1
|
Durans ADM, Napoleão-Pêgo P, Reis FCG, Dias ER, Machado LESF, Lechuga GC, Junqueira ACV, De-Simone SG, Provance DW. Chagas Disease Diagnosis with Trypanosoma cruzi-Exclusive Epitopes in GFP. Vaccines (Basel) 2024; 12:1029. [PMID: 39340059 PMCID: PMC11435546 DOI: 10.3390/vaccines12091029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 09/30/2024] Open
Abstract
Serological tests are critical tools in the fight against infectious disease. They detect antibodies produced during an adaptive immune response against a pathogen with an immunological reagent, whose antibody binding characteristics define the specificity and sensitivity of the assay. While pathogen proteins have conveniently served as reagents, their performance is limited by the natural grouping of specific and non-specific antibody binding sites, epitopes. An attractive solution is to build synthetic proteins that only contains pathogen-specific epitopes, which could theoretically reach 100% specificity. However, the genesis of de novo proteins remains a challenge. To address the uncertainty of producing a synthetic protein, we have repurposed the beta barrel of fluorescent proteins into a receptacle that can receive several epitope sequences without compromising its ability to be expressed. Here, two versions of a multiepitope protein were built using the receptacle that differ by their grouping of epitopes specific to the parasite Trypanosoma cruzi, the causative agent for Chagas disease. An evaluation of their performance as the capture reagent in ELISAs showed near-complete agreement with recommended diagnostic protocols. The results suggest that a single assay could be developed for the diagnosis of Chagas disease and that this approach could be applied to other diseases.
Collapse
Affiliation(s)
- Andressa da M Durans
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Epidemiology and Molecular Systematics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Interdisciplinary Laboratory of Medical Researchers, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Paloma Napoleão-Pêgo
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Epidemiology and Molecular Systematics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Flavia C G Reis
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Evandro R Dias
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Laboratory on Parasitic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Luciana E S F Machado
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Department of Genetics and Biology Evolution, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Guilherme C Lechuga
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Epidemiology and Molecular Systematics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Angela C V Junqueira
- Laboratory on Parasitic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Salvatore G De-Simone
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Epidemiology and Molecular Systematics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Program of Post-Graduation on Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, Brazil
| | - David W Provance
- Center for Technological Development in Health, National Institute of Science and Technology for Innovation in Neglected Population Diseases, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
- Epidemiology and Molecular Systematics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
2
|
David TI, Pestov NB, Korneenko TV, Barlev NA. Non-Immunoglobulin Synthetic Binding Proteins for Oncology. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1232-1247. [PMID: 37770391 DOI: 10.1134/s0006297923090043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 09/30/2023]
Abstract
Extensive application of technologies like phage display in screening peptide and protein combinatorial libraries has not only facilitated creation of new recombinant antibodies but has also significantly enriched repertoire of the protein binders that have polypeptide scaffolds without homology to immunoglobulins. These innovative synthetic binding protein (SBP) platforms have grown in number and now encompass monobodies/adnectins, DARPins, lipocalins/anticalins, and a variety of miniproteins such as affibodies and knottins, among others. They serve as versatile modules for developing complex affinity tools that hold promise in both diagnostic and therapeutic settings. An optimal scaffold typically has low molecular weight, minimal immunogenicity, and demonstrates resistance against various challenging conditions, including proteolysis - making it potentially suitable for peroral administration. Retaining functionality under reducing intracellular milieu is also advantageous. However, paramount to its functionality is the scaffold's ability to tolerate mutations across numerous positions, allowing for the formation of a sufficiently large target binding region. This is achieved through the library construction, screening, and subsequent expression in an appropriate system. Scaffolds that exhibit high thermodynamic stability are especially coveted by the developers of new SBPs. These are steadily making their way into clinical settings, notably as antagonists of oncoproteins in signaling pathways. This review surveys the diverse landscape of SBPs, placing particular emphasis on the inhibitors targeting the oncoprotein KRAS, and highlights groundbreaking opportunities for SBPs in oncology.
Collapse
Affiliation(s)
- Temitope I David
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
- Laboratory of Molecular Oncology, Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russia
| | - Nikolay B Pestov
- Institute of Biomedical Chemistry, Moscow, 119121, Russia.
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences, Moscow, 108819, Russia
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Tatyana V Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Nikolai A Barlev
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences, Moscow, 108819, Russia
- Institute of Cytology Russian Academy of Sciences, St.-Petersburg, 194064, Russia
- School of Medicine, Nazarbayev University, Astana, 010000, Kazakhstan
| |
Collapse
|
3
|
Wang Y, Wang X, Gao T, Lou C, Wang H, Liu Y, Cao A. Folding of Flexible Protein Fragments and Design of Nanoparticle-Based Artificial Antibody Targeting Lysozyme. J Phys Chem B 2022; 126:5045-5054. [PMID: 35763806 DOI: 10.1021/acs.jpcb.2c03200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It is generally believed that a protein's sequence solely determines its native structure, but how the long- and short-range interactions jointly determine the native structure/conformation of the protein or every local fragment of the protein is still not fully understood. Since most protein fragments are unstructured on their own, direct observation of the folding of flexible protein fragments is very difficult. Interestingly, we show that it is possible to graft the complementary-determining regions (CDRs) of antibodies onto the surface of a gold nanoparticle (AuNP) to create AuNP-based artificial antibodies (denoted as Goldbodies), such as an antilysozyme Goldbody. Goldbodies can specifically recognize the corresponding antigens like the original natural antibodies do, but direct structural evidence for the refolding or restoration of native conformation of the grafted CDRs on AuNPs is still missing and in high demand. Herein we design a new Goldbody that targets an epitope on the lysozyme different from that of the previous antilysozyme Goldbody, and the one circle of helix in the CDR makes it possible to distinguish the unfolded conformation of the free CDR and its folded conformation on AuNPs by circular dichroism (CD) spectroscopy. The refolding of flexible protein fragments on NPs provides unique evidence and inspiration for understanding the fundamental principles of protein folding.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Xinping Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Tiange Gao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Chenxi Lou
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Yuanfang Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China.,Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
4
|
Velappan N, Micheva-Viteva S, Adikari SH, Waldo GS, Lillo AM, Bradbury ARM. Selection and verification of antibodies against the cytoplasmic domain of M2 of influenza, a transmembrane protein. MAbs 2021; 12:1843754. [PMID: 33206590 PMCID: PMC7678940 DOI: 10.1080/19420862.2020.1843754] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Interactions between the cytoplasmic domains of viral transmembrane proteins and host machinery often determine the outcome of viral infection. The M2 protein of influenza A has been identified as a key player in autophagy-mediated viral replication. Here, we describe the engineering and validation of an antibody specific for the cytoplasmic domain of the M2 protein. Through phage and yeast display selection techniques, we obtained an antibody that recognizes: 1) the M2 cytoplasmic domain purified from bacterial inclusion bodies and refolded, 2) full-length M2 recombinant protein expressed in mammalian cells, and 3) native M2 protein in influenza A infected cells. This antibody can serve as a molecular tool to enhance our knowledge of protein–protein interactions between influenza A virus and the host cell machinery. We anticipate the methods described herein will further the development of antibodies specific to the cytoplasmic domains of transmembrane proteins.
