1
|
Jung E, Chung YH, Mao C, Fiering SN, Steinmetz NF. The Potency of Cowpea Mosaic Virus Particles for Cancer In Situ Vaccination Is Unaffected by the Specific Encapsidated Viral RNA. Mol Pharm 2023; 20:3589-3597. [PMID: 37294891 PMCID: PMC10530639 DOI: 10.1021/acs.molpharmaceut.3c00214] [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] [Indexed: 06/11/2023]
Abstract
Plant virus nanoparticles can be used as drug carriers, imaging reagents, vaccine carriers, and immune adjuvants in the formulation of intratumoral in situ cancer vaccines. One example is the cowpea mosaic virus (CPMV), a nonenveloped virus with a bipartite positive-strand RNA genome with each RNA packaged separately into identical protein capsids. Based on differences in their densities, the components carrying RNA-1 (6 kb) denoted as the bottom (B) component or carrying RNA-2 (3.5 kb) denoted as the middle (M) component can be separated from each other and from a top (T) component, which is devoid of any RNA. Previous preclinical mouse studies and canine cancer trials used mixed populations of CPMV (containing B, M, and T components), so it is unclear whether the particle types differ in their efficacies. It is known that the CPMV RNA genome contributes to immunostimulation by activation of TLR7. To determine whether the two RNA genomes that have different sizes and unrelated sequences cause different immune stimulation, we compared the therapeutic efficacies of B and M components and unfractionated CPMV in vitro and in mouse cancer models. We found that separated B and M particles behaved similarly to the mixed CPMV, activating innate immune cells to induce the secretion of pro-inflammatory cytokines such as IFNα, IFNγ, IL-6, and IL-12, while inhibiting immunosuppressive cytokines such as TGF-β and IL-10. In murine models of melanoma and colon cancer, the mixed and separated CPMV particles all significantly reduced tumor growth and prolonged survival with no significant difference. This shows that the specific RNA genomes similarly stimulate the immune system even though B particles have 40% more RNA than M particles; each CPMV particle type can be used as an effective adjuvant against cancer with the same efficacy as native mixed CPMV. From a translational point of view, the use of either B or M component vs the mixed CPMV formulation offers the advantage that separated B or M alone is noninfectious toward plants and thus provides agronomic safety.
Collapse
Affiliation(s)
- Eunkyeong Jung
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Young Hun Chung
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Chenkai Mao
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, New Hampshire 03755, United States
| | - Steven N Fiering
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, New Hampshire 03755, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine, Hanover, New Hampshire 03755, United States
| | - Nicole F Steinmetz
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Institute for Materials Design and Discovery, University of California San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Jung E, Mao C, Bhatia M, Koellhoffer EC, Fiering SN, Steinmetz NF. Inactivated Cowpea Mosaic Virus for In Situ Vaccination: Differential Efficacy of Formalin vs UV-Inactivated Formulations. Mol Pharm 2023; 20:500-507. [PMID: 36399598 PMCID: PMC9812890 DOI: 10.1021/acs.molpharmaceut.2c00744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cowpea mosaic virus (CPMV) has been developed as a promising nanoplatform technology for cancer immunotherapy; when applied as in situ vaccine, CPMV exhibits potent, systemic, and durable efficacy. While CPMV is not infectious to mammals, it is infectious to legumes; therefore, agronomic safety needs to be addressed to broaden the translational application of CPMV. RNA-containing formulations are preferred over RNA-free virus-like particles because the RNA and protein, each, contribute to CPMV's potent antitumor efficacy. We have previously optimized inactivation methods to develop CPMV that contains RNA but is not infectious to plants. We established that inactivated CPMV has reduced efficacy compared to untreated, native CPMV. However, a systematic comparison between native CPMV and different inactivated forms of CPMV was not done. Therefore, in this study, we directly compared the therapeutic efficacies and mechanisms of immune activation of CPMV, ultraviolet- (UV-), and formalin (Form)-inactivated CPMV to explain the differential efficacies. In a B16F10 melanoma mouse tumor model, Form-CPMV suppressed the tumor growth with prolonged survival (there were no statistical differences comparing CPMV and Form-CPMV). In comparison, UV-CPMV inhibited tumor growth significantly but not as well as Form-CPMV or CPMV. The reduced therapeutic efficacy of UV-CPMV is explained by the degree of cross-linking and aggregated state of the RNA, which renders it inaccessible for sensing by Toll-like receptor (TLR) 7/8 to activate immune responses. The mechanistic studies showed that the highly aggregated state of UV-CPMV inhibited TLR7 signaling more so than for the Form-CPMV formulation, reducing the secretion of interleukin-6 (IL-6) and interferon-α (IFN-α), cytokines associated with TLR7 signaling. These findings support the translational development of Form-CPMV as a noninfectious immunotherapeutic agent.
Collapse
Affiliation(s)
| | | | - Misha Bhatia
- Department of Nanoengineering, University of, California San Diego, La Jolla, California 92093, United, States
| | - Edward C. Koellhoffer
- Radiology, University of California San Diego, La Jolla, California 92093, United States
| | - Steven N. Fiering
- Department of Microbiology and, Immunology and Dartmouth Cancer Center, Dartmouth, Geisel School of Medicine, Hanover, New Hampshire 03755, United States
| | - Nicole F. Steinmetz
- Department of Nanoengineering, Radiology, Bioengineering, Moores Cancer Center, Center for Nano-Immuno Engineering, and Institute for Materials, Design and Discovery, University of California San Diego, La, Jolla, California 92093, United States
| |
Collapse
|
3
|
Park J, Wen AM, Gao H, Shin MD, Simon DI, Wang Y, Steinmetz NF. Designing S100A9-Targeted Plant Virus Nanoparticles to Target Deep Vein Thrombosis. Biomacromolecules 2021; 22:2582-2594. [PMID: 34060817 DOI: 10.1021/acs.biomac.1c00303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Thromboembolic conditions are a leading cause of death worldwide, and deep vein thrombosis (DVT), or occlusive venous clot formation, is a critical and rising problem that contributes to damage of vital organs, long-term complications, and life-threatening conditions such as pulmonary embolism. Early diagnosis and treatment are correlated to better prognosis. However, current technologies in these areas, such as ultrasonography for diagnostics and anticoagulants for treatment, are limited in terms of their accuracy and therapeutic windows. In this work, we investigated targeting myeloid related protein 14 (MRP-14, also known as S100A9) using plant virus-based nanoparticle carriers as a means to achieve tissue specificity aiding prognosis and therapeutic intervention. We used a combinatorial peptide library screen to identify peptide ligands that bind MRP-14. Candidates were selected and formulated as nanoparticles by using cowpea mosaic virus (CPMV) and tobacco mosaic virus (TMV). Intravascular delivery of our MRP-14-targeted nanoparticles in a murine model of DVT resulted in enhanced accumulation in the thrombi and reduced thrombus size, suggesting application of nanoparticles for molecular targeting of MRP-14 could be a promising direction for improving DVT diagnostics, therapeutics, and therefore prognosis.
