1
|
Casas-Mollano JA, Zinselmeier M, Sychla A, Smanski MJ. Efficient gene activation in plants by the MoonTag programmable transcriptional activator. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528671. [PMID: 36824723 PMCID: PMC9948947 DOI: 10.1101/2023.02.15.528671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
CRISPR/Cas-based transcriptional activators have been developed to induce gene expression in eukaryotic and prokaryotic organisms. The main advantages of CRISPR-Cas based systems is that they can achieve high levels of transcriptional activation and are very easy to program via pairing between the guide RNA and the DNA target strand. SunTag is a second-generation system that activates transcription by recruiting multiple copies of an activation domain (AD) to its target promoters. SunTag is a strong activator; however, in some species it is difficult to stably express. To overcome this problem, we designed MoonTag, a new activator that worked on the same basic principle as SunTag, but whose components are better tolerated when stably expressed in transgenic plants. We demonstrate that MoonTag is capable of inducing high levels of transcription in all plants tested. In Setaria, MoonTag is capable of inducing high levels of transcription of reporter genes as well as of endogenous genes. More important, MoonTag components are expressed in transgenic plants to high levels without any deleterious effects. MoonTag is also able to efficiently activate genes in eudicotyledonous species such as Arabidopsis and tomato. Finally, we show that MoonTag activation is functional across a range of temperatures, which is promising for potential field applications.
Collapse
Affiliation(s)
- J Armando Casas-Mollano
- Department of Biochemistry, Molecular Biology, and Biophysics and Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
- Center for Precision Plant Genomics, University of Minnesota, Saint Paul, MN 55108
| | - Matthew Zinselmeier
- Department of Biochemistry, Molecular Biology, and Biophysics and Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
- Center for Precision Plant Genomics, University of Minnesota, Saint Paul, MN 55108
- Department of Genetics, Cellular, and Developmental Biology, University of Minnesota, Saint Paul, MN 55108
| | - Adam Sychla
- Department of Biochemistry, Molecular Biology, and Biophysics and Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
- Center for Precision Plant Genomics, University of Minnesota, Saint Paul, MN 55108
| | - Michael J Smanski
- Department of Biochemistry, Molecular Biology, and Biophysics and Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108
- Center for Precision Plant Genomics, University of Minnesota, Saint Paul, MN 55108
| |
Collapse
|
2
|
Troisi M, Marini E, Abbiento V, Stazzoni S, Andreano E, Rappuoli R. A new dawn for monoclonal antibodies against antimicrobial resistant bacteria. Front Microbiol 2022; 13:1080059. [PMID: 36590399 PMCID: PMC9795047 DOI: 10.3389/fmicb.2022.1080059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial resistance (AMR) is a quickly advancing threat for human health worldwide and almost 5 million deaths are already attributable to this phenomenon every year. Since antibiotics are failing to treat AMR-bacteria, new tools are needed, and human monoclonal antibodies (mAbs) can fill this role. In almost 50 years since the introduction of the first technology that led to mAb discovery, enormous leaps forward have been made to identify and develop extremely potent human mAbs. While their usefulness has been extensively proved against viral pathogens, human mAbs have yet to find their space in treating and preventing infections from AMR-bacteria and fully conquer the field of infectious diseases. The novel and most innovative technologies herein reviewed can support this goal and add powerful tools in the arsenal of weapons against AMR.
Collapse
Affiliation(s)
- Marco Troisi
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Eleonora Marini
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Valentina Abbiento
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Samuele Stazzoni
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Emanuele Andreano
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Rino Rappuoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Fondazione Biotecnopolo di Siena, Siena, Italy
| |
Collapse
|
3
|
Uchino S, Ito Y, Sato Y, Handa T, Ohkawa Y, Tokunaga M, Kimura H. Live imaging of transcription sites using an elongating RNA polymerase II-specific probe. J Cell Biol 2022; 221:212888. [PMID: 34854870 PMCID: PMC8647360 DOI: 10.1083/jcb.202104134] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/12/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
In eukaryotic nuclei, most genes are transcribed by RNA polymerase II (RNAP2), whose regulation is a key to understanding the genome and cell function. RNAP2 has a long heptapeptide repeat (Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7), and Ser2 is phosphorylated on an elongation form. To detect RNAP2 Ser2 phosphorylation (RNAP2 Ser2ph) in living cells, we developed a genetically encoded modification-specific intracellular antibody (mintbody) probe. The RNAP2 Ser2ph-mintbody exhibited numerous foci, possibly representing transcription “factories,” and foci were diminished during mitosis and in a Ser2 kinase inhibitor. An in vitro binding assay using phosphopeptides confirmed the mintbody’s specificity. RNAP2 Ser2ph-mintbody foci were colocalized with proteins associated with elongating RNAP2 compared with factors involved in the initiation. These results support the view that mintbody localization represents the sites of RNAP2 Ser2ph in living cells. RNAP2 Ser2ph-mintbody foci showed constrained diffusional motion like chromatin, but they were more mobile than DNA replication domains and p300-enriched foci, suggesting that the elongating RNAP2 complexes are separated from more confined chromatin domains.
Collapse
Affiliation(s)
- Satoshi Uchino
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuma Ito
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuko Sato
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsuya Handa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Makio Tokunaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Kimura
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
4
|
Lin CW, Lerner RA. Antibody Libraries as Tools to Discover Functional Antibodies and Receptor Pleiotropism. Int J Mol Sci 2021; 22:4123. [PMID: 33923551 PMCID: PMC8073236 DOI: 10.3390/ijms22084123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/16/2022] Open
Abstract
Most antibodies currently in use have been selected based on their binding affinity. However, nowadays, antibodies that can not only bind but can also alter the function of cell surface signaling components are increasingly sought after as therapeutic drugs. Therefore, the identification of such functional antibodies from a large antibody library is the subject of intensive research. New methods applied to combinatorial antibody libraries now allow the isolation of functional antibodies in the cellular environment. These selected agonist antibodies have provided new insights into important issues of signal transduction. Notably, when certain antibodies bind to a given receptor, the cell fate induced by them may be the same or different from that induced by natural agonists. In addition, combined with phenotypic screening, this platform allows us to discover unexpected experimental results and explore various phenomena in cell biology, such as those associated with stem cells and cancer cells.
Collapse
Affiliation(s)
| | - Richard A. Lerner
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA;
| |
Collapse
|
5
|
Tjalsma SJD, Hori M, Sato Y, Bousard A, Ohi A, Raposo AC, Roensch J, Le Saux A, Nogami J, Maehara K, Kujirai T, Handa T, Bagés‐Arnal S, Ohkawa Y, Kurumizaka H, da Rocha ST, Żylicz JJ, Kimura H, Heard E. H4K20me1 and H3K27me3 are concurrently loaded onto the inactive X chromosome but dispensable for inducing gene silencing. EMBO Rep 2021; 22:e51989. [PMID: 33605056 PMCID: PMC7926250 DOI: 10.15252/embr.202051989] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/22/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
During X chromosome inactivation (XCI), in female placental mammals, gene silencing is initiated by the Xist long non-coding RNA. Xist accumulation at the X leads to enrichment of specific chromatin marks, including PRC2-dependent H3K27me3 and SETD8-dependent H4K20me1. However, the dynamics of this process in relation to Xist RNA accumulation remains unknown as is the involvement of H4K20me1 in initiating gene silencing. To follow XCI dynamics in living cells, we developed a genetically encoded, H3K27me3-specific intracellular antibody or H3K27me3-mintbody. By combining live-cell imaging of H3K27me3, H4K20me1, the X chromosome and Xist RNA, with ChIP-seq analysis we uncover concurrent accumulation of both marks during XCI, albeit with distinct genomic distributions. Furthermore, using a Xist B and C repeat mutant, which still shows gene silencing on the X but not H3K27me3 deposition, we also find a complete lack of H4K20me1 enrichment. This demonstrates that H4K20me1 is dispensable for the initiation of gene silencing, although it may have a role in the chromatin compaction that characterises facultative heterochromatin.
Collapse
Affiliation(s)
- Sjoerd J D Tjalsma
- Mammalian Developmental Epigenetics GroupInstitut CurieCNRS UMR3215, INSERM U934PSL UniversityParisFrance
| | - Mayako Hori
- Graduate School of Bioscience and BiotechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Yuko Sato
- Graduate School of Bioscience and BiotechnologyTokyo Institute of TechnologyYokohamaJapan
- Cell Biology CenterInstitute of Innovative ResearchTokyo Institute of TechnologyYokohamaJapan
| | - Aurelie Bousard
- Mammalian Developmental Epigenetics GroupInstitut CurieCNRS UMR3215, INSERM U934PSL UniversityParisFrance
| | - Akito Ohi
- Graduate School of Bioscience and BiotechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Ana Cláudia Raposo
- Faculdade de MedicinaInstituto de Medicina MolecularJoão Lobo AntunesUniversidade de LisboaLisboaPortugal
| | - Julia Roensch
- Mammalian Developmental Epigenetics GroupInstitut CurieCNRS UMR3215, INSERM U934PSL UniversityParisFrance
| | - Agnes Le Saux
- Mammalian Developmental Epigenetics GroupInstitut CurieCNRS UMR3215, INSERM U934PSL UniversityParisFrance
| | - Jumpei Nogami
- Division of TranscriptomicsMedical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Kazumitsu Maehara
- Division of TranscriptomicsMedical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Tomoya Kujirai
- Institute for Quantitative BiosciencesThe University of TokyoTokyoJapan
| | - Tetsuya Handa
- Cell Biology CenterInstitute of Innovative ResearchTokyo Institute of TechnologyYokohamaJapan
| | - Sandra Bagés‐Arnal
- The Novo Nordisk Foundation Center for Stem Cell BiologyCopenhagenDenmark
| | - Yasuyuki Ohkawa
- Division of TranscriptomicsMedical Institute of BioregulationKyushu UniversityFukuokaJapan
| | | | - Simão Teixeira da Rocha
- Faculdade de MedicinaInstituto de Medicina MolecularJoão Lobo AntunesUniversidade de LisboaLisboaPortugal
| | - Jan J Żylicz
- Mammalian Developmental Epigenetics GroupInstitut CurieCNRS UMR3215, INSERM U934PSL UniversityParisFrance
- The Novo Nordisk Foundation Center for Stem Cell BiologyCopenhagenDenmark
- Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Hiroshi Kimura
- Graduate School of Bioscience and BiotechnologyTokyo Institute of TechnologyYokohamaJapan
- Cell Biology CenterInstitute of Innovative ResearchTokyo Institute of TechnologyYokohamaJapan
| | - Edith Heard
- EMBL HeidelbergHeidelbergGermany
- Collège de FranceParisFrance
| |
Collapse
|
6
|
Jayanthi B, Bachhav B, Wan Z, Martinez Legaspi S, Segatori L. A platform for post-translational spatiotemporal control of cellular proteins. Synth Biol (Oxf) 2021; 6:ysab002. [PMID: 33763602 PMCID: PMC7976946 DOI: 10.1093/synbio/ysab002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Mammalian cells process information through coordinated spatiotemporal regulation of proteins. Engineering cellular networks thus relies on efficient tools for regulating protein levels in specific subcellular compartments. To address the need to manipulate the extent and dynamics of protein localization, we developed a platform technology for the target-specific control of protein destination. This platform is based on bifunctional molecules comprising a target-specific nanobody and universal sequences determining target subcellular localization or degradation rate. We demonstrate that nanobody-mediated localization depends on the expression level of the target and the nanobody, and the extent of target subcellular localization can be regulated by combining multiple target-specific nanobodies with distinct localization or degradation sequences. We also show that this platform for nanobody-mediated target localization and degradation can be regulated transcriptionally and integrated within orthogonal genetic circuits to achieve the desired temporal control over spatial regulation of target proteins. The platform reported in this study provides an innovative tool to control protein subcellular localization, which will be useful to investigate protein function and regulate large synthetic gene circuits.