Collapse
Affiliation(s)
- Nileena Velappan
- Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory , Los Alamos, NM, USA
| | - Sofiya Micheva-Viteva
- Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory , Los Alamos, NM, USA
| | - Samantha H Adikari
- Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory , Los Alamos, NM, USA
| | - Geoffrey S Waldo
- Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory , Los Alamos, NM, USA
| | - Antonietta M Lillo
- Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory , Los Alamos, NM, USA
| | | |
Collapse
|
5
|
Gomes LR, Durans AM, Napoleão-Pêgo P, Waterman JA, Freitas MS, De Sá NBR, Pereira LV, Furtado JS, Aquino RG, Machado MCR, Fintelman-Rodrigues N, Souza TML, Morel CM, Provance DW, De-Simone SG. Multiepitope Proteins for the Differential Detection of IgG Antibodies against RBD of the Spike Protein and Non-RBD Regions of SARS-CoV-2. Vaccines (Basel) 2021; 9:986. [PMID: 34579223 PMCID: PMC8473315 DOI: 10.3390/vaccines9090986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/04/2021] [Accepted: 08/28/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has exposed the extent of global connectivity and collective vulnerability to emerging diseases. From its suspected origins in Wuhan, China, it spread to all corners of the world in a matter of months. The absence of high-performance, rapid diagnostic methods that could identify asymptomatic carriers contributed to its worldwide transmission. Serological tests offer numerous benefits compared to other assay platforms to screen large populations. First-generation assays contain targets that represent proteins from SARS-CoV-2. While they could be quickly produced, each actually has a mixture of specific and non-specific epitopes that vary in their reactivity for antibodies. To generate the next generation of the assay, epitopes were identified in three SARS-Cov-2 proteins (S, N, and Orf3a) by SPOT synthesis analysis. After their similarity to other pathogen sequences was analyzed, 11 epitopes outside of the receptor-binding domain (RBD) of the spike protein that showed high reactivity and uniqueness to the virus. These were incorporated into a ß-barrel protein core to create a highly chimeric protein. Another de novo protein was designed that contained only epitopes in the RBD. In-house ELISAs suggest that both multiepitope proteins can serve as targets for high-performance diagnostic tests. Our approach to bioengineer chimeric proteins is highly amenable to other pathogens and immunological uses.
Collapse
Affiliation(s)
- Larissa R. Gomes
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
| | - Andressa M. Durans
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
- Interdisciplinary Medical Research Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Paloma Napoleão-Pêgo
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
| | - Jessica A. Waterman
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
| | - Mariana S. Freitas
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
| | - Nathalia B. R. De Sá
- AIDS & Molecular Immunology Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil;
| | - Lilian V. Pereira
- Angra dos Reis Health Department, Angra dos Reis 23906-10, Brazil; (L.V.P.); (J.S.F.); (R.G.A.)
| | - Jéssica S. Furtado
- Angra dos Reis Health Department, Angra dos Reis 23906-10, Brazil; (L.V.P.); (J.S.F.); (R.G.A.)
| | - Romário G. Aquino
- Angra dos Reis Health Department, Angra dos Reis 23906-10, Brazil; (L.V.P.); (J.S.F.); (R.G.A.)
| | | | - Natalia Fintelman-Rodrigues
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
- Immunopharmacology Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Thiago M. L. Souza
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
- Immunopharmacology Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Carlos M. Morel
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
| | - David W. Provance
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
- Interdisciplinary Medical Research Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Salvatore G. De-Simone
- FIOCRUZ, Center of Technological Development in Health (CDTS)/National Institute of Science and Technology for Innovation on Neglected Population Diseases (INCT-IDPN), Rio de Janeiro 21040-900, Brazil; (L.R.G.); (A.M.D.); (P.N.-P.); (J.A.W.); (M.S.F.); (N.F.-R.); (T.M.L.S.); (C.M.M.); (D.W.P.)
- Department of Cellular and Molecular Biology, Biology Institute, Federal Fluminense University, Niterói 24020-141, Brazil
| |
Collapse
|
6
|
Napoleão-Pêgo P, Carneiro FRG, Durans AM, Gomes LR, Morel CM, Provance DW, De-Simone SG. Performance assessment of a multi-epitope chimeric antigen for the serological diagnosis of acute Mayaro fever. Sci Rep 2021; 11:15374. [PMID: 34321560 PMCID: PMC8319364 DOI: 10.1038/s41598-021-94817-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023] Open
Abstract
Mayaro virus (MAYV), which causes mayaro fever, is endemic to limited regions of South America that may expand due to the possible involvement of Aedes spp. mosquitoes in its transmission. Its effective control will require the accurate identification of infected individuals, which has been restricted to nucleic acid-based tests due to similarities with other emerging members of the Alphavirus genus of the Togaviridae family; both in structure and clinical symptoms. Serological tests have a more significant potential to expand testing at a reasonable cost, and their performance primarily reflects that of the antigen utilized to capture pathogen-specific antibodies. Here, we describe the assembly of a synthetic gene encoding multiple copies of antigenic determinants mapped from the nsP1, nsP2, E1, and E2 proteins of MAYV that readily expressed as a stable chimeric protein in bacteria. Its serological performance as the target in ELISAs revealed a high accuracy for detecting anti-MAYV IgM antibodies. No cross-reactivity was observed with serum from seropositive individuals for dengue, chikungunya, yellow fever, Zika, and other infectious diseases as well as healthy individuals. Our data suggest that this bioengineered antigen could be used to develop high-performance serological tests for MAYV infections.
