1
|
Hengelbrock A, Probst F, Baukmann S, Uhl A, Tschorn N, Stitz J, Schmidt A, Strube J. Digital Twin for Continuous Production of Virus-like Particles toward Autonomous Operation. ACS OMEGA 2024; 9:34990-35013. [PMID: 39157157 PMCID: PMC11325504 DOI: 10.1021/acsomega.4c04985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024]
Abstract
Lentiviral vector and virus-like particle (VLP) manufacturing have been published in fed-batch upstream and batch downstream modes before. Batch downstream and continuous upstream in perfusion mode were reported as well. This study exemplifies development and validation steps for a digital twin combining a physical-chemical-based mechanistic model for all unit operations with a process analytical technology strategy in order to show the efforts and benefits of autonomous operation approaches for manufacturing scale. As the general models are available from various other biologic manufacturing studies, the main step is model calibration for the human embryo kidney cell-based VLPs with experimental quantitative validation within the Quality-by-Design (QbD) approach, including risk assessment to define design and control space. For continuous operation in perfusion mode, the main challenge is the efficient separation of large particle manifolds for VLPs and cells, including cell debris, which is of similar size. Here, innovative tangential flow filtration operations are needed to avoid fast blocking with low mechanical stress pumps. A twofold increase of productivity was achieved using simulation case studies. This increase is similar to improvements previously described for other entities like plasmid DNAs, monoclonal antibodies (mAbs), and single-chain fragments of variability (scFv) fragments. The advantages of applying a digital twin for an advanced process control strategy have proven additional productivity gains of 20% at 99.9% reliability.
Collapse
Affiliation(s)
- Alina Hengelbrock
- Institute
for Separation and Process Technology, Clausthal
University of Technology, Clausthal 38678, Zellerfeld, Germany
| | - Finja Probst
- Institute
for Separation and Process Technology, Clausthal
University of Technology, Clausthal 38678, Zellerfeld, Germany
| | - Simon Baukmann
- Institute
for Separation and Process Technology, Clausthal
University of Technology, Clausthal 38678, Zellerfeld, Germany
| | - Alexander Uhl
- Institute
for Separation and Process Technology, Clausthal
University of Technology, Clausthal 38678, Zellerfeld, Germany
| | - Natalie Tschorn
- Faculty
of Applied Natural Sciences, Technische
Hochschule Köln, Leverkusen 51379, Germany
| | - Jörn Stitz
- Faculty
of Applied Natural Sciences, Technische
Hochschule Köln, Leverkusen 51379, Germany
| | - Axel Schmidt
- Institute
for Separation and Process Technology, Clausthal
University of Technology, Clausthal 38678, Zellerfeld, Germany
| | - Jochen Strube
- Institute
for Separation and Process Technology, Clausthal
University of Technology, Clausthal 38678, Zellerfeld, Germany
| |
Collapse
|
2
|
Piranej S, Zhang L, Bazrafshan A, Marin M, Melikian GB, Salaita K. Rolosense: Mechanical Detection of SARS-CoV-2 Using a DNA-Based Motor. ACS CENTRAL SCIENCE 2024; 10:1332-1347. [PMID: 39071064 PMCID: PMC11273449 DOI: 10.1021/acscentsci.4c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 07/30/2024]
Abstract
Assays that detect viral infections play a significant role in limiting the spread of diseases such as SARS-CoV-2. Here, we present Rolosense, a virus sensing platform that leverages the motion of 5 μm DNA-based motors on RNA fuel chips to transduce the presence of viruses. Motors and chips are modified with aptamers, which are designed for multivalent binding to viral targets and lead to stalling of motion. Therefore, the motors perform a "mechanical test" of the viral target and stall in the presence of whole virions, which represents a unique mechanism of transduction distinct from conventional assays. Rolosense can detect SARS-CoV-2 spiked in artificial saliva and exhaled breath condensate with a sensitivity of 103 copies/mL and discriminates among other respiratory viruses. The assay is modular and amenable to multiplexing, as demonstrated by our one-pot detection of influenza A and SARS-CoV-2. As a proof of concept, we show that readout can be achieved using a smartphone camera with a microscopic attachment in as little as 15 min without amplification reactions. Taken together, these results show that mechanical detection using Rolosense can be broadly applied to any viral target and has the potential to enable rapid, low-cost point-of-care screening of circulating viruses.
Collapse
Affiliation(s)
- Selma Piranej
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Luona Zhang
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Alisina Bazrafshan
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Mariana Marin
- Department
of Pediatrics, Emory University School of
Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Gregory B. Melikian
- Department
of Pediatrics, Emory University School of
Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Khalid Salaita
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
3
|
Raghunath G, Abbott EH, Marin M, Wu H, Reyes Ballista JM, Brindley MA, Melikyan GB. Disruption of Transmembrane Phosphatidylserine Asymmetry by HIV-1 Incorporated SERINC5 Is Not Responsible for Virus Restriction. Biomolecules 2024; 14:570. [PMID: 38785977 PMCID: PMC11118262 DOI: 10.3390/biom14050570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Host restriction factor SERINC5 (SER5) incorporates into the HIV-1 membrane and inhibits infectivity by a poorly understood mechanism. Recently, SER5 was found to exhibit scramblase-like activity leading to the externalization of phosphatidylserine (PS) on the viral surface, which has been proposed to be responsible for SER5's antiviral activity. This and other reports that document modulation of HIV-1 infectivity by viral lipid composition prompted us to investigate the role of PS in regulating SER5-mediated HIV-1 restriction. First, we show that the level of SER5 incorporation into virions correlates with an increase in PS levels in the outer leaflet of the viral membrane. We developed an assay to estimate the PS distribution across the viral membrane and found that SER5, but not SER2, which lacks antiviral activity, abrogates PS asymmetry by externalizing this lipid. Second, SER5 incorporation diminished the infectivity of pseudoviruses produced from cells lacking a flippase subunit CDC50a and, therefore, exhibited a higher baseline level of surface-accessible PS. Finally, exogenous manipulation of the viral PS levels utilizing methyl-alpha-cyclodextrin revealed a lack of correlation between external PS and virion infectivity. Taken together, our study implies that the increased PS exposure to SER5-containing virions itself is not directly linked to HIV-1 restriction.
Collapse
Affiliation(s)
- Gokul Raghunath
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Elizabeth H. Abbott
- Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Mariana Marin
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Hui Wu
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (M.M.); (H.W.)
| | - Judith Mary Reyes Ballista
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.M.R.B.); (M.A.B.)
| | - Melinda A. Brindley
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.M.R.B.); (M.A.B.)
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Gregory B. Melikyan
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
4
|
Verma S, Chen YC, Marin M, Gillespie SE, Melikyan GB. IFITM1 and IFITM3 Proteins Inhibit the Infectivity of Progeny HIV-1 without Disrupting Envelope Glycoprotein Clusters. Viruses 2023; 15:2390. [PMID: 38140631 PMCID: PMC10748374 DOI: 10.3390/v15122390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Human interferon-induced transmembrane (IFITM) proteins inhibit the fusion of a broad spectrum of enveloped viruses, both when expressed in target cells and when present in infected cells. Upon expression in infected cells, IFITMs incorporate into progeny virions and reduce their infectivity by a poorly understood mechanism. Since only a few envelope glycoproteins (Envs) are present on HIV-1 particles, and Env clustering has been proposed to be essential for optimal infectivity, we asked if IFITM protein incorporation modulates HIV-1 Env clustering. The incorporation of two members of the IFITM family, IFITM1 and IFITM3, into HIV-1 pseudoviruses correlated with a marked reduction of infectivity. Super-resolution imaging of Env distribution on single HIV-1 pseudoviruses did not reveal significant effects of IFITMs on Env clustering. However, IFITM3 reduced the Env processing and incorporation into virions relative to the control and IFITM1-containing viruses. These results show that, in addition to interfering with the Env function, IFITM3 restricts HIV-1 Env cleavage and incorporation into virions. The lack of notable effect of IFITMs on Env clustering supports alternative restriction mechanisms, such as modification of the properties of the viral membrane.
Collapse
Affiliation(s)
- Smita Verma
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.V.); (Y.-C.C.); (M.M.)
| | - Yen-Cheng Chen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.V.); (Y.-C.C.); (M.M.)
| | - Mariana Marin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.V.); (Y.-C.C.); (M.M.)
- Children’s Hospital of Atlanta, Atlanta, GA 30322, USA
| | - Scott E. Gillespie
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.V.); (Y.-C.C.); (M.M.)
| | - Gregory B. Melikyan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.V.); (Y.-C.C.); (M.M.)
- Children’s Hospital of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Ten Eyck A, Chen YC, Gifford L, Torres-Rivera D, Dyer EL, Melikyan GB. Label-free imaging of nuclear membrane for analysis of nuclear import of viral complexes. J Virol Methods 2023; 322:114834. [PMID: 37875225 PMCID: PMC10841631 DOI: 10.1016/j.jviromet.2023.114834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 10/26/2023]
Abstract
HIV-1 enters the nucleus of non-dividing cells through the nuclear pore complex where it integrates into the host genome. The mechanism of HIV-1 nuclear import remains poorly understood. A powerful means to investigate the docking of HIV-1 at the nuclear pore and nuclear import of viral complexes is through single virus tracking in live cells. This approach necessitates fluorescence labeling of HIV-1 particles and the nuclear envelope, which may be challenging, especially in the context of primary cells. Here, we leveraged a deep neural network model for label-free visualization of the nuclear envelope using transmitted light microscopy. A training image set of cells with fluorescently labeled nuclear Lamin B1 (ground truth), along with the corresponding transmitted light images, was acquired and used to train our model to predict the morphology of the nuclear envelope in fixed cells. This protocol yielded accurate predictions of the nuclear membrane and was used in conjunction with virus infection to examine the nuclear entry of fluorescently labeled HIV-1 complexes. Analyses of HIV-1 nuclear import as a function of virus input yielded identical numbers of fluorescent viral complexes per nucleus using the ground truth and predicted nuclear membrane images. We also demonstrate the utility of predicting the nuclear envelope based on transmitted light images for multicolor fluorescence microscopy of infected cells. Importantly, we show that our model can be adapted to predict the nuclear membrane of live cells imaged at 37 °C, making this approach compatible with single virus tracking. Collectively, these findings demonstrate the utility of deep learning approaches for label-free imaging of cellular structures during early stages of virus infection.
Collapse
Affiliation(s)
- Andrew Ten Eyck
- Department of Biomedical Engineering, Georgia Institute of Technology-Emory School of Medicine, Atlanta, GA, USA
| | - Yen-Cheng Chen
- Division of Infectious Diseases, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Levi Gifford
- Division of Infectious Diseases, Department of Pediatrics, Emory University, Atlanta, GA, USA; Graduate Division of Biological and Biomedical Sciences, Biochemistry, Cell and Developmental Biology Program, Emory University, Atlanta, GA, USA
| | - Dariana Torres-Rivera
- Division of Infectious Diseases, Department of Pediatrics, Emory University, Atlanta, GA, USA; Graduate Division of Biological and Biomedical Sciences, Biochemistry, Cell and Developmental Biology Program, Emory University, Atlanta, GA, USA
| | - Eva L Dyer
- Department of Biomedical Engineering, Georgia Institute of Technology-Emory School of Medicine, Atlanta, GA, USA; Department of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gregory B Melikyan
- Department of Biomedical Engineering, Georgia Institute of Technology-Emory School of Medicine, Atlanta, GA, USA; Division of Infectious Diseases, Department of Pediatrics, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, GA, USA.
| |
Collapse
|
6
|
Sharma M, Marin M, Wu H, Prikryl D, Melikyan GB. Human Immunodeficiency Virus 1 Preferentially Fuses with pH-Neutral Endocytic Vesicles in Cell Lines and Human Primary CD4+ T-Cells. ACS NANO 2023; 17:17436-17450. [PMID: 37589658 PMCID: PMC10510587 DOI: 10.1021/acsnano.3c05508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 08/18/2023]
Abstract
Despite extensive efforts, the principal sites of productive HIV-1 entry in different target cells─plasma membrane (PM) vs endosomes─remain controversial. To delineate the site(s) of HIV-1 fusion, we implemented a triple labeling approach that involves tagging pseudoviruses with the fluid-phase viral content marker, iCherry, the viral membrane marker, DiD, and the extraviral pH sensor, ecliptic pHluorin. The viral content marker iCherry is released into the cytoplasm upon virus-cell fusion irrespective of the sites of fusion. In contrast, the extent of dilution of the membrane marker upon fusion with the PM (loss of signal) vs the endosomal membrane (no change in punctate DiD appearance) discriminates between the principal sites of viral fusion. Additionally, ecliptic pHluorin incorporated into the viral membrane reports whether virus fusion occurs in acidic endosomes. Real-time single virus imaging in living HeLa-derived cells, a CD4+ T-cell line, and activated primary human CD4+ T-cells revealed a strong (80-90%) HIV-1 preference for fusion with endosomes. Intriguingly, we observed HIV-1 fusion only with pH-neutral intracellular vesicles and never with acidified endosomes. These endocytic fusion events are likely culminating in productive infection since endocytic inhibitors, such as EIPA, Pitstop2, and Dynasore, as well as a dominant-negative dynamin-2 mutant, inhibited HIV-1 infection in HeLa-derived and primary CD4+ T-cells. Furthermore, the inhibition of endocytosis in HeLa-derived cells promoted hemifusion at the PM but abrogated complete fusion. Collectively, these data reveal that the primary HIV-1 entry pathway in diverse cell types is through fusion with pH-neutral intracellular vesicles.