Collapse
Affiliation(s)
- Brianna Jayanthi
- Systems, Synthetic and Physical Biology Graduate Program, Rice University, Houston, TX, USA
| | - Bhagyashree Bachhav
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA
| | - Zengyi Wan
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Laura Segatori
- Systems, Synthetic and Physical Biology Graduate Program, Rice University, Houston, TX, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Biosciences, Rice University, Houston, TX, USA
| |
Collapse
|
7
|
Se Thoe E, Fauzi A, Tang YQ, Chamyuang S, Chia AYY. A review on advances of treatment modalities for Alzheimer's disease. Life Sci 2021; 276:119129. [PMID: 33515559 DOI: 10.1016/j.lfs.2021.119129] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease which is mainly characterized by progressive impairment in cognition, emotion, language and memory in older population. Considering the impact of AD, formulations of pharmaceutical drugs and cholinesterase inhibitors have been widely propagated, receiving endorsement by FDA as a form of AD treatment. However, these medications were gradually discovered to be ineffective in removing the root of AD pathogenesis but merely targeting the symptoms so as to improve a patient's cognitive outcome. Hence, a search for better disease-modifying alternatives is put into motion. Having a clear understanding of the neuroprotective mechanisms and diverse properties undertaken by specific genes, antibodies and nanoparticles is central towards designing novel therapeutic agents. In this review, we provide a brief introduction on the background of Alzheimer's disease, the biology of blood-brain barrier, along with the potentials and drawbacks associated with current therapeutic treatment avenues pertaining to gene therapy, immunotherapy and nanotherapy for better diagnosis and management of Alzheimer's disease.
Collapse
Affiliation(s)
- Ewen Se Thoe
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Ayesha Fauzi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Yin Quan Tang
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Sunita Chamyuang
- School of Science, Mae Fah Luang University, Chaing Rai 57100, Thailand; Microbial Products and Innovation Research Group, Mae Fah Luang University, Chaing Rai 57100, Thailand
| | - Adeline Yoke Yin Chia
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia.
| |
Collapse
|
8
|
Abstract
RAS was identified as a human oncogene in the early 1980s and subsequently found to be mutated in nearly 30% of all human cancers. More importantly, RAS plays a central role in driving tumor development and maintenance. Despite decades of effort, there remain no FDA approved drugs that directly inhibit RAS. The prevalence of RAS mutations in cancer and the lack of effective anti-RAS therapies stem from RAS' core role in growth factor signaling, unique structural features, and biochemistry. However, recent advances have brought promising new drugs to clinical trials and shone a ray of hope in the field. Here, we will exposit the details of RAS biology that illustrate its key role in cell signaling and shed light on the difficulties in therapeutically targeting RAS. Furthermore, past and current efforts to develop RAS inhibitors will be discussed in depth.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - Imran Khan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| | - John P O'Bryan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| |
Collapse
|
9
|
Cattaneo A, Chirichella M. Targeting the Post-translational Proteome with Intrabodies. Trends Biotechnol 2018; 37:578-591. [PMID: 30577991 DOI: 10.1016/j.tibtech.2018.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022]
Abstract
The complexity of the proteome exceeds that of the genome. Post-translational modifications (PTMs) and conformational changes of proteins trigger new molecular interactions whose systematic elucidation is hampered by the lack of specific tools. PTMs are particularly relevant for epigenetic regulation of gene expression; a field of translational interest. However, state-of-the-art inhibitors used in epigenetic studies and therapies target modifier enzymes such as acetylases and deacetylases, rather than a single PTM protein per se. The systematic development of anti-PTM intrabodies, which allow targeting of intracellular proteins in the context of living cells, will help reaching a new level of precision and specificity in the description of epigenetics, paving the way to new therapeutic opportunities.
Collapse
Affiliation(s)
- Antonino Cattaneo
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri, 7 - 56126 Pisa, Italy.
| | - Michele Chirichella
- Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri, 7 - 56126 Pisa, Italy; Current address: Institute for Research in Biomedicine, Università della Svizzera italiana (USI), Via Vincenzo Vela 6, CH-6500 Bellinzona, Switzerland
| |
Collapse
|
10
|
Luz D, Shiga EA, Chen G, Quintilio W, Andrade FB, Maranhão AQ, Caetano BA, Mitsunari T, Silva MA, Rocha LB, Moro AM, Sidhu SS, Piazza RMF. Structural Changes in Stx1 Engineering Monoclonal Antibody Improves Its Functionality as Diagnostic Tool for a Rapid Latex Agglutination Test. Antibodies (Basel) 2018; 7:antib7010009. [PMID: 31544861 PMCID: PMC6698835 DOI: 10.3390/antib7010009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/10/2018] [Accepted: 01/26/2018] [Indexed: 11/16/2022] Open
Abstract
Stx1 toxin is one of the AB5 toxins of Shiga toxin-producing Escherichia coli (STEC) responsible for foodborne intoxication during outbreaks. The single-chain variable fragment (scFv) is the most common recombinant antibody format; it consists of both variable chains connected by a peptide linker with conserved specificity and affinity for antigen. The drawbacks of scFv production in bacteria are the heterologous expression, conformation and stability of the molecule, which could change the affinity for the antigen. In this work, we obtained a stable and functional scFv-Stx1 in bacteria, starting from IgG produced by hybridoma cells. After structural modifications, i.e., change in protein orientation, vector and linker, its solubility for expression in bacteria was increased as well as the affinity for its antigen, demonstrated by a scFv dissociation constant (KD) of 2.26 × 10-7 M. Also, it was able to recognize purified Stx1 and cross-reacted with Stx2 toxin by ELISA (Enzyme-Linked Immunosorbent Assay), and detected 88% of Stx1-producing strains using a rapid latex agglutination test. Thus, the scFv fragment obtained in the present work is a bacteria-produced tool for use in a rapid diagnosis test, providing an alternative for STEC diagnosis.
Collapse
Affiliation(s)
- Daniela Luz
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Emerson A Shiga
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Gang Chen
- Department of Molecular Genetics, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.
| | - Wagner Quintilio
- Laboratório de Biofármacos em Células Animais, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Fernanda B Andrade
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Andrea Q Maranhão
- Laboratório de Imunologia, Universidade de Brasília, Brasília 70910-900, Brazil.
| | - Bruna A Caetano
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Thaís Mitsunari
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Míriam A Silva
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Letícia B Rocha
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Ana M Moro
- Laboratório de Biofármacos em Células Animais, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| | - Sachdev S Sidhu
- Department of Molecular Genetics, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.
| | - Roxane M F Piazza
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, SP 05503-900, Brazil.
| |
Collapse
|
11
|
Use of peptide aptamers, cationic peptides and artificial zinc finger proteins to generate resistance to plant viruses. Curr Opin Virol 2017; 26:120-124. [PMID: 28806695 DOI: 10.1016/j.coviro.2017.07.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/22/2017] [Accepted: 07/25/2017] [Indexed: 11/22/2022]
Abstract
Various RNA/DNA viruses have caused severe infectious diseases in plants as well as animals, including humans, and been a threat to the production of agricultural crops. Therefore, prevention of plant virus infections is a major objective in crop protection. One attractive approach is to inhibit functions of viral proteins responsible for virus infections. In this review, I describe the status using such approaches to confer virus resistance to plants by three types of peptides/proteins: peptide aptamers, artificial zinc finger proteins and acidic peptides. These approaches vary in their specificity, broadness to other viruses, extent of protection and mechanisms of action. Additional ways to improve these approaches are also discussed.
Collapse
|
12
|
Sato Y, Kujirai T, Arai R, Asakawa H, Ohtsuki C, Horikoshi N, Yamagata K, Ueda J, Nagase T, Haraguchi T, Hiraoka Y, Kimura A, Kurumizaka H, Kimura H. A Genetically Encoded Probe for Live-Cell Imaging of H4K20 Monomethylation. J Mol Biol 2016; 428:3885-3902. [PMID: 27534817 DOI: 10.1016/j.jmb.2016.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 01/28/2023]
Abstract
Eukaryotic gene expression is regulated in the context of chromatin. Dynamic changes in post-translational histone modification are thought to play key roles in fundamental cellular functions such as regulation of the cell cycle, development, and differentiation. To elucidate the relationship between histone modifications and cellular functions, it is important to monitor the dynamics of modifications in single living cells. A genetically encoded probe called mintbody (modification-specific intracellular antibody), which is a single-chain variable fragment tagged with a fluorescent protein, has been proposed as a useful visualization tool. However, the efficacy of intracellular expression of antibody fragments has been limited, in part due to different environmental conditions in the cytoplasm compared to the endoplasmic reticulum where secreted proteins such as antibodies are folded. In this study, we have developed a new mintbody specific for histone H4 Lys20 monomethylation (H4K20me1). The specificity of the H4K20me1-mintbody in living cells was verified using yeast mutants and mammalian cells in which this target modification was diminished. Expression of the H4K20me1-mintbody allowed us to monitor the oscillation of H4K20me1 levels during the cell cycle. Moreover, dosage-compensated X chromosomes were visualized using the H4K20me1-mintbody in mouse and nematode cells. Using X-ray crystallography and mutational analyses, we identified critical amino acids that contributed to stabilization and/or proper folding of the mintbody. Taken together, these data provide important implications for future studies aimed at developing functional intracellular antibodies. Specifically, the H4K20me1-mintbody provides a powerful tool to track this particular histone modification in living cells and organisms.