Collapse
Affiliation(s)
- Paloma Napoleão-Pêgo
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Flávia R G Carneiro
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil.,Laboratory of Interdisplinary Medical Research (LIPMED), Oswaldo Cruz Institute (IOC), FIOCRUZ, Brazil Av 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Andressa M Durans
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil.,Laboratory of Interdisplinary Medical Research (LIPMED), Oswaldo Cruz Institute (IOC), FIOCRUZ, Brazil Av 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Larissa R Gomes
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Carlos M Morel
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil
| | - David W Provance
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil.,Laboratory of Interdisplinary Medical Research (LIPMED), Oswaldo Cruz Institute (IOC), FIOCRUZ, Brazil Av 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Salvatore G De-Simone
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil. .,Biology Institute, Federal Fluminense University, Outeiro de São Joao Batista S/N, Niterói, RJ, 24020-141, Brazil.
| |
Collapse
|
7
|
Chee SMQ, Wongsantichon J, Yi LS, Sana B, Frosi Y, Robinson RC, Ghadessy FJ. Functional display of bioactive peptides on the vGFP scaffold. Sci Rep 2021; 11:10127. [PMID: 33980885 PMCID: PMC8115314 DOI: 10.1038/s41598-021-89421-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/27/2021] [Indexed: 11/24/2022] Open
Abstract
Grafting bioactive peptides into recipient protein scaffolds can often increase their activities by conferring enhanced stability and cellular longevity. Here, we describe use of vGFP as a novel scaffold to display peptides. vGFP comprises GFP fused to a bound high affinity Enhancer nanobody that potentiates its fluorescence. We show that peptides inserted into the linker region between GFP and the Enhancer are correctly displayed for on-target interaction, both in vitro and in live cells by pull-down, measurement of target inhibition and imaging analyses. This is further confirmed by structural studies highlighting the optimal display of a vGFP-displayed peptide bound to Mdm2, the key negative regulator of p53 that is often overexpressed in cancer. We also demonstrate a potential biosensing application of the vGFP scaffold by showing target-dependent modulation of intrinsic fluorescence. vGFP is relatively thermostable, well-expressed and inherently fluorescent. These properties make it a useful scaffold to add to the existing tool box for displaying peptides that can disrupt clinically relevant protein–protein interactions.
Collapse
Affiliation(s)
- Sharon Min Qi Chee
- p53 Laboratory, A*STAR, 8A Biomedical Grove, Singapore, 138648, Singapore
| | - Jantana Wongsantichon
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Lau Sze Yi
- p53 Laboratory, A*STAR, 8A Biomedical Grove, Singapore, 138648, Singapore
| | - Barindra Sana
- p53 Laboratory, A*STAR, 8A Biomedical Grove, Singapore, 138648, Singapore
| | - Yuri Frosi
- p53 Laboratory, A*STAR, 8A Biomedical Grove, Singapore, 138648, Singapore
| | - Robert C Robinson
- School of Biomolecular Science and Engineering (BSE), Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand.,Research Institute for Interdisciplinary Science, Okayama University, Okayama, 700-8530, Japan
| | - Farid J Ghadessy
- p53 Laboratory, A*STAR, 8A Biomedical Grove, Singapore, 138648, Singapore.
| |
Collapse
|
8
|
Plaks JG, Brewer JA, Jacobsen NK, McKenna M, Uzarski JR, Lawton TJ, Filocamo SF, Kaar JL. Rosetta-Enabled Structural Prediction of Permissive Loop Insertion Sites in Proteins. Biochemistry 2020; 59:3993-4002. [PMID: 32970423 DOI: 10.1021/acs.biochem.0c00533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
While loop motifs frequently play a major role in protein function, our understanding of how to rationally engineer proteins with novel loop domains remains limited. In the absence of rational approaches, the incorporation of loop domains often destabilizes proteins, thereby requiring massive screening and selection to identify sites that can accommodate loop insertion. We developed a computational strategy for rapidly scanning the entire structure of a scaffold protein to determine the impact of loop insertion at all possible amino acid positions. This approach is based on the Rosetta kinematic loop modeling protocol and was demonstrated by identifying sites in lipase that were permissive to insertion of the LAP peptide. Interestingly, the identification of permissive sites was dependent on the contribution of the residues in the near-loop environment on the Rosetta score and did not correlate with conventional structural features (e.g., B-factors). As evidence of this, several insertion sites (e.g., following residues 17, 47-49, and 108), which were predicted and confirmed to be permissive, interrupted helices, while others (e.g., following residues 43, 67, 116, 119, and 121), which are situated in loop regions, were nonpermissive. This approach was further shown to be predictive for β-glucosidase and human phosphatase and tensin homologue (PTEN), and to facilitate the engineering of insertion sites through in silico mutagenesis. By enabling the design of loop-containing protein libraries with high probabilities of soluble expression, this approach has broad implications in many areas of protein engineering, including antibody design, improving enzyme activity, and protein modification.
Collapse
Affiliation(s)
- Joseph G Plaks
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Jeff A Brewer
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Nicole K Jacobsen
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Michael McKenna
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Joshua R Uzarski
- U.S. Army Combat Capabilities Development Command Soldier Center, Natick, Massachusetts 01760, United States
| | - Timothy J Lawton
- U.S. Army Combat Capabilities Development Command Soldier Center, Natick, Massachusetts 01760, United States
| | - Shaun F Filocamo
- U.S. Army Combat Capabilities Development Command Soldier Center, Natick, Massachusetts 01760, United States
| | - Joel L Kaar
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|
9
|
The Last Secret of Protein Folding: The Real Relationship Between Long-Range Interactions and Local Structures. Protein J 2020; 39:422-433. [PMID: 33040262 DOI: 10.1007/s10930-020-09925-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2020] [Indexed: 01/20/2023]
Abstract
The protein folding problem has been extensively studied for decades, and hundreds of thousands of protein structures have been solved. Yet, how proteins fold from a linear peptide chain to their unique 3D structures is not fully understood. With key clues having emerged unexpectedly from the field of nanoscience, a "Confined Lowest Energy Fragment" (CLEF) hypothesis was proposed. The CLEF hypothesis states that a protein chain can be divided into CLEFs, the semi-independent folding units, by a small number of key residues that form key long-range interactions. The native structure of a CLEF is the lowest energy state under the constraints of the key long-range interactions, but the native structure of the whole protein is not necessary the lowest energy state as Anfinsen's thermodynamic hypothesis suggested. The CLEF hypothesis proposes a unified CLEF mechanism for protein folding, basically a two-step process. In the first step, the favorable enthalpy of CLEFs for native structures quickly brings those residues for the key long-range interactions together, forming intermediates corresponding to the so-called hydrophobic collapse. In the second step, those collapsed key residues shuffle for the right combination to form the native key long-range interactions. The CLEF hypothesis provides a simple solution to all protein folding paradoxes, and proposes a "CLEF Age" or "Stone Age" for the prebiotic evolution of proteins.
