1
|
Fishburn AT, Florio CJ, Klaessens TN, Prince B, Adia NAB, Lopez NJ, Beesabathuni NS, Becker SS, Cherkashchenko L, Haggard Arcé ST, Hoang V, Shiu TN, Richardson RB, Evans MJ, Rückert C, Shah PS. Microcephaly protein ANKLE2 promotes Zika virus replication. mBio 2025; 16:e0268324. [PMID: 39804047 PMCID: PMC11796389 DOI: 10.1128/mbio.02683-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/26/2024] [Indexed: 02/06/2025] Open
Abstract
Orthoflaviviruses are positive-sense single-stranded RNA viruses that hijack host proteins to promote their own replication. Zika virus (ZIKV) is infamous among orthoflaviviruses for its association with severe congenital birth defects, notably microcephaly. We previously mapped ZIKV-host protein interactions and identified the interaction between ZIKV non-structural protein 4A (NS4A) and host microcephaly protein ankyrin repeat and LEM domain-containing 2 (ANKLE2). Using a fruit fly model, we showed that NS4A induced microcephaly in an ANKLE2-dependent manner. Here, we explore the role of ANKLE2 in ZIKV replication to understand the biological significance of the interaction from a viral perspective. We observe that ANKLE2 localization is drastically shifted to sites of NS4A accumulation during infection and that knockout of ANKLE2 reduces ZIKV replication in multiple human cell lines. This decrease in virus replication is coupled with a moderate increase in innate immune activation. Using microscopy, we observe dysregulated formation of virus-induced endoplasmic reticulum rearrangements in ANKLE2 knockout cells. Knockdown of the ANKLE2 ortholog in Aedes aegypti cells also decreases virus replication, suggesting ANKLE2 is a beneficial replication factor across hosts. Finally, we show that NS4A from four other orthoflaviviruses physically interacts with ANKLE2 and is also beneficial to their replication. Thus, ANKLE2 likely promotes orthoflavivirus replication by regulating membrane rearrangements that serve to accelerate viral genome replication and protect viral dsRNA from immune detection. Taken together with our previous results, our findings indicate that ZIKV and other orthoflaviviruses hijack ANKLE2 for a conserved role in replication, and this drives unique pathogenesis for ZIKV since ANKLE2 has essential roles in developing tissues.IMPORTANCEZIKV is a major concern due to its association with birth defects, including microcephaly. We previously identified a physical interaction between ZIKV NS4A and host microcephaly protein ANKLE2. Mutations in ANKLE2 cause congenital microcephaly, and NS4A induces microcephaly in an ANKLE2-dependent manner. Here, we establish the role of ANKLE2 in ZIKV replication. Depletion of ANKLE2 from cells significantly reduces ZIKV replication and disrupts virus-induced membrane rearrangements. ANKLE2's ability to promote ZIKV replication is conserved in mosquito cells and for other related mosquito-borne orthoflaviviruses. Our data point to an overall model in which ANKLE2 regulates virus-induced membrane rearrangements to accelerate orthoflavivirus replication and avoid immune detection. However, ANKLE2's unique role in ZIKV NS4A-induced microcephaly is a consequence of ZIKV infection of important developing tissues in which ANKLE2 has essential roles.
Collapse
Affiliation(s)
- Adam T. Fishburn
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Cole J. Florio
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Thomas N. Klaessens
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Brian Prince
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada, USA
| | - Neil A. B. Adia
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Nicholas J. Lopez
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | | | - Sydney S. Becker
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Liubov Cherkashchenko
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Sophia T. Haggard Arcé
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Vivian Hoang
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Traci N. Shiu
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - R. Blake Richardson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Matthew J. Evans
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada, USA
| | - Priya S. Shah
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
- Department of Chemical Engineering, University of California, Davis, California, USA
| |
Collapse
|
2
|
Champeil J, Mangion M, Gilbert R, Gaillet B. Improved Manufacturing Methods of Extracellular Vesicles Pseudotyped with the Vesicular Stomatitis Virus Glycoprotein. Mol Biotechnol 2024; 66:1116-1131. [PMID: 38182864 DOI: 10.1007/s12033-023-01007-3] [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: 07/12/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024]
Abstract
Extracellular vesicles (EV), which expose the vesicular stomatitis virus glycoprotein (VSVG) on their surface, are used for delivery of nucleic acids and proteins in human cell lines. These particles are biomanufactured using methods that are difficult to scale up. Here, we describe the development of the first EV-VSVG production process in serum-free media using polyethylenimine (PEI)-based transient transfection of HEK293 suspension cells, as well as the first EV-VSVG purification process to utilize both ultracentrifugation and chromatography. Three parameters were investigated for EV-VSVG production: cell density, DNA concentration, and DNA:PEI ratio. The best production titer was obtained with 3 × 106 cells/mL, a plasmid concentration of 2 µg/mL, and a DNA:PEI ratio of 1:4. The production kinetics of VSVG was performed and showed that the highest amount of VSVG was obtained 3 days after transfection. Addition of cell culture supplements during the transfection resulted in an increase in VSVG production, with a maximum yield obtained with 2 mM of sodium butyrate added 18 h after transfection. Moreover, the absence of EV-VSVG during cell transfection with a GFP-coding plasmid revealed to be ineffective, with no fluorescent cells. An efficient EV-VSVG purification procedure consisting of a two-step concentration by low-speed centrifugation and sucrose cushion ultracentrifugation followed by a heparin affinity chromatography purification was also developed. Purified bioactive EV-VSVG preparations were characterized and revealed that EV-VSVG are spherical particles of 176.4 ± 88.32 nm with 91.4% of protein similarity to exosomes.
Collapse
Affiliation(s)
- Juliette Champeil
- Chemical Engineering Department, Université Laval, 1065, Avenue de la Médecine, Pavillon Pouliot, Québec, QC, G1V 0A6, Canada
- PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC, H2X 3Y7, Canada
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada
| | - Mathias Mangion
- Chemical Engineering Department, Université Laval, 1065, Avenue de la Médecine, Pavillon Pouliot, Québec, QC, G1V 0A6, Canada
- PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC, H2X 3Y7, Canada
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada
| | - Rénald Gilbert
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada
- Human Health Therapeutics Research Center, National Research Council Canada, 6100, Avenue Royalmount, Montréal, Québec, H4P 2R2, Canada
| | - Bruno Gaillet
- Chemical Engineering Department, Université Laval, 1065, Avenue de la Médecine, Pavillon Pouliot, Québec, QC, G1V 0A6, Canada.
- PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC, H2X 3Y7, Canada.
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada.
| |
Collapse
|
3
|
Labisch JJ, Kassar M, Bollmann F, Valentic A, Hubbuch J, Pflanz K. Steric exclusion chromatography of lentiviral vectors using hydrophilic cellulose membranes. J Chromatogr A 2022; 1674:463148. [DOI: 10.1016/j.chroma.2022.463148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022]
|
4
|
Petit MJ, Kenaston MW, Pham OH, Nagainis AA, Fishburn AT, Shah PS. Nuclear dengue virus NS5 antagonizes expression of PAF1-dependent immune response genes. PLoS Pathog 2021; 17:e1010100. [PMID: 34797876 PMCID: PMC8641875 DOI: 10.1371/journal.ppat.1010100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/03/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Dengue virus (DENV) disruption of the innate immune response is critical to establish infection. DENV non-structural protein 5 (NS5) plays a central role in this disruption, such as antagonism of STAT2. We recently found that DENV serotype 2 (DENV2) NS5 interacts with Polymerase associated factor 1 complex (PAF1C). The primary members of PAF1C are PAF1, LEO1, CTR9, and CDC73. This nuclear complex is an emerging player in the immune response. It promotes the expression of many genes, including genes related to the antiviral, antimicrobial and inflammatory responses, through close association with the chromatin of these genes. Our previous work demonstrated that NS5 antagonizes PAF1C recruitment to immune response genes. However, it remains unknown if NS5 antagonism of PAF1C is complementary to its antagonism of STAT2. Here, we show that knockout of PAF1 enhances DENV2 infectious virion production. By comparing gene expression profiles in PAF1 and STAT2 knockout cells, we find that PAF1 is necessary to express immune response genes that are STAT2-independent. Finally, we mapped the viral determinants for the NS5-PAF1C protein interaction. We found that NS5 nuclear localization and the C-terminal region of the methyltransferase domain are required for its interaction with PAF1C. Mutation of these regions rescued the expression of PAF1-dependent immune response genes that are antagonized by NS5. In sum, our results support a role for PAF1C in restricting DENV2 replication that NS5 antagonizes through its protein interaction with PAF1C. Dengue virus (DENV) is a pathogen that infects nearly 400 million people a year and thus represents a major challenge for public health. Productive infection by DENV relies on the effective evasion of intrinsic antiviral defenses and is often accomplished through virus-host protein interactions. Here, we investigate the recently discovered interaction between DENV non-structural protein 5 (NS5) and the transcriptional regulator Polymerase associated factor 1 complex (PAF1C). Our work demonstrates PAF1C member PAF1 acts as an antiviral factor and inhibits DENV replication. In parallel, we identified immune response genes involved in intrinsic antiviral defense that depend on PAF1 for expression. We further identified the regions of NS5 required for the protein interaction with PAF1C. Breaking the NS5-PAF1C protein interaction restores the expression of PAF1-dependent immune response genes. Together, our work establishes the antiviral role of PAF1C in DENV infection and NS5 antagonism of PAF1-dependent gene expression through a virus-host protein interaction.
