code expansion technology, which has provided access to a large assortment of unnatural amino acids with novel chemical functionalities that can be site-specifically incorporated into proteins in living cells. The synergistic interplay of these two disciplines holds much promise to advance their individual progress, while creating new paradigms for synthetic biology. In this review we seek to provide an account of the recent advances at the interface of these two research areas.
Collapse
Affiliation(s)
- Rachel E Kelemen
- Department of Chemistry, Boston College, 2609 Beacon Street, 246B Merkert Chemistry Center, Chestnut Hill, MA 02467, United States
| | - Sarah B Erickson
- Department of Chemistry, Boston College, 2609 Beacon Street, 246B Merkert Chemistry Center, Chestnut Hill, MA 02467, United States
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 2609 Beacon Street, 246B Merkert Chemistry Center, Chestnut Hill, MA 02467, United States.
| |
Collapse
|
18
|
Baker AT, Aguirre-Hernández C, Halldén G, Parker AL. Designer Oncolytic Adenovirus: Coming of Age. Cancers (Basel) 2018; 10:E201. [PMID: 29904022 PMCID: PMC6025169 DOI: 10.3390/cancers10060201] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/06/2018] [Accepted: 06/11/2018] [Indexed: 12/26/2022] Open
Abstract
The licensing of talimogene laherparepvec (T-Vec) represented a landmark moment for oncolytic virotherapy, since it provided unequivocal evidence for the long-touted potential of genetically modified replicating viruses as anti-cancer agents. Whilst T-Vec is promising as a locally delivered virotherapy, especially in combination with immune-checkpoint inhibitors, the quest continues for a virus capable of specific tumour cell killing via systemic administration. One candidate is oncolytic adenovirus (Ad); it’s double stranded DNA genome is easily manipulated and a wide range of strategies and technologies have been employed to empower the vector with improved pharmacokinetics and tumour targeting ability. As well characterised clinical and experimental agents, we have detailed knowledge of adenoviruses’ mechanisms of pathogenicity, supported by detailed virological studies and in vivo interactions. In this review we highlight the strides made in the engineering of bespoke adenoviral vectors to specifically infect, replicate within, and destroy tumour cells. We discuss how mutations in genes regulating adenoviral replication after cell entry can be used to restrict replication to the tumour, and summarise how detailed knowledge of viral capsid interactions enable rational modification to eliminate native tropisms, and simultaneously promote active uptake by cancerous tissues. We argue that these designer-viruses, exploiting the viruses natural mechanisms and regulated at every level of replication, represent the ideal platforms for local overexpression of therapeutic transgenes such as immunomodulatory agents. Where T-Vec has paved the way, Ad-based vectors now follow. The era of designer oncolytic virotherapies looks decidedly as though it will soon become a reality.
Collapse
Affiliation(s)
- Alexander T Baker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Carmen Aguirre-Hernández
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Alan L Parker
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| |
Collapse
|
19
|
Lin X, Yu ACS, Chan TF. Efforts and Challenges in Engineering the Genetic Code. Life (Basel) 2017; 7:life7010012. [PMID: 28335420 PMCID: PMC5370412 DOI: 10.3390/life7010012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 12/15/2022] Open
Abstract
This year marks the 48th anniversary of Francis Crick’s seminal work on the origin of the genetic code, in which he first proposed the “frozen accident” hypothesis to describe evolutionary selection against changes to the genetic code that cause devastating global proteome modification. However, numerous efforts have demonstrated the viability of both natural and artificial genetic code variations. Recent advances in genetic engineering allow the creation of synthetic organisms that incorporate noncanonical, or even unnatural, amino acids into the proteome. Currently, successful genetic code engineering is mainly achieved by creating orthogonal aminoacyl-tRNA/synthetase pairs to repurpose stop and rare codons or to induce quadruplet codons. In this review, we summarize the current progress in genetic code engineering and discuss the challenges, current understanding, and future perspectives regarding genetic code modification.