Collapse
|
10
|
Hortua Triana MA, Márquez-Nogueras KM, Chang L, Stasic AJ, Li C, Spiegel KA, Sharma A, Li ZH, Moreno SNJ. Tagging of Weakly Expressed Toxoplasma gondii Calcium-Related Genes with High-Affinity Tags. J Eukaryot Microbiol 2018; 65:709-721. [PMID: 29672999 DOI: 10.1111/jeu.12626] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
Calcium ions regulate a diversity of cellular functions in all eukaryotes. The cytosolic Ca2+ concentration is tightly regulated at the physiological cytosolic concentration of 50-100 nm. The Toxoplasma gondii genome predicts the presence of several genes encoding potential Ca2+ channels, pumps, and transporters. Many of these genes are weakly expressed and likely tightly regulated due to their potential impact to the physiology of the cell. Endogenous tagging has been widely used to localize proteins in T. gondii but low level of expression of many of them makes visualization of tags difficult and sometimes impossible. The use of high-performance tags for labeling proteins expressed at low level is ideal for investigating the localization of these gene products. We designed a Carboxy-terminus tagging plasmid containing the previously characterized "spaghetti monster-HA" (smHA) or "spaghetti monster-MYC" (smMYC) tags. These tags consist of 10 copies of a single epitope (HA or MYC) inserted into a darkened green fluorescence protein scaffold. We localized six proteins of various levels of expression. Clonal lines were isolated and validated by PCR, western blot, and immunofluorescence analyses. Some gene products were only visible when tagged with smHA and in one case the smHA revealed a novel localization previously undetected.
Collapse
Affiliation(s)
- Miryam A Hortua Triana
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602
| | | | - Le Chang
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602
| | - Andrew J Stasic
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602
| | - Catherine Li
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602
| | - Kevin A Spiegel
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602
| | - Amrita Sharma
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602.,Department of Cellular Biology, University of Georgia, Athens, Georgia, 30602
| | - Zhu-Hong Li
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602
| | - Silvia N J Moreno
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, 30602.,Department of Cellular Biology, University of Georgia, Athens, Georgia, 30602
| |
Collapse
|
11
|
Bansal N, Zheng Z, Song LF, Pei J, Merz KM. The Role of the Active Site Flap in Streptavidin/Biotin Complex Formation. J Am Chem Soc 2018; 140:5434-5446. [PMID: 29607642 DOI: 10.1021/jacs.8b00743] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Obtaining a detailed description of how active site flap motion affects substrate or ligand binding will advance structure-based drug design (SBDD) efforts on systems including the kinases, HSP90, HIV protease, ureases, etc. Through this understanding, we will be able to design better inhibitors and better proteins that have desired functions. Herein we address this issue by generating the relevant configurational states of a protein flap on the molecular energy landscape using an approach we call MTFlex-b and then following this with a procedure to estimate the free energy associated with the motion of the flap region. To illustrate our overall workflow, we explored the free energy changes in the streptavidin/biotin system upon introducing conformational flexibility in loop3-4 in the biotin unbound ( apo) and bound ( holo) state. The free energy surfaces were created using the Movable Type free energy method, and for further validation, we compared them to potential of mean force (PMF) generated free energy surfaces using MD simulations employing the FF99SBILDN and FF14SB force fields. We also estimated the free energy thermodynamic cycle using an ensemble of closed-like and open-like end states for the ligand unbound and bound states and estimated the binding free energy to be approximately -16.2 kcal/mol (experimental -18.3 kcal/mol). The good agreement between MTFlex-b in combination with the MT method with experiment and MD simulations supports the effectiveness of our strategy in obtaining unique insights into the motions in proteins that can then be used in a range of biological and biomedical applications.
Collapse
Affiliation(s)
- Nupur Bansal
- Department of Chemistry and Department of Biochemistry and Molecular Biology , Michigan State University , 578 South Shaw Lane , East Lansing , Michigan 48824 , United States
| | - Zheng Zheng
- Department of Chemistry and Department of Biochemistry and Molecular Biology , Michigan State University , 578 South Shaw Lane , East Lansing , Michigan 48824 , United States
| | - Lin Frank Song
- Department of Chemistry and Department of Biochemistry and Molecular Biology , Michigan State University , 578 South Shaw Lane , East Lansing , Michigan 48824 , United States
| | - Jun Pei
- Department of Chemistry and Department of Biochemistry and Molecular Biology , Michigan State University , 578 South Shaw Lane , East Lansing , Michigan 48824 , United States
| | - Kenneth M Merz
- Department of Chemistry and Department of Biochemistry and Molecular Biology , Michigan State University , 578 South Shaw Lane , East Lansing , Michigan 48824 , United States.,Institute for Cyber Enabled Research , Michigan State University , 567 Wilson Road , East Lansing , Michigan 48824 , United States
| |
Collapse
|
12
|
D'Angelo S, Ferrara F, Naranjo L, Erasmus MF, Hraber P, Bradbury ARM. Many Routes to an Antibody Heavy-Chain CDR3: Necessary, Yet Insufficient, for Specific Binding. Front Immunol 2018; 9:395. [PMID: 29568296 PMCID: PMC5852061 DOI: 10.3389/fimmu.2018.00395] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/13/2018] [Indexed: 12/11/2022] Open
Abstract
Because of its great potential for diversity, the immunoglobulin heavy-chain complementarity-determining region 3 (HCDR3) is taken as an antibody molecule’s most important component in conferring binding activity and specificity. For this reason, HCDR3s have been used as unique identifiers to investigate adaptive immune responses in vivo and to characterize in vitro selection outputs where display systems were employed. Here, we show that many different HCDR3s can be identified within a target-specific antibody population after in vitro selection. For each identified HCDR3, a number of different antibodies bearing differences elsewhere can be found. In such selected populations, all antibodies with the same HCDR3 recognize the target, albeit at different affinities. In contrast, within unselected populations, the majority of antibodies with the same HCDR3 sequence do not bind the target. In one HCDR3 examined in depth, all target-specific antibodies were derived from the same VDJ rearrangement, while non-binding antibodies with the same HCDR3 were derived from many different V and D gene rearrangements. Careful examination of previously published in vivo datasets reveals that HCDR3s shared between, and within, different individuals can also originate from rearrangements of different V and D genes, with up to 26 different rearrangements yielding the same identical HCDR3 sequence. On the basis of these observations, we conclude that the same HCDR3 can be generated by many different rearrangements, but that specific target binding is an outcome of unique rearrangements and VL pairing: the HCDR3 is necessary, albeit insufficient, for specific antibody binding.