Collapse
Affiliation(s)
- Manish Sharma
- Department
of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Mariana Marin
- Department
of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Hui Wu
- Department
of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - David Prikryl
- Department
of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Gregory B. Melikyan
- Department
of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| |
Collapse
|
7
|
Piranej S, Zhang L, Bazrafshan A, Marin M, Melikyan GB, Salaita K. Rolosense: Mechanical detection of SARS-CoV-2 using a DNA-based motor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530294. [PMID: 36909543 PMCID: PMC10002644 DOI: 10.1101/2023.02.27.530294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Assays detecting viral infections play a significant role in limiting the spread of diseases such as SARS-CoV-2. Here we present Rolosense, a virus sensing platform that transduces the motion of synthetic DNA-based motors transporting 5-micron particles on RNA fuel chips. Motors and chips are modified with virus-binding aptamers that lead to stalling of motion. Therefore, motors perform a "mechanical test" of viral target and stall in the presence of whole virions which represents a unique mechanism of transduction distinct from conventional assays. Rolosense can detect SARS-CoV-2 spiked in artificial saliva and exhaled breath condensate with a sensitivity of 103 copies/mL and discriminates among other respiratory viruses. The assay is modular and amenable to multiplexing, as we demonstrated one-pot detection of influenza A and SARS-CoV-2. As a proof-of-concept, we show readout can be achieved using a smartphone camera in as little as 15 mins without any sample preparation steps. Taken together, mechanical detection using Rolosense can be broadly applied to any viral target and has the potential to enable rapid, low-cost, point-of-care screening of circulating viruses.
Collapse
Affiliation(s)
- Selma Piranej
- Department of Chemistry, Emory University, Atlanta, GA 30322 (USA)
| | - Luona Zhang
- Department of Chemistry, Emory University, Atlanta, GA 30322 (USA)
| | | | - Mariana Marin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322 (USA)
- Children’s Healthcare of Atlanta, Atlanta, Georgia 30322 (USA)
| | - Gregory B. Melikyan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322 (USA)
- Children’s Healthcare of Atlanta, Atlanta, Georgia 30322 (USA)
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA 30322 (USA)
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322 (USA)
| |
Collapse
|
8
|
Gupta R, Arora K, Roy SS, Joseph A, Rastogi R, Arora NM, Kundu PK. Platforms, advances, and technical challenges in virus-like particles-based vaccines. Front Immunol 2023; 14:1123805. [PMID: 36845125 PMCID: PMC9947793 DOI: 10.3389/fimmu.2023.1123805] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Viral infectious diseases threaten human health and global stability. Several vaccine platforms, such as DNA, mRNA, recombinant viral vectors, and virus-like particle-based vaccines have been developed to counter these viral infectious diseases. Virus-like particles (VLP) are considered real, present, licensed and successful vaccines against prevalent and emergent diseases due to their non-infectious nature, structural similarity with viruses, and high immunogenicity. However, only a few VLP-based vaccines have been commercialized, and the others are either in the clinical or preclinical phases. Notably, despite success in the preclinical phase, many vaccines are still struggling with small-scale fundamental research owing to technical difficulties. Successful production of VLP-based vaccines on a commercial scale requires a suitable platform and culture mode for large-scale production, optimization of transduction-related parameters, upstream and downstream processing, and monitoring of product quality at each step. In this review article, we focus on the advantages and disadvantages of various VLP-producing platforms, recent advances and technical challenges in VLP production, and the current status of VLP-based vaccine candidates at commercial, preclinical, and clinical levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Prabuddha K. Kundu
- Department of Research and Development, Premas Biotech Pvt Ltd., Sector IV, Industrial Model Township (IMT), Manesar, Gurgaon, India
| |
Collapse
|
9
|
Raghunath G, Chen YC, Marin M, Wu H, Melikyan GB. SERINC5-Mediated Restriction of HIV-1 Infectivity Correlates with Resistance to Cholesterol Extraction but Not with Lipid Order of Viral Membrane. Viruses 2022; 14:v14081636. [PMID: 35893701 PMCID: PMC9332783 DOI: 10.3390/v14081636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 12/13/2022] Open
Abstract
Serine incorporator 5 (SER5) is a protein that upon incorporation into virions inhibits HIV-1 infectivity by interfering with the ability of the Env glycoprotein to promote viral fusion. The mechanisms by which SER5 antagonizes HIV-1 fusion are not well understood. A recent study of SER5's structure revealed a lipid-binding pocket, suggesting the ability to sequester lipids. This finding, along with the well-documented modulation of HIV-1 infectivity by viral lipids, especially cholesterol, prompted our examination of SER5's effect on the general lipid order of the HIV-1 membrane. Pseudoviruses bearing the SER5-sensitive HXB2-Env and containing SER5 or SER2, a control protein that lacks antiviral activity, were analyzed using two distinct lipid-order probes. We show that SER5 incorporation does not noticeably affect the lipid order of pseudoviruses. Although viral cholesterol extraction reduces HIV-1 infectivity, SER5+ viruses are less sensitive to cholesterol extraction than the control samples. In contrast, the virus' sensitivity to cholesterol oxidation was not affected by SER5 incorporation. The hydrolytic release of sphingomyelin-sequestered cholesterol had a minimal impact on the apparent resistance to cholesterol extraction. Based on these results, we propose that a subpopulation of more stable Env glycoproteins responsible for the residual infectivity of SER5+ viruses is less sensitive to the cholesterol content of the viral membrane.
Collapse
Affiliation(s)
- Gokul Raghunath
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (Y.-C.C.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Yen-Cheng Chen
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (Y.-C.C.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Mariana Marin
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (Y.-C.C.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Hui Wu
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (Y.-C.C.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Gregory B. Melikyan
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA 30322, USA; (G.R.); (Y.-C.C.); (M.M.); (H.W.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
10
|
Kirschman J, Marin M, Chen YC, Chen J, Herschhorn A, Smith AB, Melikyan GB. SERINC5 Restricts HIV-1 Infectivity by Promoting Conformational Changes and Accelerating Functional Inactivation of Env. Viruses 2022; 14:1388. [PMID: 35891369 PMCID: PMC9323560 DOI: 10.3390/v14071388] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/18/2022] [Accepted: 06/23/2022] [Indexed: 12/16/2022] Open
Abstract
SERINC5 incorporates into HIV-1 particles and inhibits the ability of Env glycoprotein to mediate virus-cell fusion. SERINC5-resistance maps to Env, with primary isolates generally showing greater resistance than laboratory-adapted strains. Here, we examined a relationship between the inhibition of HIV-1 infectivity and the rate of Env inactivation using a panel of SERINC5-resistant and -sensitive HIV-1 Envs. SERINC5 incorporation into pseudoviruses resulted in a faster inactivation of sensitive compared to resistant Env strains. A correlation between fold reduction in infectivity and the rate of inactivation was also observed for multiple Env mutants known to stabilize and destabilize the closed Env structure. Unexpectedly, most mutations disfavoring the closed Env conformation rendered HIV-1 less sensitive to SERINC5. In contrast, functional inactivation of SERINC5-containing viruses was significantly accelerated in the presence of a CD4-mimetic compound, suggesting that CD4 binding sensitizes Env to SERINC5. Using a small molecule inhibitor that selectively targets the closed Env structure, we found that, surprisingly, SERINC5 increases the potency of this compound against a laboratory-adapted Env which prefers a partially open conformation, indicating that SERINC5 may stabilize the closed trimeric Env structure. Our results reveal a complex effect of SERINC5 on Env conformational dynamics that promotes Env inactivation and is likely responsible for the observed restriction phenotype.
Collapse
Affiliation(s)
- Junghwa Kirschman
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (J.K.); (M.M.); (Y.-C.C.)
| | - Mariana Marin
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (J.K.); (M.M.); (Y.-C.C.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Yen-Cheng Chen
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (J.K.); (M.M.); (Y.-C.C.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Junhua Chen
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.); (A.B.S.III)
| | - Alon Herschhorn
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Amos B. Smith
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.); (A.B.S.III)
| | - Gregory B. Melikyan
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; (J.K.); (M.M.); (Y.-C.C.)
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Abstract
The development and adoption of digital twins (DT) for Quality-by-Design (QbD)-based processes with flexible operating points within a proven acceptable range (PAR) and automation through Advanced Process Control (APC) with Process Analytical Technology (PAT) instead of conventional process execution based on offline analytics and inflexible process set points is one of the great challenges in modern biotechnology. Virus-like particles (VLPs) are part of a line of innovative drug substances (DS). VLPs, especially those based on human immunodeficiency virus (HIV), HIV-1 Gag VLPs, have very high potential as a versatile vaccination platform, allowing for pseudotyping with heterologous envelope proteins, e.g., the S protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As enveloped VLPs, optimal process control with minimal hold times is essential. This study demonstrates, for the first time, the use of a digital twin for the overall production process of HIV-1 Gag VLPs from cultivation, clarification, and purification to lyophilization. The accuracy of the digital twins is in the range of 0.8 to 1.4% in depth filtration (DF) and 4.6 to 5.2% in ultrafiltration/diafiltration (UFDF). The uncertainty due to variability in the model parameter determination is less than 4.5% (DF) and less than 3.8% (UFDF). In the DF, a prediction of the final filter capacity was demonstrated from as low as 5.8% (9mbar) of the final transmembrane pressure (TMP). The scale-up based on DT in chromatography shows optimization potential in productivity up to a factor of 2. The schedule based on DT and PAT for APC has been compared to conventional process control, and hold-time and process duration reductions by a factor of 2 have been achieved. This work lays the foundation for the short-term validation of the DT and PAT for APC in an automated S7 process environment and the conversion from batch to continuous production.
Collapse
|
12
|
Process Design and Optimization towards Digital Twins for HIV-Gag VLP Production in HEK293 Cells, including Purification. Processes (Basel) 2022. [DOI: 10.3390/pr10020419] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Despite great efforts to develop a vaccine against human immunodeficiency virus (HIV), which causes AIDS if untreated, no approved HIV vaccine is available to date. A promising class of vaccines are virus-like particles (VLPs), which were shown to be very effective for the prevention of other diseases. In this study, production of HI-VLPs using different 293F cell lines, followed by a three-step purification of HI-VLPs, was conducted. The quality-by-design-based process development was supported by process analytical technology (PAT). The HI-VLP concentration increased 12.5-fold while >80% purity was achieved. This article reports on the first general process development and optimization up to purification. Further research will focus on process development for polishing and formulation up to lyophilization. In addition, process analytical technology and process modeling for process automation and optimization by digital twins in the context of quality-by-design framework will be developed.