Collapse
Affiliation(s)
- Marine J. Petit
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, United States of America
- Department of Chemical Engineering, University of California, Davis, California, United States of America
| | - Matthew W. Kenaston
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, United States of America
| | - Oanh H. Pham
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, United States of America
| | - Ariana A. Nagainis
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, United States of America
- Department of Chemical Engineering, University of California, Davis, California, United States of America
| | - Adam T. Fishburn
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, United States of America
| | - Priya S. Shah
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, United States of America
- Department of Chemical Engineering, University of California, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
5
|
Tagging and Capturing of Lentiviral Vectors Using Short RNAs. Int J Mol Sci 2021; 22:ijms221910263. [PMID: 34638603 PMCID: PMC8508951 DOI: 10.3390/ijms221910263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/19/2021] [Indexed: 11/22/2022] Open
Abstract
Lentiviral (LV) vectors have emerged as powerful tools for transgene delivery ex vivo but in vivo gene therapy applications involving LV vectors have faced a number of challenges, including the low efficiency of transgene delivery, a lack of tissue specificity, immunogenicity to both the product encoded by the transgene and the vector, and the inactivation of the vector by the human complement cascade. To mitigate these issues, several engineering approaches, involving the covalent modification of vector particles or the incorporation of specific protein domains into the vector’s envelope, have been tested. Short synthetic oligonucleotides, including aptamers bound to the surface of LV vectors, may provide a novel means with which to retarget LV vectors to specific cells and to shield these vectors from neutralization by sera. The purpose of this study was to develop strategies to tether nucleic acid sequences, including short RNA sequences, to LV vector particles in a specific and tight fashion. To bind short RNA sequences to LV vector particles, a bacteriophage lambda N protein-derived RNA binding domain (λN), fused to the measles virus hemagglutinin protein, was used. The λN protein bound RNA sequences bearing a boxB RNA hairpin. To test this approach, we used an RNA aptamer specific to the human epidermal growth factor receptor (EGFR), which was bound to LV vector particles via an RNA scaffold containing a boxB RNA motif. The results obtained confirmed that the EGFR-specific RNA aptamer bound to cells expressing EGFR and that the boxB containing the RNA scaffold was bound specifically to the λN RNA binding domain attached to the vector. These results show that LV vectors can be equipped with nucleic acid sequences to develop improved LV vectors for in vivo applications.
Collapse
|
6
|
Shahryari A, Burtscher I, Nazari Z, Lickert H. Engineering Gene Therapy: Advances and Barriers. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alireza Shahryari
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- School of Medicine Department of Human Genetics Technical University of Munich Klinikum Rechts der Isar 81675 München Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
- Stem Cell Research Center Golestan University of Medical Sciences Gorgan 49341‐74515 Iran
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
| | - Zahra Nazari
- Department of Biology School of Basic Sciences Golestan University Gorgan 49361‐79142 Iran
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- School of Medicine Department of Human Genetics Technical University of Munich Klinikum Rechts der Isar 81675 München Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
| |
Collapse
|
7
|
Minami SA, Shah PS. Transient light-activated gene expression in Chinese hamster ovary cells. BMC Biotechnol 2021; 21:13. [PMID: 33541329 PMCID: PMC7863527 DOI: 10.1186/s12896-021-00670-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/13/2021] [Indexed: 12/05/2022] Open
Abstract
Background Chinese hamster ovary (CHO) cells are widely used for industrial production of biopharmaceuticals. Many genetic, chemical, and environmental approaches have been developed to modulate cellular pathways to improve titers. However, these methods are often irreversible or have off-target effects. Development of techniques which are precise, tunable, and reversible will facilitate temporal regulation of target pathways to maximize titers. In this study, we investigate the use of optogenetics in CHO cells. The light-activated CRISPR-dCas9 effector (LACE) system was first transiently transfected to express eGFP in a light-inducible manner. Then, a stable system was tested using lentiviral transduction. Results Transient transfections resulted in increasing eGFP expression as a function of LED intensity, and activation for 48 h yielded up to 4-fold increased eGFP expression compared to cells kept in the dark. Fluorescence decreased once the LACE system was deactivated, and a protein half-life of 14.9 h was calculated, which is in agreement with values reported in the literature. In cells stably expressing the LACE system, eGFP expression was confirmed, but there was no significant increase in expression following light activation. Conclusions Taken together, these results suggest that optogenetics can regulate CHO cell cultures, but development of stable cell lines requires optimized expression levels of the LACE components to maintain high dynamic range. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-021-00670-1.
Collapse
Affiliation(s)
- Shiaki A Minami
- Department of Chemical Engineering, University of California, Davis, USA
| | - Priya S Shah
- Department of Chemical Engineering, University of California, Davis, USA. .,Department of Microbiology and Molecular Genetics, University of California, Davis, USA.
| |
Collapse
|
8
|
Labisch JJ, Bollmann F, Wolff MW, Pflanz K. A new simplified clarification approach for lentiviral vectors using diatomaceous earth improves throughput and safe handling. J Biotechnol 2020; 326:11-20. [PMID: 33301854 DOI: 10.1016/j.jbiotec.2020.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/22/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Lentiviral vectors have proven their great potential to serve as a DNA delivery tool for gene modified cell therapy and gene therapy applications. The downstream processing of these vectors is however still a great challenge, particularly because of the low stability of the virus. Harvesting and clarification are critical and until now insufficiently characterized steps for lentivirus processing. To address this bottleneck, we analyzed whether lentiviral vectors produced by transient transfection of HEK293 T/17 SF suspension cells can be efficiently clarified with a lab-scale method with the filter aid diatomaceous earth (DE) and bioburden reducing membrane filters achieving high lentivirus recoveries. Using a design of experiment approach we found that higher DE concentrations are advantageous for a higher turbidity reduction and shorter filtration times, but at the same time LV titer decreases with increasing DE concentration. A DE concentration of 9 g/L was identified with a DoE as a robust set-point. Clarification with DE was compared with for lab-scale traditionally employed centrifugation and subsequent bioburden reduction filtration of viral vectors. The use of DE allows to perform a harvest and clarification process, which does not only facilitate faster and safer virus handling, but enables a lower material consumption due to the extremely increased filter capacity, thus representing an efficient and robust lab-scale clarification process.
Collapse
Affiliation(s)
- Jennifer J Labisch
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstr. 5, 30167, Hannover, Germany; Research & Development, Sartorius Stedim Biotech GmbH, August-Spindler-Str. 11, 37079, Goettingen, Germany.