Collapse
Affiliation(s)
| | | | | | | | - Peter Hraber
- Los Alamos National Laboratory, Los Alamos, NM, United States
| | | |
Collapse
|
13
|
Artificial antibody created by conformational reconstruction of the complementary-determining region on gold nanoparticles. Proc Natl Acad Sci U S A 2017; 115:E34-E43. [PMID: 29255034 PMCID: PMC5776806 DOI: 10.1073/pnas.1713526115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mimicking protein-like specific interactions and functions has been a long-pursued goal in nanotechnology. The key challenge is to precisely organize nonfunctional surface groups on nanoparticles into specific 3D conformations to function in a concerted and orchestrated manner. Here, we develop a method to graft the complementary-determining regions of natural antibodies onto nanoparticles and reconstruct their “active” conformation to create nanoparticle-based artificial antibodies that recognize the corresponding antigens. Our work demonstrates that it is possible to create functions on nanoparticles by conformational engineering, namely tuning flexible surface groups into specific conformations. Our straightforward strategy could be used further to create other artificial antibodies for various applications and provides a new tool to understand the structure and folding of natural proteins. To impart biomedical functions to nanoparticles (NPs), the common approach is to conjugate functional groups onto NPs by dint of the functions of those groups per se. It is still beyond current reach to create protein-like specific interactions and functions on NPs by conformational engineering of nonfunctional groups on NPs. Here, we develop a conformational engineering method to create an NP-based artificial antibody, denoted “Goldbody,” through conformational reconstruction of the complementary-determining regions (CDRs) of natural antibodies on gold NPs (AuNPs). The seemingly insurmountable task of controlling the conformation of the CDR loops, which are flexible and nonfunctional in the free form, was accomplished unexpectedly in a simple way. Upon anchoring both terminals of the free CDR loops on AuNPs, we managed to reconstruct the “active” conformation of the CDR loops by tuning the span between the two terminals and, as a result, the original specificity was successfully reconstructed on the AuNPs. Two Goldbodies have been created by this strategy to specifically bind with hen egg white lysozyme and epidermal growth factor receptor, with apparent affinities several orders of magnitude stronger than that of the original natural antibodies. Our work demonstrates that it is possible to create protein-like functions on NPs in a protein-like way, namely by tuning flexible surface groups to the correct conformation. Given the apparent merits, including good stability, of Goldbodies, we anticipate that a category of Goldbodies could be created to target different antigens and thus used as substitutes for natural antibodies in various applications.
Collapse
|
14
|
Glanville J, D'Angelo S, Khan TA, Reddy ST, Naranjo L, Ferrara F, Bradbury ARM. Deep sequencing in library selection projects: what insight does it bring? Curr Opin Struct Biol 2016; 33:146-60. [PMID: 26451649 DOI: 10.1016/j.sbi.2015.09.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/19/2015] [Accepted: 09/17/2015] [Indexed: 11/17/2022]
Abstract
High throughput sequencing is poised to change all aspects of the way antibodies and other binders are discovered and engineered. Millions of available sequence reads provide an unprecedented sampling depth able to guide the design and construction of effective, high quality naïve libraries containing tens of billions of unique molecules. Furthermore, during selections, high throughput sequencing enables quantitative tracing of enriched clones and position-specific guidance to amino acid variation under positive selection during antibody engineering. Successful application of the technologies relies on specific PCR reagent design, correct sequencing platform selection, and effective use of computational tools and statistical measures to remove error, identify antibodies, estimate diversity, and extract signatures of selection from the clone down to individual structural positions. Here we review these considerations and discuss some of the remaining challenges to the widespread adoption of the technology.
Collapse
Affiliation(s)
- J Glanville
- Program in Computational and Systems Immunology, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - S D'Angelo
- University of New Mexico Comprehensive Cancer Center, and Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - T A Khan
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - S T Reddy
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - L Naranjo
- Bioscience division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - F Ferrara
- University of New Mexico Comprehensive Cancer Center, and Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - A R M Bradbury
- Bioscience division, Los Alamos National Laboratory, Los Alamos, NM, USA.
| |
Collapse
|
15
|
Pacheco S, Béhar G, Maillasson M, Mouratou B, Pecorari F. Affinity transfer to the archaeal extremophilic Sac7d protein by insertion of a CDR. Protein Eng Des Sel 2015; 27:431-8. [PMID: 25301962 DOI: 10.1093/protein/gzu042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Artificially transforming a scaffold protein into binders often consists of introducing diversity into its natural binding region by directed mutagenesis. We have previously developed the archaeal extremophilic Sac7d protein as a scaffold to derive affinity reagents (Affitins) by randomization of only a flat surface, or a flat surface and two short loops with natural lengths. Short loops are believed to contribute to stability of extremophilic proteins, and loop extension has been reported detrimental for the thermal and chemical stabilities of mesophilic proteins. In this work, we wanted to evaluate the possibility of designing target-binding proteins based on Sac7d by using a complementary determining region (CDR). To this aim, we inserted into three different loops a 10 residues CDR from the cAb-Lys3 anti-lysozyme camel antibody. The chimeras obtained were as stable as wild-type (WT) Sac7d at extreme pH and their structural integrity was supported. Chimeras were thermally stable, but with T(m)s from 60.9 to 66.3°C (cf. 91°C for Sac7d) which shows that loop extension is detrimental for thermal stability of Sac7d. The loop 3 enabled anti-lysozyme activity. These results pave the way for the use of CDR(s) from antibodies and/or extended randomized loop(s) to increase the potential of binding of Affitins.