Collapse
|
13
|
Sublingual Immunization with Chimeric C1q/CD40 Ligand/HIV Virus-like Particles Induces Strong Mucosal Immune Responses against HIV. Vaccines (Basel) 2021; 9:vaccines9111236. [PMID: 34835167 PMCID: PMC8618657 DOI: 10.3390/vaccines9111236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Development of a vaccine that can elicit robust HIV specific antibody responses in the mucosal compartments is desired for effective prevention of HIV via sexual transmission. However, the current mucosal vaccines have either poor immunogenicity when administered orally or invite safety concerns when administered intranasally. Sublingual immunization has received more attention in recent years based on its efficiency in inducing systemic and mucosal immune responses in both mucosal and extra-mucosal tissues. To facilitate the transport of the immunogen across the sub-mucosal epithelial barrier, we found that CD91, the receptor of C1q, is prevalently expressed in the sublingual mucosal lining, and thus, a modified chimeric C1q surface conjugated CD40L/HIV VLP was generated. The ability of this chimeric C1q/CD40L/HIV VLP to bind, cross the epithelial layer, access and activate the sub-mucosal layer dendritic cells (DCs), and ultimately induce enhanced mucosal and systemic immune responses against HIV is evaluated in this study. We found that C1q/CD40L/HIV VLPs have enhanced binding, increased transport across the epithelial layer, and upregulate DC activation markers as compared to CD40L/HIV VLPs alone. Mice immunized with C1q/CD40L/HIV VLPs by sublingual administration showed higher levels of IgA salivary antibodies against both HIV Gag and Env than mice immunized with CD40L/HIV VLPs. Moreover, sublingual immunization with C1q/CD40L/HIV VLPs induced more Env- and Gag-specific IFN-γ producing T cells than the CD40L/HIV VLPs group. Interestingly, C1q/CD40L/HIV VLP immunization can also induce more mucosal homing T cells than that in CD40L/HIV VLP group. Our data suggest that incorporation of C1q to CD40L/HIV VLPs is a promising novel strategy and that the sublingual immunization can be a favorite immunization route for HIV mucosal vaccines.
Collapse
|
14
|
The late endosome-resident lipid bis(monoacylglycero)phosphate is a cofactor for Lassa virus fusion. PLoS Pathog 2021; 17:e1009488. [PMID: 34492091 PMCID: PMC8448326 DOI: 10.1371/journal.ppat.1009488] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/17/2021] [Accepted: 08/25/2021] [Indexed: 11/20/2022] Open
Abstract
Arenavirus entry into host cells occurs through a low pH-dependent fusion with late endosomes that is mediated by the viral glycoprotein complex (GPC). The mechanisms of GPC-mediated membrane fusion and of virus targeting to late endosomes are not well understood. To gain insights into arenavirus fusion, we examined cell-cell fusion induced by the Old World Lassa virus (LASV) GPC complex. LASV GPC-mediated cell fusion is more efficient and occurs at higher pH with target cells expressing human LAMP1 compared to cells lacking this cognate receptor. However, human LAMP1 is not absolutely required for cell-cell fusion or LASV entry. We found that GPC-induced fusion progresses through the same lipid intermediates as fusion mediated by other viral glycoproteins–a lipid curvature-sensitive intermediate upstream of hemifusion and a hemifusion intermediate downstream of acid-dependent steps that can be arrested in the cold. Importantly, GPC-mediated fusion and LASV pseudovirus entry are specifically augmented by an anionic lipid, bis(monoacylglycero)phosphate (BMP), which is highly enriched in late endosomes. This lipid also specifically promotes cell fusion mediated by Junin virus GPC, an unrelated New World arenavirus. We show that BMP promotes late steps of LASV fusion downstream of hemifusion–the formation and enlargement of fusion pores. The BMP-dependence of post-hemifusion stages of arenavirus fusion suggests that these viruses evolved to use this lipid as a cofactor to selectively fuse with late endosomes. Pathogenic arenaviruses pose a serious health threat. The viral envelope glycoprotein GPC mediates attachment to host cells and drives virus entry via endocytosis and low pH-dependent fusion within late endosomes. Understanding the host factors and processes that are essential for arenavirus fusion may identify novel therapeutic targets. To delineate the mechanism of arenavirus entry, we examined cell-cell fusion induced by the Old World Lassa virus GPC proteins at low pH. Lassa GPC-mediated fusion was augmented by the human LAMP1 receptor and progressed through lipid curvature-sensitive intermediates, such as hemifusion (merger of contacting leaflets of viral and cell membrane without the formation of a fusion pore). We found that most GPC-mediated fusion events were off-path hemifusion structures and that the transition from hemifusion to full fusion and fusion pore enlargement were specifically promoted by an anionic lipid, bis(monoacylglycero)phosphate, which is highly enriched in late endosomes. This lipid also specifically promotes fusion of unrelated New World Junin arenavirus. Our results imply that arenaviruses evolved to use bis(monoacylglycero)phosphate to enter cells from late endosomes.
Collapse
|
15
|
Lewis PE, Poteet EC, Liu D, Chen C, LaBranche CC, Stanfield-Oakley SA, Montefiori DC, Ferrari G, Yao Q. CTLA-4 Blockade, during HIV Virus-Like Particles Immunization, Alters HIV-Specific B-Cell Responses. Vaccines (Basel) 2020; 8:E284. [PMID: 32517277 PMCID: PMC7349993 DOI: 10.3390/vaccines8020284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 12/23/2022] Open
Abstract
Studies have shown that blockade of CTLA-4 promoted the expansion of germinal center B-cells in viral infection or immunization with model antigens. Few studies have evaluated the immunological consequences of CTLA-4 blockade during immunization against relevant vaccine candidates. Here, we investigated the effects of CTLA-4 blockade on HIV virus-like particles (VLPs) vaccination in a C57BL/6J mouse model. We found that CTLA-4 blockade during HIV VLP immunization resulted in increased CD4+ T-cell activation, promoted the expansion of HIV envelope (Env)-specific follicular helper T cell (Tfh) cells, and significantly increased HIV Gag- and Env-specific IgG with higher avidity and antibody-dependent cellular cytotoxicity (ADCC) capabilities. Furthermore, after only a single immunization, CTLA-4 blockade accelerated T-cell dependent IgG class switching and the induction of significantly high serum levels of the B-cell survival factor, A proliferation-inducing ligand (APRIL). Although no significant increase in neutralizing antibodies was observed, increased levels of class-switched Env- and Gag-specific IgG are indicative of increased polyclonal B-cell activation, which demonstrated the ability to mediate and enhance ADCC in this study. Altogether, our findings show that CTLA-4 blockade can increase the levels of HIV antigen-specific B-cell and antigen-specific Tfh cell activity and impact humoral immune responses when combined with a clinically relevant HIV VLP-based vaccine.
Collapse
Affiliation(s)
- Phoebe E. Lewis
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (P.E.L.); (E.C.P.); (D.L.); (C.C.)
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ethan C. Poteet
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (P.E.L.); (E.C.P.); (D.L.); (C.C.)
| | - Dongliang Liu
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (P.E.L.); (E.C.P.); (D.L.); (C.C.)
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (P.E.L.); (E.C.P.); (D.L.); (C.C.)
| | - Celia C. LaBranche
- Duke Human Vaccine Institute, Departments of Medicine, Immunology, Surgery, and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708, USA; (C.C.L.); (S.A.S.-O.); (D.C.M.); (G.F.)
| | - Sherry A. Stanfield-Oakley
- Duke Human Vaccine Institute, Departments of Medicine, Immunology, Surgery, and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708, USA; (C.C.L.); (S.A.S.-O.); (D.C.M.); (G.F.)
| | - David C. Montefiori
- Duke Human Vaccine Institute, Departments of Medicine, Immunology, Surgery, and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708, USA; (C.C.L.); (S.A.S.-O.); (D.C.M.); (G.F.)
| | - Guido Ferrari
- Duke Human Vaccine Institute, Departments of Medicine, Immunology, Surgery, and Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708, USA; (C.C.L.); (S.A.S.-O.); (D.C.M.); (G.F.)
| | - Qizhi Yao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (P.E.L.); (E.C.P.); (D.L.); (C.C.)
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| |
Collapse
|
16
|
Abstract
Baculoviruses are arthropod-specific, enveloped viruses with circular, supercoiled double-stranded deoxyribonucleic acid genomes. While many viruses are studied to seek solutions for their adverse impact on human, veterinary, and plant health, the study of baculoviruses was stimulated initially by their potential utility to control insect pests. Later, the utility of baculovirus as gene expression vectors was evidenced leading to numerous applications. Several strategies are employed to obtain recombinant viruses that express large quantities of heterologous proteins. A major step forward was the development of bacmid technology (the construction of bacterial artificial chromosomes containing the genome of the baculovirus) which allows the manipulation of the baculovirus genome in bacteria. With this technology, foreign genes can be introduced into the bacmid by homologous and site-directed recombination or by transposition. Baculoviruses have been used to explore fundamental questions in molecular biology such as the nature of programmed cell-death. Moreover, the ability of baculoviruses to transduce mammalian cells led to the consideration of their use as gene-therapy and vaccine vectors. Strategies for genetic engineering of baculoviruses have been developed to meet the requirements of new application areas. Display of foreign proteins on the surface of virions or in nucleocapsid structures, the assembly of expressed proteins to form virus-like particles or protein complexes have been explored and validated as vaccines. The aim of this chapter is to update the areas of application of the baculoviruses in protein expression, alternative vaccine designs and gene therapy of infectious diseases and genetic disorders. Finally, we review the baculovirus-derived products on the market and in the pipeline for biomedical and veterinary use.
Collapse
|
17
|
Durous L, Rosa-Calatrava M, Petiot E. Advances in influenza virus-like particles bioprocesses. Expert Rev Vaccines 2019; 18:1285-1300. [DOI: 10.1080/14760584.2019.1704262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Laurent Durous
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Emma Petiot
- Virologie et Pathologie Humaine - VirPath team - Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| |
Collapse
|
18
|
Cervera L, Gòdia F, Tarrés-Freixas F, Aguilar-Gurrieri C, Carrillo J, Blanco J, Gutiérrez-Granados S. Production of HIV-1-based virus-like particles for vaccination: achievements and limits. Appl Microbiol Biotechnol 2019; 103:7367-7384. [DOI: 10.1007/s00253-019-10038-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022]
|
19
|
Marin M, Kushnareva Y, Mason CS, Chanda SK, Melikyan GB. HIV-1 Fusion with CD4+ T cells Is Promoted by Proteins Involved in Endocytosis and Intracellular Membrane Trafficking. Viruses 2019; 11:v11020100. [PMID: 30691001 PMCID: PMC6409670 DOI: 10.3390/v11020100] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 01/23/2019] [Indexed: 01/23/2023] Open
Abstract
The HIV-1 entry pathway into permissive cells has been a subject of debate. Accumulating evidence, including our previous single virus tracking results, suggests that HIV-1 can enter different cell types via endocytosis and CD4/coreceptor-dependent fusion with endosomes. However, recent studies that employed indirect techniques to infer the sites of HIV-1 entry into CD4+ T cells have concluded that endocytosis does not contribute to infection. To assess whether HIV-1 enters these cells via endocytosis, we probed the role of intracellular trafficking in HIV-1 entry/fusion by a targeted shRNA screen in a CD4+ T cell line. We performed a screen utilizing a direct virus-cell fusion assay as readout and identified several host proteins involved in endosomal trafficking/maturation, including Rab5A and sorting nexins, as factors regulating HIV-1 fusion and infection. Knockdown of these proteins inhibited HIV-1 fusion irrespective of coreceptor tropism, without altering the CD4 or coreceptor expression, or compromising the virus’ ability to mediate fusion of two adjacent cells initiated by virus-plasma membrane fusion. Ectopic expression of Rab5A in non-permissive cells harboring Rab5A shRNAs partially restored the HIV-cell fusion. Together, these results implicate endocytic machinery in productive HIV-1 entry into CD4+ T cells.