Collapse
Affiliation(s)
- Xiao Lin
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, NT, Hong Kong, China.
| | - Allen Chi Shing Yu
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, NT, Hong Kong, China.
| | - Ting Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, NT, Hong Kong, China.
| |
Collapse
|
20
|
Carvalho SB, Freire JM, Moleirinho MG, Monteiro F, Gaspar D, Castanho MARB, Carrondo MJT, Alves PM, Bernardes GJL, Peixoto C. Bioorthogonal Strategy for Bioprocessing of Specific-Site-Functionalized Enveloped Influenza-Virus-Like Particles. Bioconjug Chem 2016; 27:2386-2399. [PMID: 27652605 PMCID: PMC5080633 DOI: 10.1021/acs.bioconjchem.6b00372] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
![]()
Virus-like
particles (VLPs) constitute a promising platform in
vaccine development and targeted drug delivery. To date, most applications
use simple nonenveloped VLPs as human papillomavirus or hepatitis
B vaccines, even though the envelope is known to be critical to retain
the native protein folding and biological function. Here, we present
tagged enveloped VLPs (TagE-VLPs) as a valuable strategy for the downstream
processing and monitoring of the in vivo production of specific-site-functionalized
enveloped influenza VLPs. This two-step procedure allows bioorthogonal
functionalization of azide-tagged nascent influenza type A hemagglutinin
proteins in the envelope of VLPs through a strain-promoted [3 + 2]
alkyne–azide cycloaddition reaction. Importantly, labeling
does not influence VLP production and allows for construction of functionalized
VLPs without deleterious effects on their biological function. Refined
discrimination and separation between VLP and baculovirus, the major
impurity of the process, is achieved when this technique is combined
with flow cytometry analysis, as demonstrated by atomic force microscopy.
TagE-VLPs is a versatile tool broadly applicable to the production,
monitoring, and purification of functionalized enveloped VLPs for
vaccine design trial runs, targeted drug delivery, and molecular imaging.
Collapse
Affiliation(s)
- Sofia B Carvalho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa , Avenida da República, 2780-157 Oeiras, Portugal.,iBET, Instituto de Biologia Experimental e Tecnológica , Apartado 12, 2780-901 Oeiras, Portugal
| | - João M Freire
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Mafalda G Moleirinho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa , Avenida da República, 2780-157 Oeiras, Portugal.,iBET, Instituto de Biologia Experimental e Tecnológica , Apartado 12, 2780-901 Oeiras, Portugal
| | - Francisca Monteiro
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa , Avenida da República, 2780-157 Oeiras, Portugal.,iBET, Instituto de Biologia Experimental e Tecnológica , Apartado 12, 2780-901 Oeiras, Portugal
| | - Diana Gaspar
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Manuel J T Carrondo
- iBET, Instituto de Biologia Experimental e Tecnológica , Apartado 12, 2780-901 Oeiras, Portugal.,Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa , 2829-516 Monte da Caparica, Portugal
| | - Paula M Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa , Avenida da República, 2780-157 Oeiras, Portugal.,iBET, Instituto de Biologia Experimental e Tecnológica , Apartado 12, 2780-901 Oeiras, Portugal
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa , Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.,Department of Chemistry, University of Cambridge , Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Cristina Peixoto
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa , Avenida da República, 2780-157 Oeiras, Portugal.,iBET, Instituto de Biologia Experimental e Tecnológica , Apartado 12, 2780-901 Oeiras, Portugal
| |
Collapse
|
21
|
Urquhart T, Daub E, Honek JF. Bioorthogonal Modification of the Major Sheath Protein of Bacteriophage M13: Extending the Versatility of Bionanomaterial Scaffolds. Bioconjug Chem 2016; 27:2276-2280. [PMID: 27626459 DOI: 10.1021/acs.bioconjchem.6b00460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
With a mass of ∼1.6 × 107 Daltons and composed of approximately 2700 proteins, bacteriophage M13 has been employed as a molecular scaffold in bionanomaterials fabrication. In order to extend the versatility of M13 in this area, residue-specific unnatural amino acid incorporation was employed to successfully display azide functionalities on specific solvent-exposed positions of the pVIII major sheath protein of this bacteriophage. Employing a combination of engineered mutants of the gene coding for the pVIII protein, the methionine (Met) analog, l-azidohomoalanine (Aha), and a suitable Escherichia coli Met auxotroph for phage production, conditions were developed to produce M13 bacteriophage labeled with over 350 active azides (estimated by fluorescent dye labeling utilizing a strain-promoted azide-alkyne cycloaddition) and capable of azide-selective attachment to 5 nm gold nanoparticles as visualized by transmission electron microscopy. The capability of this system to undergo dual labeling utilizing both chemical acylation and bioorthogonal cycloaddition reactions was also verified. The above stratagem should prove particularly advantageous in the preparation of assemblies of larger and more complex molecular architectures based on the M13 building block.