| | - Franziska Bollmann
- Research & Development, Sartorius Stedim Biotech GmbH, August-Spindler-Str. 11, 37079, Goettingen, Germany
| | - Michael W Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen (THM), Wiesenstr. 14, 35390, Giessen, Germany
| | - Karl Pflanz
- Research & Development, Sartorius Stedim Biotech GmbH, August-Spindler-Str. 11, 37079, Goettingen, Germany
| |
Collapse
|
9
|
Experimental Evolution Generates Novel Oncolytic Vesicular Stomatitis Viruses with Improved Replication in Virus-Resistant Pancreatic Cancer Cells. J Virol 2020; 94:JVI.01643-19. [PMID: 31694943 PMCID: PMC7000975 DOI: 10.1128/jvi.01643-19] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/03/2019] [Indexed: 12/13/2022] Open
Abstract
Vesicular stomatitis virus (VSV)-based oncolytic viruses are promising agents against pancreatic ductal adenocarcinoma (PDAC). However, some PDAC cell lines are resistant to VSV. Here, using a directed viral evolution approach, we generated novel oncolytic VSVs with an improved ability to replicate in virus-resistant PDAC cell lines, while remaining highly attenuated in nonmalignant cells. Two independently evolved VSVs obtained 2 identical VSV glycoprotein mutations, K174E and E238K. Additional experiments indicated that these acquired G mutations improved VSV replication, at least in part due to improved virus attachment to SUIT-2 cells. Importantly, no deletions or mutations were found in the virus-carried transgenes in any of the passaged viruses. Our findings demonstrate long-term genomic stability of complex VSV recombinants carrying large transgenes and support further clinical development of oncolytic VSV recombinants as safe therapeutics for cancer. Vesicular stomatitis virus (VSV) based oncolytic viruses are promising agents against various cancers. We have shown that pancreatic ductal adenocarcinoma (PDAC) cell lines exhibit great diversity in susceptibility and permissibility to VSV. Here, using a directed evolution approach with our two previously described oncolytic VSV recombinants, VSV-p53wt and VSV-p53-CC, we generated novel oncolytic VSVs with an improved ability to replicate in virus-resistant PDAC cell lines. VSV-p53wt and VSV-p53-CC encode a VSV matrix protein (M) with a ΔM51 mutation (M-ΔM51) and one of two versions of a functional human tumor suppressor, p53, fused to a far-red fluorescent protein, eqFP650. Each virus was serially passaged 32 times (which accounts for more than 60 viral replication cycles) on either the SUIT-2 (moderately resistant to VSV) or MIA PaCa-2 (highly permissive to VSV) human PDAC cell lines. While no phenotypic changes were observed for MIA PaCa-2-passaged viruses, both SUIT-2-passaged VSV-p53wt and VSV-p53-CC showed improved replication in SUIT-2 and AsPC-1, another human PDAC cell line also moderately resistant to VSV, while remaining highly attenuated in nonmalignant cells. Surprisingly, two identical VSV glycoprotein (VSV-G) mutations, K174E and E238K, were identified in both SUIT-2-passaged viruses. Additional experiments indicated that the acquired G mutations improved VSV replication, at least in part due to improved virus attachment to SUIT-2 cells. Importantly, no mutations were found in the M-ΔM51 protein, and no deletions or mutations were found in the p53 or eqFP650 portions of virus-carried transgenes in any of the passaged viruses, demonstrating long-term genomic stability of complex VSV recombinants carrying large transgenes. IMPORTANCE Vesicular stomatitis virus (VSV)-based oncolytic viruses are promising agents against pancreatic ductal adenocarcinoma (PDAC). However, some PDAC cell lines are resistant to VSV. Here, using a directed viral evolution approach, we generated novel oncolytic VSVs with an improved ability to replicate in virus-resistant PDAC cell lines, while remaining highly attenuated in nonmalignant cells. Two independently evolved VSVs obtained 2 identical VSV glycoprotein mutations, K174E and E238K. Additional experiments indicated that these acquired G mutations improved VSV replication, at least in part due to improved virus attachment to SUIT-2 cells. Importantly, no deletions or mutations were found in the virus-carried transgenes in any of the passaged viruses. Our findings demonstrate long-term genomic stability of complex VSV recombinants carrying large transgenes and support further clinical development of oncolytic VSV recombinants as safe therapeutics for cancer.
Collapse
|
10
|
Khurana L, ElGindi M, Tilstam PV, Pantouris G. Elucidating the role of an immunomodulatory protein in cancer: From protein expression to functional characterization. Methods Enzymol 2019; 629:307-360. [PMID: 31727247 DOI: 10.1016/bs.mie.2019.05.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Several fundamental discoveries made over the last two decades, in the field of cancer biology, have increased our understanding of the complex tumor micro- and macroenvironments. This has shifted the current empirical cancer therapies to more rationalized treatments targeting immunomodulatory proteins. From the point of identification, a protein target undergoes several interrogations, which are necessary to truly define its druggability. Here, we outline some basic steps that can be followed for in vitro characterization of a potential immunomodulatory protein target. We describe procedures for recombinant protein expression and purification including key annotations on protein cloning, expression systems, purification strategies and protein characterization using structural and biochemical approaches. For functional characterization, we provide detailed protocols for using flow-cytometric techniques in cell lines or primary cells to study protein expression profiles, proliferation, apoptosis and cell-cycle changes. This multilevel approach can provide valuable, in-depth understanding of any protein target with potential immunomodulatory effects.
Collapse
Affiliation(s)
- Leepakshi Khurana
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT, United States
| | - Mei ElGindi
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Pathricia V Tilstam
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Georgios Pantouris
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT, United States; Department of Chemistry, University of the Pacific, Stockton, CA, United States.
| |
Collapse
|
11
|
Olgun HB, Tasyurek HM, Sanlioglu AD, Sanlioglu S. High-Grade Purification of Third-Generation HIV-Based Lentiviral Vectors by Anion Exchange Chromatography for Experimental Gene and Stem Cell Therapy Applications. Methods Mol Biol 2019; 1879:347-365. [PMID: 30006865 DOI: 10.1007/7651_2018_154] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lentiviral vectors (LVs) have been increasingly used in clinical gene therapy applications particularly due to their efficient gene transfer ability, lack of interference from preexisting viral immunity, and long-term gene expression they provide. Purity of LVs is essential in in vivo applications, for a high therapeutic benefit with minimum toxicity. Accordingly, laboratory scale production of LVs frequently involves transient cotransfection of 293T cells with packaging and transfer plasmids in the presence of CaPO4. After clearance of the cellular debris by low-speed centrifugation and filtration, lentivectors are usually concentrated by high-speed ultracentrifugation in sucrose cushion. Concentrated viral samples are then purified by anion exchange chromatography (AEX) after benzonase treatment to remove the residual cellular DNA. Here, we describe an improved practical method for LV purification using AEX, useful for experimental studies concerning gene and stem cell therapy.
Collapse
Affiliation(s)
- Hazal Banu Olgun
- Human Gene and Cell Therapy Center of Akdeniz University Hospitals and Clinics, Antalya, Turkey
| | - Hale M Tasyurek
- Human Gene and Cell Therapy Center of Akdeniz University Hospitals and Clinics, Antalya, Turkey
| | - Ahter D Sanlioglu
- Human Gene and Cell Therapy Center of Akdeniz University Hospitals and Clinics, Antalya, Turkey
| | - Salih Sanlioglu
- Human Gene and Cell Therapy Center of Akdeniz University Hospitals and Clinics, Antalya, Turkey.
| |
Collapse
|
12
|
Mekkaoui L, Parekh F, Kotsopoulou E, Darling D, Dickson G, Cheung GW, Chan L, MacLellan-Gibson K, Mattiuzzo G, Farzaneh F, Takeuchi Y, Pule M. Lentiviral Vector Purification Using Genetically Encoded Biotin Mimic in Packaging Cell. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:155-165. [PMID: 30547049 PMCID: PMC6258877 DOI: 10.1016/j.omtm.2018.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 10/17/2018] [Indexed: 11/17/2022]
Abstract
Lentiviral vectors (LVs) have recently witnessed an increasing demand in research and clinical applications. Their current purification processes represent the main bottleneck in their widespread use, as the methods used are cumbersome and yield low recoveries. We aimed to develop a one-step method to specifically purify LVs, with high yields and reduced levels of impurities, using the biotin-streptavidin system. Herein, packaging HEK293T cells were genetically engineered with a cyclical biotin-mimicking peptide displayed on a CD8α stalk, termed cTag8. LVs were modified with cTag8 by its passive incorporation onto viral surfaces during budding, without viral protein engineering or hindrance on infectivity. Expression of cTag8 on LVs allowed complete capture of infectious particles by streptavidin magnetic beads. As cTag8 binds streptavidin in the nanomolar range, the addition of micromolar concentrations of biotin resulted in the release of captured LVs by competitive elution, with overall yields of ≥60%. Analysis of eluted LVs revealed high purity with a >3-log and 2-log reduction in DNA contamination and host cell proteins, respectively. This one-step purification was also tested for scalable vector processing using monolith affinity chromatography, with an encouraging preliminary overall yield of 20%. This method will be of valuable use for both research and clinical applications of LVs.
Collapse
Affiliation(s)
- Leila Mekkaoui
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Farhaan Parekh
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | | | - David Darling
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Glenda Dickson
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Gordon W. Cheung
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Lucas Chan
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Kirsty MacLellan-Gibson
- National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK
| | - Giada Mattiuzzo
- National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK
| | - Farzin Farzaneh
- School of Cancer & Pharmaceutical Sciences, King’s College London, Molecular Medicine Group, The Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK
| | - Yasuhiro. Takeuchi
- National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QC, UK
- Division of Infection and Immunity, University College London, Rayne Building, 5 University Street, London WC1E 6JF, UK
| | - Martin Pule
- UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
- Corresponding author: Martin Pule, UCL Cancer Institute, University College London, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6BT, UK.
| |
Collapse
|
13
|
Sinn PL, Hwang BY, Li N, Ortiz JLS, Shirazi E, Parekh KR, Cooney AL, Schaffer DV, McCray PB. Novel GP64 envelope variants for improved delivery to human airway epithelial cells. Gene Ther 2017; 24:674-679. [PMID: 28880020 PMCID: PMC5759328 DOI: 10.1038/gt.2017.78] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023]
Abstract
Lentiviral vectors pseudotyped with the baculovirus envelope protein GP64 transduce primary cultures of human airway epithelia (HAE) at their apical surface. Our goal in this study was to harness a directed evolution approach to develop a novel envelope glycoprotein with increased transduction properties for HAE. Using error-prone PCR, a library of GP64 mutants was generated and used to prepare a diverse pool of lentiviral virions pseudotyped with GP64 variants. The library was serially passaged on HAE and three GP64 mutations were recovered. Single-, double- and the triple-combination mutant envelope glycoproteins were compared with wild-type GP64 for their ability to transduce HAE. Our results suggest that lentiviral vectors pseudotyped with evolved GP64 transduced HAE with greater efficiency than wild-type GP64. This effect was not observed in primary cultures of porcine airway epithelial cells, suggesting that the directed evolution protocol was species specific. In summary, our studies indicate that serial passage of a GP64 mutant library yielded specific variants with improved HAE cell tropism, yielding tools with the potential to improve the success of gene therapy for airway diseases.