Collapse
Affiliation(s)
- Mariana Marin
- Department of Pediatric, Division of Infectious Diseases, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, USA.
| | - Yulia Kushnareva
- Functional Genomics Center, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | - Caleb S Mason
- Department of Pediatric, Division of Infectious Diseases, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, USA.
| | - Sumit K Chanda
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Gregory B Melikyan
- Department of Pediatric, Division of Infectious Diseases, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322, USA.
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| |
Collapse
|
20
|
Zaitseva E, Zaitsev E, Melikov K, Arakelyan A, Marin M, Villasmil R, Margolis LB, Melikyan GB, Chernomordik LV. Fusion Stage of HIV-1 Entry Depends on Virus-Induced Cell Surface Exposure of Phosphatidylserine. Cell Host Microbe 2017; 22:99-110.e7. [PMID: 28704658 PMCID: PMC5558241 DOI: 10.1016/j.chom.2017.06.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/13/2017] [Accepted: 06/22/2017] [Indexed: 12/18/2022]
Abstract
HIV-1 entry into host cells starts with interactions between the viral envelope glycoprotein (Env) and cellular CD4 receptors and coreceptors. Previous work has suggested that efficient HIV entry also depends on intracellular signaling, but this remains controversial. Here we report that formation of the pre-fusion Env-CD4-coreceptor complexes triggers non-apoptotic cell surface exposure of the membrane lipid phosphatidylserine (PS). HIV-1-induced PS redistribution depends on Ca2+ signaling triggered by Env-coreceptor interactions and involves the lipid scramblase TMEM16F. Externalized PS strongly promotes Env-mediated membrane fusion and HIV-1 infection. Blocking externalized PS or suppressing TMEM16F inhibited Env-mediated fusion. Exogenously added PS promoted fusion, with fusion dependence on PS being especially strong for cells with low surface density of coreceptors. These findings suggest that cell-surface PS acts as an important cofactor that promotes the fusogenic restructuring of pre-fusion complexes and likely focuses the infection on cells conducive to PS signaling.
Collapse
Affiliation(s)
- Elena Zaitseva
- Section on Membrane Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Eugene Zaitsev
- Section on Membrane Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kamran Melikov
- Section on Membrane Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anush Arakelyan
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Mariana Marin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Rafael Villasmil
- Flow Cytometry Core, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Leonid B Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gregory B Melikyan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Leonid V Chernomordik
- Section on Membrane Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Huang X, Zhu Q, Huang X, Yang L, Song Y, Zhu P, Zhou P. In vivo electroporation in DNA-VLP prime-boost preferentially enhances HIV-1 envelope-specific IgG2a, neutralizing antibody and CD8 T cell responses. Vaccine 2017; 35:2042-2051. [DOI: 10.1016/j.vaccine.2017.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/24/2017] [Accepted: 03/03/2017] [Indexed: 01/14/2023]
|
22
|
Adjuvanting a Simian Immunodeficiency Virus Vaccine with Toll-Like Receptor Ligands Encapsulated in Nanoparticles Induces Persistent Antibody Responses and Enhanced Protection in TRIM5α Restrictive Macaques. J Virol 2017; 91:JVI.01844-16. [PMID: 27928002 DOI: 10.1128/jvi.01844-16] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/23/2016] [Indexed: 12/16/2022] Open
Abstract
Our previous work has shown that antigens adjuvanted with ligands specific for Toll-like receptor 4 (TLR4) and TLR7/8 encapsulated in poly(lactic-co-glycolic) acid (PLGA)-based nanoparticles (NPs) induce robust and durable immune responses in mice and macaques. We investigated the efficacy of these NP adjuvants in inducing protective immunity against simian immunodeficiency virus (SIV). Rhesus macaques (RMs) were immunized with NPs containing TLR4 and TLR7/8 agonists mixed with soluble recombinant SIVmac239-derived envelope (Env) gp140 and Gag p55 (protein) or with virus-like particles (VLPs) containing SIVmac239 Env and Gag. NP-adjuvanted vaccines induced robust innate responses, antigen-specific antibody responses of a greater magnitude and persistence, and enhanced plasmablast responses compared to those achieved with alum-adjuvanted vaccines. NP-adjuvanted vaccines induced antigen-specific, long-lived plasma cells (LLPCs), which persisted in the bone marrow for several months after vaccination. NP-adjuvanted vaccines induced immune responses that were associated with enhanced protection against repeated low-dose, intravaginal challenges with heterologous SIVsmE660 in animals that carried TRIM5α restrictive alleles. The protection induced by immunization with protein-NP correlated with the prechallenge titers of Env-specific IgG antibodies in serum and vaginal secretions. However, no such correlate was apparent for immunization with VLP-NP or alum as the adjuvant. Transcriptional profiling of peripheral blood mononuclear cells isolated within the first few hours to days after primary vaccination revealed that NP-adjuvanted vaccines induced a molecular signature similar to that induced by the live attenuated yellow fever viral vaccine. This systems approach identified early blood transcriptional signatures that correlate with Env-specific antibody responses in vaginal secretions and protection against infection. These results demonstrate the adjuvanticity of the NP adjuvant in inducing persistent and protective antibody responses against SIV in RMs with implications for the design of vaccines against human immunodeficiency virus (HIV). IMPORTANCE The results of the RV144 HIV vaccine trial, which demonstrated a rapid waning of protective immunity with time, have underscored the need to develop strategies to enhance the durability of protective immune responses. Our recent work in mice has highlighted the capacity of nanoparticle-encapsulated TLR ligands (NP) to induce potent and durable antibody responses that last a lifetime in mice. In the present study, we evaluated the ability of these NP adjuvants to promote robust and durable protective immune responses against SIV in nonhuman primates. Our results demonstrate that immunization of rhesus macaques with NP adjuvants mixed with soluble SIV Env or a virus-like particle form of Env (VLP) induces potent and durable Env-specific antibody responses in the serum and in vaginal secretions. These responses were superior to those induced by alum adjuvant, and they resulted in enhanced protection against a low-dose intravaginal challenge with a heterologous strain of SIV in animals with TRIM5a restrictive alleles. These results highlight the potential for such NP TLR L adjuvants in promoting robust and durable antibody responses against HIV in the next generation of HIV immunogens currently being developed.
Collapse
|
23
|
Abstract
We describe the development and potential use of various designs of recombinant HIV-1 envelope glycoprotein trimers that mimic the structure of the virion-associated spike, which is the target for neutralizing antibodies. The goal of trimer development programs is to induce broadly neutralizing antibodies with the potential to intervene against multiple circulating HIV-1 strains. Among the topics we address are the designs of various constructs; how native-like trimers can be produced and purified; the properties of such trimers in vitro and their immunogenicity in various animals; and the immunization strategies that may lead to the eventual elicitation of broadly neutralizing antibodies. In summary, native-like trimers are a now a platform for structure- and immunology-based design improvements that could eventually yield immunogens of practical value for solving the long-standing HIV-1 vaccine problem.
Collapse
Affiliation(s)
- Rogier W. Sanders
- Department of Microbiology and ImmunologyWeill Medical College of Cornell UniversityNew YorkNYUSA
- Department of Medical MicrobiologyAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - John P. Moore
- Department of Microbiology and ImmunologyWeill Medical College of Cornell UniversityNew YorkNYUSA
| |
Collapse
|
24
|
Abstract
Although viruses are simple biological systems, they are capable of evolving highly efficient techniques for infecting cells, expressing their genomes, and generating new copies of themselves. It is possible to genetically manipulate most of the different classes of known viruses in order to produce recombinant viruses that express foreign proteins. Recombinant viruses have been used in gene therapy to deliver selected genes into higher organisms, in vaccinology and immunotherapy, and as important research tools to study the structure and function of these proteins. Virus-like particles (VLPs) are multiprotein structures that mimic the organization and conformation of authentic native viruses but lack the viral genome. They have been applied not only as prophylactic and therapeutic vaccines but also as vehicles in drug and gene delivery and, more recently, as tools in nanobiotechnology. In this chapter, basic and advanced features of viruses and VLPs are presented and their major applications are discussed. The different production platforms based on animal cell technology are explained, and their main challenges and future perspectives are explored. The implications of large-scale production of viruses and VLPs are discussed in the context of process control, monitoring, and optimization. The main upstream and downstream technical challenges are identified and discussed accordingly.
Collapse
|
25
|
Hicar MD, Chen X, Sulli C, Barnes T, Goodman J, Sojar H, Briney B, Willis J, Chukwuma VU, Kalams SA, Doranz BJ, Spearman P, Crowe JE. Human Antibodies that Recognize Novel Immunodominant Quaternary Epitopes on the HIV-1 Env Protein. PLoS One 2016; 11:e0158861. [PMID: 27411063 PMCID: PMC4943599 DOI: 10.1371/journal.pone.0158861] [Citation(s) in RCA: 7] [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: 08/09/2015] [Accepted: 06/23/2016] [Indexed: 11/19/2022] Open
Abstract
Numerous broadly neutralizing antibodies (Abs) target epitopes that are formed or enhanced during mature HIV envelope formation (i.e. quaternary epitopes). Generally, it is thought that Env epitopes that induce broadly neutralizing Abs are difficult to access and poorly immunogenic because of the characteristic oligomerization, conformational flexibility, sequence diversity and extensive glycosylation of Env protein. To enhance for isolation of quaternary epitope-targeting Abs (QtAbs), we previously used HIV virus-like particles (VLPs) to bind B cells from long-term non-progressor subjects to identify a panel of monoclonal Abs. When expressed as recombinant full-length Abs, a subset of these novel Abs exhibited the binding profiles of QtAbs, as they either failed to bind to monomeric Env protein or showed much higher affinity for Env trimers and VLPs. These QtAbs represented a significant proportion of the B-cell response identified with VLPs. The Ab genes of these clones were highly mutated, but they did not neutralize common HIV strains. We sought to further define the epitopes targeted by these QtAbs. Competition-binding and mapping studies revealed these Abs targeted four separate epitopes; they also failed to compete for binding by Abs to known major neutralizing epitopes. Detailed epitope mapping studies revealed that two of the four epitopes were located in the gp41 subunit of Env. These QtAbs bound pre-fusion forms of antigen and showed differential binding kinetics depending on whether oligomers were produced as recombinant gp140 trimers or as full-length Env incorporated into VLPs. Antigenic regions within gp41 present unexpectedly diverse structural epitopes, including these QtAb epitopes, which may be targeted by the naturally occurring Ab response to HIV infection.
Collapse
Affiliation(s)
- Mark D. Hicar
- Departments of Pediatrics, University at Buffalo, Buffalo, New York, United States of America
- Departments of Microbiology and Immunology, University at Buffalo, Buffalo, New York, United States of America
| | - Xuemin Chen
- Departments of Pediatrics, Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Chidananda Sulli
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Trevor Barnes
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Jason Goodman
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Hakimuddin Sojar
- Departments of Pediatrics, University at Buffalo, Buffalo, New York, United States of America
| | - Bryan Briney
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jordan Willis
- The Program in Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Valentine U. Chukwuma
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Spyros A. Kalams
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- The Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Benjamin J. Doranz
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Paul Spearman
- Departments of Pediatrics, Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States of America
| | - James E. Crowe
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
26
|
|
27
|
Kessans SA, Linhart MD, Meador LR, Kilbourne J, Hogue BG, Fromme P, Matoba N, Mor TS. Immunological Characterization of Plant-Based HIV-1 Gag/Dgp41 Virus-Like Particles. PLoS One 2016; 11:e0151842. [PMID: 26986483 PMCID: PMC4795674 DOI: 10.1371/journal.pone.0151842] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/04/2016] [Indexed: 02/08/2023] Open
Abstract
It is widely anticipated that a prophylactic vaccine may be needed to control the HIV/AIDS epidemic worldwide. Despite over two decades of research, a vaccine against HIV-1 remains elusive, although a recent clinical trial has shown promising results. Recent studies have focused on highly conserved domains within HIV-1 such as the membrane proximal external region (MPER) of the envelope glycoprotein, gp41. MPER has been shown to play critical roles in mucosal transmission of HIV-1, though this peptide is poorly immunogenic on its own. Here we provide evidence that plant-produced HIV-1 enveloped virus-like particles (VLPs) consisting of Gag and a deconstructed form of gp41 comprising the MPER, transmembrane, and cytoplasmic domains (Dgp41) provides an effective platform to display MPER for use as an HIV vaccine candidate. Prime-boost strategies combining systemic and mucosal priming with systemic boosting using two different vaccine candidates (VLPs and CTB-MPR--a fusion of MPER and the B-subunit of cholera toxin) were investigated in BALB/c mice. Serum antibody responses against both the Gag and gp41 antigens were elicited when systemically primed with VLPs. These responses could be recalled following systemic boosting with VLPs. In addition, mucosal priming with VLPs allowed for a boosting response against Gag and gp41 when boosted with either candidate. Importantly, the VLPs also induced Gag-specific CD4 and CD8 T-cell responses. This report on the immunogenicity of plant-based Gag/Dgp41 VLPs may represent an important milestone on the road towards a broadly efficacious and inexpensive subunit vaccine against HIV-1.