Collapse
Affiliation(s)
- Taylor Urquhart
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue, Waterloo, Ontario, Canada N2L 3G1
| | - Elisabeth Daub
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue, Waterloo, Ontario, Canada N2L 3G1
| | - John Frank Honek
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo , 200 University Avenue, Waterloo, Ontario, Canada N2L 3G1
| |
Collapse
|
22
|
Kelemen RE, Mukherjee R, Cao X, Erickson SB, Zheng Y, Chatterjee A. A Precise Chemical Strategy To Alter the Receptor Specificity of the Adeno-Associated Virus. Angew Chem Int Ed Engl 2016; 55:10645-9. [PMID: 27483453 DOI: 10.1002/anie.201604067] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/29/2016] [Indexed: 12/12/2022]
Abstract
The ability to target the adeno-associated virus (AAV) to specific types of cells, by altering the cell-surface receptor it binds, is desirable to generate safe and efficient therapeutic vectors. Chemical attachment of receptor-targeting agents onto the AAV capsid holds potential to alter its tropism, but is limited by the lack of site specificity of available conjugation strategies. The development of an AAV production platform is reported that enables incorporation of unnatural amino acids (UAAs) into specific sites on the virus capsid. Incorporation of an azido-UAA enabled site-specific attachment of a cyclic-RGD peptide onto the capsid, retargeting the virus to the αv β3 integrin receptors, which are overexpressed in tumor vasculature. Retargeting ability was site-dependent, underscoring the importance of achieving site-selective capsid modification. This work provides a general chemical approach to introduce various receptor binding agents onto the AAV capsid with site selectivity to generate optimized vectors with engineered infectivity.
Collapse
Affiliation(s)
- Rachel E Kelemen
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, 02467, USA
| | - Raja Mukherjee
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, 02467, USA
| | - Xiaofu Cao
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, 02467, USA
| | - Sarah B Erickson
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, 02467, USA
| | - Yunan Zheng
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, 02467, USA
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
23
|
Kelemen RE, Mukherjee R, Cao X, Erickson SB, Zheng Y, Chatterjee A. A Precise Chemical Strategy To Alter the Receptor Specificity of the Adeno-Associated Virus. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201604067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rachel E. Kelemen
- Department of Chemistry; Boston College; 2609 Beacon Street Chestnut Hill MA 02467 USA
| | - Raja Mukherjee
- Department of Chemistry; Boston College; 2609 Beacon Street Chestnut Hill MA 02467 USA
| | - Xiaofu Cao
- Department of Chemistry; Boston College; 2609 Beacon Street Chestnut Hill MA 02467 USA
| | - Sarah B. Erickson
- Department of Chemistry; Boston College; 2609 Beacon Street Chestnut Hill MA 02467 USA
| | - Yunan Zheng
- Department of Chemistry; Boston College; 2609 Beacon Street Chestnut Hill MA 02467 USA
| | - Abhishek Chatterjee
- Department of Chemistry; Boston College; 2609 Beacon Street Chestnut Hill MA 02467 USA
| |
Collapse
|
24
|
Oum YH, Desai TM, Marin M, Melikyan GB. Click labeling of unnatural sugars metabolically incorporated into viral envelope glycoproteins enables visualization of single particle fusion. J Virol Methods 2016; 233:62-71. [PMID: 27033181 DOI: 10.1016/j.jviromet.2016.02.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 02/11/2016] [Indexed: 10/22/2022]
Abstract
Enveloped viruses infect target cells by fusing their membrane with cellular membrane through a process that is mediated by specialized viral glycoproteins. The inefficient and highly asynchronous nature of viral fusion complicates studies of virus entry on a population level. Single virus imaging in living cells has become an important tool for delineating the entry pathways and for mechanistic studies of viral fusion. We have previously demonstrated that incorporation of fluorescent labels into the viral membrane and trapping fluorescent proteins in the virus interior enables the visualization of single virus fusion in living cells. Here, we implement a new approach to non-invasively label the viral membrane glycoproteins through metabolic incorporation of unnatural sugars followed by click-reaction with organic fluorescent dyes. This approach allows for efficient labeling of diverse viral fusion glycoproteins on the surface of HIV pseudoviruses. Incorporation of a content marker into surface-labeled viral particles enables sensitive detection of single virus fusion with live cells.