Collapse
Affiliation(s)
- Yuko Sato
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| | - Tomoya Kujirai
- Laboratory of Structural Biology, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Ritsuko Arai
- Cell Architecture Laboratory, Structural Biology Center, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Haruhiko Asakawa
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Chizuru Ohtsuki
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Naoki Horikoshi
- Laboratory of Structural Biology, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Kazuo Yamagata
- Faculty of Biology-Oriented Science and Technology, Kindai University, Kinokawa City, Wakayama 649-6493, Japan
| | - Jun Ueda
- Center for Education in Laboratory Animal Research, Chubu University, Kasugai, Aichi 487-8501, Japan
| | - Takahiro Nagase
- Public Relations Team, Kazusa DNA Research Institute, Chiba 292-0818, Japan
| | - Tokuko Haraguchi
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan; Advanced ICT Research Institute, National Institute of Information and Communications Technology (NICT), Kobe 651-2492, Japan
| | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan; Advanced ICT Research Institute, National Institute of Information and Communications Technology (NICT), Kobe 651-2492, Japan
| | - Akatsuki Kimura
- Cell Architecture Laboratory, Structural Biology Center, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Hitoshi Kurumizaka
- Laboratory of Structural Biology, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Hiroshi Kimura
- Cell Biology Unit, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| |
Collapse
|
13
|
Bieli D, Alborelli I, Harmansa S, Matsuda S, Caussinus E, Affolter M. Development and Application of Functionalized Protein Binders in Multicellular Organisms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 325:181-213. [DOI: 10.1016/bs.ircmb.2016.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
14
|
Waraho-Zhmayev D, Meksiriporn B, Portnoff AD, DeLisa MP. Optimizing recombinant antibodies for intracellular function using hitchhiker-mediated survival selection. Protein Eng Des Sel 2014; 27:351-8. [PMID: 25225416 DOI: 10.1093/protein/gzu038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The 'hitchhiker' mechanism of the bacterial twin-arginine translocation pathway has previously been adapted as a genetic selection for detecting pairwise protein interactions in the cytoplasm of living Escherichia coli cells. Here, we extended this method, called FLI-TRAP, for rapid isolation of intracellular antibodies (intrabodies) in the single-chain Fv format that possess superior traits simply by demanding bacterial growth on high concentrations of antibiotic. Following just a single round of survival-based enrichment using FLI-TRAP, variants of an intrabody against the dimerization domain of the yeast Gcn4p transcription factor were isolated having significantly greater intracellular stability that translated to yield enhancements of >10-fold. Likewise, an intrabody specific for the non-amyloid component region of α-synuclein was isolated that has ~8-fold improved antigen-binding affinity. Collectively, our results illustrate the potential of the FLI-TRAP method for intracellular stabilization and affinity maturation of intrabodies, all without the need for purification or immobilization of the antigen.
Collapse
Affiliation(s)
- Dujduan Waraho-Zhmayev
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, 126 Pracha-utid Road, Bangmod, Toongkru, Bangkok 10140, Thailand
| | | | - Alyse D Portnoff
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Matthew P DeLisa
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
15
|
Mazuc E, Guglielmi L, Bec N, Parez V, Hahn CS, Mollevi C, Parrinello H, Desvignes JP, Larroque C, Jupp R, Dariavach P, Martineau P. In-cell intrabody selection from a diverse human library identifies C12orf4 protein as a new player in rodent mast cell degranulation. PLoS One 2014; 9:e104998. [PMID: 25122211 PMCID: PMC4133367 DOI: 10.1371/journal.pone.0104998] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 07/14/2014] [Indexed: 01/04/2023] Open
Abstract
The high specificity of antibodies for their antigen allows a fine discrimination of target conformations and post-translational modifications, making antibodies the first choice tool to interrogate the proteome. We describe here an approach based on a large-scale intracellular expression and selection of antibody fragments in eukaryotic cells, so-called intrabodies, and the subsequent identification of their natural target within living cell. Starting from a phenotypic trait, this integrated system allows the identification of new therapeutic targets together with their companion inhibitory intrabody. We applied this system in a model of allergy and inflammation. We first cloned a large and highly diverse intrabody library both in a plasmid and a retroviral eukaryotic expression vector. After transfection in the RBL-2H3 rat basophilic leukemia cell line, we performed seven rounds of selection to isolate cells displaying a defect in FcεRI-induced degranulation. We used high throughput sequencing to identify intrabody sequences enriched during the course of selection. Only one intrabody was common to both plasmid and retroviral selections, and was used to capture and identify its target from cell extracts. Mass spectrometry analysis identified protein RGD1311164 (C12orf4), with no previously described function. Our data demonstrate that RGD1311164 is a cytoplasmic protein implicated in the early signaling events following FcεRI-induced cell activation. This work illustrates the strength of the intrabody-based in-cell selection, which allowed the identification of a new player in mast cell activation together with its specific inhibitor intrabody.
Collapse
Affiliation(s)
- Elsa Mazuc
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
| | - Laurence Guglielmi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
| | - Nicole Bec
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
| | - Vincent Parez
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
| | - Chang S. Hahn
- Sanofi-Aventis, Bridgewater, New Jersey, United States of America
| | - Caroline Mollevi
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
| | - Hugues Parrinello
- MGX-Montpellier GenomiX, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Christian Larroque
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
| | - Ray Jupp
- Sanofi-Aventis, Bridgewater, New Jersey, United States of America
| | - Piona Dariavach
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
- Université Montpellier2, Montpellier, France
- * E-mail: (PD); (PM)
| | - Pierre Martineau
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM, U896, Montpellier, France
- Université Montpellier1, Montpellier, France
- ICM, Institut régional du Cancer Montpellier, Montpellier, France
- * E-mail: (PD); (PM)
| |
Collapse
|
16
|
Kaiser PD, Maier J, Traenkle B, Emele F, Rothbauer U. Recent progress in generating intracellular functional antibody fragments to target and trace cellular components in living cells. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1933-1942. [PMID: 24792387 DOI: 10.1016/j.bbapap.2014.04.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 04/16/2014] [Accepted: 04/21/2014] [Indexed: 02/04/2023]
Abstract
In biomedical research there is an ongoing demand for new technologies, which help to elucidate disease mechanisms and provide the basis to develop novel therapeutics. In this context a comprehensive understanding of cellular processes and their pathophysiology based on reliable information on abundance, localization, posttranslational modifications and dynamic interactions of cellular components is indispensable. Besides their significant impact as therapeutic molecules, antibodies are arguably the most powerful research tools to study endogenous proteins and other cellular components. However, for cellular diagnostics their use is restricted to endpoint assays using fixed and permeabilized cells. Alternatively, live cell imaging using fluorescent protein-tagged reporters is widely used to study protein localization and dynamics in living cells. However, only artificially introduced chimeric proteins are visualized, whereas the endogenous proteins, their posttranslational modifications as well as non-protein components of the cell remain invisible and cannot be analyzed. To overcome these limitations, traceable intracellular binding molecules provide new opportunities to perform cellular diagnostics in real time. In this review we summarize recent progress in the generation of intracellular and cell penetrating antibodies and their application to target and trace cellular components in living cells. We highlight recent advances in the structural formulation of recombinant antibody formats, reliable screening protocols and sophisticated cellular targeting technologies and propose that such intrabodies will become versatile research tools for real time cell-based diagnostics including target validation and live cell imaging. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.
Collapse
Affiliation(s)
- Philipp D Kaiser
- Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany; Department of Pharmaceutical Biotechnology, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Julia Maier
- Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany; Department of Pharmaceutical Biotechnology, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Bjoern Traenkle
- Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany; Department of Pharmaceutical Biotechnology, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Felix Emele
- Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany; Department of Pharmaceutical Biotechnology, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Ulrich Rothbauer
- Natural and Medical Sciences Institute at the University of Tuebingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany; Department of Pharmaceutical Biotechnology, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany.
| |
Collapse
|
17
|
Sato M, Sawahata R, Sakuma C, Takenouchi T, Kitani H. Single domain intrabodies against WASP inhibit TCR-induced immune responses in transgenic mice T cells. Sci Rep 2013; 3:3003. [PMID: 24141565 PMCID: PMC3801110 DOI: 10.1038/srep03003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/04/2013] [Indexed: 11/26/2022] Open
Abstract
Intrabody technology provides a novel approach to decipher the molecular mechanisms of protein function in cells. Single domains composed of only the variable regions (VH or VL) of antibodies are the smallest recombinant antibody fragments to be constructed thus far. In this study, we developed transgenic (Tg) mice expressing the VH or VL single domains derived from a monoclonal antibody raised against the N-terminal domain of Wiskott–Aldrich syndrome protein (WASP), which is an adaptor molecule in immune cells. In T cells from anti-WASP VH and VL single domain Tg mice, interleukin-2 production induced by T cell receptor (TCR) stimulation were impaired, and specific interaction between the WASP N-terminal domain and the Fyn SH3 domain was strongly inhibited by masking the binding sites in WASP. These results strongly suggest that the VH/VL single domain intrabodies are sufficient to knockdown the domain function of target proteins in the cytosol.
Collapse
Affiliation(s)
- Mitsuru Sato
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, 1-2 Ohwashi, Tsukuba, Ibaraki 305-8634, Japan
| | | | | | | | | |
Collapse
|
18
|
Feng J, Guo H, Li S, Lu T. A study of the mechanism of the chaperone-like function of an scFv of human creatine kinase by computer simulation. PLoS One 2013; 8:e62147. [PMID: 23637984 PMCID: PMC3634753 DOI: 10.1371/journal.pone.0062147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 03/18/2013] [Indexed: 11/19/2022] Open
Abstract
A new application of antibodies is to use them as macromolecular chaperones. Protein antigens usually have multiple epitopes, thus, there may be a plurality of antibodies binding to one antigen. However, not all antibodies that bind to one antigen could act as a chaperone. Experiments show that some screened anti-human creatine kinase single chain antibodies (scFV) could assist in the folding and stabilizing of the enzyme, while others could not. We built the model of the single chain antibody (scFv-A4) that increased the stability of human creatine kinase (HCK) by the homology modeling method. Epitopes of human creatine kinase were predicted by computer and then the binding of scFv-A4 and HCK was modeled with computer. The calculation results were further combined with the peptide array membrane experiment results to obtain reliable models for the scFv-A4-HCK complex. Based on the above study we gave an explanation about how scFv-A4 could act as a macromolecular chaperone assisting the folding of HCK. This study provides an approach for predicting antigen-antibody binding mode and also a useful theoretical guidance for the study of antibodies' chaperone-like function.