Collapse
Affiliation(s)
- Sarah A. Kessans
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Mark D. Linhart
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Lydia R. Meador
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Jacquelyn Kilbourne
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Brenda G. Hogue
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Petra Fromme
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona, United States of America
| | - Nobuyuki Matoba
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Tsafrir S. Mor
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
28
|
Hicar MD, Chen X, Kalams SA, Sojar H, Landucci G, Forthal DN, Spearman P, Crowe JE. Low frequency of broadly neutralizing HIV antibodies during chronic infection even in quaternary epitope targeting antibodies containing large numbers of somatic mutations. Mol Immunol 2015; 70:94-103. [PMID: 26748387 DOI: 10.1016/j.molimm.2015.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022]
Abstract
Neutralizing antibodies (Abs) are thought to be a critical component of an appropriate HIV vaccine response. It has been proposed that Abs recognizing conformationally dependent quaternary epitopes on the HIV envelope (Env) trimer may be necessary to neutralize diverse HIV strains. A number of recently described broadly neutralizing monoclonal Abs (mAbs) recognize complex and quaternary epitopes. Generally, many such Abs exhibit extensive numbers of somatic mutations and unique structural characteristics. We sought to characterize the native antibody (Ab) response against circulating HIV focusing on such conformational responses, without a prior selection based on neutralization. Using a capture system based on VLPs incorporating cleaved envelope protein, we identified a selection of B cells that produce quaternary epitope targeting Abs (QtAbs). Similar to a number of broadly neutralizing Abs, the Ab genes encoding these QtAbs showed extensive numbers of somatic mutations. However, when expressed as recombinant molecules, these Abs failed to neutralize virus or mediate ADCVI activity. Molecular analysis showed unusually high numbers of mutations in the Ab heavy chain framework 3 region of the variable genes. The analysis suggests that large numbers of somatic mutations occur in Ab genes encoding HIV Abs in chronically infected individuals in a non-directed, stochastic, manner.
Collapse
Affiliation(s)
- Mark D Hicar
- Department of Pediatrics, University at Buffalo, Buffalo, NY 14222, United States; Department of Microbiology and Immunology, University at Buffalo, Buffalo, NY 14222, United States
| | - Xuemin Chen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Spyros A Kalams
- Department of Pathology, Microbiology and Immunology Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Medicine, Vanderbilt, University Medical Center, Nashville, TN 37232, United States
| | - Hakimuddin Sojar
- Department of Pediatrics, University at Buffalo, Buffalo, NY 14222, United States
| | - Gary Landucci
- Department of Medicine, University of California, Irvine, CA 92668, United States
| | - Donald N Forthal
- Department of Medicine, University of California, Irvine, CA 92668, United States
| | - Paul Spearman
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, United States; Department of Pathology, Microbiology and Immunology Vanderbilt University Medical Center, Nashville, TN 37232, United States; Children's Healthcare of Atlanta, Atlanta, GA 30329, United States
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology Vanderbilt University Medical Center, Nashville, TN 37232, United States; The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States.
| |
Collapse
|
29
|
Optimized production of HIV-1 virus-like particles by transient transfection in CAP-T cells. Appl Microbiol Biotechnol 2015; 100:3935-47. [PMID: 26685677 DOI: 10.1007/s00253-015-7213-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 02/07/2023]
Abstract
HIV-1 virus-like particles (VLPs) have great potential as new-generation vaccines. The novel CAP-T cell line is used for the first time to produce Gag-GFP HIV-1 VLPs by means of polyethylenimine (PEI)-mediated transient transfection. CAP-T cells are adapted to grow to high cell densities in serum-free medium, and are able to express complex recombinant proteins with human post-translational modifications. Furthermore, this cell line is easily transfected with PEI, which offers the flexibility to rapidly generate and screen a number of candidates in preclinical studies. Transient transfection optimization of CAP-T cells has been performed systematically in this work. It is determined that for optimal production, cells need to be growing at mid-exponential phase, Protein Expression Medium (PEM) medium has to be added post-transfection, and cells can be transfected by independent addition of DNA and PEI with no prior complexation. A Box-Behnken experimental design is used to optimize cell density at time of transfection, DNA/cell and PEI/cell ratios. The optimal conditions determined are transfection at a density of 3.3E + 06 cells/mL with 0.5 pg of DNA/cell and 3 pg of PEI/cell. Using the optimized protocol, 6 × 10(10) VLP/mL are obtained, demonstrating that CAP-T is a highly efficient cell line for the production of HIV-1 VLPs and potentially other complex viral-based biotherapeutics.
Collapse
|
30
|
Poteet E, Lewis P, Li F, Zhang S, Gu J, Chen C, Ho SO, Do T, Chiang S, Fujii G, Yao Q. A Novel Prime and Boost Regimen of HIV Virus-Like Particles with TLR4 Adjuvant MPLA Induces Th1 Oriented Immune Responses against HIV. PLoS One 2015; 10:e0136862. [PMID: 26312747 PMCID: PMC4552547 DOI: 10.1371/journal.pone.0136862] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 08/10/2015] [Indexed: 01/11/2023] Open
Abstract
HIV virus-like particles (VLPs) present the HIV envelope protein in its native conformation, providing an ideal vaccine antigen. To enhance the immunogenicity of the VLP vaccine, we sought to improve upon two components; the route of administration and the additional adjuvant. Using HIV VLPs, we evaluated sub-cheek as a novel route of vaccine administration when combined with other conventional routes of immunization. Of five combinations of distinct prime and boost sequences, which included sub-cheek, intranasal, and intradermal routes of administration, intranasal prime and sub-cheek boost (IN+SC) resulted in the highest HIV-specific IgG titers among the groups tested. Using the IN+SC regimen we tested the adjuvant VesiVax Conjugatable Adjuvant Lipid Vesicles (CALV) + monophosphoryl lipid A (MPLA) at MPLA concentrations of 0, 7.5, 12.5, and 25 μg/dose in combination with our VLPs. Mice that received 12.5 or 25 μg/dose MPLA had the highest concentrations of Env-specific IgG2c (20.7 and 18.4 μg/ml respectively), which represents a Th1 type of immune response in C57BL/6 mice. This was in sharp contrast to mice which received 0 or 7.5 μg MPLA adjuvant (6.05 and 5.68 μg/ml of IgG2c respectively). In contrast to IgG2c, MPLA had minor effects on Env-specific IgG1; therefore, 12.5 and 25 μg/dose of MPLA induced the optimal IgG1/IgG2c ratio of 1.3. Additionally, the percentage of germinal center B cells increased significantly from 15.4% in the control group to 31.9% in the CALV + 25 μg MPLA group. These mice also had significantly more IL-2 and less IL-4 Env-specific CD8+ T cells than controls, correlating with an increased percentage of Env-specific central memory CD4+ and CD8+ T cells. Our study shows the strong potential of IN+SC as an efficacious route of administration and the effectiveness of VLPs combined with MPLA adjuvant to induce Env specific Th1-oriented HIV-specific immune responses.
Collapse
Affiliation(s)
- Ethan Poteet
- Michael E. DeBakey Department of Surgery, Division of Surgical Research, Baylor College of Medicine, Houston, TX, 77030, United States of America
| | - Phoebe Lewis
- Michael E. DeBakey Department of Surgery, Division of Surgical Research, Baylor College of Medicine, Houston, TX, 77030, United States of America
| | - Feng Li
- Michael E. DeBakey Department of Surgery, Division of Surgical Research, Baylor College of Medicine, Houston, TX, 77030, United States of America
| | - Sheng Zhang
- Michael E. DeBakey Department of Surgery, Division of Surgical Research, Baylor College of Medicine, Houston, TX, 77030, United States of America
| | - Jianhua Gu
- Houston Methodist Research Institute, Houston, TX, 77030, United States of America
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Division of Surgical Research, Baylor College of Medicine, Houston, TX, 77030, United States of America
| | - Sam On Ho
- Molecular Express, Inc., Rancho Domínguez, CA, 90220, United States of America
| | - Thai Do
- Molecular Express, Inc., Rancho Domínguez, CA, 90220, United States of America
| | - SuMing Chiang
- Molecular Express, Inc., Rancho Domínguez, CA, 90220, United States of America
| | - Gary Fujii
- Molecular Express, Inc., Rancho Domínguez, CA, 90220, United States of America
| | - Qizhi Yao
- Michael E. DeBakey Department of Surgery, Division of Surgical Research, Baylor College of Medicine, Houston, TX, 77030, United States of America
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX, 77030, United States of America
- * E-mail:
| |
Collapse
|
31
|
Mucosal Immunization with Newcastle Disease Virus Vector Coexpressing HIV-1 Env and Gag Proteins Elicits Potent Serum, Mucosal, and Cellular Immune Responses That Protect against Vaccinia Virus Env and Gag Challenges. mBio 2015. [PMID: 26199332 PMCID: PMC4513081 DOI: 10.1128/mbio.01005-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Newcastle disease virus (NDV) avirulent strain LaSota was used to coexpress gp160 Env and p55 Gag from a single vector to enhance both Env-specific and Gag-specific immune responses. The optimal transcription position for both Env and Gag genes in the NDV genome was determined by generating recombinant NDV (rNDV)-Env-Gag (gp160 located between the P and M genes and Gag between the HN and L genes), rNDV-Gag-Env (Gag located between the P and M genes and gp160 between the HN and L genes), rNDV-Env/Gag (gp160 followed by Gag located between the P and M genes), and rNDV-Gag/Env (Gag followed by gp160 located between the P and M genes). All the recombinant viruses replicated at levels similar to those seen with parental NDV in embryonated chicken eggs and in chicken fibroblast cells. Both gp160 and Gag proteins were expressed at high levels in cell culture, with gp160 found to be incorporated into the envelope of NDV. The Gag and Env proteins expressed by all the recombinants except rNDV-Env-Gag self-assembled into human immunodeficiency virus type 1 (HIV-1) virus-like particles (VLPs). Immunization of guinea pigs by the intranasal route with these rNDVs produced long-lasting Env- and Gag-specific humoral immune responses. The Env-specific humoral and mucosal immune responses and Gag-specific humoral immune responses were higher in rNDV-Gag/Env and rNDV-Env/Gag than in the other recombinants. rNDV-Gag/Env and rNDV-Env/Gag were also more efficient in inducing cellular as well as protective immune responses to challenge with vaccinia viruses expressing HIV-1 Env and Gag in mice. These results suggest that vaccination with a single rNDV coexpressing Env and Gag represents a promising strategy to enhance immunogenicity and protective efficacy against HIV. A safe and effective vaccine that can induce both systemic and mucosal immune responses is needed to control HIV-1. In this study, we showed that coexpression of Env and Gag proteins of HIV-1 performed using a single Newcastle disease virus (NDV) vector led to the formation of HIV-1 virus-like particles (VLPs). Immunization of guinea pigs with recombinant NDVs (rNDVs) elicited potent long-lasting systemic and mucosal immune responses to HIV. Additionally, the rNDVs were efficient in inducing cellular immune responses to HIV and protective immunity to challenge with vaccinia viruses expressing HIV Env and Gag in mice. These results suggest that the use of a single NDV expressing Env and Gag proteins simultaneously is a novel strategy to develop a safe and effective vaccine against HIV.