Collapse
Affiliation(s)
- Yoon Hyeun Oum
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, USA
| | - Tanay M Desai
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, USA
| | - Mariana Marin
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, USA
| | - Gregory B Melikyan
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
25
|
Debarba JA, Monteiro KM, Moura H, Barr JR, Ferreira HB, Zaha A. Identification of Newly Synthesized Proteins by Echinococcus granulosus Protoscoleces upon Induction of Strobilation. PLoS Negl Trop Dis 2015; 9:e0004085. [PMID: 26393918 PMCID: PMC4578768 DOI: 10.1371/journal.pntd.0004085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022] Open
Abstract
Background The proteins responsible for the key molecular events leading to the structural changes between the developmental stages of Echinococcus granulosus remain unknown. In this work, azidohomoalanine (AHA)-specific labeling was used to identify proteins expressed by E. granulosus protoscoleces (PSCs) upon the induction of strobilar development. Methodology/Principal Findings The in vitro incorporation of AHA with different tags into newly synthesized proteins (NSPs) by PSCs was analyzed using SDS-PAGE and confocal microscopy. The LC-MS/MS analysis of AHA-labeled NSPs by PSCs undergoing strobilation allowed for the identification of 365 proteins, of which 75 were differentially expressed in comparison between the presence or absence of strobilation stimuli and 51 were expressed exclusively in either condition. These proteins were mainly involved in metabolic, regulatory and signaling processes. Conclusions/Significance After the controlled-labeling of proteins during the induction of strobilar development, we identified modifications in protein expression. The changes in the metabolism and the activation of control and signaling pathways may be important for the correct parasite development and be target for further studies. In the life cycle of the parasite Echinococcus granulosus, hydatid cysts produce the pre-adult form, which has the ability to either differentiate into an adult worm (strobilation) or dedifferentiate into a secondary hydatid cyst. We used different protein tags that allowed for the visualization and purification of proteins produced specifically after the induction of strobilar development to identify proteins that might be involved in this process (temporally controlled and context-dependent). As a result, we found proteins that are involved in important processes during development, such as energy metabolism, control pathways and cellular communication. We believe that these results will be useful for the development of scientific approaches to controlling and preventing cystic hydatid disease.
Collapse
Affiliation(s)
- João Antonio Debarba
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, and Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Karina Mariante Monteiro
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, and Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Biologia Molecular e Biotecnologia, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Hercules Moura
- Biological Mass Spectrometry Laboratory, Clinical Chemistry Branch, Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - John R. Barr
- Biological Mass Spectrometry Laboratory, Clinical Chemistry Branch, Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Henrique Bunselmeyer Ferreira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, and Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Biologia Molecular e Biotecnologia, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Arnaldo Zaha
- Programa de Pós-Graduação em Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Laboratório de Biologia Molecular de Cestódeos, and Laboratório de Genômica Estrutural e Funcional, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Biologia Molecular e Biotecnologia, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- * E-mail:
| |
Collapse
|
26
|
Lim SI, Hahn YS, Kwon I. Site-specific albumination of a therapeutic protein with multi-subunit to prolong activity in vivo. J Control Release 2015; 207:93-100. [PMID: 25862515 PMCID: PMC4430413 DOI: 10.1016/j.jconrel.2015.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/03/2015] [Accepted: 04/05/2015] [Indexed: 11/21/2022]
Abstract