Collapse
Affiliation(s)
- Jooneon Park
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Amy M Wen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Huiyun Gao
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Matthew D Shin
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Daniel I Simon
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Yunmei Wang
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States.,Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States.,Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States.,Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, California 92093, United States.,Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, United States.,Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
4
|
Shukla S, Wang C, Beiss V, Cai H, Washington T, Murray AA, Gong X, Zhao Z, Masarapu H, Zlotnick A, Fiering S, Steinmetz NF. The unique potency of Cowpea mosaic virus (CPMV) in situ cancer vaccine. Biomater Sci 2020; 8:5489-5503. [PMID: 32914796 PMCID: PMC8086234 DOI: 10.1039/d0bm01219j] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunosuppressive tumor microenvironment enables cancer to resist immunotherapies. We have established that intratumoral administration of plant-derived Cowpea mosaic virus (CPMV) nanoparticles as an in situ vaccine overcomes the local immunosuppression and stimulates a potent anti-tumor response in several mouse cancer models and canine patients. CPMV does not infect mammalian cells but acts as a danger signal that leads to the recruitment and activation of innate and subsequently, adaptive immune cells. In the present study we addressed whether other icosahedral viruses or virus-like particles (VLPs) of plant, bacteriophage and mammalian origin can be similarly employed as intratumoral immunotherapy. Our results indicate that CPMV in situ vaccine outperforms Cowpea chlorotic mottle virus (CCMV), Physalis mosaic virus (PhMV), Sesbania mosaic virus (SeMV), bacteriophage Qβ VLPs, or Hepatitis B virus capsids (HBVc). Furthermore, ex vivo and in vitro assays reveal unique features of CPMV that makes it an inherently stronger immune stimulant.
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Chao Wang
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Veronique Beiss
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Hui Cai
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Torus Washington
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA.
| | - Abner A Murray
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xingjian Gong
- Department of Bioengineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhongchao Zhao
- Molecular and Cellular Biochemistry Department, Indiana University Bloomington, IN 47405, USA
| | - Hema Masarapu
- Department of Virology, Sri Venkateswara University, Tirupati 517502, India
| | - Adam Zlotnick
- Molecular and Cellular Biochemistry Department, Indiana University Bloomington, IN 47405, USA
| | - Steven Fiering
- Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA. and Department of Bioengineering, University of California-San Diego, La Jolla, CA 92039, USA and Department of Radiology, University of California-San Diego, La Jolla, CA 92039, USA and Moores Cancer Center, University of California-San Diego, La Jolla, CA 92039, USA and Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| |
Collapse
|
5
|
Wang C, Beiss V, Steinmetz NF. Cowpea Mosaic Virus Nanoparticles and Empty Virus-Like Particles Show Distinct but Overlapping Immunostimulatory Properties. J Virol 2019; 93:e00129-19. [PMID: 31375592 PMCID: PMC6803287 DOI: 10.1128/jvi.00129-19] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 07/16/2019] [Indexed: 12/30/2022] Open
Abstract
Cowpea mosaic virus (CPMV) is a plant virus that has been developed for multiple biomedical and nanotechnology applications, including immunotherapy. Two key platforms are available: virus nanoparticles (VNPs) based on the complete CMPV virion, including the genomic RNA, and virus-like nanoparticles (VLPs) based on the empty CPMV (eCPMV) virion. It is unclear whether these platforms differ in terms of immunotherapeutic potential. We therefore compared their physicochemical properties and immunomodulatory activities following in situ vaccination of an aggressive ovarian tumor mouse model (ID8-Defb29/Vegf-A). In physicochemical terms, CPMV and eCPMV were very similar, and both significantly increased the survival of tumor-bearing mice and showed promising antitumor efficacy. However, they demonstrated distinct yet overlapping immunostimulatory effects due to the presence of virus RNA in wild-type particles, indicating their suitability for different immunotherapeutic strategies. Specifically, we found that the formulations had similar effects on most secreted cytokines and immune cells, but the RNA-containing CPMV particles were uniquely able to boost populations of potent antigen-presenting cells, such as tumor-infiltrating neutrophils and activated dendritic cells. Our results will facilitate the development of CPMV and eCPMV as immunotherapeutic vaccine platforms with tailored responses.IMPORTANCE The engagement of antiviral effector responses caused by viral infection is essential when using viruses or virus-like particles (VLPs) as an immunotherapeutic agent. Here, we compare the chemophysical and immunostimulatory properties of wild-type cowpea mosaic virus (CPMV) (RNA containing) and eCPMV (RNA-free VLPs) produced from two expression systems (agrobacterium-based plant expression system and baculovirus-insect cell expression). CPMV and eCPMV could each be developed as novel adjuvants to overcome immunosuppression and thus promote tumor regression in ovarian cancer (and other tumor types). To our knowledge, this is the first study to define the immunotherapeutic differences between CPMV and eCPMV, which is essential for the further development of biomedical applications for plant viruses and the selection of rational combinations of immunomodulatory reagents.
Collapse
Affiliation(s)
- Chao Wang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Veronique Beiss
- Department of NanoEngineering, University of California, San Diego, La Jolla, California, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Zheng Y, Lee PW, Wang C, Thomas LD, Stewart PL, Steinmetz NF, Pokorski JK. Freeze-Drying To Produce Efficacious CPMV Virus-like Particles. NANO LETTERS 2019; 19:2099-2105. [PMID: 30801195 PMCID: PMC7272238 DOI: 10.1021/acs.nanolett.9b00300] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In situ cancer vaccination that uses immune stimulating agents is revolutionizing the way that cancer is treated. In this realm, viruses and noninfectious virus-like particles have gained significant traction in reprogramming the immune system to recognize and eliminate malignancies. Recently, cowpea mosaic virus-like particles (VLPs) have shown exceptional promise in their ability to fight a variety of cancers. However, the current methods used to produce CPMV VLPs rely on agroinfiltration in plants. These protocols remain complicated and labor intensive and have the potential to introduce unwanted immunostimulatory agents, like lipopolysaccharides. This Letter describes a simple "post-processing" method to remove RNA from wild-type CPMV, while retaining the structure and function of the capsid. Lyophilization was able to eject encapsulated RNA to form lyo-eCPMV and, when purified, eliminated nearly all traces of encapsulated RNA. Lyo-eCPMV was characterized by cryo-electron microscopy single particle reconstruction to confirm the structural integrity of the viral capsid. Finally, lyo-eCPMV showed equivalent anticancer efficacy as eCPMV, produced by agroinfiltration, when using an invasive melanoma model. These results describe a straightforward method to prepare CPMV VLPs from infectious virions.
Collapse
Affiliation(s)
- Yi Zheng
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
| | - Parker W. Lee
- Department of Macromolecular Science and Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Chao Wang
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
| | - Linda D. Thomas
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio 44106, United States
- School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Phoebe L. Stewart
- Department of Pharmacology and Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, Cleveland, Ohio 44106, United States
- School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Jonathan K. Pokorski
- Department of NanoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Macromolecular Science and Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
7
|
Garmann RF, Knobler CM, Gelbart WM. Protocol for Efficient Cell-Free Synthesis of Cowpea Chlorotic Mottle Virus-Like Particles Containing Heterologous RNAs. Methods Mol Biol 2019; 1776:249-265. [PMID: 29869247 DOI: 10.1007/978-1-4939-7808-3_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report a protocol for efficient cell-free synthesis of cowpea chlorotic mottle virus (CCMV)-like particles containing a broad range of lengths and sequences of RNA. Our protocol starts with a purified stock of wild-type CCMV (protocols for harvesting and purifying the virus are detailed elsewhere) and features three basic steps: disassembly of the CCMV and purification of the capsid protein (CP) from the viral RNA; coassembly of the purified CP and an RNA of choice; and characterization of the assembly products. We highlight several key factors that increase the yield of the assembly reaction: the CP should be uncleaved and sufficiently free of viral RNA; the length of the RNA should be between about 100 and 4000 nucleotides; and the stoichiometry of CP and RNA should be 6-1 by mass. Additionally, we point out that separating the assembly reaction into multiple steps-by successively lowering the ionic strength and then the pH of the assembly buffers-results in the highest yields of well-formed, nuclease-resistant, CCMV-like particles. Finally, we describe methods for characterizing the assembly products using native agarose gel electrophoresis and negative-stain transmission electron microscopy.