Collapse
|
32
|
Kondo N, Marin M, Kim JH, Desai TM, Melikyan GB. Distinct requirements for HIV-cell fusion and HIV-mediated cell-cell fusion. J Biol Chem 2015; 290:6558-73. [PMID: 25589785 DOI: 10.1074/jbc.m114.623181] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Whether HIV-1 enters cells by fusing with the plasma membrane or with endosomes is a subject of active debate. The ability of HIV-1 to mediate fusion between adjacent cells, a process referred to as "fusion-from-without" (FFWO), shows that this virus can fuse with the plasma membrane. To compare FFWO occurring at the cell surface with HIV-cell fusion through a conventional entry route, we designed an experimental approach that enabled the measurements of both processes in the same sample. The following key differences were observed. First, a very small fraction of viruses fusing with target cells participated in FFWO. Second, whereas HIV-1 fusion with adherent cells was insensitive to actin inhibitors, post-CD4/coreceptor binding steps during FFWO were abrogated. A partial dependence of HIV-cell fusion on actin remodeling was observed in CD4(+) T cells, but this effect appeared to be due to the actin dependence of virus uptake. Third, deletion of the cytoplasmic tail of HIV-1 gp41 dramatically enhanced the ability of the virus to promote FFWO, while having a modest effect on virus-cell fusion. Distinct efficiencies and actin dependences of FFWO versus HIV-cell fusion are consistent with the notion that, except for a minor fraction of particles that mediate fusion between the plasma membranes of adjacent cells, HIV-1 enters through an endocytic pathway. We surmise, however, that cell-cell contacts enabling HIV-1 fusion with the plasma membrane could be favored at the sites of high density of target cells, such as lymph nodes.
Collapse
Affiliation(s)
- Naoyuki Kondo
- From the Division of Pediatric Infectious Diseases, Emory University Children's Center, Atlanta and
| | - Mariana Marin
- From the Division of Pediatric Infectious Diseases, Emory University Children's Center, Atlanta and
| | - Jeong Hwa Kim
- From the Division of Pediatric Infectious Diseases, Emory University Children's Center, Atlanta and
| | - Tanay M Desai
- From the Division of Pediatric Infectious Diseases, Emory University Children's Center, Atlanta and
| | - Gregory B Melikyan
- From the Division of Pediatric Infectious Diseases, Emory University Children's Center, Atlanta and Children's Healthcare of Atlanta, Atlanta, Georgia 30322
| |
Collapse
|
33
|
Tong T, Crooks ET, Osawa K, Robinson JE, Barnes M, Apetrei C, Binley JM. Multi-Parameter Exploration of HIV-1 Virus-Like Particles as Neutralizing Antibody Immunogens in Guinea Pigs, Rabbits and Macaques. Virology 2014; 456-457:55-69. [PMID: 24882891 PMCID: PMC4037872 DOI: 10.1016/j.virol.2014.03.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/26/2014] [Accepted: 03/13/2014] [Indexed: 12/18/2022]
Abstract
Virus-like particles (VLPs) offer a platform to test the hypothesis that, since antibody binding to native envelope glycoprotein (Env) trimers results in HIV-1 neutralization, that native Env trimers presented in membranes may be useful for inducing neutralizing antibodies (nAbs) in a vaccine setting. So far, VLPs have not fulfilled this potential. Here, using a "shotgun" approach, we evaluated a wide cross-section of variables in a series of VLP immunizations. We identified 3 tentative leads. First, that VLP doses may not have been sufficient for optimal nAb induction. Second, that dampening the antigenicity of non-functional Env (for example uncleaved gp160) using either protease digests or IgG masking may be useful. Third, that guinea pig sera preferentially target non-conserved epitopes and exhibit relatively high background activity, suggesting that rabbits may be preferable as small animal vaccine models. Recent immunogenicity studies in rabbits appear to bear out all 3 of these leads.
Collapse
Affiliation(s)
- Tommy Tong
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA
| | - Ema T. Crooks
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA
| | - Keiko Osawa
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA
| | | | - Mary Barnes
- Tulane National Primate Research Center, 18703 Three Rivers Rd, Covington, LA 70433, USA
| | - Cristian Apetrei
- Tulane National Primate Research Center, 18703 Three Rivers Rd, Covington, LA 70433, USA
| | - James M. Binley
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA
| |
Collapse
|
34
|
Kiss G, Chen X, Brindley MA, Campbell P, Afonso CL, Ke Z, Holl JM, Guerrero-Ferreira RC, Byrd-Leotis LA, Steel J, Steinhauer DA, Plemper RK, Kelly DF, Spearman PW, Wright ER. Capturing enveloped viruses on affinity grids for downstream cryo-electron microscopy applications. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2014; 20:164-74. [PMID: 24279992 PMCID: PMC4073796 DOI: 10.1017/s1431927613013937] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Electron microscopy (EM), cryo-electron microscopy (cryo-EM), and cryo-electron tomography (cryo-ET) are essential techniques used for characterizing basic virus morphology and determining the three-dimensional structure of viruses. Enveloped viruses, which contain an outer lipoprotein coat, constitute the largest group of pathogenic viruses to humans. The purification of enveloped viruses from cell culture presents certain challenges. Specifically, the inclusion of host-membrane-derived vesicles, the complete destruction of the viruses, and the disruption of the internal architecture of individual virus particles. Here, we present a strategy for capturing enveloped viruses on affinity grids (AG) for use in both conventional EM and cryo-EM/ET applications. We examined the utility of AG for the selective capture of human immunodeficiency virus virus-like particles, influenza A, and measles virus. We applied nickel-nitrilotriacetic acid lipid layers in combination with molecular adaptors to selectively adhere the viruses to the AG surface. This further development of the AG method may prove essential for the gentle and selective purification of enveloped viruses directly onto EM grids for ultrastructural analyses.
Collapse
Affiliation(s)
- Gabriella Kiss
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
| | - Xuemin Chen
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
| | - Melinda A. Brindley
- Center for Inflammation, Immunity & Infection. Georgia State University. Atlanta, GA 30303
| | - Patricia Campbell
- Department of Microbiology and Immunology. Emory University School of Medicine. GA 30322
| | - Claudio L. Afonso
- USDA, ARS, Southeast Poultry Research Laboratory, Athens, Georgia, USA
| | - Zunlong Ke
- School of Biology, Georgia Institute of Technology, Atlanta GA 30332
| | - Jens M. Holl
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
| | - Ricardo C. Guerrero-Ferreira
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
| | - Lauren A. Byrd-Leotis
- Department of Microbiology and Immunology. Emory University School of Medicine. GA 30322
| | - John Steel
- Department of Microbiology and Immunology. Emory University School of Medicine. GA 30322
| | - David A. Steinhauer
- Department of Microbiology and Immunology. Emory University School of Medicine. GA 30322
| | - Richard K. Plemper
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
- Center for Inflammation, Immunity & Infection. Georgia State University. Atlanta, GA 30303
| | | | - Paul W. Spearman
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
| | - Elizabeth R. Wright
- Division of Pediatric Infectious Diseases. Department of Pediatrics. Emory University School of Medicine. Children’s Healthcare of Atlanta. Atlanta, GA 30322
- To whom correspondence should be addressed. ; Tel. (+1) 404-727-4665; Fax (+1) 404-727-9223
| |
Collapse
|
35
|
Gutiérrez-Granados S, Cervera L, Gòdia F, Carrillo J, Segura MM. Development and validation of a quantitation assay for fluorescently tagged HIV-1 virus-like particles. J Virol Methods 2013; 193:85-95. [DOI: 10.1016/j.jviromet.2013.05.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/24/2013] [Accepted: 05/08/2013] [Indexed: 02/03/2023]
|
36
|
Wallace A, West K, Rothman AL, Ennis FA, Lu S, Wang S. Post-translational intracellular trafficking determines the type of immune response elicited by DNA vaccines expressing Gag antigen of Human Immunodeficiency Virus Type 1 (HIV-1). Hum Vaccin Immunother 2013; 9:2095-102. [PMID: 23941868 DOI: 10.4161/hv.26009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In the current study, immune responses induced by Gag DNA vaccines with different designs were evaluated in Balb/C mice. The results demonstrated that the DNA vaccine with the full length wild type gag gene (Wt-Gag) mainly produced Gag antigens intracellularly and induced a higher level of cell-mediated immune (CMI) responses, as measured by IFN-gamma ELISPOT, intracellular cytokine staining (ICS), and cytotoxic T lymphocytes (CTL) assays against a dominant CD8(+) T cell epitope (AMQMLKETI). In contrast, the addition of a tissue plasminogen activator (tPA) leader sequence significantly improved overall Gag protein expression/secretion and Gag-specific antibody responses; however, Gag-specific CMI responses were decreased. The mutation of zinc-finger motif changed Gag protein expression patterns and reduced the ability to generate both CMI and antibody responses against Gag. These findings indicate that the structure and post-translational processing of antigens expressed by DNA vaccines play a critical role in eliciting optimal antibody or CMI responses.
Collapse
Affiliation(s)
- Aaron Wallace
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA
| | - Kim West
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA; Center for Infectious Diseases and Vaccine Research; University of Massachusetts Medical School; Worcester, MA USA
| | - Alan L Rothman
- Center for Infectious Diseases and Vaccine Research; University of Massachusetts Medical School; Worcester, MA USA
| | - Francis A Ennis
- Center for Infectious Diseases and Vaccine Research; University of Massachusetts Medical School; Worcester, MA USA
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines; Department of Medicine; University of Massachusetts Medical School; Worcester, MA USA
| |
Collapse
|
37
|
Padilla-Parra S, Marin M, Gahlaut N, Suter R, Kondo N, Melikyan GB. Fusion of mature HIV-1 particles leads to complete release of a gag-GFP-based content marker and raises the intraviral pH. PLoS One 2013; 8:e71002. [PMID: 23951066 PMCID: PMC3739801 DOI: 10.1371/journal.pone.0071002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 06/26/2013] [Indexed: 12/30/2022] Open
Abstract
By imaging the release of a GFP-based viral content marker produced upon virus maturation, we have previously found that HIV-1 fuses with endosomes. In contrast, fusion at the cell surface did not progress beyond a lipid mixing stage (hemifusion). However, recent evidence suggesting that free GFP can be trapped within the mature HIV-1 capsid raises concerns that this content marker may not be released immediately after the formation of a fusion pore. To determine whether a significant portion of GFP is trapped in the mature capsid, we first permeabilized the viral membrane with saponin. The overwhelming majority of pseudoviruses fully released GFP while the remaining particles exhibited partial loss or no loss of content. The extent of GFP release correlated with HIV-1 maturation, implying that incomplete Gag processing, but not GFP entrapment by mature capsids, causes partial content release. Next, we designed a complementary assay for visualizing pore formation by monitoring the intraviral pH with an additional pH-sensitive fluorescent marker. The loss of GFP through saponin-mediated pores was associated with a concomitant increase in the intraviral pH due to equilibration with the pH of an external buffer. We next imaged single HIV-cell fusion and found that these events were manifested in a highly correlated loss of content and increase in the intraviral pH, as it equilibrated with the cytosolic pH. Fused or saponin-permeabilized pseudoviruses that partially lost GFP did not release the remaining content marker under conditions expected to promote the capsid dissociation. We were thus unable to detect significant entrapment of GFP by the mature HIV-1 capsid. Together, our results validate the use of the GFP-based content marker for imaging single virus fusion and inferring the sites of HIV-1 entry.