Collapse
Affiliation(s)
- Jianyu Feng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Hong Guo
- College of Mathematics and Computer Science, Fuzhou University, Fuzhou, China
| | - Sen Li
- Beijing Key Laboratory of Genetic Engineering Drugs and Biotechnology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Tun Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| |
Collapse
|
19
|
Generation and characterization of a novel recombinant antibody against LMP1-TES1 of Epstein-Barr virus isolated by phage display. Viruses 2013; 5:1131-42. [PMID: 23609879 PMCID: PMC3705269 DOI: 10.3390/v5041131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/11/2013] [Accepted: 04/15/2013] [Indexed: 01/19/2023] Open
Abstract
Latent Membrane Protein 1 (LMP1) is a primary target for controlling tumorigenesis in Epstein-Barr virus related malignancies; in this study, we aimed to develop a specific antibody against the TES1 domain of the oncogenic LMP1. We screened a full human naïve Fab phage library against TES1 peptide, which consisted of C terminal-activating regions proximal 44 amino acids. After three rounds of panning, enrichment and testing by phage ELISA and further analyzed by DNA sequencing, we selected a phage clone with the highest affinity to LMP1-TES1 and designated it as htesFab. The positive clone was expressed in Escherichia coli and the purified htesFab was characterized for its binding specificity and affinity to LMP1. ELISA, immunofluorescence and FACS analysis confirmed that htesFab could recognize LMP1 TES1 both in vitro and in LMP1 expressing HNE2-LMP1 cells. Furthermore, MTT assay showed that htesFab inhibited the proliferation of HNE2-LMP1 cells in a dose-dependent manner. In summary, this study reported the isolation and characterization of human Fab, which specifically targets the C terminal region/TES1 of LMP1, and has potential to be developed as novel tool for the diagnosis and therapy of Epstein-Barr virus related carcinoma.
Collapse
|
20
|
Matsui H, Sakurai F, Katayama K, Abe Y, Machitani M, Kurachi S, Tachibana M, Mizuguchi H. A targeted adenovirus vector displaying a human fibronectin type III domain-based monobody in a fiber protein. Biomaterials 2013; 34:4191-4201. [PMID: 23473963 DOI: 10.1016/j.biomaterials.2013.02.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 02/17/2013] [Indexed: 11/17/2022]
Abstract
A major drawback of adenovirus (Ad) vectors is their nonspecific transduction into various types of cells or tissue after in vivo application, which might lead to unexpected toxicity and tissue damage. To overcome this problem, we developed a fiber-mutant Ad vector displaying a monobody specific for epidermal growth factor receptor (EGFR) or vascular endothelial growth factor receptor 2 (VEGFR2) in the C-terminus of the knobless fiber protein derived from T4 phage fibritin. A monobody, which is a single domain antibody mimic based on the tenth human fibronectin type III domain scaffold with a structure similar to the variable domains of antibodies, would be suitable as a targeting molecule for display on the Ad capsid proteins because of its highly stable structure even under reducing conditions and low molecular weight (approximately 10 kDa). Surface plasmon resonance (SPR) analysis revealed that the monobody-displaying Ad vector specifically bound to the targeted molecules, leading to significant increases in cellular binding and transduction efficiencies in the targeted cells. Transduction with the monobody-displaying Ad vectors was significantly inhibited in the presence of the Fc-chimera protein of EGFR and VEGFR2. This monobody-displaying Ad vector would be a crucial resource for targeted gene therapy.
Collapse
Affiliation(s)
- Hayato Matsui
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kazufumi Katayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasuhiro Abe
- Laboratory of Biopharmaceutical Research (Pharmaceutical Proteomics), National Institute of Biomedical Innovation, Osaka, Japan
| | - Mitsuhiro Machitani
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shinnosuke Kurachi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | - Masashi Tachibana
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan; Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan.
| |
Collapse
|
21
|
Canton I, Massignani M, Patikarnmonthon N, Chierico L, Robertson J, Renshaw SA, Warren NJ, Madsen JP, Armes SP, Lewis AL, Battaglia G. Fully synthetic polymer vesicles for intracellular delivery of antibodies in live cells. FASEB J 2012; 27:98-108. [PMID: 23033321 DOI: 10.1096/fj.12-212183] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is an emerging need both in pharmacology and within the biomedical industry to develop new tools to target intracellular mechanisms. The efficient delivery of functionally active proteins within cells is potentially a powerful research strategy, especially through the use of antibodies. In this work, we report on a nanovector for the efficient encapsulation and delivery of antibodies into live cells with no significant loss of cell viability or any deleterious effect on cell metabolic activity. This delivery system is based on poly[2-(methacryloyloxy)ethyl phosphorylcholine]-block-[2-(diisopropylamino)ethyl methacrylate] (PMPC-PDPA), a pH-sensitive diblock copolymer that self-assembles to form nanometer-sized vesicles, also known as polymersomes, at physiological pH. Polymersomes can successfully deliver relatively high antibody payloads within different types of live cells. We demonstrate that these antibodies can target their respective epitope showing immunolabeling of γ-tubulin, actin, Golgi protein, and the transcription factor NF-κB in live cells. Finally, we demonstrate that intracellular delivery of antibodies can control specific subcellular events, as well as modulate cell activity and proinflammatory processes.
Collapse
Affiliation(s)
- Irene Canton
- The Krebs Institute, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Haus-Cohen M, Assaraf YG, Binyamin L, Benhar I, Reiter Y. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. J Gastrointest Oncol 2012; 109:750-8. [PMID: 14999785 DOI: 10.1002/ijc.20037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Serum carbohydrate antigen (CA 19-9) is the most common tumor marker assessed in pancreatic cancer patients; nevertheless few articles have comprehensively evaluated the evidence for its utility in pancreatic cancer management. METHODS Literature search was performed using Medline with keywords "pancreatic cancer", "tumor markers", "CA 19-9", "diagnosis", "screening", "prognosis", "resectability" and "recurrence". All English language articles pertaining to the role of CA 19-9 in pancreatic cancer were critically analyzed to determine its utility as a biomarker for pancreatic cancer. RESULTS Serum CA 19-9 is the most extensively validated pancreatic cancer biomarker with multiple clinical applications. CA 19-9 serum levels have a sensitivity and specificity of 79-81% and 82-90% respectively for the diagnosis of pancreatic cancer in symptomatic patients; but are not useful as a screening marker because of low positive predictive value (0.5-0.9%). Pre-operative CA 19-9 serum levels provide useful prognostic information as patients with normal levels (<37 U/mL) have a prolonged median survival (32-36 months) compared to patients with elevated levels (>37 U/mL) (12-15 months). A CA 19-9 serum level of <100 U/mL implies likely resectable disease whereas levels >100 U/mL suggest unresectablity or metastatic disease. Normalization or a decrease in post-operative CA 19-9 serum levels by ≥20-50% from baseline following surgical resection or chemotherapy is associated with prolonged survival compared to failure of CA 19-9 serum levels to normalize or an increase. Important limitations to CA 19-9 serum level evaluation in pancreatic cancer include poor sensitivity, false negative results in Lewis negative phenotype (5-10%) and increased false positivity in the presence of obstructive jaundice (10-60%). CONCLUSIONS CA 19-9 is the most extensively studied and validated serum biomarker for the diagnosis of pancreatic cancer in symptomatic patients. CA 19-9 serum levels can provide important information with regards to prognosis, overall survival, and response to chemotherapy as well as predict post-operative recurrence. However, non-specific expression in several benign and malignant diseases, false negative results in Lewis negative genotype and an increased false positive results in the presence of obstructive jaundice severely limit the universal applicability of serum CA 19-9 levels in pancreatic cancer management.
Collapse
Affiliation(s)
- Maya Haus-Cohen
- Department of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 32000, Israel
| | | | | | | | | |
Collapse
|
23
|
Zakri AM, Ziegler A, Commandeur U, Fischer R, Torrance L. In vivo expression and binding activity of scFv-RWAV, which recognizes the coat protein of tomato leaf curl New Delhi virus (family Geminiviridae). Arch Virol 2012; 157:1291-9. [PMID: 22491815 DOI: 10.1007/s00705-012-1310-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 03/15/2012] [Indexed: 11/26/2022]
Abstract
Recombinant antibodies expressed in plants have the potential to interrupt virus infections by blocking essential stages of the infection cycle. Here, we show that the expression of a recombinant single-chain variable fragment (scFv) that recognizes the coat protein of tomato leaf curl New Delhi virus (ToLCNDV) in vitro can also bind to a recombinant coat protein in vivo in the reducing environment of the plant cytosol. The scFv and its target were both expressed as fluorescent protein fusions, one incorporating green fluorescent protein (GFP) and the other DsRed. We found that the incorporation of a nuclear localization signal into the scFv construct resulted in the nuclear import of the antibody-antigen complex, as shown by colocalization of the two fluorescent signals. This demonstrates that recombinant antibodies can be targeted to the nucleus and will bind to geminivirus coat proteins therein, allowing the virus infection cycle to be interrupted during its critical replicative phase.
Collapse
Affiliation(s)
- Adel M Zakri
- Institute for Molecular Biotechnology (Biology VII), RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany.
| | | | | | | | | |
Collapse
|
24
|
Karlsson AJ, Lim HK, Xu H, Rocco MA, Bratkowski MA, Ke A, DeLisa MP. Engineering antibody fitness and function using membrane-anchored display of correctly folded proteins. J Mol Biol 2011; 416:94-107. [PMID: 22197376 DOI: 10.1016/j.jmb.2011.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/06/2011] [Accepted: 12/09/2011] [Indexed: 10/14/2022]
Abstract
A hallmark of the bacterial twin-arginine translocation (Tat) pathway is its ability to export folded proteins. Here, we discovered that overexpressed Tat substrate proteins form two distinct, long-lived translocation intermediates that are readily detected by immunolabeling methods. Formation of the early translocation intermediate Ti-1, which exposes the N- and C-termini to the cytoplasm, did not require an intact Tat translocase, a functional Tat signal peptide, or a correctly folded substrate. In contrast, formation of the later translocation intermediate, Ti-2, which exhibits a bitopic topology with the N-terminus in the cytoplasm and C-terminus in the periplasm, was much more particular, requiring an intact translocase, a functional signal peptide, and a correctly folded substrate protein. The ability to directly detect Ti-2 intermediates was subsequently exploited for a new protein engineering technology called MAD-TRAP (membrane-anchored display for Tat-based recognition of associating proteins). Through the use of just two rounds of mutagenesis and screening with MAD-TRAP, the intracellular folding and antigen-binding activity of a human single-chain antibody fragment were simultaneously improved. This approach has several advantages for library screening, including the unique involvement of the Tat folding quality control mechanism that ensures only native-like proteins are displayed, thus eliminating poorly folded sequences from the screening process.