Collapse
Affiliation(s)
- Rees F Garmann
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| | - Charles M Knobler
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - William M Gelbart
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA.,California NanoSystems Institute, UCLA, Los Angeles, CA, USA.,Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| |
Collapse
|
8
|
Berardi A, Baldelli Bombelli F, Thuenemann EC, Lomonossoff GP. Viral nanoparticles can elude protein barriers: exploiting rather than imitating nature. NANOSCALE 2019; 11:2306-2316. [PMID: 30662985 DOI: 10.1039/c8nr09067j] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Protein-corona formation in body fluids and/or entrapment of nanoparticles in protein matrices (e.g. food and mucus) can hinder the delivery of nanoparticles, irrespective of the route of administration. Here we demonstrate that certain viral nanoparticles (VNPs) can evade the adhesion of a broad panel of macromolecules from several biological milieus. We also show that the permeability of VNPs through mucin gels is far superior to that of synthetic nanoparticles. The non-sticky nature of VNPs implies that they will be able to readily cross most non-specific protein and glycoprotein barriers encountered, ubiquitously, upon administration through mucosal, and non-mucosal routes.
Collapse
Affiliation(s)
- Alberto Berardi
- Department of Pharmaceutical Sciences and Pharmaceutics, Faculty of Pharmacy, Applied Science Private University, Amman 11931, Jordan.
| | | | | | | |
Collapse
|
9
|
Kruse I, Peyret H, Saxena P, Lomonossoff GP. Encapsidation of Viral RNA in Picornavirales: Studies on Cowpea Mosaic Virus Demonstrate Dependence on Viral Replication. J Virol 2019; 93:e01520-18. [PMID: 30355698 PMCID: PMC6321914 DOI: 10.1128/jvi.01520-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/19/2018] [Indexed: 12/17/2022] Open
Abstract
To elucidate the linkage between replication and encapsidation in Picornavirales, we have taken advantage of the bipartite nature of a plant-infecting member of this order, cowpea mosaic virus (CPMV), to decouple the two processes. RNA-free virus-like particles (empty virus-like particles [eVLPs]) can be generated by transiently coexpressing the RNA-2-encoded coat protein precursor (VP60) with the RNA-1-encoded 24,000-molecular-weight (24K) protease, in the absence of the replication machinery (K. Saunders, F. Sainsbury, and G. P. Lomonossoff, Virology 393:329-337, 2009, https://doi.org/10.1016/j.virol.2009.08.023). We have made use of the ability to produce assembled capsids of CPMV in the absence of replication to examine the putative linkage between RNA replication and packaging in the Picornavirales We have created a series of mutant RNA-1 and RNA-2 molecules and have assessed the effects of the mutations on both the replication and packaging of the viral RNAs. We demonstrate that mutations that affect replication have a concomitant impact on encapsidation and that RNA-1-mediated replication is required for encapsidation of both RNA-1 and RNA-2. This close coupling between replication and encapsidation provides a means for the specific packaging of viral RNAs. Moreover, we demonstrate that this feature of CPMV can be used to specifically encapsidate custom RNA by placing a sequence of choice between the RNA-2 sequences required for replication.IMPORTANCE The mechanism whereby members of the order Picornavirales specifically package their genomic RNAs is poorly understood. Research with monopartite members of the order, such as poliovirus, indicated that packaging is linked to replication, although the presence of "packaging signals" along the length of the viral RNA has also been suggested. Thanks to the bipartite nature of the CPMV genome, which allows the manipulation of RNA-1 without modifying RNA-2, we show here that this specificity is due to a functional link between the two processes of viral replication and encapsidation. This has important implications for our understanding of the fundamental molecular biology of Picornavirales and opens the door to novel research and therapeutic applications in the field of custom RNA packaging and delivery technologies.
Collapse
Affiliation(s)
- Inga Kruse
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | - Hadrien Peyret
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | - Pooja Saxena
- Department of Biological Chemistry, John Innes Centre, Norwich, United Kingdom
| | | |
Collapse
|
10
|
Berardi A, Evans DJ, Baldelli Bombelli F, Lomonossoff GP. Stability of plant virus-based nanocarriers in gastrointestinal fluids. NANOSCALE 2018; 10:1667-1679. [PMID: 29231944 PMCID: PMC5804478 DOI: 10.1039/c7nr07182e] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/04/2017] [Indexed: 05/17/2023]
Abstract
Cowpea mosaic virus (CPMV) is a plant virus which is being extensively investigated as a drug delivery and vaccine nanocarrier for parenteral administration. However, to date little is known about the suitability of plant-based nanocarriers for oral delivery. In this study, the colloidal (i.e. aggregation), physical (i.e. denaturation) and chemical (i.e. digestion of the polypeptides) stability of CPMV and its empty virus-like particles (eVLPs) in conditions resembling the gastrointestinal fluids were evaluated. The nanoparticles were incubated in various simulated gastric and intestinal fluids and in pig gastric and intestinal fluids. CPMV and eVLPs had similar stabilities. In simulated gastric media, they were stable at pH ≥ 2.5. At lower pH destabilisation of the particle structure occurred, which, in turn, rendered the polypeptides extremely sensitive to pepsin digestion. However, both CPMV and eVLPs were stable in simulated intestinal fluids, in pig gastric fluids and in pig intestinal fluids. Thus CPMV, despite being a protein-based nanoparticle, was much more resistant to the harsh GI conditions than soluble proteins. Remarkably, both CPMV and eVLPs incubated in pig gastric and intestinal fluids were not subject to protein adsorption, with no formation of a detectable protein corona. The lack of a protein corona on CPMV and eVLP surfaces in GI fluids would imply that, if orally administered, these nanoparticles could maintain their native surface characteristics; thus, their biological interactions would remain predictable and unchanged. In summary, CPMV and eVLPs can be considered promising nanocarriers for applications requiring oral delivery, given their chemical, physical and colloidal stability and lack of protein adsorption from the environment in most of the tested conditions.
Collapse
Affiliation(s)
- Alberto Berardi
- Department of Pharmaceutical Sciences and Pharmaceutics, Faculty of Pharmacy, Applied Science Private University, Amman 11931, Jordan. and Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - David J Evans
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Francesca Baldelli Bombelli
- Laboratory of Supramolecular and BioNano Materials (SupraBioNanoLab), Department of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Milano, Italy
| | - George P Lomonossoff
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| |
Collapse
|
11
|
Soto CM, Dressick WJ. Dual-Functionalized Virus-Gold Nanoparticle Clusters for Biosensing. Methods Mol Biol 2018; 1776:533-552. [PMID: 29869264 DOI: 10.1007/978-1-4939-7808-3_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Metallic nanoscale 3D architectures concentrate electromagnetic energy at precise spatial locations to enable sensing and photocatalysis applications. We have developed solution-based methods to reproducibly fabricate 3D gold nanostructures useful as efficient surface-enhanced Raman spectroscopy (SERS) biosensors. Virus capsids were recruited as templates to assemble gold nanoparticles on their surfaces at well-defined locations to prepare the nanoscale 3D structures. Cowpea mosaic virus (CPMV) and its variants were selected as specific templates due to their high symmetry, scalability, and stability, which have proven useful in materials science applications. While the methods described herein were optimized for the CPMV capsids, they also provide a useful starting point for researchers who are working toward the nanoassembly of metal nanoparticles on other protein scaffolds.