Collapse
Affiliation(s)
- Sergi Padilla-Parra
- Division of Pediatric Infectious Diseases, Emory University Children’s Center, Atlanta, Georgia, United States of America
| | - Mariana Marin
- Division of Pediatric Infectious Diseases, Emory University Children’s Center, Atlanta, Georgia, United States of America
| | - Nivriti Gahlaut
- Division of Pediatric Infectious Diseases, Emory University Children’s Center, Atlanta, Georgia, United States of America
| | - Rolf Suter
- Division of Pediatric Infectious Diseases, Emory University Children’s Center, Atlanta, Georgia, United States of America
| | - Naoyuki Kondo
- Division of Pediatric Infectious Diseases, Emory University Children’s Center, Atlanta, Georgia, United States of America
| | - Gregory B. Melikyan
- Division of Pediatric Infectious Diseases, Emory University Children’s Center, Atlanta, Georgia, United States of America
- Children’s Healthcare of Atlanta, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
38
|
Kessans SA, Linhart MD, Matoba N, Mor T. Biological and biochemical characterization of HIV-1 Gag/dgp41 virus-like particles expressed in Nicotiana benthamiana. PLANT BIOTECHNOLOGY JOURNAL 2013; 11:681-90. [PMID: 23506331 PMCID: PMC3688661 DOI: 10.1111/pbi.12058] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/10/2013] [Accepted: 01/27/2013] [Indexed: 05/29/2023]
Abstract
The transmembrane HIV-1 envelope protein gp41 has been shown to play critical roles in the viral mucosal transmission and infection of CD4⁺ cells. Gag is a structural protein configuring the enveloped viral particles and has been suggested to constitute a target of the cellular immunity that may control viral load. We hypothesized that HIV enveloped virus-like particles (VLPs) consisting of Gag and a deconstructed form of gp41 comprising the membrane proximal external, transmembrane and cytoplasmic domains (dgp41) could be expressed in plants. To this end, plant-optimized HIV-1 genes were constructed and expressed in Nicotiana benthamiana by stable transformation, or transiently using a Tobamovirus-based expression system or a combination of both. Our results of biophysical, biochemical and electron microscopy characterization demonstrates that plant cells could support not only the formation of enveloped HIV-1 Gag VLPs, but also the accumulation of VLPs that incorporated dgp41. These findings provide further impetus for the journey towards a broadly efficacious and inexpensive subunit vaccine against HIV-1.
Collapse
Affiliation(s)
- Sarah A Kessans
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
| | - Mark D Linhart
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
| | - Nobuyuki Matoba
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
- Owensboro Cancer Research ProgramOwensboro, KY, USA
- James Graham Brown Cancer Center and Department of Pharmacology & Toxicology, University of Louisville School of MedicineLouisville, KY, USA
| | - Tsafrir Mor
- School of Life Sciences and The Biodesign Institute, Arizona State UniversityTempe, AZ, USA
| |
Collapse
|
39
|
Cervera L, Gutiérrez-Granados S, Martínez M, Blanco J, Gòdia F, Segura MM. Generation of HIV-1 Gag VLPs by transient transfection of HEK 293 suspension cell cultures using an optimized animal-derived component free medium. J Biotechnol 2013; 166:152-65. [DOI: 10.1016/j.jbiotec.2013.05.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 01/08/2023]
|
40
|
Tomo N, Goto T, Morikawa Y. Trans-packaging of human immunodeficiency virus type 1 genome into Gag virus-like particles in Saccharomyces cerevisiae. Microb Cell Fact 2013; 12:28. [PMID: 23530915 PMCID: PMC3623794 DOI: 10.1186/1475-2859-12-28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/15/2013] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Yeast is recognized as a generally safe microorganism and is utilized for the production of pharmaceutical products, including vaccines. We previously showed that expression of human immunodeficiency virus type 1 (HIV-1) Gag protein in Saccharomyces cerevisiae spheroplasts released Gag virus-like particles (VLPs) extracellularly, suggesting that the production system could be used in vaccine development. In this study, we further establish HIV-1 genome packaging into Gag VLPs in a yeast cell system. RESULTS The nearly full-length HIV-1 genome containing the entire 5' long terminal repeat, U3-R-U5, did not transcribe gag mRNA in yeast. Co-expression of HIV-1 Tat, a transcription activator, did not support the transcription. When the HIV-1 promoter U3 was replaced with the promoter for the yeast glyceraldehyde-3-phosphate dehydrogenase gene, gag mRNA transcription was restored, but no Gag protein expression was observed. Co-expression of HIV-1 Rev, a factor that facilitates nuclear export of gag mRNA, did not support the protein synthesis. Progressive deletions of R-U5 and its downstream stem-loop-rich region (SL) to the gag start ATG codon restored Gag protein expression, suggesting that a highly structured noncoding RNA generated from the R-U5-SL region had an inhibitory effect on gag mRNA translation. When a plasmid containing the HIV-1 genome with the R-U5-SL region was coexpressed with an expression plasmid for Gag protein, the HIV-1 genomic RNA was transcribed and incorporated into Gag VLPs formed by Gag protein assembly, indicative of the trans-packaging of HIV-1 genomic RNA into Gag VLPs in a yeast cell system. The concentration of HIV-1 genomic RNA in Gag VLPs released from yeast was approximately 500-fold higher than that in yeast cytoplasm. The deletion of R-U5 to the gag gene resulted in the failure of HIV-1 RNA packaging into Gag VLPs, indicating that the packaging signal of HIV-1 genomic RNA present in the R-U5 to gag region functions similarly in yeast cells. CONCLUSIONS Our data indicate that selective trans-packaging of HIV-1 genomic RNA into Gag VLPs occurs in a yeast cell system, analogous to a mammalian cell system, suggesting that yeast may provide an alternative packaging system for lentiviral RNA.
Collapse
Affiliation(s)
- Naoki Tomo
- Kitasato Institute for Life Sciences and Graduate School for Infection Control, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo, 108-8641, Japan
| | | | | |
Collapse
|
41
|
Chen Q, Lai H. Plant-derived virus-like particles as vaccines. Hum Vaccin Immunother 2013; 9:26-49. [PMID: 22995837 PMCID: PMC3667944 DOI: 10.4161/hv.22218] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 09/06/2012] [Accepted: 09/13/2012] [Indexed: 02/07/2023] Open
Abstract
Virus-like particles (VLPs) are self-assembled structures derived from viral antigens that mimic the native architecture of viruses but lack the viral genome. VLPs have emerged as a premier vaccine platform due to their advantages in safety, immunogenicity, and manufacturing. The particulate nature and high-density presentation of viral structure proteins on their surface also render VLPs as attractive carriers for displaying foreign epitopes. Consequently, several VLP-based vaccines have been licensed for human use and achieved significant clinical and economical success. The major challenge, however, is to develop novel production platforms that can deliver VLP-based vaccines while significantly reducing production times and costs. Therefore, this review focuses on the essential role of plants as a novel, speedy and economical production platform for VLP-based vaccines. The advantages of plant expression systems are discussed in light of their distinctive posttranslational modifications, cost-effectiveness, production speed, and scalability. Recent achievements in the expression and assembly of VLPs and their chimeric derivatives in plant systems as well as their immunogenicity in animal models are presented. Results of human clinical trials demonstrating the safety and efficacy of plant-derived VLPs are also detailed. Moreover, the promising implications of the recent creation of "humanized" glycosylation plant lines as well as the very recent approval of the first plant-made biologics by the U. S. Food and Drug Administration (FDA) for plant production and commercialization of VLP-based vaccines are discussed. It is speculated that the combined potential of plant expression systems and VLP technology will lead to the emergence of successful vaccines and novel applications of VLPs in the near future.
Collapse
Affiliation(s)
- Qiang Chen
- Center for Infectious Diseases and Vaccinology, Biodesign Institute at Arizona State University, Tempe, AZ USA.
| | | |
Collapse
|
42
|
Kondo N, Melikyan GB. Intercellular adhesion molecule 1 promotes HIV-1 attachment but not fusion to target cells. PLoS One 2012; 7:e44827. [PMID: 22970312 PMCID: PMC3435301 DOI: 10.1371/journal.pone.0044827] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/07/2012] [Indexed: 12/24/2022] Open
Abstract
Incorporation of intercellular adhesion molecule 1 (ICAM-1) into HIV-1 particles is known to markedly enhance the virus binding and infection of cells expressing lymphocyte function-associated antigen-1 (LFA-1). At the same time, ICAM-1 has been reported to exert a less pronounced effect on HIV-1 fusion with lymphoid cells. Here we examined the role of ICAM-1/LFA-1 interactions in productive HIV-1 entry into lymphoid cells using a direct virus-cell fusion assay. ICAM-1 promoted HIV-1 attachment to cells in a temperature-dependent manner. It exerted a marginal effect on virus binding in the cold, but enhanced binding up to 4-fold at physiological temperature. ICAM-1-independent attachment in the cold was readily reversible upon subsequent incubation at elevated temperature, whereas ICAM-1-bearing particles were largely retained by cells. The better virus retention resulted in a proportional increase in HIV-1 internalization and fusion, suggesting that ICAM-1 did not specifically accelerate endocytosis or fusion steps. We also measured the rates of CD4 engagement, productive endocytosis and HIV-endosome fusion using specific fusion inhibitors. These rates were virtually independent of the presence of ICAM-1 in viral particles. Importantly, irrespective of the presence of ICAM-1, HIV-1 escaped from the low temperature block, which stopped virus endocytosis and fusion, much later than from a membrane-impermeant fusion inhibitor targeting surface-accessible particles. This result, along with the complete inhibition of HIV-1 fusion by a small molecule dynamin inhibitor, implies this virus enters lymphoid cells used in this study via endocytosis and that this pathway is not altered by the viral ICAM-1. Our data highlight the role of ICAM-1 in stabilizing the HIV-1 attachment to LFA-1 expressing cells, which leads to a proportional enhancement of the receptor-mediated uptake and fusion with endosomes.
Collapse
Affiliation(s)
- Naoyuki Kondo
- Division of Pediatric Infectious Diseases, Emory Children's Center, Atlanta, Georgia, United States of America
| | - Gregory B. Melikyan
- Division of Pediatric Infectious Diseases, Emory Children's Center, Atlanta, Georgia, United States of America
- Children's Healthcare of Atlanta, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
43
|
Li L, Wang XH, Banerjee S, Volsky B, Williams C, Virland D, Nadas A, Seaman MS, Chen X, Spearman P, Zolla-Pazner S, Gorny MK. Different pattern of immunoglobulin gene usage by HIV-1 compared to non-HIV-1 antibodies derived from the same infected subject. PLoS One 2012; 7:e39534. [PMID: 22761815 PMCID: PMC3382572 DOI: 10.1371/journal.pone.0039534] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 05/23/2012] [Indexed: 12/15/2022] Open
Abstract
A biased usage of immunoglobulin (Ig) genes is observed in human anti-HIV-1 monoclonal antibodies (mAbs) resulting probably from compensation to reduced usage of the VH3 family genes, while the other alternative suggests that this bias usage is due to antigen requirements. If the antigen structure is responsible for the preferential usage of particular Ig genes, it may have certain implications for HIV vaccine development by the targeting of particular Ig gene-encoded B cell receptors to induce neutralizing anti-HIV-1 antibodies. To address this issue, we have produced HIV-1 specific and non-HIV-1 mAbs from an infected individual and analyzed the Ig gene usage. Green-fluorescence labeled virus-like particles (VLP) expressing HIV-1 envelope (Env) proteins of JRFL and BaL and control VLPs (without Env) were used to select single B cells for the production of 68 recombinant mAbs. Ten of these mAbs were HIV-1 Env specific with neutralizing activity against V3 and the CD4 binding site, as well as non-neutralizing mAbs to gp41. The remaining 58 mAbs were non-HIV-1 Env mAbs with undefined specificities. Analysis revealed that biased usage of Ig genes was restricted only to anti-HIV-1 but not to non-HIV-1 mAbs. The VH1 family genes were dominantly used, followed by VH3, VH4, and VH5 among anti-HIV-1 mAbs, while non-HIV-1 specific mAbs preferentially used VH3 family genes, followed by VH4, VH1 and VH5 families in a pattern identical to Abs derived from healthy individuals. This observation suggests that the biased usage of Ig genes by anti-HIV-1 mAbs is driven by structural requirements of the virus antigens rather than by compensation to any depletion of VH3 B cells due to autoreactive mechanisms, according to the gp120 superantigen hypothesis.