Collapse
Affiliation(s)
- Sabino Pacheco
- CRCNA - UMR 892 INSERM, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France 6299 CNRS, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France University of Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France Institut Pasteur, CNRS UMR 3528, Unité de Microbiologie Structurale, 25 rue du Dr. Roux, 72724 Paris Cedex 15, France
| | - Ghislaine Béhar
- CRCNA - UMR 892 INSERM, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France 6299 CNRS, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France University of Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France
| | - Mike Maillasson
- CRCNA - UMR 892 INSERM, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France 6299 CNRS, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France University of Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France Plate-forme IMPACT Biogenouest, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France
| | - Barbara Mouratou
- CRCNA - UMR 892 INSERM, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France 6299 CNRS, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France University of Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France
| | - Frédéric Pecorari
- CRCNA - UMR 892 INSERM, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France 6299 CNRS, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France University of Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes, Cedex 1, France
| |
Collapse
|
16
|
High-performance probes for light and electron microscopy. Nat Methods 2015; 12:568-76. [PMID: 25915120 PMCID: PMC4573404 DOI: 10.1038/nmeth.3365] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/24/2015] [Indexed: 12/12/2022]
Abstract
We describe an engineered family of highly antigenic molecules based on GFP-like fluorescent proteins. These molecules contain numerous copies of peptide epitopes and simultaneously bind IgG antibodies at each location. These 'spaghetti monster' fluorescent proteins (smFPs) distributed well in neurons, notably into small dendrites, spines and axons. smFP immunolabeling localized weakly expressed proteins not well resolved with traditional epitope tags. By varying epitope and scaffold, we generated a diverse family of mutually orthogonal antigens. In cultured neurons and mouse and fly brains, smFP probes allowed robust, orthogonal multicolor visualization of proteins, cell populations and neuropil. smFP variants complement existing tracers and greatly increase the number of simultaneous imaging channels, and they performed well in advanced preparations such as array tomography, super-resolution fluorescence imaging and electron microscopy. In living cells, the probes improved single-molecule image tracking and increased yield for RNA-seq. These probes facilitate new experiments in connectomics, transcriptomics and protein localization.
Collapse
|
17
|
Kadonosono T, Yabe E, Furuta T, Yamano A, Tsubaki T, Sekine T, Kuchimaru T, Sakurai M, Kizaka-Kondoh S. A fluorescent protein scaffold for presenting structurally constrained peptides provides an effective screening system to identify high affinity target-binding peptides. PLoS One 2014; 9:e103397. [PMID: 25084350 PMCID: PMC4118881 DOI: 10.1371/journal.pone.0103397] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/01/2014] [Indexed: 11/18/2022] Open
Abstract
Peptides that have high affinity for target molecules on the surface of cancer cells are crucial for the development of targeted cancer therapies. However, unstructured peptides often fail to bind their target molecules with high affinity. To efficiently identify high-affinity target-binding peptides, we have constructed a fluorescent protein scaffold, designated gFPS, in which structurally constrained peptides are integrated at residues K131-L137 of superfolder green fluorescent protein. Molecular dynamics simulation supported the suitability of this site for presentation of exogenous peptides with a constrained structure. gFPS can present 4 to 12 exogenous amino acids without a loss of fluorescence. When gFPSs presenting human epidermal growth factor receptor type 2 (HER2)-targeting peptides were added to the culture medium of HER2-expressing cells, we could easily identify the peptides with high HER2-affinity and -specificity based on gFPS fluorescence. In addition, gFPS could be expressed on the yeast cell surface and applied for a high-throughput screening. These results demonstrate that gFPS has the potential to serve as a powerful tool to improve screening of structurally constrained peptides that have a high target affinity, and suggest that it could expedite the one-step identification of clinically applicable cancer cell-binding peptides.
Collapse
Affiliation(s)
- Tetsuya Kadonosono
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
| | - Etsuri Yabe
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
| | - Tadaomi Furuta
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama City, Japan
| | - Akihiro Yamano
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
| | - Takuya Tsubaki
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
| | - Takuya Sekine
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
| | - Takahiro Kuchimaru
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
| | - Minoru Sakurai
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama City, Japan
| | - Shinae Kizaka-Kondoh
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama City, Japan
- * E-mail:
| |
Collapse
|
18
|
Björnerås J, Kurnik M, Oliveberg M, Gräslund A, Mäler L, Danielsson J. Direct detection of neuropeptide dynorphin A binding to the second extracellular loop of the κ opioid receptor using a soluble protein scaffold. FEBS J 2014; 281:814-24. [PMID: 24616919 DOI: 10.1111/febs.12626] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The molecular determinants for selectivity of ligand binding to membrane receptors are of key importance for the understanding of cellular signalling, as well as for rational therapeutic intervention. In the present study, we target the interaction between the κ opioid receptor (KOR) and its native peptide ligand dynorphin A (DynA) using solution state NMR spectroscopy, which is generally made difficult by the sheer size of membrane bound receptors. Our method is based on 'transplantation' of an extracellular loop of KOR into a 'surrogate' scaffold; in this case, a soluble β-barrel. Our results corroborate the general feasibility of the method, showing that the inserted receptor segment has negligible effects on the properties of the scaffold protein, at the same time as maintaining an ability to bind its native DynA ligand. Upon DynA binding, only small induced chemical shift changes of the KOR loop were observed, whereas chemical shift changes of DynA and NMR paramagnetic relaxation data show conclusively that the peptide interacts with the inserted loop. The binding interface is composed of a disordered part of the KOR loop and involves both electrostatic and hydrophobic interactions. Even so, simultaneous effects along the DynA sequence upon binding show that control of the recognition is a concerted event.
Collapse
|
19
|
Lee J, Kim HJ, Roh J, Seo Y, Kim M, Jun HR, Pham CD, Kwon MH. Functional consequences of complementarity-determining region deactivation in a multifunctional anti-nucleic acid antibody. J Biol Chem 2013; 288:35877-85. [PMID: 24155236 PMCID: PMC3861637 DOI: 10.1074/jbc.m113.508499] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many murine monoclonal anti-DNA antibodies (Abs) derived from mice models for systemic lupus erythematosus have additional cell-penetration and/or nucleic acid-hydrolysis properties. Here, we examined the influence of deactivating each complementarity-determining region (CDR) within a multifunctional anti-nucleic acid antibody (Ab) that possesses these activities, the catalytic 3D8 single chain variable fragment (scFv). CDR-deactivated 3D8 scFv variants were generated by replacing all of the amino acids within each CDR with Gly/Ser residues. The structure of 3D8 scFv accommodated single complete CDR deactivations. Different functional activities of 3D8 scFv were affected differently depending on which CDR was deactivated. The only exception was CDR1, located within the light chain (LCDR1); deactivation of LCDR1 abolished all of the functional activities of 3D8 scFv. A hybrid Ab, HW6/3D8L1, in which the LCDR1 from an unrelated Ab (HW6) was replaced with the LCDR1 from 3D8, acquired all activities associated with the 3D8 scFv. These results suggest that the activity of a multifunctional 3D8 scFv Ab can be modulated by single complete CDR deactivation and that the LCDR1 plays a crucial role in maintaining Ab properties. This study presents a new approach for determining the role of individual CDRs in multifunctional Abs with important implications for the future of Ab engineering.