Collapse
Affiliation(s)
- Amy J Karlsson
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Romer T, Leonhardt H, Rothbauer U. Engineering antibodies and proteins for molecular in vivo imaging. Curr Opin Biotechnol 2011; 22:882-7. [DOI: 10.1016/j.copbio.2011.06.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/01/2011] [Accepted: 06/06/2011] [Indexed: 12/31/2022]
|
26
|
Schiefner A, Chatwell L, Körner J, Neumaier I, Colby DW, Volkmer R, Wittrup KD, Skerra A. A disulfide-free single-domain V(L) intrabody with blocking activity towards huntingtin reveals a novel mode of epitope recognition. J Mol Biol 2011; 414:337-55. [PMID: 21968397 DOI: 10.1016/j.jmb.2011.09.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/19/2011] [Accepted: 09/20/2011] [Indexed: 10/17/2022]
Abstract
We present the crystal structure and biophysical characterization of a human V(L) [variable domain immunoglobulin (Ig) light chain] single-domain intrabody that binds to the huntingtin (Htt) protein and has been engineered for antigen recognition in the absence of its intradomain disulfide bond, otherwise conserved in the Ig fold. Analytical ultracentrifugation demonstrated that the αHtt-V(L) 12.3 domain is a stable monomer under physiological conditions even at concentrations >20 μM. Using peptide SPOT arrays, we identified the minimal binding epitope to be EKLMKAFESLKSFQ, comprising the N-terminal residues 5-18 of Htt and including the first residue of the poly-Gln stretch. X-ray structural analysis of αHtt-V(L) both as apo protein and in the presence of the epitope peptide revealed several interesting insights: first, the role of mutations acquired during the combinatorial selection process of the αHtt-V(L) 12.3 domain-initially starting from a single-chain Fv fragment-that are responsible for its stability as an individually soluble Ig domain, also lacking the disulfide bridge, and second, a previously unknown mode of antigen recognition, revealing a novel paratope. The Htt epitope peptide adopts a purely α-helical structure in the complex with αHtt-V(L) and is bound at the base of the complementarity-determining regions (CDRs) at the concave β-sheet that normally gives rise to the interface between the V(L) domain and its paired V(H) (variable domain Ig heavy chain) domain, while only few interactions with CDR-L1 and CDR-L3 are formed. Notably, this noncanonical mode of antigen binding may occur more widely in the area of in vitro selected antibody fragments, including other Ig-like scaffolds, possibly even if a V(H) domain is present.
Collapse
Affiliation(s)
- André Schiefner
- Munich Center for Integrated Protein Science and Lehrstuhl für Biologische Chemie, Technische Universität München, 85350 Freising-Weihenstephan, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
State of the art in tumor antigen and biomarker discovery. Cancers (Basel) 2011; 3:2554-96. [PMID: 24212823 PMCID: PMC3757432 DOI: 10.3390/cancers3022554] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 05/24/2011] [Accepted: 05/27/2011] [Indexed: 12/22/2022] Open
Abstract
Our knowledge of tumor immunology has resulted in multiple approaches for the treatment of cancer. However, a gap between research of new tumors markers and development of immunotherapy has been established and very few markers exist that can be used for treatment. The challenge is now to discover new targets for active and passive immunotherapy. This review aims at describing recent advances in biomarkers and tumor antigen discovery in terms of antigen nature and localization, and is highlighting the most recent approaches used for their discovery including “omics” technology.
Collapse
|
28
|
Intrabody Expression in Mammalian Cells. ANTIBODY EXPRESSION AND PRODUCTION 2011. [PMCID: PMC7120103 DOI: 10.1007/978-94-007-1257-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
The intracellular expression of antibodies or antibody fragments (intrabodies) in different compartments of mammalian cells allows to block or modulate the function of endogenous molecules. Intrabodies can alter protein folding, protein-protein, protein-DNA, protein-RNA interactions and protein modification. They can induce a phenotypic knockout and work as neutralizing agents by direct binding to the target antigen, by diverting its intracellular traffic or by inhibiting its association with binding partners. They have been largely employed as research tools and are emerging as therapeutic molecules for the treatment of human diseases as viral pathologies, cancer and misfolding diseases. The fast growing bio-market of recombinant antibodies provides intrabodies with enhanced binding specificity, stability and solubility, together with lower immunogenicity, for their use in therapy. This chapter describes the crucial aspects required to express intrabodies in different intracellular compartments of mammalian cells, their various modes of action and gives an update on the applications of intrabodies in human diseases.
Collapse
|
29
|
Gal-Tanamy M, Zemel R, Bachmatov L, Jangra RK, Shapira A, Villanueva R, Yi M, Lemon SM, Benhar I, Tur-Kaspa R. Inhibition of protease-inhibitor-resistant hepatitis C virus replicons and infectious virus by intracellular intrabodies. Antiviral Res 2010; 88:95-106. [PMID: 20705106 PMCID: PMC4418563 DOI: 10.1016/j.antiviral.2010.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 06/16/2010] [Accepted: 08/04/2010] [Indexed: 12/15/2022]
Abstract
Hepatitis C virus (HCV) infection is a common cause of chronic liver disease and a serious threat to human health. The HCV NS3/4A serine protease is necessary for viral replication and innate immune evasion, and represents a well-validated target for specific antiviral therapy. We previously reported the isolation of single-chain antibodies (scFvs) that inhibit NS3/4A protease activity in vitro. Expressed intracellularly (intrabodies), these scFvs blocked NS3-mediated proliferation of NS3-transfected cells. Here we show that anti-NS3 scFvs suppress HCV RNA replication when expressed intracellularly in Huh7 hepatoma cells bearing either subgenomic or genome-length HCV RNA replicons. The expression of intrabodies directed against NS3 inhibited the autonomous amplification of HCV replicons resistant to small-molecule inhibitors of the NS3/4A protease, and replicons derived from different HCV genotypes. The combination of intrabodies and interferon-α had an additive inhibitory effect on RNA replication in the replicon model. Intrabody expression also inhibited production of infectious HCV in a cell culture system. The NS3 protease activity was inhibited by the intrabodies in NS3-expressing cells. In contrast, cell-free synthesis of HCV RNA by preformed replicase complexes was not inhibited by intrabodies, suggesting that the major mode of inhibition of viral replication is inhibition of NS3/4A protease activity and subsequent suppression of viral polyprotein processing.
Collapse
Affiliation(s)
- Meital Gal-Tanamy
- Molecular Hepatology Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv Israel
- Center for Hepatitis Research, Institute for Human Infections and Immunity, and the Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Romy Zemel
- Molecular Hepatology Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Larissa Bachmatov
- Molecular Hepatology Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Rohit K. Jangra
- Center for Hepatitis Research, Institute for Human Infections and Immunity, and the Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Assaf Shapira
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv Israel
| | - Rodrigo Villanueva
- Center for Hepatitis Research, Institute for Human Infections and Immunity, and the Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - MinKyung Yi
- Center for Hepatitis Research, Institute for Human Infections and Immunity, and the Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Stanley M. Lemon
- Center for Hepatitis Research, Institute for Human Infections and Immunity, and the Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv Israel
| | - Ran Tur-Kaspa
- Molecular Hepatology Research Laboratory, Felsenstein Medical Research Center, Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
- Department of Medicine D and Liver Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| |
Collapse
|
30
|
Grimm S, Lundberg E, Yu F, Shibasaki S, Vernet E, Skogs M, Nygren PÅ, Gräslund T. Selection and characterisation of affibody molecules inhibiting the interaction between Ras and Raf in vitro. N Biotechnol 2010; 27:766-73. [PMID: 20674812 DOI: 10.1016/j.nbt.2010.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 07/16/2010] [Accepted: 07/23/2010] [Indexed: 10/19/2022]
Abstract
Development of molecules with the ability to selectively inhibit particular protein-protein interactions is important in providing tools for understanding cell biology. In this work, we describe efforts to select small Ras- and Raf-specific three-helix bundle affibody binding proteins capable of inhibiting the interaction between H-Ras and Raf-1, from a combinatorial library displayed on bacteriophage. Target-specific variants with typically high nanomolar or low micromolar affinities (K(D)) could be selected successfully against both proteins, as shown by dot blot, ELISA and real-time biospecific interaction analyses. Affibody molecule variants selected against H-Ras were shown to bind epitopes overlapping each other at a site that differed from that at which H-Ras interacts with Raf-1. In contrast, an affibody molecule isolated during selection against Raf-1 was shown to effectively inhibit the interaction between H-Ras and Raf-1 in a dose-dependent manner. Possible intracellular applications of the selected affibody molecules are discussed.
Collapse
Affiliation(s)
- Sebastian Grimm
- Division of Molecular Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kvam E, Sierks MR, Shoemaker CB, Messer A. Physico-chemical determinants of soluble intrabody expression in mammalian cell cytoplasm. Protein Eng Des Sel 2010; 23:489-98. [PMID: 20378699 PMCID: PMC2865363 DOI: 10.1093/protein/gzq022] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 03/02/2010] [Accepted: 03/12/2010] [Indexed: 12/25/2022] Open
Abstract
Soluble antibody fragments are desirable not only as potential therapeutic and diagnostic agents for extracellular targets but also as 'intrabodies' for functional genomics, proteomics and gene therapy inside cells. However, antibody fragments are notoriously aggregation-prone when expressed intracellularly, due in part to unfavorable redox potential and macromolecular crowding in cell cytoplasm. Only a small proportion of intrabodies are soluble in cytoplasm and little is known about the sequence determinants that confer such stability. By comparing the cytoplasmic expression of several related human single-chain variable fragments and camelid V(HH)s in mammalian cells, we report that intrabody solubility is highly influenced by CDR content and is improved by an overall negative charge at cytoplasmic pH and reduced hydrophilicity. We hypothesize that ionic repulsion and weak hydrophobic interactions compensate, to different extents, for impaired disulfide bond formation in cytoplasm, thereby decreasing the risk for intrabody aggregation. As proof of principle, we demonstrate that the soluble expression of an aggregation-prone positively charged intrabody is modestly enhanced via cis or trans acidification using highly charged peptide tags (3XFLAG tag, SV40 NLS). These findings suggest that simple sequence analysis and electrostatic manipulation may aid in predicting and engineering solubility-enhanced intrabodies from antibody libraries for intracellular use.