Collapse
Affiliation(s)
- Liuzhe Li
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Xiao-Hong Wang
- Veterans Affairs New York Harbor Healthcare System, New York, New York, United States of America
| | - Sagarika Banerjee
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Barbara Volsky
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Constance Williams
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Diana Virland
- Veterans Affairs New York Harbor Healthcare System, New York, New York, United States of America
| | - Arthur Nadas
- Institute of Environmental Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Michael S. Seaman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xuemin Chen
- Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
| | - Paul Spearman
- Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
| | - Susan Zolla-Pazner
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- Veterans Affairs New York Harbor Healthcare System, New York, New York, United States of America
| | - Miroslaw K. Gorny
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
HIV-1 virus-like particles produced by stably transfected Drosophila S2 cells: a desirable vaccine component. J Virol 2012; 86:7662-76. [PMID: 22553333 DOI: 10.1128/jvi.07164-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of a successful vaccine against human immunodeficiency virus type 1 (HIV-1) likely requires immunogens that elicit both broadly neutralizing antibodies against envelope spikes and T cell responses that recognize multiple viral proteins. HIV-1 virus-like particles (VLP), because they display authentic envelope spikes on the particle surface, may be developed into such immunogens. However, in one way or the other current systems for HIV-1 VLP production have many limitations. To overcome these, in the present study we developed a novel strategy to produce HIV-1 VLP using stably transfected Drosophila S2 cells. We cotransfected S2 cells with plasmids encoding HIV-1 envelope, Gag, and Rev proteins and a selection marker. After stably transfected S2 clones were established, HIV-1 VLP and their immunogenicity in mice were carefully evaluated. Here, we report that HIV-1 envelope proteins are properly cleaved, glycosylated, and incorporated into VLP with Gag. The amount of VLP released into culture supernatants is comparable to those produced by insect cells infected with recombinant baculoviruses. Moreover, cryo-electron microscopy tomography revealed average 17 spikes per purified VLP, and antigenic epitopes on the spikes were recognized by the broadly neutralizing antibodies 2G12, b12, VRC01, and 4E10 but not by PG16. Finally, mice primed with DNA and boosted with VLP in the presence of CpG exhibited anti-envelope antibody responses, including ELISA-binding, neutralizing, antibody-dependent cell-mediated cytotoxicity and antibody-dependent cell-mediated viral inhibition, as well as envelope and Gag-specific CD8 T cell responses. Thus, we conclude that HIV-1 VLP produced by the S2 expression system has many desirable features to be developed into a vaccine component against HIV-1.
Collapse
|
45
|
HIV-1 virus-like particles bearing pure env trimers expose neutralizing epitopes but occlude nonneutralizing epitopes. J Virol 2012; 86:3574-87. [PMID: 22301141 DOI: 10.1128/jvi.06938-11] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hypothetically, since native HIV-1 Env trimers are exclusively recognized by neutralizing antibodies, they might induce the neutralizing antibodies in a vaccine setting. This idea has not been evaluated due to the difficulty of separating trimers from nonfunctional Env (uncleaved gp160 and gp41 stumps). The latter are immunodominant and induce nonneutralizing antibodies. We previously showed that nonfunctional Env can be selectively cleared from virus-like particle (VLP) surfaces by enzyme digests (E. T. Crooks, T. Tong(,) K. Osawa, and J. M. Binley, J.Virol. 85:5825, 2011). Here, we investigated the effects of these digests on the antigenicity of VLPs and their sensitivity to neutralization. Before digestion, WT VLPs (bearing wild-type Env) and UNC VLPs (bearing uncleaved gp160) were recognized by various Env-specific monoclonal antibodies (MAbs), irrespective of their neutralizing activity, a result which is consistent with the presence of nonfunctional Env. After digestion, only neutralizing MAbs recognized WT VLPs, consistent with selective removal of nonfunctional Env (i.e., "trimer VLPs"). Digests eliminated the binding of all MAbs to UNC VLPs, again consistent with removal of nonfunctional Env. An exception was MAb 2F5, which weakly bound to digested UNC VLPs and bald VLPs (bearing no Env), perhaps due to lipid cross-reactivity. Trimer VLPs were infectious, and their neutralization sensitivity was largely comparable to that of undigested WT VLPs. However, they were ∼100-fold more sensitive to the MAbs 4E10 and Z13e1, suggesting increased exposure of the gp41 base. Importantly, a scatterplot analysis revealed a strong correlation between MAb binding and neutralization of trimer VLPs. This suggests that trimer VLPs bear essentially pure native trimer that should allow its unfettered evaluation in a vaccine setting.
Collapse
|
46
|
Makarova N, Zhao C, Zhang Y, Bhosle S, Suppiah S, Rhea JM, Kozyr N, Arnold RS, Ly H, Molinaro RJ, Parslow TG, Hunter E, Liotta D, Petros J, Blackwell JL. Antibody responses against xenotropic murine leukemia virus-related virus envelope in a murine model. PLoS One 2011; 6:e18272. [PMID: 21494670 PMCID: PMC3071813 DOI: 10.1371/journal.pone.0018272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 02/24/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Xenotropic murine leukemia virus-related virus (XMRV) was recently discovered to be the first human gammaretrovirus that is associated with chronic fatigue syndrome and prostate cancer (PC). Although a mechanism for XMRV carcinogenesis is yet to be established, this virus belongs to the family of gammaretroviruses well known for their ability to induce cancer in the infected hosts. Since its original identification XMRV has been detected in several independent investigations; however, at this time significant controversy remains regarding reports of XMRV detection/prevalence in other cohorts and cell type/tissue distribution. The potential risk of human infection, coupled with the lack of knowledge about the basic biology of XMRV, warrants further research, including investigation of adaptive immune responses. To study immunogenicity in vivo, we vaccinated mice with a combination of recombinant vectors expressing codon-optimized sequences of XMRV gag and env genes and virus-like particles (VLP) that had the size and morphology of live infectious XMRV. RESULTS Immunization elicited Env-specific binding and neutralizing antibodies (NAb) against XMRV in mice. The peak titers for ELISA-binding antibodies and NAb were 1:1024 and 1:464, respectively; however, high ELISA-binding and NAb titers were not sustained and persisted for less than three weeks after immunizations. CONCLUSIONS Vaccine-induced XMRV Env antibody titers were transiently high, but their duration was short. The relatively rapid diminution in antibody levels may in part explain the differing prevalences reported for XMRV in various prostate cancer and chronic fatigue syndrome cohorts. The low level of immunogenicity observed in the present study may be characteristic of a natural XMRV infection in humans.
Collapse
Affiliation(s)
- Natalia Makarova
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Chunxia Zhao
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Yuanyuan Zhang
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Sushma Bhosle
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Suganthi Suppiah
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Jeanne M. Rhea
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Natalia Kozyr
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Rebecca S. Arnold
- Department of Urology, Emory University, Atlanta, Georgia, United States of America
| | - Hinh Ly
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ross J. Molinaro
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Core Laboratories Emory University Hospital Midtown, Emory University, Atlanta, Georgia, United States of America
| | - Tristram G. Parslow
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Eric Hunter
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Dennis Liotta
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - John Petros
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, United States of America
- Department of Urology, Emory University, Atlanta, Georgia, United States of America
- Department of Hematology-Oncology, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Jerry L. Blackwell
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Division of Infectious Diseases, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
47
|
Roldão A, Mellado MCM, Castilho LR, Carrondo MJT, Alves PM. Virus-like particles in vaccine development. Expert Rev Vaccines 2011; 9:1149-76. [PMID: 20923267 DOI: 10.1586/erv.10.115] [Citation(s) in RCA: 591] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Virus-like particles (VLPs) are multiprotein structures that mimic the organization and conformation of authentic native viruses but lack the viral genome, potentially yielding safer and cheaper vaccine candidates. A handful of prophylactic VLP-based vaccines is currently commercialized worldwide: GlaxoSmithKline's Engerix (hepatitis B virus) and Cervarix (human papillomavirus), and Merck and Co., Inc.'s Recombivax HB (hepatitis B virus) and Gardasil (human papillomavirus) are some examples. Other VLP-based vaccine candidates are in clinical trials or undergoing preclinical evaluation, such as, influenza virus, parvovirus, Norwalk and various chimeric VLPs. Many others are still restricted to small-scale fundamental research, despite their success in preclinical tests. This article focuses on the essential role of VLP technology in new-generation vaccines against prevalent and emergent diseases. The implications of large-scale VLP production are discussed in the context of process control, monitorization and optimization. The main up- and down-stream technical challenges are identified and discussed accordingly. Successful VLP-based vaccine blockbusters are briefly presented concomitantly with the latest results from clinical trials and the recent developments in chimeric VLP-based technology for either therapeutic or prophylactic vaccination.
Collapse
Affiliation(s)
- António Roldão
- Instituto de Tecnologia Química e Biológica/Universidade Nova de Lisboa, Apartado 127, P-2781-901, Oeiras, Portugal
| | | | | | | | | |
Collapse
|
48
|
Domingo-Espín J, Unzueta U, Saccardo P, Rodríguez-Carmona E, Corchero JL, Vázquez E, Ferrer-Miralles N. Engineered biological entities for drug delivery and gene therapy protein nanoparticles. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 104:247-98. [PMID: 22093221 PMCID: PMC7173510 DOI: 10.1016/b978-0-12-416020-0.00006-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of genetic engineering techniques has speeded up the growth of the biotechnological industry, resulting in a significant increase in the number of recombinant protein products on the market. The deep knowledge of protein function, structure, biological interactions, and the possibility to design new polypeptides with desired biological activities have been the main factors involved in the increase of intensive research and preclinical and clinical approaches. Consequently, new biological entities with added value for innovative medicines such as increased stability, improved targeting, and reduced toxicity, among others have been obtained. Proteins are complex nanoparticles with sizes ranging from a few nanometers to a few hundred nanometers when complex supramolecular interactions occur, as for example, in viral capsids. However, even though protein production is a delicate process that imposes the use of sophisticated analytical methods and negative secondary effects have been detected in some cases as immune and inflammatory reactions, the great potential of biodegradable and tunable protein nanoparticles indicates that protein-based biotechnological products are expected to increase in the years to come.
Collapse
Affiliation(s)
- Joan Domingo-Espín
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| | - Ugutz Unzueta
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| | - Paolo Saccardo
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| | - Escarlata Rodríguez-Carmona
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| | - José Luís Corchero
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| | - Esther Vázquez
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| | - Neus Ferrer-Miralles
- Institute for Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Barcelona, Spain
| |
Collapse
|
49
|
A plasmid DNA immunogen expressing fifteen protein antigens and complex virus-like particles (VLP+) mimicking naturally occurring HIV. Vaccine 2011; 29:744-53. [DOI: 10.1016/j.vaccine.2010.11.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 11/04/2010] [Accepted: 11/08/2010] [Indexed: 11/17/2022]
|
50
|
Roldão A, Silva A, Mellado M, Alves P, Carrondo M. Viruses and Virus-Like Particles in Biotechnology. COMPREHENSIVE BIOTECHNOLOGY 2011. [PMCID: PMC7151966 DOI: 10.1016/b978-0-08-088504-9.00072-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Although viruses are simple biological systems, they are capable of evolving highly efficient techniques for infecting cells, expressing their genomes, and generating new copies of themselves. It is possible to genetically manipulate most of the different classes of known viruses in order to produce recombinant viruses that express foreign proteins. Recombinant viruses have been used in gene therapy to deliver selected genes into higher organisms, in vaccinology and immunotherapy, and as important research tools to study the structure and function of these proteins. Virus-like particles (VLPs) are multiprotein structures that mimic the organization and conformation of authentic native viruses but lack the viral genome. They have been applied not only as prophylactic and therapeutic vaccines but also as vehicles in drug and gene delivery and, more recently, as tools in nanobiotechnology. In this article, basic and advanced features of viruses and VLPs are presented and their major applications are discussed. The different production platforms based on animal cell technology are explained, and their main challenges and future perspectives are explored. The implications of large-scale production of viruses and VLPs are discussed in the context of process control, monitorization, and optimization. The main upstream and downstream technical challenges are identified and discussed accordingly.
Collapse
|