Collapse
Affiliation(s)
- Jiyeon Lee
- From the Department of Biomedical Sciences, Graduate School, Ajou University, San 5, Woncheon-dong, Yeongtong-gu, Suwon 443-749, South Korea and
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Cotham VC, Wine Y, Brodbelt JS. Selective 351 nm photodissociation of cysteine-containing peptides for discrimination of antigen-binding regions of IgG fragments in bottom-up liquid chromatography-tandem mass spectrometry workflows. Anal Chem 2013; 85:5577-85. [PMID: 23641966 DOI: 10.1021/ac400851x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Despite tremendous inroads in the development of more sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) strategies for mass spectrometry-based proteomics, there remains a significant need for enhancing the selectivity of MS/MS-based workflows for streamlined analysis of complex biological mixtures. Here, a novel LC-MS/MS platform based on 351 nm ultraviolet photodissociation (UVPD) is presented for the selective analysis of cysteine-peptide subsets in complex protein digests. Cysteine-selective UVPD is mediated through the site-specific conjugation of reduced cysteine residues with a 351 nm active chromogenic Alexa Fluor 350 (AF350) maleimide tag. Only peptides containing the AF350 chromophore undergo photodissociation into extensive arrays of b- and y-type fragment ions, thus providing a facile means for differentiating cysteine-peptide targets from convoluting peptide backgrounds. With the use of this approach in addition to strategic proteolysis, the selective analysis of diagnostic heavy-chain complementarity determining regions (CDRs) of single-chain antibody (scAb) fragments is demonstrated.
Collapse
Affiliation(s)
- Victoria C Cotham
- Department of Chemistry and Biochemistry, The University of Texas at Austin, 1 University Station A5300, Austin, TX 78712, USA
| | | | | |
Collapse
|
21
|
Li Y. Conformational sampling in template-free protein loop structure modeling: an overview. Comput Struct Biotechnol J 2013; 5:e201302003. [PMID: 24688696 PMCID: PMC3962101 DOI: 10.5936/csbj.201302003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/23/2013] [Accepted: 01/28/2013] [Indexed: 01/04/2023] Open
Abstract
Accurately modeling protein loops is an important step to predict three-dimensional structures as well as to understand functions of many proteins. Because of their high flexibility, modeling the three-dimensional structures of loops is difficult and is usually treated as a "mini protein folding problem" under geometric constraints. In the past decade, there has been remarkable progress in template-free loop structure modeling due to advances of computational methods as well as stably increasing number of known structures available in PDB. This mini review provides an overview on the recent computational approaches for loop structure modeling. In particular, we focus on the approaches of sampling loop conformation space, which is a critical step to obtain high resolution models in template-free methods. We review the potential energy functions for loop modeling, loop buildup mechanisms to satisfy geometric constraints, and loop conformation sampling algorithms. The recent loop modeling results are also summarized.
Collapse
Affiliation(s)
- Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA 23529, USA
| |
Collapse
|
22
|
Petrovskaya LE, Shingarova LN, Kryukova EA, Boldyreva EF, Yakimov SA, Guryanova SV, Novoseletsky VN, Dolgikh DA, Kirpichnikov MP. Construction of TNF-binding proteins by grafting hypervariable regions of F10 antibody on human fibronectin domain scaffold. BIOCHEMISTRY (MOSCOW) 2012; 77:62-70. [PMID: 22339634 DOI: 10.1134/s0006297912010075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor (TNF) plays a key role in the pathogenesis of various diseases. To study the possibility of constructing TNF-binding proteins by grafting hypervariable regions of immunoglobulins (CDR), we have replaced amino acid sequences of loops from the tenth type III domain of human fibronectin ((10)Fn3) by amino acid sequences of CDR from the light and heavy chains of the anti-TNF antibody F10. The assessment of TNF-binding properties of the resulting proteins by ELISA has revealed the highest activity of Hd3 containing sequences CDR-H1 and CDR-H2 of the antibody F10 and of Hd2 containing sequences CDR-H1 and CDR-H3. The proteins constructed by us on the fibronectin domain scaffold specifically bound TNF during Western blotting and also weakened its cytotoxic effect on L929 line cells. The highest neutralizing activity was demonstrated by the proteins Hd2 and Hd3, which induced, respectively, 10- and 50-fold increase in the EC(50) of TNF.
Collapse
Affiliation(s)
- L E Petrovskaya
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Inoue H, Iihara A, Takahashi H, Shimada I, Ishida I, Maeda Y. Affinity transfer to a human protein by CDR3 grafting of camelid VHH. Protein Sci 2011; 20:1971-81. [PMID: 21910150 DOI: 10.1002/pro.734] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/22/2011] [Accepted: 08/26/2011] [Indexed: 11/10/2022]
Abstract
VHH is the binding domain of the IgG heavy chain. Some VHHs have an extremely long CDR3 that contributes to antigen binding. We studied the antigen binding ability of CDR3 by grafting a CDR3 from an antigen-binding VHH onto a nonbinding VHH. cAb-CA05-(1RI8), the CDR3-grafted VHH, had an antigen-binding ability. To find a human scaffold protein acceptable for VHH CDR3 grafting, we focused on the conserved structure of VHH, especially the N-terminal and C-terminal amino acid residues of the CDR3 loop and the Cys residue of CDR1. Human origin protein structures with the same orientation were searched in PDB and ubiquitin was selected. Ubi-(1RI8), the CDR3-grafted ubiquitin, had antigen-binding ability, though the affinity was relatively low compared to cAb-CA05-(1RI8). The thermodynamic parameters of Ubi-(1RI8) binding to HEWL were different from cAb-CA05-(1RI8). Hydrogen-deuterium exchange experiments showed decreased stability around the CDR3 grafting region of Ubi-(1RI8), which might explain the decreased antigen-binding ability and the differences in thermodynamic properties. We concluded that the orientation of the CDR3 sequence of Ubi-(1RI8) could not be reconstructed correctly.