Collapse
Affiliation(s)
- Carissa M Soto
- Center for Bio/Molecular Science and Engineering, US Naval Research Laboratory, Washington, DC, USA. .,Materials and Applications Division, US Bureau of Engraving and Printing, Washington, DC, USA.
| | - Walter J Dressick
- Center for Bio/Molecular Science and Engineering, US Naval Research Laboratory, Washington, DC, USA
| |
Collapse
|
12
|
Narayanan KB, Han SS. Icosahedral plant viral nanoparticles - bioinspired synthesis of nanomaterials/nanostructures. Adv Colloid Interface Sci 2017; 248:1-19. [PMID: 28916111 DOI: 10.1016/j.cis.2017.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 10/18/2022]
Abstract
Viral nanotechnology utilizes virus nanoparticles (VNPs) and virus-like nanoparticles (VLPs) of plant viruses as highly versatile platforms for materials synthesis and molecular entrapment that can be used in the nanotechnological fields, such as in next-generation nanoelectronics, nanocatalysis, biosensing and optics, and biomedical applications, such as for targeting, therapeutic delivery, and non-invasive in vivo imaging with high specificity and selectivity. In particular, plant virus capsids provide biotemplates for the production of novel nanostructured materials with organic/inorganic moieties incorporated in a very precise and controlled manner. Interestingly, capsid proteins of spherical plant viruses can self-assemble into well-organized icosahedral three-dimensional (3D) nanoscale multivalent architectures with high monodispersity and structural symmetry. Using viral genetic and protein engineering of icosahedral viruses with a variety of sizes, the interior, exterior and the interfaces between coat protein (CP) subunits can be manipulated to fabricate materials with a wide range of desirable properties allowing for biomineralization, encapsulation, infusion, controlled self-assembly, and multivalent ligand display of nanoparticles or molecules for varied applications. In this review, we discuss the various functional nanomaterials/nanostructures developed using the VNPs and VLPs of different icosahedral plant viruses and their nano(bio)technological and nanomedical applications.
Collapse
|
13
|
Cho CF, Yu L, Nsiama TK, Kadam AN, Raturi A, Shukla S, Amadei GA, Steinmetz NF, Luyt LG, Lewis JD. Viral nanoparticles decorated with novel EGFL7 ligands enable intravital imaging of tumor neovasculature. NANOSCALE 2017; 9:12096-12109. [PMID: 28799610 PMCID: PMC5770569 DOI: 10.1039/c7nr02558k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Angiogenesis is a dynamic process fundamental to the development of solid tumors. Epidermal growth factor-like domain 7 (EGFL7) is a protein whose expression is restricted to endothelial cells undergoing active remodeling that has emerged as a key mediator of this process. EGFL7 expression is associated with poor outcome in several cancers, making it a promising target for imaging or therapeutic strategies. Here, EGFL7 is explored as a molecular target for active neovascularization. Using a combinatorial peptide screening approach, we describe the discovery and characterization of a novel high affinity EGFL7-binding peptide, E7p72, that specifically targets human endothelial cells. Viral nanoparticles decorated with E7p72 peptides specifically target tumor-associated neovasculature with high specificity as assessed by intravital imaging. This work highlights the value of EGFL7 as a target for angiogenic vessels and opens the door for novel targeted therapeutic approaches.
Collapse
Affiliation(s)
- Choi-Fong Cho
- Translational Prostate Cancer Research Group, University of Alberta, 5-142C Katz Group Building, 114th St and 87th Ave, Edmonton, AB T6G 2E1, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lebedev N, Griva I, Dressick WJ, Phelps J, Johnson JE, Meshcheriakova Y, Lomonossoff GP, Soto CM. A virus-based nanoplasmonic structure as a surface-enhanced Raman biosensor. Biosens Bioelectron 2016; 77:306-14. [DOI: 10.1016/j.bios.2015.09.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/10/2015] [Accepted: 09/13/2015] [Indexed: 02/05/2023]
|
15
|
Spa SJ, Bunschoten A, Rood MTM, Peters RJB, Koster AJ, van Leeuwen FWB. Orthogonal Functionalization of Ferritin via Supramolecular Re‐Assembly. Eur J Inorg Chem 2015. [DOI: 10.1002/ejic.201500386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Silvia J. Spa
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands, http://www.interventionalmolecularimaging.com
| | - Anton Bunschoten
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands, http://www.interventionalmolecularimaging.com
| | - Marcus T. M. Rood
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands, http://www.interventionalmolecularimaging.com
| | - Ruud J. B. Peters
- RIKILT‐BU Contaminants and Toxins, Wageningen University, 6708 WB Wageningen, The Netherlands
| | - Abraham J. Koster
- Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Fijs W. B. van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands, http://www.interventionalmolecularimaging.com
- Laboratory of Bio Nano Technology, Wageningen University, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
16
|
Wen AM, Steinmetz NF. The aspect ratio of nanoparticle assemblies and the spatial arrangement of ligands can be optimized to enhance the targeting of cancer cells. Adv Healthc Mater 2014; 3:1739-44. [PMID: 24729309 DOI: 10.1002/adhm.201400141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Amy M. Wen
- Department of Biomedical Engineering; Schools of Medicine and Engineering; Case Western Reserve University; 10900 Euclid Avenue Cleveland Ohio 44106 USA
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering; Schools of Medicine and Engineering; Case Western Reserve University; 10900 Euclid Avenue Cleveland Ohio 44106 USA
- Department of Radiology; Schools of Medicine and Engineering; Case Western Reserve University; 10900 Euclid Avenue Cleveland Ohio 44106 USA
- Department of Materials Science and Engineering; Schools of Medicine and Engineering; Case Western Reserve University; 10900 Euclid Avenue Cleveland Ohio 44106 USA
- Department of Macromolecular Science and Engineering; Schools of Medicine and Engineering; Case Western Reserve University; 10900 Euclid Avenue Cleveland Ohio 44106 USA
| |
Collapse
|
17
|
Schoonen L, van Hest JCM. Functionalization of protein-based nanocages for drug delivery applications. NANOSCALE 2014; 6:7124-41. [PMID: 24860847 DOI: 10.1039/c4nr00915k] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Traditional drug delivery strategies involve drugs which are not targeted towards the desired tissue. This can lead to undesired side effects, as normal cells are affected by the drugs as well. Therefore, new systems are now being developed which combine targeting functionalities with encapsulation of drug cargo. Protein nanocages are highly promising drug delivery platforms due to their perfectly defined structures, biocompatibility, biodegradability and low toxicity. A variety of protein nanocages have been modified and functionalized for these types of applications. In this review, we aim to give an overview of different types of modifications of protein-based nanocontainers for drug delivery applications.