Collapse
Affiliation(s)
- PL Sinn
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, University of Iowa, Iowa City, IA, USA
| | - B-Y Hwang
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - N Li
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, University of Iowa, Iowa City, IA, USA
| | - JLS Ortiz
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - E Shirazi
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - KR Parekh
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA and
| | - AL Cooney
- Pappajohn Biomedical Institute and the Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, University of Iowa, Iowa City, IA, USA
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - DV Schaffer
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - PB McCray
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Pappajohn Biomedical Institute and the Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, University of Iowa, Iowa City, IA, USA
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
14
|
Chu Y, Oum YH, Carrico IS. Surface modification via strain-promoted click reaction facilitates targeted lentiviral transduction. Virology 2015; 487:95-103. [PMID: 26499046 DOI: 10.1016/j.virol.2015.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 11/29/2022]
Abstract
As a result of their ability to integrate into the genome of both dividing and non-dividing cells, lentiviruses have emerged as a promising vector for gene delivery. Targeted gene transduction of specific cells and tissues by lentiviral vectors has been a major goal, which has proven difficult to achieve. We report a novel targeting protocol that relies on the chemoselective attachment of cancer specific ligands to unnatural glycans on lentiviral surfaces. This strategy exhibits minimal perturbation on virus physiology and demonstrates remarkable flexibility. It allows for targeting but can be more broadly useful with applications such as vector purification and immunomodulation.
Collapse
Affiliation(s)
- Yanjie Chu
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| | - Yoon Hyeun Oum
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| | - Isaac S Carrico
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA; Institute of Chemical Biology and Drug Discovery, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA.
| |
Collapse
|
15
|
Engineering foot-and-mouth disease virus serotype O IND R2/1975 for one-step purification by immobilized metal affinity chromatography. Biologicals 2015; 43:390-8. [DOI: 10.1016/j.biologicals.2015.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/11/2015] [Accepted: 06/01/2015] [Indexed: 11/22/2022] Open
|
16
|
Bugaj LJ, Spelke DP, Mesuda CK, Varedi M, Kane RS, Schaffer DV. Regulation of endogenous transmembrane receptors through optogenetic Cry2 clustering. Nat Commun 2015; 6:6898. [PMID: 25902152 PMCID: PMC4408875 DOI: 10.1038/ncomms7898] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 03/11/2015] [Indexed: 12/27/2022] Open
Abstract
Transmembrane receptors are the predominant conduit through which cells sense and transduce extracellular information into intracellular biochemical signals. Current methods to control and study receptor function, however, suffer from poor resolution in space and time and often employ receptor overexpression, which can introduce experimental artifacts. We report a genetically-encoded approach, termed Clustering Indirectly using Cryptochrome 2 (CLICR), for spatiotemporal control over endogenous transmembrane receptor activation, enabled through the optical regulation of target receptor clustering and downstream signaling using non-covalent interactions with engineered Arabidopsis Cryptochrome 2 (Cry2). CLICR offers a modular platform to enable photocontrol of the clustering of diverse transmembrane receptors including FGFR, PDGFR, and integrins in multiple cell types including neural stem cells. Furthermore, light-inducible manipulation of endogenous receptor tyrosine kinase (RTK) activity can modulate cell polarity and establish phototaxis in fibroblasts. The resulting spatiotemporal control over cellular signaling represents a powerful new optogenetic framework for investigating and controlling cell function and fate.
Collapse
Affiliation(s)
- L J Bugaj
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.,The UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA
| | - D P Spelke
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.,The UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA
| | - C K Mesuda
- Department of Chemical Engineering, University of California, Berkeley, Berkeley, California 94720, USA
| | - M Varedi
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California 94720, USA
| | - R S Kane
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | - D V Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.,The UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.,Department of Chemical Engineering, University of California, Berkeley, Berkeley, California 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California 94720, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720, USA
| |
Collapse
|
17
|
Fritz AL, Adil MM, Mao SR, Schaffer DV. cAMP and EPAC Signaling Functionally Replace OCT4 During Induced Pluripotent Stem Cell Reprogramming. Mol Ther 2015; 23:952-963. [PMID: 25666918 DOI: 10.1038/mt.2015.28] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/03/2015] [Indexed: 12/17/2022] Open
Abstract
The advent of induced pluripotent stem cells--generated via the ectopic overexpression of reprogramming factors such as OCT4, SOX2, KLF4, and C-MYC (OSKM) in a differentiated cell type--has enabled groundbreaking research efforts in regenerative medicine, disease modeling, and drug discovery. Although initial studies have focused on the roles of nuclear factors, increasing evidence highlights the importance of signal transduction during reprogramming. By utilizing a quantitative, medium-throughput screen to initially identify signaling pathways that could potentially replace individual transcription factors during reprogramming, we initially found that several pathways--such as Notch, Smoothened, and cyclic AMP (cAMP) signaling--were capable of generating alkaline phosphatase positive colonies in the absence of OCT4, the most stringently required Yamanaka factor. After further investigation, we discovered that cAMP signal activation could functionally replace OCT4 to induce pluripotency, and results indicate that the downstream exchange protein directly activated by cAMP (EPAC) signaling pathway rather than protein kinase A (PKA) signaling is necessary and sufficient for this function. cAMP signaling may reduce barriers to reprogramming by contributing to downstream epithelial gene expression, decreasing mesenchymal gene expression, and increasing proliferation. Ultimately, these results elucidate mechanisms that could lead to new reprogramming methodologies and advance our understanding of stem cell biology.
Collapse
Affiliation(s)
- Ashley L Fritz
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - Maroof M Adil
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - Sunnie R Mao
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA; Department of Bioengineering, University of California, Berkeley, California, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA.
| |
Collapse
|
18
|
Enhanced cellular secretion of AAV2 by expression of foreign viral envelope proteins. Biochem Eng J 2015. [DOI: 10.1016/j.bej.2014.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Fritz AL, Mao SR, West MG, Schaffer DV. A medium-throughput analysis of signaling pathways involved in early stages of stem cell reprogramming. Biotechnol Bioeng 2014; 112:209-19. [PMID: 25065366 DOI: 10.1002/bit.25336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/28/2014] [Accepted: 07/07/2014] [Indexed: 02/06/2023]
Abstract
The induction of pluripotency from adult cells has enormous potential in regenerative medicine. While initial efforts to study mechanisms and improve efficiency of induced pluripotent stem cell (iPSC) reprogramming focused on the direct roles of transcriptional regulators, increasing evidence indicates that cellular signal transduction pathways can modulate this process. Here, we present a medium-throughput system to study the effect of signaling pathways on the early stages of reprogramming. We generated a set of lentiviral vectors encoding 38 genes that upregulate or downregulate major signal transduction pathways and quantified each signaling factor's effect on reprogramming. This approach confirmed the role of several factors previously implicated in reprogramming, as well as identified several GTPases-factors that to date have not been largely studied in reprogramming-that improve or hinder iPSC reprogramming. In addition, this methodology is useful in determining new targets for enhancing pluripotency reprogramming, lineage reprogramming, and/or cell differentiation.