Collapse
Affiliation(s)
- Erik Kvam
- New York State Department of Health, Wadsworth Center/ David Axelrod Institute, 120 New Scotland Ave., PO Box 22002, Albany, NY 12201-2002, USA
- Department of Biomedical Sciences, University at Albany, Albany, NY, USA
| | - Michael R. Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, AZ, USA
| | - Charles B. Shoemaker
- Department of Biomedical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | - Anne Messer
- New York State Department of Health, Wadsworth Center/ David Axelrod Institute, 120 New Scotland Ave., PO Box 22002, Albany, NY 12201-2002, USA
- Department of Biomedical Sciences, University at Albany, Albany, NY, USA
| |
Collapse
|
32
|
Schmidthals K, Helma J, Zolghadr K, Rothbauer U, Leonhardt H. Novel antibody derivatives for proteome and high-content analysis. Anal Bioanal Chem 2010; 397:3203-8. [PMID: 20372881 PMCID: PMC2911542 DOI: 10.1007/s00216-010-3657-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 03/13/2010] [Accepted: 03/13/2010] [Indexed: 11/28/2022]
Abstract
The understanding of cellular processes and their pathophysiological alterations requires comprehensive data on the abundance, distribution, modification, and interaction of all cellular components. On the one hand, artificially introduced fluorescent fusion proteins provide information about their distribution and dynamics in living cells but not about endogenous factors. On the other hand, antibodies can detect endogenous proteins, posttranslational modifications, and other cellular components but mostly in fixed and permeabilized cells. Here we highlight a new technology based on the antigen-binding domain of heavy-chain antibodies (V(H)H) from Camelidae. These extremely stable V(H)H domains can be produced in bacteria, coupled to matrices, and used for affinity purification and proteome studies. Alternatively, these V(H)H domains can be fused with fluorescent proteins and expressed in living cells. These fluorescent antigen-binding proteins called "chromobodies" can be used to detect and trace proteins and other cellular components in vivo. Chromobodies can, in principle, detect any antigenic structure, including posttranslational modifications, and thereby dramatically expand the quality and quantity of information that can be gathered in high-content analysis. Depending on the epitope chosen, chromobodies can also be used to modulate protein function in living cells.
Collapse
Affiliation(s)
- Katrin Schmidthals
- Department of Biology II, Ludwig Maximilians University, Grosshaderner Str. 2, 82152 Martinsried, Germany
| | | | | | | | | |
Collapse
|
33
|
Inhibition of human cytomegalovirus replication via peptide aptamers directed against the nonconventional nuclear localization signal of the essential viral replication factor pUL84. J Virol 2009; 83:11902-13. [PMID: 19740994 DOI: 10.1128/jvi.01378-09] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The UL84 open reading frame of human cytomegalovirus encodes an essential multifunctional regulatory protein that is thought to act in the nucleus as an initiator of lytic viral replication. Nuclear trafficking of pUL84 is facilitated by a complex nonconventional nuclear localization signal (NLS) that mediates its interaction with the cellular importin-alpha/beta pathway. Since binding of pUL84 to importin-alpha proteins mechanistically differs from that of cellular proteins containing a classical NLS, we assumed that specific interference with the nuclear import of pUL84 might be possible and that this could constitute a novel principle for antiviral therapy. In order to test this hypothesis, we employed peptide aptamer technology and isolated several peptide aptamers from a randomized peptide expression library that specifically bind with high affinity to the unconventional pUL84 NLS under intracellular conditions. Coimmunoprecipitation experiments confirmed these interactions in mammalian cells, and the antiviral potential of the identified peptide aptamers was determined using three independent experimental approaches. (i) Infection experiments with a recombinant human cytomegalovirus expressing green fluorescent protein demonstrated 50 to 60% decreased viral replication in primary human fibroblasts stably expressing pUL84-specific aptamers. (ii) A 50 to 70% reduction of viral plaque formation, as well as a 70 to 90% inhibition of virus release in the presence of pUL84-specific aptamers, was observed. (iii) Immunofluorescence analyses revealed a shift from an almost exclusively nuclear pUL84 staining pattern to a nucleocytoplasmic distribution upon coexpression of the identified molecules, indicating that interference with the nuclear import of pUL84 contributes to the observed antiviral activity of the identified pUL84-binding aptamer molecules.
Collapse
|
34
|
Seo MJ, Jeong KJ, Leysath CE, Ellington AD, Iverson BL, Georgiou G. Engineering antibody fragments to fold in the absence of disulfide bonds. Protein Sci 2009; 18:259-67. [PMID: 19177559 DOI: 10.1002/pro.31] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Disulfide bonds play a critical role in the stabilization of the immunoglobulin beta-sandwich sandwich. Under reducing conditions, such as those that prevail in the cytoplasm, disulfide bonds do not normally form and as a result most antibodies expressed in that compartment (intrabodies) accumulate in a misfolded and inactive state. We have developed a simple method for the quantitative isolation of antibody fragments that retain full activity under reducing conditions from large mutant libraries. In E. coli, inactivation of the cysteine oxidoreductase DsbA abolishes protein oxidation in the periplasm, which leads to the accumulation of scFvs and other disulfide-containing proteins in a reduced form. Libraries of mutant scFvs were tethered onto the inner membrane of dsbA cells and mutants that could bind fluorescently labeled antigen in the reducing periplasm were screened by Anchored Periplasmic Expression (APEx; Harvey et al., Proc Natl Acad Sci USA 2004;101:9193-9198.). Using this approach, we isolated scFv antibody variants that are fully active when expressed in the cytoplasm or when the four Cys residues that normally form disulfides are substituted by Ser residues.
Collapse
Affiliation(s)
- Min Jeong Seo
- Department of Chemical Engineering, University of Texas, Austin, 78712, USA
| | | | | | | | | | | |
Collapse
|
35
|
Loftus JC, Yang Z, Tran NL, Kloss J, Viso C, Berens ME, Lipinski CA. The Pyk2 FERM domain as a target to inhibit glioma migration. Mol Cancer Ther 2009; 8:1505-14. [PMID: 19509258 DOI: 10.1158/1535-7163.mct-08-1055] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The invasion of malignant glioma cells into the surrounding normal brain precludes effective clinical treatment. In this report, we investigated the role of the NH(2)-terminal FERM domain in the regulation of the promigratory function of Pyk2. We report that the substitution of residues that constitute a small cleft on the surface of the F3 module of the FERM domain do not significantly alter Pyk2 expression but result in the loss of Pyk2 phosphorylation. A monoclonal antibody, designated 12A10, specifically targeting the Pyk2 FERM domain was generated and recognizes an epitope located on the beta5C-alpha1C surface of the F3 module of the FERM domain. Amino acid substitutions in the F3 module that resulted in the loss of Pyk2 phosphorylation also inhibited the binding of 12A10, suggesting that the 12A10 epitope overlaps a site that plays a role in Pyk2 activity. Conjugation of 12A10 to a membrane transport peptide led to intracellular accumulation and inhibition of glioma cell migration in a concentration-dependent manner. A single chain Fv fragment of 12A10 was stable when expressed in the intracellular environment, interacted directly with Pyk2, reduced Pyk2 phosphorylation, and inhibited glioma cell migration in vitro. Stable intracellular expression of the 12A10 scFv significantly extended survival in a glioma xenograft model. Together, these data substantiate a central role for the FERM domain in regulation of Pyk2 activity and identify the F3 module as a novel target to inhibit Pyk2 activity and inhibit glioma progression.
Collapse
Affiliation(s)
- Joseph C Loftus
- Mayo Clinic Arizona, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Kvam E, Nannenga BL, Wang MS, Jia Z, Sierks MR, Messer A. Conformational targeting of fibrillar polyglutamine proteins in live cells escalates aggregation and cytotoxicity. PLoS One 2009; 4:e5727. [PMID: 19492089 PMCID: PMC2683928 DOI: 10.1371/journal.pone.0005727] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 04/29/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Misfolding- and aggregation-prone proteins underlying Parkinson's, Huntington's and Machado-Joseph diseases, namely alpha-synuclein, huntingtin, and ataxin-3 respectively, adopt numerous intracellular conformations during pathogenesis, including globular intermediates and insoluble amyloid-like fibrils. Such conformational diversity has complicated research into amyloid-associated intracellular dysfunction and neurodegeneration. To this end, recombinant single-chain Fv antibodies (scFvs) are compelling molecular tools that can be selected against specific protein conformations, and expressed inside cells as intrabodies, for investigative and therapeutic purposes. METHODOLOGY/PRINCIPAL FINDINGS Using atomic force microscopy (AFM) and live-cell fluorescence microscopy, we report that a human scFv selected against the fibrillar form of alpha-synuclein targets isomorphic conformations of misfolded polyglutamine proteins. When expressed in the cytoplasm of striatal cells, this conformation-specific intrabody co-localizes with intracellular aggregates of misfolded ataxin-3 and a pathological fragment of huntingtin, and enhances the aggregation propensity of both disease-linked polyglutamine proteins. Using this intrabody as a tool for modulating the kinetics of amyloidogenesis, we show that escalating aggregate formation of a pathologic huntingtin fragment is not cytoprotective in striatal cells, but rather heightens oxidative stress and cell death as detected by flow cytometry. Instead, cellular protection is achieved by suppressing aggregation using a previously described intrabody that binds to the amyloidogenic N-terminus of huntingtin. Analogous cytotoxic results are observed following conformational targeting of normal or polyglutamine-expanded human ataxin-3, which partially aggregate through non-polyglutamine domains. CONCLUSIONS/SIGNIFICANCE These findings validate that the rate of aggregation modulates polyglutamine-mediated intracellular dysfunction, and caution that molecules designed to specifically hasten aggregation may be detrimental as therapies for polyglutamine disorders. Moreover, our findings introduce a novel antibody-based tool that, as a consequence of its general specificity for fibrillar conformations and its ability to function intracellularly, offers broad research potential for a variety of human amyloid diseases.
Collapse
Affiliation(s)
- Erik Kvam
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, University at Albany, Albany, New York, United States of America
| | - Brent L. Nannenga
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Min S. Wang
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Zongjian Jia
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Michael R. Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Anne Messer
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, University at Albany, Albany, New York, United States of America
| |
Collapse
|
37
|
Meli G, Visintin M, Cannistraci I, Cattaneo A. Direct in Vivo Intracellular Selection of Conformation-sensitive Antibody Domains Targeting Alzheimer's Amyloid-β Oligomers. J Mol Biol 2009; 387:584-606. [DOI: 10.1016/j.jmb.2009.01.061] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 01/27/2009] [Accepted: 01/28/2009] [Indexed: 12/21/2022]
|
38
|
Cytoplasmic expression and specific binding of the VH/VL single domain intrabodies in transfected NIH3T3 cells. Exp Mol Pathol 2008; 86:51-6. [PMID: 19094983 DOI: 10.1016/j.yexmp.2008.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Accepted: 11/14/2008] [Indexed: 11/21/2022]
Abstract
Intracellularly expressed antibody fragments (intrabodies) have been utilized as powerful tools not only for clinical applications but also for the functional analysis of proteins inside the cell. Among several types of intrabodies developed so far, single domain types composed of only the variable regions (V(H) or V(L)) of antibodies are the smallest and thus the easiest to design. In this study, four types of single domain intrabodies were evaluated against a cytosolic protein, Wiskott-Aldrich syndrome protein (WASP), in gene-transfected NIH3T3 cells. These single domains were composed of the V(H) and V(L) region with or without their leader sequences. Although these single domains were expressed at similar levels in NIH3T3 cells, the binding activity to the cytosolic target was higher in the single domain constructs with leader sequences. These results suggest the usefulness of the single domain intrabody constructs to analyze the functional domains of cytosolic proteins in cells.