Collapse
Affiliation(s)
- Lise Schoonen
- Institute of Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | | |
Collapse
|
18
|
Aljabali AAA, Evans DJ. Templated mineralization by charge-modified cowpea mosaic virus. Methods Mol Biol 2014; 1108:89-95. [PMID: 24243242 DOI: 10.1007/978-1-62703-751-8_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Templated mineralization of virus particles provides routes to narrowly dispersed nanoparticles that are not readily prepared by other means. The templated mineralization of metal or metal oxide on the external surface of wild-type cowpea mosaic virus (CPMV), a plant virus, is facilitated by increasing the external surface negative charge. This is achieved by the chemical modification of surface lysine groups by succinic anhydride. Hence, for example, treatment of charge-modified CPMV succinamate with a 1:2 mixture of iron(II) and iron(III) salts, followed by raising the pH to 10.2, led to the formation of narrowly dispersed, CPMV-templated, magnetite (Fe3O4) nanoparticles.
Collapse
|
19
|
Lua LHL, Connors NK, Sainsbury F, Chuan YP, Wibowo N, Middelberg APJ. Bioengineering virus-like particles as vaccines. Biotechnol Bioeng 2013; 111:425-40. [PMID: 24347238 DOI: 10.1002/bit.25159] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/23/2013] [Accepted: 11/12/2013] [Indexed: 12/12/2022]
Abstract
Virus-like particle (VLP) technology seeks to harness the optimally tuned immunostimulatory properties of natural viruses while omitting the infectious trait. VLPs that assemble from a single protein have been shown to be safe and highly efficacious in humans, and highly profitable. VLPs emerging from basic research possess varying levels of complexity and comprise single or multiple proteins, with or without a lipid membrane. Complex VLP assembly is traditionally orchestrated within cells using black-box approaches, which are appropriate when knowledge and control over assembly are limited. Recovery challenges including those of adherent and intracellular contaminants must then be addressed. Recent commercial VLPs variously incorporate steps that include VLP in vitro assembly to address these problems robustly, but at the expense of process complexity. Increasing research activity and translation opportunity necessitate bioengineering advances and new bioprocessing modalities for efficient and cost-effective production of VLPs. Emerging approaches are necessarily multi-scale and multi-disciplinary, encompassing diverse fields from computational design of molecules to new macro-scale purification materials. In this review, we highlight historical and emerging VLP vaccine approaches. We overview approaches that seek to specifically engineer a desirable immune response through modular VLP design, and those that seek to improve bioprocess efficiency through inhibition of intracellular assembly to allow optimal use of existing purification technologies prior to cell-free VLP assembly. Greater understanding of VLP assembly and increased interdisciplinary activity will see enormous progress in VLP technology over the coming decade, driven by clear translational opportunity.
Collapse
Affiliation(s)
- Linda H L Lua
- Protein Expression Facility, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | | | | | | | | | | |
Collapse
|
20
|
Yildiz I, Lee KL, Chen K, Shukla S, Steinmetz NF. Infusion of imaging and therapeutic molecules into the plant virus-based carrier cowpea mosaic virus: cargo-loading and delivery. J Control Release 2013; 172:568-78. [PMID: 23665254 PMCID: PMC3815978 DOI: 10.1016/j.jconrel.2013.04.023] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 02/04/2023]
Abstract
This work is focused on the development of a plant virus-based carrier system for cargo delivery, specifically 30nm-sized cowpea mosaic virus (CPMV). Whereas previous reports described the engineering of CPMV through genetic or chemical modification, we report a non-covalent infusion technique that facilitates efficient cargo loading. Infusion and retention of 130-155 fluorescent dye molecules per CPMV using DAPI (4',6-diamidino-2-phenylindole dihydrochloride), propidium iodide (3,8-diamino-5-[3-(diethylmethylammonio)propyl]-6-phenylphenanthridinium diiodide), and acridine orange (3,6-bis(dimethylamino)acridinium chloride), as well as 140 copies of therapeutic payload proflavine (PF, acridine-3,6-diamine hydrochloride), is reported. Loading is achieved through interaction of the cargo with the CPMV's encapsidated RNA molecules. The loading mechanism is specific; empty RNA-free eCPMV nanoparticles could not be loaded. Cargo-infused CPMV nanoparticles remain chemically active, and surface lysine residues were covalent modified with dyes leading to the development of dual-functional CPMV carrier systems. We demonstrate cargo-delivery to a panel of cancer cells (cervical, breast, and colon): CPMV nanoparticles enter cells via the surface marker vimentin, the nanoparticles target the endolysosome, where the carrier is degraded and the cargo is released allowing imaging and/or cell killing. In conclusion, we demonstrate cargo-infusion and delivery to cells; the methods discussed provide a useful means for functionalization of CPMV toward its application as drug and/or contrast agent delivery vehicle.
Collapse
Affiliation(s)
- Ibrahim Yildiz
- Department of Biomedical Engineering, Case Western Reserve University, Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Karin L. Lee
- Department of Biomedical Engineering, Case Western Reserve University, Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Kevin Chen
- Department of Biomedical Engineering, Case Western Reserve University, Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University, Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Radiology, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Materials Science and Engineering, Case Western Reserve University, School of Engineering, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Wen AM, Shukla S, Saxena P, Aljabali AA, Yildiz I, Dey S, Mealy JE, Yang AC, Evans DJ, Lomonossoff GP, Steinmetz NF. Interior engineering of a viral nanoparticle and its tumor homing properties. Biomacromolecules 2012; 13:3990-4001. [PMID: 23121655 PMCID: PMC3525095 DOI: 10.1021/bm301278f] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of multifunctional nanoparticles for medical applications is of growing technological interest. A single formulation containing imaging and/or drug moieties that is also capable of preferential uptake in specific cells would greatly enhance diagnostics and treatments. There is growing interest in plant-derived viral nanoparticles (VNPs) and establishing new platform technologies based on these nanoparticles inspired by nature. Cowpea mosaic virus (CPMV) serves as the standard model for VNPs. Although exterior surface modification is well-known and has been comprehensively studied, little is known of interior modification. Additional functionality conferred by the capability for interior engineering would be of great benefit toward the ultimate goal of targeted drug delivery. Here, we examined the capacity of empty CPMV (eCPMV) particles devoid of RNA to encapsulate a wide variety of molecules. We systematically investigated the conjugation of fluorophores, biotin affinity tags, large molecular weight polymers such as poly(ethylene glycol) (PEG), and various peptides through targeting reactive cysteines displayed selectively on the interior surface. Several methods are described that mutually confirm specific functionalization of the interior. Finally, CPMV and eCPMV were labeled with near-infrared fluorophores and studied side-by-side in vitro and in vivo. Passive tumor targeting via the enhanced permeability and retention effect and optical imaging were confirmed using a preclinical mouse model of colon cancer. The results of our studies lay the foundation for the development of the eCPMV platform in a range of biomedical applications.