Collapse
Affiliation(s)
- Ashley L Fritz
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, 94720
| | | | | | | |
Collapse
|
20
|
Hughes AJ, Spelke DP, Xu Z, Kang CC, Schaffer DV, Herr AE. Single-cell western blotting. Nat Methods 2014; 11:749-55. [PMID: 24880876 PMCID: PMC4077215 DOI: 10.1038/nmeth.2992] [Citation(s) in RCA: 314] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 04/28/2014] [Indexed: 02/06/2023]
Abstract
To measure cell-to-cell variation in protein-mediated functions, we developed an approach to conduct ∼10(3) concurrent single-cell western blots (scWesterns) in ∼4 h. A microscope slide supporting a 30-μm-thick photoactive polyacrylamide gel enables western blotting: settling of single cells into microwells, lysis in situ, gel electrophoresis, photoinitiated blotting to immobilize proteins and antibody probing. We applied this scWestern method to monitor single-cell differentiation of rat neural stem cells and responses to mitogen stimulation. The scWestern quantified target proteins even with off-target antibody binding, multiplexed to 11 protein targets per single cell with detection thresholds of <30,000 molecules, and supported analyses of low starting cell numbers (∼200) when integrated with FACS. The scWestern overcomes limitations of antibody fidelity and sensitivity in other single-cell protein analysis methods and constitutes a versatile tool for the study of complex cell populations at single-cell resolution.
Collapse
Affiliation(s)
- Alex J Hughes
- 1] Department of Bioengineering, University of California (UC) Berkeley, Berkeley, California, USA. [2] California Institute for Quantitative Biosciences, UC Berkeley, Berkeley, California, USA. [3] [4]
| | - Dawn P Spelke
- 1] Department of Bioengineering, University of California (UC) Berkeley, Berkeley, California, USA. [2] California Institute for Quantitative Biosciences, UC Berkeley, Berkeley, California, USA. [3] The UC Berkeley-UC San Francisco Graduate Program in Bioengineering, UC Berkeley, Berkeley, California, USA. [4]
| | - Zhuchen Xu
- 1] Department of Bioengineering, University of California (UC) Berkeley, Berkeley, California, USA. [2] California Institute for Quantitative Biosciences, UC Berkeley, Berkeley, California, USA
| | - Chi-Chih Kang
- 1] Department of Bioengineering, University of California (UC) Berkeley, Berkeley, California, USA. [2] California Institute for Quantitative Biosciences, UC Berkeley, Berkeley, California, USA
| | - David V Schaffer
- 1] Department of Bioengineering, University of California (UC) Berkeley, Berkeley, California, USA. [2] California Institute for Quantitative Biosciences, UC Berkeley, Berkeley, California, USA. [3] The UC Berkeley-UC San Francisco Graduate Program in Bioengineering, UC Berkeley, Berkeley, California, USA. [4] Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, California, USA. [5] Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, USA
| | - Amy E Herr
- 1] Department of Bioengineering, University of California (UC) Berkeley, Berkeley, California, USA. [2] California Institute for Quantitative Biosciences, UC Berkeley, Berkeley, California, USA. [3] The UC Berkeley-UC San Francisco Graduate Program in Bioengineering, UC Berkeley, Berkeley, California, USA
| |
Collapse
|
21
|
Systematic improvement of lentivirus transduction protocols by antibody fragments fused to VSV-G as envelope glycoprotein. Biomaterials 2014; 35:4204-12. [DOI: 10.1016/j.biomaterials.2014.01.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 01/22/2014] [Indexed: 12/22/2022]
|
22
|
Characteristics of oncolytic vesicular stomatitis virus displaying tumor-targeting ligands. J Virol 2013; 87:13543-55. [PMID: 24089573 DOI: 10.1128/jvi.02240-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We sought proof of principle that tumor-targeting ligands can be displayed on the surface of vesicular stomatitis virus (VSV) by engineering its glycoprotein. Here, we successfully rescued VSVs displaying tumor vasculature-targeting ligands. By using a rational approach, we investigated various feasible insertion sites on the G protein of VSV (VSV-G) for display of tumor vasculature-targeting ligands, cyclic RGD (cRGD) and echistatin. We found seven sites on VSV-G that tolerated insertion of the 9-residue cRGD peptide, two of which could tolerate insertion of the 49-amino acid echistatin domain. All of the ligand-displaying viruses replicated as well as the parental virus. In vitro studies demonstrated that the VSV-echistatin viruses specifically bound to targeted integrins. Since the low-density lipoprotein receptor (LDLR) was recently identified as a major receptor for VSV, we investigated the entry of ligand-displaying viruses after masking LDLR. The experiment showed that the modified viruses can enter the cell independently of LDLR, whereas entry of unmodified virus is significantly blocked by a specific monoclonal antibody against LDLR. Both parental and ligand-displaying viruses displayed equal oncolytic efficacies in a syngeneic mouse myeloma model. We further demonstrated that single-chain antibody fragments against tumor-specific antigens can be inserted at the N terminus of the G protein and that corresponding replication-competent VSVs can be rescued efficiently. Overall, we demonstrated that functional tumor-targeting ligands can be displayed on replication-competent VSVs without perturbing viral growth and oncolytic efficacy. This study provides a rational foundation for the future development of fully retargeted oncolytic VSVs.
Collapse
|
23
|
Segura MM, Mangion M, Gaillet B, Garnier A. New developments in lentiviral vector design, production and purification. Expert Opin Biol Ther 2013; 13:987-1011. [PMID: 23590247 DOI: 10.1517/14712598.2013.779249] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Lentiviruses are a very potent class of viral vectors for which there is presently a rapidly growing interest for a number of gene therapy. However, their construction, production and purification need to be performed according to state-of-the-art techniques in order to obtain sufficient quantities of high purity material of any usefulness and safety. AREAS COVERED The recent advances in the field of recombinant lentivirus vector design, production and purification will be reviewed with an eye toward its utilization for gene therapy. Such a review should be helpful for the potential user of this technology. EXPERT OPINION The principal hurdles toward the use of recombinant lentivirus as a gene therapy vector are the low titer at which it is produced as well as the difficulty to purify it at an acceptable level without degrading it. The recent advances in the bioproduction of this vector suggest these issues are about to be resolved, making the retrovirus gene therapy a mature technology.
Collapse
Affiliation(s)
- Maria Mercedes Segura
- Chemical Engineering Department, Universitat Autònoma de Barcelona, Campus Bellaterra, Cerdanyola del Vallès (08193), Barcelona, Spain
| | | | | | | |
Collapse
|
24
|
Engineering a serum-resistant and thermostable vesicular stomatitis virus G glycoprotein for pseudotyping retroviral and lentiviral vectors. Gene Ther 2013; 20:807-15. [PMID: 23364315 PMCID: PMC3735647 DOI: 10.1038/gt.2013.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 11/13/2012] [Accepted: 12/17/2012] [Indexed: 01/19/2023]
Abstract
Vesicular stomatitis virus G glycoprotein (VSV-G) is the most widely used envelope protein for retroviral and lentiviral vector pseudotyping; however, serum inactivation of VSV-G pseudotyped vectors is a significant challenge for in vivo gene delivery. To address this problem, we conducted directed evolution of VSV-G to increase its resistance to human serum neutralization. After six selection cycles, numerous common mutations were present. Based on their location within VSV-G, we analyzed whether substitutions in several surface exposed residues could endow viral vectors with higher resistance to serum. S162T, T230N, and T368A mutations enhanced serum resistance, and additionally K66T, T368A, and E380K substitutions increased the thermostability of VSV-G pseudotyped retroviral vectors, an advantageous byproduct of the selection strategy. Analysis of a number of combined mutants revealed that VSV-G harboring T230N + T368A or K66T + S162T + T230N + T368A mutations exhibited both higher in vitro resistance to human serum and higher thermostability, as well as enhanced resistance to rabbit and mouse serum. Finally, lentiviral vectors pseudotyped with these variants were more resistant to human serum in a murine model. These serum-resistant and thermostable VSV-G variants may aid the application of retroviral and lentiviral vectors to gene therapy.
Collapse
|
25
|
Astrocytes regulate adult hippocampal neurogenesis through ephrin-B signaling. Nat Neurosci 2012; 15:1399-406. [PMID: 22983209 PMCID: PMC3458152 DOI: 10.1038/nn.3212] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/09/2012] [Indexed: 12/18/2022]
Abstract
Neurogenesis in the adult hippocampus involves activation of quiescent neural stem cells (NSCs) to yield transiently amplifying NSCs and progenitors, and ultimately neurons that affect learning and memory. This process is tightly controlled by microenvironmental cues, though few endogenous factors are known to regulate neuronal differentiation. While astrocytes have been implicated, their role in juxtacrine (i.e. cell-cell contact-dependent) signaling within NSC niches has not been investigated. We show that ephrin-B2 presented from rodent hippocampal astrocytes regulates neurogenesis in vivo. Furthermore, clonal analysis in NSC fate-mapping studies reveals a novel role for ephrin-B2 in instructing neuronal differentiation. Additionally, ephrin-B2 signaling, transduced by EphB4 receptors on NSCs, activates β-catenin in vitro and in vivo independent of Wnt signaling and upregulates proneural transcription factors. Ephrin-B2+ astrocytes thus promote neuronal differentiation of adult NSCs through juxtacrine signaling, findings that advance our understanding of adult neurogenesis and may have future regenerative medicine implications.