Collapse
|
39
|
Zhou C, Przedborski S. Intrabody and Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2008; 1792:634-42. [PMID: 18834937 DOI: 10.1016/j.bbadis.2008.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 08/22/2008] [Accepted: 09/03/2008] [Indexed: 10/21/2022]
Abstract
The intrabody technology has become a promising therapeutic avenue for a variety of incurable diseases. This technology is an intracellular application of gene-engineered antibodies, aimed at ablating the abnormal function of intracellular molecules. Parkinson's disease (PD) is a common neurodegenerative disease with no cure. Recent studies have explored possible intrabody applications against alpha-synuclein (alpha-syn), whose misfolding is believed to cause a familial form of PD. Here, we review the origin, production, and therapeutic mechanisms of intrabodies and the potential of intrabody protection against alpha-syn toxicity. Furthermore, we propose possible intrabody applications against leucine-rich repeat kinase 2 (LRRK2), whose mutations are the most frequent known cause of familial and sporadic PD.
Collapse
Affiliation(s)
- Chun Zhou
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
40
|
Adenovirus targeting to HLA-A1/MAGE-A1-positive tumor cells by fusing a single-chain T-cell receptor with minor capsid protein IX. Gene Ther 2008; 15:978-89. [PMID: 18323790 DOI: 10.1038/gt.2008.26] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenovirus vectors have great potential in cancer gene therapy. Targeting of cancer-testis (CT) antigens, which are specifically presented at the surface of tumor cells by human leukocyte antigen (HLA) class I molecules, is an attractive option. In this study, a single-chain T-cell receptor (scTCR) directed against the CT antigen melanoma-associated antigen (MAGE)-A1 in complex with the HLA class I molecule of haplotype HLA-A1 is fused with the C terminus of the adenovirus minor capsid protein IX. Propagation of a protein-IX (pIX)-gene-deleted human adenovirus 5 (HAdV-5) vector on cells that constitutively express the pIXscTCR fusion protein yielded viral particles with the pIXscTCR fusion protein incorporated in their capsid. Generated particles specifically transduced melanoma cell lines expressing the HLA-A1/MAGE-A1 target complex with at least 10-fold higher efficiency than control viruses. Whereas loading of HLA-A1-positive cells with MAGE-A1 peptides leads to enhanced transduction of the cells, the efficiency of virus transduction is strongly reduced if the HLA-A1 molecules are not accessible at the target cell. Taken together, these data provide proof of principle that pIXscTCR fusions can be used to target HAdV-5 vectors to tumor cells expressing intracellular CT antigens.
Collapse
|
41
|
Abstract
Combining exquisite specificity and high antigen-binding affinity, intrabodies have been used as a biotechnological tool to interrupt, modulate, or define the functions of a wide range of target antigens at the posttranslational level. An intrabody is an antibody that has been designed to be expressed intracellularly and can be directed to a specific target antigen present in various subcellular locations including the cytosol, nucleus, endoplasmic reticulum (ER), mitochondria, peroxisomes, plasma membrane and trans-Golgi network (TGN) through in frame fusion with intracellular trafficking/localization peptide sequences. Although intrabodies can be expressed in different forms, the most commonly used format is a singlechain antibody (scFv Ab) created by joining the antigen-binding variable domains of heavy and light chain with an interchain linker (ICL), most often the 15 amino acid linker (GGGGS)(3) between the variable heavy (VH) and variable light (VL) chains. Intrabodies have been used in research of cancer, HIV, autoimmune disease, neurodegenerative disease, and transplantation. Clinical application of intrabodies has mainly been hindered by the availability of robust gene delivery system(s) including target cell directed gene delivery. This review will discuss several methods of intrabody selection, different strategies of cellular targeting, and recent successful examples of intrabody applications. Taking advantage of the high specificity and affinity of an antibody for its antigen, and of the virtually unlimited diversity of antigen-binding variable domains available for molecular targeting, intrabody techniques are emerging as promising tools to generate phenotypic knockouts, to manipulate biological processes, and to obtain a more thorough understanding of functional genomics.
Collapse
|
42
|
A protein silencing switch by ligand-induced proteasome-targeting intrabodies. J Mol Biol 2007; 374:641-54. [PMID: 17950312 DOI: 10.1016/j.jmb.2007.09.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 09/14/2007] [Accepted: 09/18/2007] [Indexed: 11/24/2022]
Abstract
The selective knock-down of cellular proteins has proven useful for in vivo studies of protein function and RNAi methods are readily available for this purpose. However, interfering directly at the protein level may have distinct advantages, with the intracellular targeting of antibodies (intrabodies) representing an attractive option, although not a general one. We demonstrate a novel, general strategy named suicide (or silencing) intrabody technology (SIT), based on the inducible degradation of intrabodies, which are equipped with proteasome-targeting sequences and thus converted into suicide intrabodies. We show that suicide intrabodies are able to redirect the target cellular proteins upon stimulus administration to the proteolytic machinery, thus resulting in selective protein knock-down. Remarkably, suicide intrabody acts in a catalytic fashion. SIT is a ligand-inducible strategy, potentially applicable to any protein of interest and does not require the engineering of cellular proteolytic enzymes. SIT represents a general approach to confer "neutralizing" properties to any intrabody, a valuable feature, given the present impossibility to select a priori intrinsically neutralizing antibodies. This knock-down strategy, together with available methods to isolate functional intrabodies, should allow the large-scale investigation of intracellular protein networks.
Collapse
|
43
|
Contreras-Martínez LM, DeLisa MP. Intracellular Ribosome Display Via SecM Translation Arrest as a Selection for Antibodies with Enhanced Cytosolic Stability. J Mol Biol 2007; 372:513-24. [PMID: 17669427 DOI: 10.1016/j.jmb.2007.06.070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2007] [Revised: 06/14/2007] [Accepted: 06/26/2007] [Indexed: 10/23/2022]
Abstract
Ribosome display is a powerful approach for affinity and stability maturation of recombinant antibodies. However, since ribosome display is performed entirely in vitro, there are several limitations to this approach including technical challenges associated with: (i) efficiently expressing and stalling antibodies on ribosomes using cell-free translation mixtures; and (ii) folding of antibodies in buffers where the concentration and composition of factors varies from that found in the intracellular milieu. We have developed a novel method for intracellular ribosome display that takes advantage of the recently discovered Escherichia coli SecM translation arrest mechanism. Specifically, we provide the first evidence that the encoding mRNA of SecM-stalled heterologous proteins remains stably attached to ribosomes, thereby enabling creation of stalled antibody-ribosome-mRNA (ARM) complexes entirely inside of living cells. Since ARM complexes faithfully maintain a genotype-phenotype link between the arrested antibody and its encoding mRNA, we demonstrate that this method is ideally suited for isolating stability-enhanced single-chain variable fragment (scFv) antibodies that are efficiently folded and functional in the bacterial cytoplasm.
Collapse
|
44
|
Gilch S, Kehler C, Schätzl HM. Peptide Aptamers Expressed in the Secretory Pathway Interfere with Cellular PrPSc Formation. J Mol Biol 2007; 371:362-73. [PMID: 17574575 DOI: 10.1016/j.jmb.2007.05.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Revised: 05/11/2007] [Accepted: 05/17/2007] [Indexed: 11/22/2022]
Abstract
Prion diseases are rare and obligatory fatal neurodegenerative disorders caused by the accumulation of a misfolded isoform (PrPSc) of the host-encoded prion protein (PrPc). Prophylactic and therapeutic regimens against prion diseases are very limited. To extend such strategies we selected peptide aptamers binding to PrP from a combinatorial peptide library presented on the Escherichia coli thioredoxin A (trxA) protein as a scaffold. In a yeast two-hybrid screen employing full-length murine PrP (aa 23-231) as a bait we identified three peptide aptamers that reproducibly bind to PrP. Treatment of prion-infected cells with recombinantly expressed aptamers added to the culture medium abolished PrPSc conversion with an IC50 between 350 and 700 nM. For expression in eukaryotic cells, peptide aptamers were fused to an N-terminal signal peptide for entry of the secretory pathway. The C terminus was modified by a glycosyl-phosphatidyl-inositol-(GPI) anchoring signal, a KDEL retention motif and the transmembrane and cytosolic domain of LAMP-I, respectively. These peptide aptamers retained their binding properties to PrPc and, depending on peptide sequence and C-terminal modification, interfered with endogenous PrPSc conversion upon expression in prion-infected cells. Notably, infection of cell cultures could be prevented by expression of KDEL peptide aptamers. For the first time, we show that trxA-based peptide aptamers can be targeted to the secretory pathway, thereby not losing the affinity for their target protein. Beside their inhibitory effect on prion conversion, these molecules could be used as fundament for rational drug design.
Collapse
Affiliation(s)
- Sabine Gilch
- Institute of Virology, Prion Research Group, Technical University Munich, Trogerstr. 30, 81675 Munich, Germany
| | | | | |
Collapse
|
45
|
Doorbar J, Griffin H. Intrabody strategies for the treatment of human papillomavirus-associated disease. Expert Opin Biol Ther 2007; 7:677-89. [PMID: 17477805 DOI: 10.1517/14712598.7.5.677] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human papillomaviruses (HPVs) are associated with a variety of epithelial lesions, including benign genital warts and cervical intraepithelial neoplasia. Both cause significant morbidity in the general population, with cervical intraepithelial neoplasia progressing to cervical cancer in a subset of women who cannot resolve their infection. At present, there are no antiviral agents for the treatment of genital HPV infections, with many lesions requiring surgical intervention. Although other approaches are available for the treatment of genital warts, HPV infection cannot usually be cured and lesion recurrence is often a problem. A growing understanding of the molecular biology of HPV infection has identified several viral protein functions that may serve as drug targets. Among these are the HPV E1 and E2 proteins, which are necessary for viral genome replication and partitioning, and the E6 and E7 proteins, which are necessary for cell proliferation and apoptotic inhibition. With the exception of E1, these proteins lack enzymatic activity and achieve their effects by interacting with cellular proteins. Protein-protein interactions are in general quite difficult to inhibit using conventional small molecule drugs, but are amenable to inhibition using intracellular antibodies or intrabodies, which bind the viral proteins and sterically inhibit their association with cellular partners. The lack of homology between viral and cellular proteins, and the fact that HPV infections can be treated topically, makes them particularly well suited to the intrabody approach. This review covers the various strategies that are being considered for the treatment of HPV infections and the different intrabody formats that have been used to inhibit HPV function in model systems. The clinical utility of the approach is considered alongside the general difficulties of using protein molecules as intracellular therapeutics.