Albumin fusion/conjugation (albumination) has been an effective method to prolong in vivo half-life of therapeutic proteins. However, its broader application to proteins with complex folding pathway or multi-subunit is restricted by incorrect folding, poor expression, heterogeneity, and loss of native activity of the proteins linked to albumin. We hypothesized that the site-specific conjugation of albumin to a permissive site of a target protein will expand the utilities of albumin as a therapeutic activity extender to proteins with a complex structure. We show here the genetic incorporation of a non-natural amino acid (NNAA) followed by chemoselective albumin conjugation to prolong therapeutic activity in vivo. Urate oxidase (Uox), a therapeutic enzyme for treatment of hyperuricemia, is a homotetramer with multiple surface lysines, limiting conventional approaches for albumination. Incorporation of p-azido-l-phenylalanine into two predetermined positions of Uox allowed site-specific linkage of dibenzocyclooctyne-derivatized human serum albumin (HSA) through strain-promoted azide-alkyne cycloaddition (SPAAC). The bio-orthogonality of SPAAC resulted in the production of a chemically well-defined conjugate, Uox-HSA, with a retained enzymatic activity. Uox-HSA had a half-life of 8.8 h in mice, while wild-type Uox had a half-life of 1.3 h. The AUC increased 5.5-fold (1657 vs. 303 mU/mL x h). These results clearly demonstrated that site-specific albumination led to the prolonged enzymatic activity of Uox in vivo. Site-specific albumination enabled by NNAA incorporation and orthogonal chemistry demonstrates its promise for the development of long-acting protein therapeutics with high potency and safety.
Collapse
Affiliation(s)
- Sung In Lim
- Department of Chemical Engineering, University of Virginia, VA 22904, United States
| | - Young S Hahn
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA 22908, United States
| | - Inchan Kwon
- Department of Chemical Engineering, University of Virginia, VA 22904, United States; School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, Republic of Korea.
| |
Collapse
|
27
|
McKay CS, Finn MG. Click chemistry in complex mixtures: bioorthogonal bioconjugation. CHEMISTRY & BIOLOGY 2014; 21:1075-101. [PMID: 25237856 PMCID: PMC4331201 DOI: 10.1016/j.chembiol.2014.09.002] [Citation(s) in RCA: 584] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 01/18/2023]
Abstract
The selective chemical modification of biological molecules drives a good portion of modern drug development and fundamental biological research. While a few early examples of reactions that engage amine and thiol groups on proteins helped establish the value of such processes, the development of reactions that avoid most biological molecules so as to achieve selectivity in desired bond-forming events has revolutionized the field. We provide an update on recent developments in bioorthogonal chemistry that highlights key advances in reaction rates, biocompatibility, and applications. While not exhaustive, we hope this summary allows the reader to appreciate the rich continuing development of good chemistry that operates in the biological setting.
Collapse
Affiliation(s)
- Craig S McKay
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - M G Finn
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
28
|
O'Donovan L, De Bank PA. A hydrazide-anchored dendron scaffold for chemoselective ligation strategies. Org Biomol Chem 2014; 12:7290-6. [DOI: 10.1039/c4ob00870g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We report the design and synthesis of a dendron scaffold, enabling the chemoselective decoration of target molecules with multiple copies of functional species, such as peptides, via a hydrazone bond.
Collapse
Affiliation(s)
- Liz O'Donovan
- Department of Pharmacy and Pharmacology and Centre for Regenerative Medicine
- University of Bath
- Bath, UK
| | - Paul A. De Bank
- Department of Pharmacy and Pharmacology and Centre for Regenerative Medicine
- University of Bath
- Bath, UK
| |
Collapse
|
29
|
Lu Q, Ye X, Liu F, Zhao Y, Qin J, Liang M, Fang C, Chen HZ. Homologous recombination-based adenovirus vector system for tumor cell-specific gene delivery. Cancer Biol Ther 2013; 14:728-35. [PMID: 23792576 DOI: 10.4161/cbt.25090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cancer gene therapy requires tumor-specific delivery and expression of a transgene to maximize antitumor efficacy and minimize side effects. In this study, we developed a new tumor-targeting, homologous recombination-based adenovirus vector system, HRAVS. HRAVS is composed of two adenovirus vectors, Ad.CMV.IR containing reverse sequence (IR) and a CMV promoter and Ad.IR.EGFP comprising the report gene EGFP and IR. For improved viral DNA replication and transgene expression, the E1a gene was added to HRAVS to generate the enhanced HRAVS, EHRAVS, which consists of Ad.CMV.IR and Ad.IR.EGFP/E1a. The optimal vector composition ratio of Ad.CMV.IR to Ad.IR.EGFP or Ad.IR.EGFP/E1a was identified as 30:70 based on EGFP expression efficiency in tumor cells. The transgene expression of HRAVS and EHRAVS was efficiently and specifically activated in tumor cells only and not in normal cells. Moreover, compared with HRAVS, EHRAVS infection led to higher virus yields and transgene expression and higher toxicity to tumor cells, and these results could be related to the involvement of E1a genes. The results in present study suggest the need for in vivo antitumor study using these new dual-Ad vector systems based on the homologous recombination.