Collapse
|
26
|
Abstract
Gene therapy vectors based on murine retroviruses have now been in clinical trials for over 20 years. During that time, a variety of novel vector pseudotypes were developed in an effort to improve gene transfer. Lentiviral vectors are now in clinical trials and a similar evolution of vector technology is anticipated. These modifications present challenges for those producing large-scale clinical materials. This chapter discusses approaches to process development for novel lentiviral vectors, highlight considerations, and methods to be incorporated into the development schema.
Collapse
Affiliation(s)
- Anna Leath
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | |
Collapse
|
27
|
Asuri P, Bartel MA, Vazin T, Jang JH, Wong TB, Schaffer DV. Directed evolution of adeno-associated virus for enhanced gene delivery and gene targeting in human pluripotent stem cells. Mol Ther 2012; 20:329-38. [PMID: 22108859 PMCID: PMC3277219 DOI: 10.1038/mt.2011.255] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 10/27/2011] [Indexed: 12/12/2022] Open
Abstract
Efficient approaches for the precise genetic engineering of human pluripotent stem cells (hPSCs) can enhance both basic and applied stem cell research. Adeno- associated virus (AAV) vectors are of particular interest for their capacity to mediate efficient gene delivery to and gene targeting in various cells. However, natural AAV serotypes offer only modest transduction of human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), which limits their utility for efficiently manipulating the hPSC genome. Directed evolution is a powerful means to generate viral vectors with novel capabilities, and we have applied this approach to create a novel AAV variant with high gene delivery efficiencies (~50%) to hPSCs, which are importantly accompanied by a considerable increase in gene-targeting frequencies, up to 0.12%. While this level is likely sufficient for numerous applications, we also show that the gene-targeting efficiency mediated by an evolved AAV variant can be further enhanced (>1%) in the presence of targeted double- stranded breaks (DSBs) generated by the co-delivery of artificial zinc finger nucleases (ZFNs). Thus, this study demonstrates that under appropriate selective pressures, AAV vectors can be created to mediate efficient gene targeting in hPSCs, alone or in the presence of ZFN- mediated double-stranded DNA breaks.
Collapse
Affiliation(s)
- Prashanth Asuri
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720-1462, USA
| | | | | | | | | | | |
Collapse
|
28
|
Padmashali RM, Andreadis ST. Engineering fibrinogen-binding VSV-G envelope for spatially- and cell-controlled lentivirus delivery through fibrin hydrogels. Biomaterials 2011; 32:3330-9. [PMID: 21296411 DOI: 10.1016/j.biomaterials.2011.01.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 01/13/2011] [Indexed: 01/20/2023]
Abstract
We recently demonstrated that fibrin hydrogels can be used as vehicles for efficient lentivirus gene delivery. Gene transfer in fibrin gels was strongly dependent on matrix degradation by target cells but a fraction of lentiviral particles diffused out of the gels over time compromising spatial control of gene transfer. To overcome this challenge, we engineered lentiviral particles that bind covalently to fibrin during polymerization. To this end, we fused into the viral envelope glycoprotein (VSV-G) peptide domains that are recognized by factor XIII and protease cleavage sites that are recognized by plasmin. Lentivirus pseudotyped with the modified envelopes bound to fibrinogen in a factor XIII dose dependent manner and was released upon plasmin treatment. The peptide/VSV-G fusion envelope variants did not compromise the transduction efficiency of the resulting virus except when lacking any flexible linkers separating the peptide from the VSV-G envelope. Diffusion of virus from the gels decreased dramatically, especially at high concentrations of FXIII, even for fibrin gels with low fibrinogen concentration that were loaded with high titer virus. Lentivirus arrays prepared with fibrin-conjugated lentivirus yielded highly efficient gene transfer that was confined to virus-containing fibrin spots. As a result, signal/noise ratio increased and cross-contamination between neighboring sites was minimal. Finally, in addition to lentivirus microarrays this strategy may be used to achieve spatially-controlled gene transfer for therapeutic applications.
Collapse
Affiliation(s)
- Roshan M Padmashali
- Bioengineering Laboratory, 908 Furnas Hall, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260-4200, USA
| | | |
Collapse
|
29
|
An evolved adeno-associated viral variant enhances gene delivery and gene targeting in neural stem cells. Mol Ther 2011; 19:667-75. [PMID: 21224831 DOI: 10.1038/mt.2010.287] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gene delivery to, and gene targeting in, stem cells would be a highly enabling technology for basic science and biomedical application. Adeno-associated viral (AAV) vectors have demonstrated the capacity for efficient delivery to numerous cells, but their application to stem cells has been limited by low transduction efficiency. Due to their considerable advantages, however, engineering AAV delivery systems to enhance gene delivery to stem cells may have an impact in stem cell biology and therapy. Therefore, using several diverse AAV capsid libraries-including randomly mutagenized, DNA shuffled, and random peptide insertion variants-we applied directed evolution to create a "designer" AAV vector with enhanced delivery efficiency for neural stem cells (NSCs). A novel AAV variant, carrying an insertion of a selected peptide sequence on the surface of the threefold spike within the heparin-binding site, emerged from this evolution. Importantly, this evolved AAV variant mediated efficient gene delivery to rat, mouse, and human NSCs, as well as efficient gene targeting within adult NSCs, and it is thus promising for applications ranging from basic stem cell biology to clinical translation.
Collapse
|
30
|
Segura MM, Kamen AA, Garnier A. Overview of current scalable methods for purification of viral vectors. Methods Mol Biol 2011; 737:89-116. [PMID: 21590394 DOI: 10.1007/978-1-61779-095-9_4] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As a result of the growing interest in the use of viruses for gene therapy and vaccines, many virus-based products are being developed. The manufacturing of viruses poses new challenges for process developers and regulating authorities that need to be addressed to ensure quality, efficacy, and safety of the final product. The design of suitable purification strategies will depend on a multitude of variables including the vector production system and the nature of the virus. In this chapter, we provide an overview of the most commonly used purification methods for viral gene therapy vectors. Current chromatography options available for large-scale purification of γ-retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes simplex virus, baculovirus, and poxvirus vectors are presented.
Collapse
Affiliation(s)
- María Mercedes Segura
- Department of Biochemistry and Molecular Biology, Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | | | | |
Collapse
|
31
|
Peltier J, Conway A, Keung AJ, Schaffer DV. Akt increases sox2 expression in adult hippocampal neural progenitor cells, but increased sox2 does not promote proliferation. Stem Cells Dev 2010; 20:1153-61. [PMID: 21028992 DOI: 10.1089/scd.2010.0130] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple extracellular factors have been shown to modulate adult hippocampal neural progenitor cell (NPC) proliferation and self-renewal, and we have previously shown that Akt is an important mediator of the effects of these extracellular factors on NPC proliferation and differentiation. However, very little work has investigated how and whether Akt is involved in maintaining the multipotency of these cells. Here we demonstrate that Akt promotes expression of Sox2, a core transcription factor important for the self-renewal of NPCs. Retroviral-mediated overexpression of wild-type Akt increased Sox2 protein expression, particularly under conditions that promote cell differentiation, whereas Akt inhibition decreased Sox2. Similarly, quantitative reverse transcription (RT)-PCR in differentiating cultures indicated that Akt rescued Sox2 mRNA to levels present under conditions that promote cell proliferation. Additionally, pharmacological inhibition of Akt did not affect Sox2 protein levels in cells constitutively expressing Sox2 from a retroviral vector, indicating that Akt does not affect Sox2 protein stability. Further, in contrast to Akt overexpression, Sox2 overexpression does not increase NPC viable cell number or proliferation yet does inhibit differentiation. Collectively, these results indicate that Akt promotes cell proliferation and maintenance of a multipotent state via two downstream paths.
Collapse
Affiliation(s)
- Joseph Peltier
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720-3220, USA
| | | | | | | |
Collapse
|
32
|
Peltier J, Schaffer DV. Viral packaging and transduction of adult hippocampal neural progenitors. Methods Mol Biol 2010; 621:103-16. [PMID: 20405362 DOI: 10.1007/978-1-60761-063-2_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Genetic manipulation of adult hippocampal neural progenitor cells is a useful technique for exploring gene function through gain of function and loss of function mutations or RNAi. Furthermore, the introduction of new genes can "re-program" progenitor cell behavior to force a desired lineage in signaling environments that are not normally permissive for that cell fate. Additionally, by using a systems biology approach, neural progenitors can even be taught new behaviors and responses to signaling. In this chapter, we describe protocols for retroviral and lentiviral packaging and transduction of progenitors. These techniques are important for studying the role of various genes in progenitor fate choice.