Collapse
Affiliation(s)
- John Doorbar
- National Institute for Medical Research, Division of Virology, The Ridgeway, Mill Hill, London, NW7 1AA, UK.
| | | |
Collapse
|
46
|
Fang CY, Chiang CY, Pan YR, Tse KP, Chang YS, Chang HY. Modulation of Epstein-Barr virus latent membrane protein 1 activity by intrabodies. Intervirology 2007; 50:254-63. [PMID: 17460414 DOI: 10.1159/000101996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 02/14/2007] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE The Epstein-Barr virus (EBV) has been implicated in the development of many human neoplasias including B lymphoma and nasopharyngeal carcinoma. EBV latent membrane protein 1 (LMP1) is essential to virus-induced B cell immortalization and the downregulation of cell adhesion molecules that increases cell motility. Therefore, identifying LMP1 activity modulation methods may lead to the development of new therapies for LMP1-positive tumors. METHODS This study uses a phage display single-chain variable fragments (scFvs) library to screen recombinant antibodies specific to the LMP1 C terminal region. A total of 45 individual clones were obtained, and these scFvs were cloned as intrabodies and transfected into LMP1-positive cells. RESULTS One of the scFv clones, designated H3, was capable of reducing LMP1-mediated NF-kappaB activation in HEK293 cells. Immunofluorescence and co-immunoprecipitation studies show that scFv H3 could interact with LMP1 in vivo. In addition, expression of scFv H3 intrabody could reduce cell motility in MDCK-LMP1 cells in the transwell migration assay. CONCLUSION These data indicate that scFv H3 intrabody can inhibit LMP1 functions in epithelial cells and may be useful for attenuating the LMP1 function in LMP1-positive tumors.
Collapse
Affiliation(s)
- Chih Yeu Fang
- Institute of Molecular Medicine, National Tsing Hua University, Hsin-Chu, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
47
|
Vellinga J, De Vrij J, Myhre S, Uil T, Martineau P, Lindholm L, Hoeben RC. Efficient incorporation of a functional hyper-stable single-chain antibody fragment protein-IX fusion in the adenovirus capsid. Gene Ther 2007; 14:664-70. [PMID: 17268536 PMCID: PMC2233715 DOI: 10.1038/sj.gt.3302908] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 12/03/2006] [Accepted: 12/03/2006] [Indexed: 11/08/2022]
Abstract
Recombinant adenoviruses are frequently used as gene transfer vehicles for therapeutic gene delivery. Strategies to amend their tropism include the incorporation of polypeptides with high affinity for cellular receptors. Single-chain antibodies have a great potential to achieve such cell type specificity. In this study, we evaluated the efficiency of incorporation of a single-chain antibody fused with the adenovirus minor capsid protein IX in the capsid of adenovirus type 5 vectors. To this end, the codons for the single-chain antibody fragments (scFv) 13R4 were fused with those encoding of pIX via a 75-Angstrom spacer sequence. The 13R4 is a hyper-stable single-chain antibody directed against beta-galactosidase, which was selected for its capacity to fold correctly in a reducing environment such as the cytoplasm. A lentiviral vector was used to stably express the pIX.flag.75.13R4.MYC.HIS fusion gene in 911 helper cells. Upon propagation of pIX-gene deleted human adenovirus-5 vectors on these cells, the pIX-fusion protein was efficiently incorporated in the capsid. Here, the 13R4 scFv was functional as was evident from its capacity to bind its ligand beta-galactosidase. These data demonstrate that the minor capsid protein IX can be used as an anchor for incorporation of single-chain antibodies in the capsids of adenovirus vectors.
Collapse
Affiliation(s)
- Jort Vellinga
- Department of Molecular Cell Biology,
Leiden University Medical CenterDepartment of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg
20, 2300 RC Leiden,The Netherlands,FR
| | - Jeroen De Vrij
- Department of Molecular Cell Biology,
Leiden University Medical CenterDepartment of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg
20, 2300 RC Leiden,The Netherlands,FR
| | | | - Taco Uil
- Department of Molecular Cell Biology,
Leiden University Medical CenterDepartment of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg
20, 2300 RC Leiden,The Netherlands,FR
| | - Pierre Martineau
- CPBS, Centre de pharmacologie et biotechnologies pour la santé
CNRS : UMR5160BIO-RADUniversité Montpellier IUniversité Montpellier II - Sciences et Techniques du LanguedocFaculté de Pharmacie - bat. I
15 Av Charles Flahault - BP 14491
34093 MONTPELLIER CEDEX 5,FR
| | | | - Rob C. Hoeben
- Department of Molecular Cell Biology,
Leiden University Medical CenterDepartment of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg
20, 2300 RC Leiden,The Netherlands,FR
| |
Collapse
|
48
|
Hesse A, Kosmides D, Kontermann RE, Nettelbeck DM. Tropism modification of adenovirus vectors by peptide ligand insertion into various positions of the adenovirus serotype 41 short-fiber knob domain. J Virol 2006; 81:2688-99. [PMID: 17192304 PMCID: PMC1865974 DOI: 10.1128/jvi.02722-06] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Recombinant adenoviruses have emerged as promising agents in therapeutic gene transfer, genetic vaccination, and viral oncolysis. Therapeutic applications of adenoviruses, however, would benefit substantially from targeted virus cell entry, for example, into cancer or immune cells, as opposed to the broad tropism that adenoviruses naturally possess. Such tropism modification of adenoviruses requires the deletion of their natural cell binding properties and the incorporation of cell binding ligands. The short fibers of subgroup F adenoviruses have recently been suggested as a tool for genetic adenovirus detargeting based on the reduced infectivity of corresponding adenovectors with chimeric fibers in vitro and in vivo. The goal of our study was to determine functional insertion sites for peptide ligands in the adenovirus serotype 41 (Ad41) short fiber knob. With a model peptide, CDCRGDCFC, we could demonstrate that ligand incorporation into three of five analyzed loops of the knob, namely, EG, HI, and IJ, is feasible without a loss of fiber trimerization. The resulting adenovectors showed enhanced infectivity for various cell types, which was superior to that of viruses with the same peptide fused to the fiber C terminus. Strategies to further augment gene transfer efficacy by extension of the fiber shaft, insertion of tandem copies of the ligand peptide, or extension of the ligand-flanking linkers failed, indicating that precise ligand positioning is pivotal. Our study establishes that internal ligand incorporation into a short-shafted adenovirus fiber is feasible and suggests the Ad41 short fiber with ligand insertion into the top (IJ loop) or side (EG and HI loops) of the knob domain as a novel platform for genetic targeting of therapeutic adenoviruses.
Collapse
Affiliation(s)
- Andrea Hesse
- Virotherapy Lab, Department of Dermatology, University Hospital Erlasngen, Hartmannstrasse 14, Erlangen, Germany
| | | | | | | |
Collapse
|
49
|
Vaccaro P, Pavoni E, Monteriù G, Andrea P, Felici F, Minenkova O. Efficient display of scFv antibodies on bacteriophage lambda. J Immunol Methods 2006; 310:149-58. [PMID: 16497320 DOI: 10.1016/j.jim.2006.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 10/17/2005] [Accepted: 01/11/2006] [Indexed: 10/25/2022]
Abstract
In the present work we demonstrate the efficient display of functional scFv antibodies on the bacteriophage lambda capsid. A single-chain (scFv) anti-CEA antibody gene was cloned in two different vectors to obtain fusion of the scFv antibody to the N- or C-terminus of the bacteriophage lambda capsid protein D (gpD). Lambda bacteriophage assembly occurs in the reducing environment of the cytoplasm; despite this the lambda-displayed anti-CEA antibody fragments retain the capacity to recognize the antigen, indicating correct single-chain antibody folding. Efficient production of functional scFv exposed on lambda capsid with viable antigen binding specificity allowed us to study and compare the capacity of display, the stability of recombinant antibody expression and the assembly efficiency of bacteriophage particles decorated with recombinant antibody fused to the amino- or carboxy-terminus of lambda D protein.
Collapse
|
50
|
Griffin H, Elston R, Jackson D, Ansell K, Coleman M, Winter G, Doorbar J. Inhibition of Papillomavirus Protein Function in Cervical Cancer Cells by Intrabody Targeting. J Mol Biol 2006; 355:360-78. [PMID: 16324714 DOI: 10.1016/j.jmb.2005.10.077] [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] [Received: 08/03/2005] [Revised: 10/19/2005] [Accepted: 10/28/2005] [Indexed: 01/08/2023]
Abstract
Papillomaviruses (HPVs) are a major cause of human disease, and are responsible for approximately half a million cases of cervical cancer each year. HPVs also cause genital warts, and are the most common sexually transmitted disease in many countries. Despite their importance, there are currently no specific antivirals that are active against HPVs. Papillomavirus protein function is mediated largely by protein-protein interactions, which are difficult to inhibit using conventional approaches. To circumvent these problems, we have prepared an scFv library, and have used this to isolate high-affinity binding molecules that may stearically hinder the association of E6 with p53 and prevent E6-mediated p53 degradation in cervical cancer cells. One of the molecules isolated from the library (GTE6-1), had an affinity for 16E6 of 60nM, and bound within the first zinc finger of the protein. GTE6-1 was able to associate with non-denatured E6 following expression in mammalian cells and could inhibit E6-mediated p53 degradation in in vitro assays. E6-mediated p53 degradation is essential for the continuous growth of cervical cancer cells caused by HPV16. To examine the potential of GTE6-1 as an inhibitor of E6 function in such cells, the molecule was expressed in scFv, diabody and triabody formats in a number of cell lines that are driven to proliferate by the HPV16 oncogenes E6 and E7, including the cervical cancer cell line SiHa. In contrast to small E6-binding peptides containing the ELLG E6-binding motif, GTE6-1 expression lead to changes in nuclear structure, the appearance of apoptosis markers, and an elevation in the levels of p53. No effects were seen with a control scFv molecule, or when GTE6-1 was expressed in cells that are driven to proliferate by simian virus 40 (SV40) T-antigen. Given the accessibility of HPV-associated lesions to topical therapy, our results suggest that large interfering molecules such as intrabodies may be useful inhibitors of viral protein-protein interactions and be particularly appropriate for the treatment of HPV-associated disease.
Collapse
Affiliation(s)
- Heather Griffin
- Division of Virology, National Institute for Medical Research, London NW7 1AA, UK
| | | | | | | | | | | | | |
Collapse
|