Collapse
Affiliation(s)
- Amy M. Wen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Pooja Saxena
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Alaa A.A. Aljabali
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Ibrahim Yildiz
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Sourav Dey
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Joshua E. Mealy
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Alice C. Yang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - David J. Evans
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - George P. Lomonossoff
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Department of Radiology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Department of Materials Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
22
|
Agrawal A, Manchester M. Differential uptake of chemically modified cowpea mosaic virus nanoparticles in macrophage subpopulations present in inflammatory and tumor microenvironments. Biomacromolecules 2012; 13:3320-6. [PMID: 22963597 PMCID: PMC3590107 DOI: 10.1021/bm3010885] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There remains a tremendous need to develop targeted therapeutics that can both image and localize the toxic effects of chemotherapeutics and antagonists on diseased tissue while reducing adverse systemic effects. These needs have fostered the development of a nanotechnology-based approach that can combine targeting and toxicity potential. In this study, CPMV nanoparticles were chemically modified with the dye Alexa Flour 488 and were also tandemly modified with PEG1000 followed by AF488; and the derivatized nanoparticles were subsequently added to macrophages stimulated with either LPS (M1) or IL-4 (M2). Previously published studies have shown that M1/M2 macrophages are both present in an inflammatory microenvironment (such as a tumor microenvironment and atherosclerosis) and play opposing yet balancing roles; M2 macrophages have a delayed and progressive onset in the tumor microenvironment (concomitant with an immunosuppression of M1 macrophages). In this study, we show higher uptake of CPMV-AF488 and CPMV-PEG-AF488 by M2 macrophages compared to M1 macrophages. M1 macrophages showed no uptake of CPMV-PEG-AF488. More specifically, M2 macrophages are known to be up-regulated in early atherosclerosis plaque. Indeed, previous work showed that M2 macrophages in plaque also correlate with CPMV internalization. These studies emphasize the potential effectiveness of CPMV as a tailored vehicle for targeting tumor macrophages involved in cancer metastasis or vascular inflammation and further highlight the potential of CPMV in targeted therapeutics against other diseases.
Collapse
Affiliation(s)
- Arpita Agrawal
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, La Jolla, CA 92093, USA
| | | |
Collapse
|
23
|
Aljabali AAA, Barclay JE, Steinmetz NF, Lomonossoff GP, Evans DJ. Controlled immobilisation of active enzymes on the cowpea mosaic virus capsid. NANOSCALE 2012; 4:5640-5. [PMID: 22865109 DOI: 10.1039/c2nr31485a] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Immobilisation of horseradish peroxidase (HRP) and glucose oxidase (GOX) via covalent attachment of modified enzyme carbohydrate to the exterior of the cowpea mosaic virus (CPMV) capsid gave high retention of enzymatic activity. The number of enzymes bound per virus was determined to be about eleven for HRP and 2-3 for GOX. This illustrates that relatively large biomacromolecules can be readily coupled to the virus surface using simple conjugation strategies. Virus-biomacromolecule hybrids have great potential for uses in catalysis, diagnostic assays or biosensors.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | | | | | | | | |
Collapse
|
24
|
Taghavian O, Mandal MK, Steinmetz NF, Rasche S, Spiegel H, Fischer R, Schillberg S. A potential nanobiotechnology platform based on infectious bursal disease subviral particles. RSC Adv 2012. [PMID: 28638593 DOI: 10.1039/c2ra00857b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We describe a novel nanobiotechnology platform based on subviral particles derived from infectious bursal disease virus (IBD-SVPs). The major virus coat protein VP2 assembles into spherical, 23 nm SVPs when expressed as a heterologous protein in the yeast Pichia pastoris. We recovered up to 38 mg of IBD-SVPs at > 95% purity from 1 L of recombinant yeast culture. The purified particles were able to tolerate organic solvents up to 20% concentration (ethanol or dimethylsulfoxide), they resisted temperatures up to 65 °C and remained stable over a wide pH range (2.5-9.0). We achieved bioconjugation to the amine groups of lysine residues and to the carboxyl groups of aspartic and glutamic acid residues, allowing the functionalization of IBD-SVPs with biotin. The accessibility of surface amine groups was measured using Alexa Fluor 488 N-hydroxysuccinimide (NHS) ester, an amine-selective fluorescent dye, revealing that approximately 60 dye molecules were attached to the surface of each particle. IBD-SVPs can therefore be exploited as a robust and versatile nanoscaffold to display diverse functional ligands.
Collapse
Affiliation(s)
- Omid Taghavian
- Fraunhofer IME, Forckenbeckstraβe 6, 52074, Aachen, Germany
| | - Manoj K Mandal
- Fraunhofer IME, Forckenbeckstraβe 6, 52074, Aachen, Germany
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Stefan Rasche
- Fraunhofer IME, Forckenbeckstraβe 6, 52074, Aachen, Germany
| | - Holger Spiegel
- Fraunhofer IME, Forckenbeckstraβe 6, 52074, Aachen, Germany
| | - Rainer Fischer
- Fraunhofer IME, Forckenbeckstraβe 6, 52074, Aachen, Germany.,Institut für Molekulare Biotechnologie, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | | |
Collapse
|
25
|
Abstract
The capsids of most plant viruses are simple and robust structures consisting of multiple copies of one or a few types of protein subunit arranged with either icosahedral or helical symmetry. In many cases, capsids can be produced in large quantities either by the infection of plants or by the expression of the subunit(s) in a variety of heterologous systems. In view of their relative simplicity, stability and ease of production, plant virus particles or virus-like particles (VLPs) have attracted attention as potential reagents for applications in bionanotechnology. As a result, plant virus particles have been subjected to both genetic and chemical modification, have been used to encapsulate foreign material and have, themselves, been incorporated into supramolecular structures.
Collapse
|
26
|
Sainsbury F, Saunders K, Aljabali AAA, Evans DJ, Lomonossoff GP. Peptide-Controlled Access to the Interior Surface of Empty Virus Nanoparticles. Chembiochem 2011; 12:2435-40. [DOI: 10.1002/cbic.201100482] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Indexed: 11/07/2022]
|
27
|
Steinmetz NF, Ablack AL, Hickey JL, Ablack J, Manocha B, Mymryk JS, Luyt LG, Lewis JD. Intravital imaging of human prostate cancer using viral nanoparticles targeted to gastrin-releasing Peptide receptors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2011; 7:1664-72. [PMID: 21520408 PMCID: PMC3163449 DOI: 10.1002/smll.201000435] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 09/27/2010] [Indexed: 05/02/2023]
Abstract
Multivalent nanoparticles have several key advantages in terms of solubility, binding avidity, and uptake, making them particularly well suited to molecular imaging applications. Herein is reported the stepwise synthesis and characterization of NIR viral nanoparticles targeted to gastrin-releasing peptide receptors that are over-expressed in human prostate cancers. The pan-bombesin analogue, [β-Ala11, Phe13, Nle14]bombesin-(7-14), is conjugated to cowpea mosaic virus particles functionalized with an NIR dye (Alexa Fluor 647) and polyethylene glycol (PEG) using the copper(I)-catalyzed azide-alkyne cycloaddition reaction. Targeting and uptake in human PC-3 prostate cells is demonstrated in vitro. Tumor homing is observed using human prostate tumor xenografts on the chicken chorioallantoic membrane model using intravital imaging. Further development of this viral nanoparticle platform may open the door to potential clinical noninvasive molecular imaging strategies.