Collapse
Affiliation(s)
- Joseph Peltier
- Department of Chemical Engineering, University of California, Berkeley, CA, USA
| | | |
Collapse
|
33
|
Leonard JN, Shah PS, Burnett JC, Schaffer DV. HIV evades RNA interference directed at TAR by an indirect compensatory mechanism. Cell Host Microbe 2008; 4:484-94. [PMID: 18996348 PMCID: PMC2742160 DOI: 10.1016/j.chom.2008.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 08/11/2008] [Accepted: 09/08/2008] [Indexed: 02/06/2023]
Abstract
HIV can rapidly evolve when placed under selective pressure, including immune surveillance or the administration of antiretroviral drugs. Typically, a variant protein allows HIV to directly evade the selective pressure. Similarly, HIV has escaped suppression by RNA interference (RNAi) directed against viral RNAs by acquiring mutations at the target region that circumvent RNAi-mediated inhibition while conserving necessary viral functions. However, when we directed RNAi against the viral TAR hairpin, which plays an indispensable role in viral transcription, resistant strains were recovered, but none carried a mutation at the target site. Instead, we isolated several strains carrying promoter mutations that indirectly compensated for the RNAi by upregulating viral transcription. Combining RNAi with the application of an antiviral drug blocked replication of such mutants. Evolutionary tuning of viral transcriptional regulation may serve as a general evasion mechanism that may be targeted to improve the efficacy of antiviral therapy.
Collapse
Affiliation(s)
- Joshua N Leonard
- Department of Chemical Engineering and the Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
34
|
Kameyama Y, Kawabe Y, Ito A, Kamihira M. Antibody-dependent gene transduction using gammaretroviral and lentiviral vectors pseudotyped with chimeric vesicular stomatitis virus glycoprotein. J Virol Methods 2008; 153:49-54. [DOI: 10.1016/j.jviromet.2008.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 06/09/2008] [Accepted: 06/10/2008] [Indexed: 10/21/2022]
|
35
|
Abstract
Viruses can be engineered to efficiently deliver exogenous genes, but their natural gene delivery properties often fail to meet human therapeutic needs. Therefore, engineering viral vectors with new properties, including enhanced targeting abilities and resistance to immune responses, is a growing area of research. This review discusses protein engineering approaches to generate viral vectors with novel gene delivery capabilities. Rational design of viral vectors has yielded successful advances in vitro, and to an extent in vivo. However, there is often insufficient knowledge of viral structure-function relationships to reengineer existing functions or create new capabilities, such as virus-cell interactions, whose molecular basis is distributed throughout the primary sequence of the viral proteins. Therefore, high-throughput library and directed evolution methods offer alternative approaches to engineer viral vectors with desired properties. Parallel and integrated efforts in rational and library-based design promise to aid the translation of engineered viral vectors toward the clinic.
Collapse
Affiliation(s)
- David V. Schaffer
- The Department o f Chemical Engineering, the Department of Bioengineering, and The Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720-3220;
| | - James T. Koerber
- The Department o f Chemical Engineering, the Department of Bioengineering, and The Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720-3220;
| | - Kwang-il Lim
- The Department o f Chemical Engineering, the Department of Bioengineering, and The Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720-3220;
| |
Collapse
|
36
|
Transposon-based mutagenesis generates diverse adeno-associated viral libraries with novel gene delivery properties. Methods Mol Biol 2008; 434:161-70. [PMID: 18470644 DOI: 10.1007/978-1-60327-248-3_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The engineering of novel properties and functions into viral vectors for improved gene delivery remains a barrier to the development of efficient, customized gene delivery vehicles. Rational methods for designing improved viral vectors are often experimentally challenging and laborious, particularly when knowledge of viral structure-function relationships is limited. As an alternative, high-throughput libraries may be rapidly and efficiently selected for viral variants with a desired function. Here we describe a transposon-based insertional mutagenesis approach to generate large diverse adeno- associated viral (AAV) libraries containing a randomly located peptide. Briefly, a selectable marker is randomly inserted throughout the AAV2 cap gene and the resulting "bookmarked' AAV cap gene is cloned into an AAV packaging vector. The selectable marker is then replaced with a defined oligonucleotide, and the final AAV library is used to package a diverse pool of AAV virions, which can used for functional selection.
Collapse
|
37
|
Abstract
Retroviral vectors have been widely used for research and clinical trials in gene therapy because of their high transduction efficiency. Retroviruses interact with target cells through their surface molecules (i.e., envelope proteins) and cellular receptors, which limit the susceptibility of target cells to retroviral vectors. Murine leukemia retrovirus (MuLV) pseudotyped with vesicular stomatitis virus G glycoprotein (VSV-G) overcomes the species barrier and is more resistant to mechanical and biochemical inactivation. A cell line producing VSV-G pseudotyped MuLV vector can be established by transfecting 293T cells expressing Gag, Pol, and VSV-G (293 GPG cell line) with a retroviral vector plasmid. Transduction potency of the resulting VSV-G pseudotyped MuLV retroviral supernatant can be quantified by titration, electron microscopy (EM), and the reverse transcriptase (RT) assay. These protocols provide methods to prepare and quantify a pseudotyped retroviral vector with high transduction rates for most types of target cells.
Collapse
Affiliation(s)
- Hong Yu
- Department of Surgery, Vascular Biology Institute, University of Miami School of Medicine, Miami, FL, USA
| | | |
Collapse
|
38
|
Cockrell AS, Kafri T. Gene delivery by lentivirus vectors. Mol Biotechnol 2007; 36:184-204. [PMID: 17873406 DOI: 10.1007/s12033-007-0010-8] [Citation(s) in RCA: 225] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/28/2022]
Abstract
The capacity to efficiently transduce nondividing cells, shuttle large genetic payloads, and maintain stable long-term transgene expression are attributes that have brought lentiviral vectors to the forefront of gene delivery vehicles for research and therapeutic applications in a clinical setting. Our discussion initiates with advances in lentiviral vector development and how these sophisticated lentiviral vectors reflect improvements in safety, regarding the prevention of replication competent lentiviruses (RCLs), vector mobilization, and insertional mutagenesis. Additionally, we describe conventional molecular regulatory systems to manage gene expression levels in a spatial and temporal fashion in the context of a lentiviral vector. State of the art technology for lentiviral vector production by transient transfection and packaging cell lines are explicitly presented with current practices used for concentration, purification, titering, and determining the safety of a vector stock. We summarize lentiviral vector applications that have received a great deal of attention in recent years including the generation of transgenic animals and the stable delivery of RNA interference molecules. Concluding remarks address some of the successes in preclinical animals, and the recent transition of lentiviral vectors to human clinical trials as therapy for a variety of infectious and genetic diseases.
Collapse
Affiliation(s)
- Adam S Cockrell
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
39
|
Jang JH, Lim KI, Schaffer DV. Library selection and directed evolution approaches to engineering targeted viral vectors. Biotechnol Bioeng 2007; 98:515-24. [PMID: 17614321 DOI: 10.1002/bit.21541] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Gene therapy, to delivery of genetic material to a patient for therapeutic benefit, has significant promise for translating basic knowledge of disease mechanism into biomedical treatments. The clinical development of the field has been slowed, however, by the need for improvements in the properties and capabilities of gene delivery vehicles. Vehicles based on viruses offer the potential for efficient gene delivery, but because viruses did not evolve to serve human therapeutic needs, many of their properties require significant improvement, including their safety, efficiency, and capacity for targeted gene delivery. Since viruses are highly complex biological entities, engineering such properties at the molecular level can be challenging. However, there has been significant progress in developing approaches that mimic the mechanisms by which viruses arose in the first place. In particular, library-based selection, the generation of one diverse genetic library and selection for new properties, and directed evolution, based on the multiple rounds of library generation and selection for iterative improvement of function, have strong potential in engineering novel properties into these complex biomolecular assemblies. This review will discuss progress in the application of peptide display, library selection, and directed evolution technologies toward engineering vectors based on retrovirus, adeno-associated virus, and adenovirus that are capable of targeted delivery to specific cell types. In addition to creating biomedically useful products, these approaches have future potential to yield novel insights into viral structure-function relationships.
Collapse
Affiliation(s)
- Jae-Hyung Jang
- Department of Chemical Engineering and Helen Wills Neuroscience Institute, University of California, 201 Gilman Hall, Berkeley, California 94720, USA
| | | | | |
Collapse
|
40
|
Ashton RS, Peltier J, Fasano CA, O'Neill A, Leonard J, Temple S, Schaffer DV, Kane RS. High-throughput screening of gene function in stem cells using clonal microarrays. Stem Cells 2007; 25:2928-35. [PMID: 17673524 DOI: 10.1634/stemcells.2007-0468] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We describe a microarray-based approach for the high-throughput screening of gene function in stem cells and demonstrate the potential of this method by growing and isolating clonal populations of both adult and embryonic neural stem cells. Clonal microarrays are constructed by seeding a population of cells at clonal density on micropatterned surfaces generated using soft lithographic microfabrication techniques. Clones of interest can be isolated after assaying in parallel for various cellular processes and functions, including proliferation, signal transduction, and differentiation. We demonstrate the compatibility of the technique with both gain- and loss-of-function studies using cell populations infected with cDNA libraries or DNA constructs that induce RNA interference. The infection of cells with a library prior to seeding and the compact but isolated growth of clonal cell populations will facilitate the screening of large libraries in a wide variety of mammalian cells, including those that are difficult to transfect by conventional methods.