Collapse
Affiliation(s)
- Qin Lu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Rubino FA, Oum YH, Rajaram L, Chu Y, Carrico IS. Chemoselective modification of viral surfaces via bioorthogonal click chemistry. J Vis Exp 2012:e4246. [PMID: 22929552 DOI: 10.3791/4246] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The modification of virus particles has received a significant amount of attention for its tremendous potential for impacting gene therapy, oncolytic applications and vaccine development. Current approaches to modifying viral surfaces, which are mostly genetics-based, often suffer from attenuation of virus production, infectivity and cellular transduction. Using chemoselective click chemistry, we have developed a straightforward alternative approach which sidesteps these issues while remaining both highly flexible and accessible. The goal of this protocol is to demonstrate the effectiveness of using bioorthogonal click chemistry to modify the surface of adenovirus type 5 particles. This two-step process can be used both therapeutically or analytically, as it allows for chemoselective ligation of targeting molecules, dyes or other molecules of interest onto proteins pre-labeled with azide tags. The three major advantages of this method are that (1) metabolic labeling demonstrates little to no impact on viral fitness, (2) a wide array of effector ligands can be utilized, and (3) it is remarkably fast, reliable and easy to access. In the first step of this procedure, adenovirus particles are produced bearing either azidohomoalanine (Aha, a methionine surrogate) or the unnatural sugar O-linked N-azidoacetylglucosamine (O-GlcNAz), both of which contain the azide (-N3) functional group. After purification of the azide-modified virus particles, an alkyne probe containing the fluorescent TAMRA moiety is ligated in a chemoselective manner to the pre-labeled proteins or glycoproteins. Finally, an SDS-PAGE analysis is performed to demonstrate the successful ligation of the probe onto the viral capsid proteins. Aha incorporation is shown to label all viral capsid proteins (Hexon, Penton and Fiber), while O-GlcNAz incorporation results in labeling of Fiber only. In this evolving field, multiple methods for azide-alkyne ligation have been successfully developed; however only the two we have found to be most convenient are demonstrated herein - strain-promoted azide-alkyne cycloaddition (SPAAC) and copper-catalyzed azide-alkyne cycloaddition (CuAAC) under deoxygenated atmosphere.
Collapse
|
31
|
Oum YH, Carrico IS. Altering adenoviral tropism via click modification with ErbB specific ligands. Bioconjug Chem 2012; 23:1370-6. [PMID: 22681483 DOI: 10.1021/bc200477z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Methods for targeting oncolytic viruses can increase efficacy and accelerate development. Genetic engineering, the predominant method for changing vector tropism, is limited in scope and often represents the bottleneck for vector development. Metabolic incorporation of an unnatural azido sugar, O-GlcNAz, at a specific site on the adenoviral surface allows chemoselective attachment of affibodies for Her2 or EGF receptors. Modification with these high-affinity, high-selectivity proteins is straightforward and readily generalizable, demonstrates minimal impact on virus physiology, and affords significant increases in gene delivery to cancer cells. As a result, this method has significant potential to increase the efficacy of next-generation viral vectors.
Collapse
Affiliation(s)
- Yoon Hyeun Oum
- Department of Chemistry, State University of New York at Stony Brook , Stony Brook, NY 11794-3400, USA
| | | |
Collapse
|