Collapse
Affiliation(s)
| | | | | | - Jailal Ablack
- Department of Oncology, London Regional Cancer Program, 790 Commissioners Rd. E., London, Ontario N6A 4L6, Canada
| | - Bhavik Manocha
- Translational Prostate Cancer Research Group London Regional Cancer Program, Room A4-823, 790 Commissioners Rd. E., London, Ontario N6A 4L6, Canada
| | - Joe S. Mymryk
- Department of Oncology, London Regional Cancer Program, 790 Commissioners Rd. E., London, Ontario N6A 4L6, Canada
| | - Leonard G. Luyt
- Department of Chemistry University of Western Ontario London, Ontario N6A 5C1, Canada
| | | |
Collapse
|
28
|
Huang RK, Steinmetz NF, Fu CY, Manchester M, Johnson JE. Transferrin-mediated targeting of bacteriophage HK97 nanoparticles into tumor cells. Nanomedicine (Lond) 2011; 6:55-68. [PMID: 21182418 DOI: 10.2217/nnm.10.99] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
AIMS Next-generation targeted nanodevices are currently under development for imaging and therapeutic applications. We engineered HK97 viral nanoparticles (VNPs) for tumor cell-specific targeting. METHODS A combination of genetic and chemical engineering methods were developed and applied to generate dual-labeled HK97 cysteine mutant particles displaying transferrin and fluorescent labels. The targeting properties of transferrin-conjugated VNPs were evaluated by in vitro experiments using different cancer cell lines. RESULTS We found that HK97-transferrin formulations were indeed targeted to cancer cells in vitro via the transferrin receptor. These studies highlight the utility and facilitate the further development of HK97-based VNPs.
Collapse
Affiliation(s)
- Rick K Huang
- Department of Molecular Biology & Center for Integrative Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
29
|
Soto CM, Ratna BR. Virus hybrids as nanomaterials for biotechnology. Curr Opin Biotechnol 2010; 21:426-38. [PMID: 20688511 DOI: 10.1016/j.copbio.2010.07.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/06/2010] [Accepted: 07/06/2010] [Indexed: 12/24/2022]
Abstract
The current review describes advances in the field of bionanotechnology in which viruses are used to fabricate nanomaterials. Viruses are introduced as protein cages, scaffolds, and templates for the production of biohybrid nanostructured materials where organic and inorganic molecules are incorporated in a precise and a controlled fashion. Genetic engineering enables the insertion or replacement of selected amino acids on virus capsids for uses from bioconjugation to crystal growth. The variety of nanomaterials generated in rod-like and spherical viruses is highlighted for tobacco mosaic virus (TMV), M13 bacteriophage, cowpea chlorotic mottle virus (CCMV), and cowpea mosaic virus (CPMV). Functional biohybrid nanomaterials find applications in biosensing, memory devices, nanocircuits, light-harvesting systems, and nanobatteries.
Collapse
Affiliation(s)
- Carissa M Soto
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave. SW, Washington, DC 20375, USA.
| | | |
Collapse
|
30
|
|
31
|
Steinmetz NF, Manchester M. PEGylated viral nanoparticles for biomedicine: the impact of PEG chain length on VNP cell interactions in vitro and ex vivo. Biomacromolecules 2010; 10:784-92. [PMID: 19281149 DOI: 10.1021/bm8012742] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PEGylation is an effective strategy for reducing biospecific interactions for pharmaceuticals. The plant virus Cowpea mosaic virus (CPMV) has been studied for potential nanobiomedical applications by virtue of its natural interactions with mammalian endothelial cells. To investigate the degree of PEGylation required to retarget CPMV-based formulations to other destinations, two CPMV-PEG formulations, CPMV-PEG1000 (P1) and CPMV-PEG2000 (P2) were tested. Modeling suggested that the PEG chains were displayed as flattened mushrooms on the particle with an estimated surface grafting area of 0.53% for P1 and 0.83% for P2. Only the P2 formulation effectively shielded the particles from interacting with cells or tissues, suggesting that either key interacting regions on the particle surface were blocked or that a sufficient hydration shell had been generated to inhibit cellular interactions. The large CPMV surface area available after PEGylation allows further attachment of imaging and therapeutic molecules to the particle to generate multifunctionality.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of Cell Biology, Center for Integrative Molecular Biosciences, The Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
32
|
Steinmetz NF, Hong V, Spoerke ED, Lu P, Breitenkamp K, Finn MG, Manchester M. Buckyballs meet viral nanoparticles: candidates for biomedicine. J Am Chem Soc 2010; 131:17093-5. [PMID: 19904938 DOI: 10.1021/ja902293w] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fullerenes such as C(60) show promise as functional components in several emerging technologies. For biomedical applications, C(60) has been used in gene- and drug-delivery vectors, as imaging agents, and as photosensitizers in cancer therapy. A major drawback of C(60) for bioapplications is its insolubility in water. To overcome this limitation, we covalently attached C(60) derivatives to Cowpea mosaic virus and bacteriophage Qbeta virus-like particles, which are examples of naturally occurring viral nanoparticle (VNP) structures that have been shown to be promising candidates for biomedicine. Two different labeling strategies were employed, giving rise to water-soluble, stable VNP-C(60) and VNP-PEG-C(60) conjugates. Samples were characterized using a combination of transmission electron microscopy, scanning transmission electron microscopy (STEM), gel electrophoresis, size-exclusion chromatography, dynamic light scattering, and Western blotting. "Click" chemistry bioconjugation using a poly(ethylene glycol) (PEG)-modified propargyl-O-PEG-C(60) derivative gave rise to high loadings of fullerene on the VNP surface, as indicated by the imaging of individual C(60) units using STEM. The cellular uptake of dye-labeled VNP-PEG-C(60) complexes in a human cancer cell line was found by confocal microscopy to be robust, showing that cell internalization was not inhibited by the attached C(60) units. These results open the door for the development of novel therapeutic devices with potential applications in photoactivated tumor therapy.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of Cell Biology, Center for Integrative Molecular Biosciences, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Saunders K, Sainsbury F, Lomonossoff GP. Efficient generation of cowpea mosaic virus empty virus-like particles by the proteolytic processing of precursors in insect cells and plants. Virology 2009; 393:329-37. [PMID: 19733890 DOI: 10.1016/j.virol.2009.08.023] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 08/06/2009] [Accepted: 08/17/2009] [Indexed: 10/20/2022]
Abstract
To elucidate the mechanism of formation of cowpea mosaic virus (CPMV) particles, RNA-2-encoded precursor proteins were expressed in Spodoptera frugiperda cells. Processing of the 105K and 95K polyproteins in trans to give the mature Large (L) and Small (S) coat proteins required both the 32K proteinase cofactor and the 24K proteinase itself, while processing of VP60, consisting of the fused L-S protein, required only the 24K proteinase. Release of the L and S proteins resulted in the formation of virus-like particles (VLPs), showing that VP60 can act as a precursor of virus capsids. Processing of VP60 expressed in plants also led to efficient production of VLPs. Analysis of the VLPs produced by the action of the 24K proteinase on precursors showed that they were empty (RNA-free). This has important implications for the use of CPMV VLPs in biotechnology and nanotechnology as it will permit the use of noninfectious particles.
Collapse
Affiliation(s)
- Keith Saunders
- Department of Biological Chemistry, John Innes Centre, Colney Lane, Norwich NR4 7UH, UK
| | | | | |
Collapse
|
34
|
Abstract
CPMV (cowpea mosaic virus), a plant virus, is a naturally occurring sphere-like nanoparticle, and is used as a synthon and/or template in bionanoscience. The virions formed by CPMV can be regarded as programmable nanobuilding blocks with a diameter of ∼30 nm. A range of molecules have been attached to this viral nanoscaffold, yielding stable nanoparticles that display multiple copies of the desired molecule. It has been shown that, in addition to surface amine groups, surface carboxy groups are also addressable, and a procedure has been developed that enables introduction of reactive thiols at the virion surface that avoids virus aggregation. Furthermore, the virions can be functionalized to form electroactive nanoparticles. Methods for the construction of arrays and multilayers, using a layer-by-layer approach, have been established. As proof of concept, for example, CPMV particles have been immobilized on surfaces and arranged in defined layers. Engineered variants of CPMV can be used as templates for mineralization with, for example, silica to give monodisperse robust silica nanoparticles of ∼32 nm. SIRV2 (Sulfolobus islandicus rod-shaped virus 2), is a robust archaeal virus, resistant to high temperature and low pH. SIRV2 can act as a template for site-selective and spatially controlled chemical modification. Both the ends and the body of the virus, or the ends only, can be chemically addressed; SIRV2 can be regarded as a structurally unique nanobuilding block.