Collapse
Affiliation(s)
- Randolph S Ashton
- The Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Koerber JT, Jang JH, Yu JH, Kane RS, Schaffer DV. Engineering adeno-associated virus for one-step purification via immobilized metal affinity chromatography. Hum Gene Ther 2007; 18:367-78. [PMID: 17437357 DOI: 10.1089/hum.2006.139] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Adeno-associated virus (AAV) is a promising vehicle for gene therapy, which will rely on the generation of high-titer, high-purity recombinant vectors. However, numerous purification protocols can involve challenging optimization or scalability issues, and most AAV serotypes do not bind heparin or sialic acid, used for AAV2/3 or AAV4/5 purification, requiring the development of new chromatography strategies. Immobilized metal affinity chromatography (IMAC) allows for robust protein purification via affinity tags such as the hexahistidine (His(6)) sequence. Through the combination of a diverse AAV2 library and rational peptide insertions, we have located an optimal His(6) tag insertion site within the viral capsid. This mutant and a related AAV8 variant can be purified from clarified cell lysate in a single gravity column step at infectious particle yields exceeding 90%. Furthermore, injection of IMAC-purified vector into the brain demonstrates that it mediates high-efficiency gene delivery in vivo, equivalent to that of wild-type capsid, with minimal immune cell activation. This affinity chromatography method may offer advantages in ease of purification, final vector purity, and process scalability. Moreover, a combined rational design and high-throughput library selection approach can aid in the design of enhanced viral gene delivery vectors.
Collapse
Affiliation(s)
- James T Koerber
- Department of Chemical Engineering and Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | | | | | | | | |
Collapse
|
42
|
Whitehead TA, Boonyaratanakornkit BB, Höllrigl V, Clark DS. A filamentous molecular chaperone of the prefoldin family from the deep-sea hyperthermophile Methanocaldococcus jannaschii. Protein Sci 2007; 16:626-34. [PMID: 17384227 PMCID: PMC2203346 DOI: 10.1110/ps.062599907] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Prefoldin is a molecular chaperone found in the domains eukarya and archaea that acts in conjunction with Group II chaperonin to correctly fold other nascent proteins. Previously, our group identified a putative single subunit of prefoldin, gamma PFD, that was up-regulated in response to heat stress in the hyperthermophilic archaeon Methanocaldococcus jannaschii. In order to characterize this protein, we subcloned and expressed it and the other two prefoldin subunits from M. jannaschii, alpha and beta PFD, into Eschericia coli and characterized the proteins. Whereas alpha and beta PFD readily assembled into the expected hexamer, gamma PFD would not assemble with either protein. Instead, gamma PFD forms long filaments of defined dimensions measuring 8.5 nm x 1.7-3.5 nm and lengths exceeding 1 microm. Filamentous gamma PFD acts as a molecular chaperone through in vitro assays, in a manner comparable to PFD. A possible molecular model for filament assembly is discussed.
Collapse
Affiliation(s)
- Timothy A Whitehead
- Department of Chemical Engineering, University of California, Berkeley, Berkeley, California 94720, USA
| | | | | | | |
Collapse
|
43
|
Peltier J, O'Neill A, Schaffer DV. PI3K/Akt and CREB regulate adult neural hippocampal progenitor proliferation and differentiation. Dev Neurobiol 2007; 67:1348-61. [PMID: 17638387 DOI: 10.1002/dneu.20506] [Citation(s) in RCA: 347] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The phosphoinositide 3-OH kinase (PI3K)/Akt pathway has been implicated in regulating several important cellular processes, including apoptosis, survival, proliferation, and metabolism. Using both pharmacological and genetic means, we demonstrate here that PI3K/Akt plays a crucial role in the proliferation of adult hippocampal neural progenitor cells. PI3K/Akt transduces intracellular signals from multiple mitogens, including basic fibroblast growth factor (FGF-2), Sonic hedgehog (Shh), and insulin-like growth factor 1 (IGF-1). In addition, retroviral vector-mediated over-expression of wild type Akt increased cell proliferation, while a dominant negative Akt inhibited proliferation. Furthermore, wild type Akt over-expression reduced glial (GFAP) and neuronal (beta-tubulin III) marker expression during differentiation, indicating that it inhibits cell differentiation. We also show that activation of the cAMP response element binding protein (CREB), which occurs in cells stimulated by FGF-2, is limited when Akt signaling is inhibited, demonstrating a link between Akt and CREB. Over-expression of wild type CREB increases progenitor proliferation, whereas dominant negative CREB only slightly decreases proliferation. These results indicate that PI3K/Akt signaling integrates extracellular signaling information to promote cellular proliferation and inhibit differentiation in adult neural progenitors.
Collapse
Affiliation(s)
- Joseph Peltier
- Department of Chemical Engineering, University of California, Berkeley, California 94720, USA
| | | | | |
Collapse
|
44
|
Yu JH, Schaffer DV. High-throughput, library-based selection of a murine leukemia virus variant to infect nondividing cells. J Virol 2006; 80:8981-8. [PMID: 16940510 PMCID: PMC1563944 DOI: 10.1128/jvi.00615-06] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gammaretroviruses, such as murine leukemia virus (MLV), are functionally distinguished from lentiviruses, such as human immunodeficiency virus, by their inability to infect nondividing cells. Attempts to engineer this property into MLV have been hindered by an incomplete understanding of early events in the viral life cycle. We utilized a transposon-based method to generate saturated peptide insertion libraries of MLV gag-pol variants with nuclear localization signals randomly incorporated throughout these overlapping genes. High-throughput selection of the libraries via iterative retroviral infection of nondividing cells led to the identification of a novel variant that successfully transduced growth-arrested cells. Vector packaging by cotransfection of the gag-pol.NLS variant with wild-type gag-pol produced high-titer virions capable of infecting neurons in vitro and in vivo. The capacity of mutant virions to transduce nondividing cells could help to elucidate incompletely understood mechanisms of the viral life cycle and greatly broaden the gene therapy applications of retroviral vectors. Furthermore, the ability to engineer key intracellular viral infection steps has potential implications for the understanding, design, and control of other post-entry events. Finally, this method of library generation and selection for a desired phenotype directly in a mammalian system can be readily expanded to address other challenges in protein engineering.
Collapse
Affiliation(s)
- Julie H Yu
- Department of Chemical Engineering, University of California-Berkeley, 201 Gilman Hall, Berkeley, CA 94720-1462, USA
| | | |
Collapse
|
45
|
Dreja H, Piechaczyk M. The effects of N-terminal insertion into VSV-G of an scFv peptide. Virol J 2006; 3:69. [PMID: 16948856 PMCID: PMC1564393 DOI: 10.1186/1743-422x-3-69] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Accepted: 09/02/2006] [Indexed: 11/10/2022] Open
Abstract
Recombinant retroviruses, including lentiviruses, are the most widely used vectors for both in vitro and in vivo stable gene transfer. However, the inability to selectively deliver transgenes into cells of interest limits the use of this technology. Due to its wide tropism, stability and ability to pseudotype a range of viral vectors, vesicular stomatitis virus G protein (VSV-G) is the most commonly used pseudotyping protein. Here, we attempted to engineer this protein for targeting purposes. Chimaeric VSV-G proteins were constructed by linking a cell-directing single-chain antibody (scFv) to its N-terminal. We show that the chimaeric VSV-G molecules can integrate into retroviral and lentiviral particles. HIV-1 particles pseudotyped with VSV-G linked to an scFv against human Major Histocompatibility Complex class I (MHC-I) bind strongly and specifically to human cells. Also, this novel molecule preferentially drives lentiviral transduction of human cells, although the titre is considerably lower that viruses pseudotyped with VSV-G. This is likely due to the inefficient fusion activity of the modified protein. To our knowledge, this is the first report where VSV-G was successfully engineered to include a large (253 amino acids) exogenous peptide and where attempts were made to change the infection profile of VSV-G pseudotyped vectors.
Collapse
Affiliation(s)
- Hanna Dreja
- Institut de Génétique Moléculaire de Montpellier, UMR 5535, IFR122, CNRS, France
| | - Marc Piechaczyk
- Institut de Génétique Moléculaire de Montpellier, UMR 5535, IFR122, CNRS, France
| |
Collapse
|