Collapse
|
35
|
Abedin MJ, Liepold L, Suci P, Young M, Douglas T. Synthesis of a cross-linked branched polymer network in the interior of a protein cage. J Am Chem Soc 2009; 131:4346-54. [PMID: 19317506 PMCID: PMC2752147 DOI: 10.1021/ja8079862] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A goal of biomimetic chemistry is to use the hierarchical architecture inherent in biological systems to guide the synthesis of functional three-dimensional structures. Viruses and other highly symmetrical protein cage architectures provide defined scaffolds to initiate hierarchical structure assembly. Here we demonstrate that a cross-linked branched polymer can be initiated and synthesized within the interior cavity of a protein cage architecture. Creating this polymer network allows for the spatial control of pendant reactive sites and dramatically increases the stability of the cage architecture. This material was generated by the sequential coupling of multifunctional monomers using click chemistry to create a branched cross-linked polymer network. Analysis of polymer growth by mass spectrometry demonstrated that the polymer was initiated at the interior surface of the cage at genetically introduced cysteine reactive sites. The polymer grew as expected to generation 2.5 where it was limited by the size constraints of the cavity. The polymer network was fully cross-linked across protein subunits that make up the cage and extended the thermal stability for the cage to at least 120 degrees C. The introduced reactive centers were shown to be active and their number density increased with increasing generation. This synthetic approach provides a new avenue for creating defined polymer networks, spatially constrained by a biological template.
Collapse
Affiliation(s)
- Md Joynal Abedin
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana 59717
- Center for Bio-Inspired Nanomaterials, Montana State University, Bozeman, Montana 59717
| | - Lars Liepold
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana 59717
- Center for Bio-Inspired Nanomaterials, Montana State University, Bozeman, Montana 59717
| | - Peter Suci
- Department of Plant Sciences, Montana State University, Bozeman, Montana 59717
- Center for Bio-Inspired Nanomaterials, Montana State University, Bozeman, Montana 59717
| | - Mark Young
- Department of Plant Sciences, Montana State University, Bozeman, Montana 59717
- Center for Bio-Inspired Nanomaterials, Montana State University, Bozeman, Montana 59717
| | - Trevor Douglas
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana 59717
- Center for Bio-Inspired Nanomaterials, Montana State University, Bozeman, Montana 59717
| |
Collapse
|
36
|
Abstract
Protein capsids derived from viruses may be modified by methods, generated, isolated, and purified on large scales with relative ease. In recent years, methods for their chemical derivatization have been employed to broaden the properties and functions accessible to investigators desiring monodisperse, atomic-resolution structures on the nanometer scale. Here we review the reactions and methods used in these endeavors, including the modification of lysine, cysteine, and tyrosine side chains, as well as the installation of unnatural amino acids, with particular attention to the special challenges imposed by the polyvalency and size of virus-based scaffolds.
Collapse
Affiliation(s)
- E Strable
- Dynavax Technologies Corp., Berkeley, CA 94710-2753, USA
| | | |
Collapse
|
37
|
Steinmetz NF, Findlay KC, Noel TR, Parker R, Lomonossoff GP, Evans DJ. Layer-by-layer assembly of viral nanoparticles and polyelectrolytes: the film architecture is different for spheres versus rods. Chembiochem 2008; 9:1662-70. [PMID: 18536062 DOI: 10.1002/cbic.200800070] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The development of tuneable thin film assemblies that contain (bio)nanoparticles is an emerging field in nanobiosciences/nanotechnology. Our research focuses on the utilisation of viral nanoparticles (VNPs) as tools and building blocks for materials science. In previous reports we studied multilayered arrays of chemically modified cowpea mosaic virus (CPMV) particles and linker molecules. To extend these studies and to gain more insights into the architecture of the arrays, we report here on the construction of multilayered assemblies of native plant viral particles and polyelectrolytes. We specifically addressed the question of whether the shape of the VNPs influences the overall structures of the arrays. To study this, we have chosen two particles with similar surface properties but different shapes: CPMV was used as a sphere-like VNP, and tobacco mosaic virus (TMV) served as a rod-shaped VNP. The multilayers were self-assembled on solid supports through electrostatic interactions. Multilayer build-up was followed by quartz crystal microbalance with dissipation monitoring and UV/Vis spectroscopy. Scanning electron microscopy was used to characterize the topologies of the thin films. Our studies show that shape indeed matters. Incorporation of CPMV in alternating arrays of VNPs and polyelectrolytes is demonstrated; in stark contrast, TMV particles were found to be excluded from the arrays, and floated atop the architecture in an ordered structure.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of Biological Chemistry, John Innes Centre, Colney, Norwich, NR47UH, UK.
| | | | | | | | | | | |
Collapse
|
38
|
Steinmetz NF, Bock E, Richter RP, Spatz JP, Lomonossoff GP, Evans DJ. Assembly of multilayer arrays of viral nanoparticles via biospecific recognition: a quartz crystal microbalance with dissipation monitoring study. Biomacromolecules 2008; 9:456-62. [PMID: 18197628 DOI: 10.1021/bm700797b] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The development of multilayered thin film assemblies containing (bio)molecules is driven by the need to miniaturize sensors, reactors, and biochips. Viral nanoparticles (VNPs) have become popular nanobuilding blocks for material fabrication, and our research has focused on the well-characterized plant virus Cowpea mosaic virus (CPMV). In a previous study, we have reported the construction of multilayer VNP assemblies. Here we extend these studies by providing further details on the formation and properties of arrays that are made by the alternating deposition of biotinylated CPMV particles and streptavidin molecules. Array formation was followed in real time by a quartz crystal microbalance with dissipation monitoring. Our data provide indications that multiple interactions between biotin and streptavidin not only promote the assembly of a multilayered structure but also generate cross-links within each layer of CPMV particles. The degree of intralayer and interlayer cross-linking and hence the mechanical properties and order of the array can be modulated by the grafting density and spacer length of the biotin moieties on the CPMV particles.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, United Kingdom.
| | | | | | | | | | | |
Collapse
|
39
|
Evans DJ. The bionanoscience of plant viruses: templates and synthons for new materials. ACTA ACUST UNITED AC 2008. [DOI: 10.1039/b804305a] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
40
|
Abstract
Bionanoscience/technology sits at the interface of chemistry, biology, physics, materials science, engineering and medicine and involves the exploitation of biomaterials, devices or methodologies on the nanoscale. One sub-field of bionanoscience/technology is concerned with the exploitation of biomaterials in the fabrication of new nano-materials and/or -devices. In this Perspective we describe examples of how plant viruses, focusing particularly on cowpea mosaic virus, a naturally occurring pre-formed sphere-like nanoparticle, are being used as templates and/or building blocks in bionanoscience and indicate their potential for future application.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Colney, Norwich, United Kingdom
| | | |
Collapse
|