Collapse
|
24
|
Velappan N, Fisher HE, Pesavento E, Chasteen L, D’Angelo S, Kiss C, Longmire M, Pavlik P, Bradbury ARM. A comprehensive analysis of filamentous phage display vectors for cytoplasmic proteins: an analysis with different fluorescent proteins. Nucleic Acids Res 2010; 38:e22. [PMID: 19955231 PMCID: PMC2831335 DOI: 10.1093/nar/gkp809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 09/14/2009] [Indexed: 01/11/2023] Open
Abstract
Filamentous phage display has been extensively used to select proteins with binding properties of specific interest. Although many different display platforms using filamentous phage have been described, no comprehensive comparison of their abilities to display similar proteins has been conducted. This is particularly important for the display of cytoplasmic proteins, which are often poorly displayed with standard filamentous phage vectors. In this article, we have analyzed the ability of filamentous phage to display a stable form of green fluorescent protein and modified variants in nine different display vectors, a number of which have been previously proposed as being suitable for cytoplasmic protein display. Correct folding and display were assessed by phagemid particle fluorescence, and with anti-GFP antibodies. The poor correlation between phagemid particle fluorescence and recognition of GFP by antibodies, indicates that proteins may fold correctly without being accessible for display. The best vector used a twin arginine transporter leader to transport the displayed protein to the periplasm, and a coil-coil arrangement to link the displayed protein to g3p. This vector was able to display less robust forms of GFP, including ones with inserted epitopes, as well as fluorescent proteins of the Azami green series. It was also functional in mock selection experiments.
Collapse
|
25
|
Mining protein loops using a structural alphabet and statistical exceptionality. BMC Bioinformatics 2010; 11:75. [PMID: 20132552 PMCID: PMC2833150 DOI: 10.1186/1471-2105-11-75] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 02/04/2010] [Indexed: 12/21/2022] Open
Abstract
Background Protein loops encompass 50% of protein residues in available three-dimensional structures. These regions are often involved in protein functions, e.g. binding site, catalytic pocket... However, the description of protein loops with conventional tools is an uneasy task. Regular secondary structures, helices and strands, have been widely studied whereas loops, because they are highly variable in terms of sequence and structure, are difficult to analyze. Due to data sparsity, long loops have rarely been systematically studied. Results We developed a simple and accurate method that allows the description and analysis of the structures of short and long loops using structural motifs without restriction on loop length. This method is based on the structural alphabet HMM-SA. HMM-SA allows the simplification of a three-dimensional protein structure into a one-dimensional string of states, where each state is a four-residue prototype fragment, called structural letter. The difficult task of the structural grouping of huge data sets is thus easily accomplished by handling structural letter strings as in conventional protein sequence analysis. We systematically extracted all seven-residue fragments in a bank of 93000 protein loops and grouped them according to the structural-letter sequence, named structural word. This approach permits a systematic analysis of loops of all sizes since we consider the structural motifs of seven residues rather than complete loops. We focused the analysis on highly recurrent words of loops (observed more than 30 times). Our study reveals that 73% of loop-lengths are covered by only 3310 highly recurrent structural words out of 28274 observed words). These structural words have low structural variability (mean RMSd of 0.85 Å). As expected, half of these motifs display a flanking-region preference but interestingly, two thirds are shared by short (less than 12 residues) and long loops. Moreover, half of recurrent motifs exhibit a significant level of amino-acid conservation with at least four significant positions and 87% of long loops contain at least one such word. We complement our analysis with the detection of statistically over-represented patterns of structural letters as in conventional DNA sequence analysis. About 30% (930) of structural words are over-represented, and cover about 40% of loop lengths. Interestingly, these words exhibit lower structural variability and higher sequential specificity, suggesting structural or functional constraints. Conclusions We developed a method to systematically decompose and study protein loops using recurrent structural motifs. This method is based on the structural alphabet HMM-SA and not on structural alignment and geometrical parameters. We extracted meaningful structural motifs that are found in both short and long loops. To our knowledge, it is the first time that pattern mining helps to increase the signal-to-noise ratio in protein loops. This finding helps to better describe protein loops and might permit to decrease the complexity of long-loop analysis. Detailed results are available at http://www.mti.univ-paris-diderot.fr/publication/supplementary/2009/ACCLoop/.
Collapse
|
26
|
Development of GFP-based biosensors possessing the binding properties of antibodies. Proc Natl Acad Sci U S A 2009; 106:11895-900. [PMID: 19574456 DOI: 10.1073/pnas.0902828106] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Proteins that can bind specifically to targets that also have an intrinsic property allowing for easy detection could facilitate a multitude of applications. While the widely used green fluorescent protein (GFP) allows for easy detection, attempts to insert multiple binding loops into GFP to impart affinity for a specific target have been met with limited success because of the structural sensitivity of the GFP chromophore. In this study, directed evolution using a surrogate loop approach and yeast surface display yielded a family of GFP scaffolds capable of accommodating 2 proximal, randomized binding loops. The library of potential GFP-based binders or ''GFAbs'' was subsequently mined for GFAbs capable of binding to protein targets. Identified GFAbs bound with nanomolar affinity and required binding contributions from both loops indicating the advantage of a dual loop GFAb platform. Finally, GFAbs were solubly produced and used as fluorescence detection reagents to demonstrate their utility.
Collapse
|
27
|
Boschek CB, Apiyo DO, Soares TA, Engelmann HE, Pefaur NB, Straatsma TP, Baird CL. Engineering an ultra-stable affinity reagent based on Top7. Protein Eng Des Sel 2009; 22:325-32. [DOI: 10.1093/protein/gzp007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
28
|
Kiss C, Temirov J, Chasteen L, Waldo GS, Bradbury AR. Directed evolution of an extremely stable fluorescent protein. Protein Eng Des Sel 2009; 22:313-23. [DOI: 10.1093/protein/gzp006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
29
|
Chen SS, Yang YM, Barankiewicz TJ. Selection of IgE-binding aptameric green fluorescent protein (Ap-GFP) by the ribosome display (RD) platform. Biochem Biophys Res Commun 2008; 374:409-14. [PMID: 18619414 DOI: 10.1016/j.bbrc.2008.06.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2008] [Accepted: 06/25/2008] [Indexed: 11/26/2022]
Abstract
GFP-Ckappa fusion protein was previously shown selectable on ribosome display platform with solid phase antibodies against GFP determinant [Y.-M. Yang, T.J. Barankiewicz, M. He, M. Taussig, S.-S. Chen, Selection of antigenic markers on a GFP-Ckappa fusion scaffold with high sensitivity by eukaryotic ribosome display, Biochem. Biophys. Res. Commun. 359 (2007) 251-257]. Herein, we show that members of aptameric peptide library constructed within the site 6 and site 8/9 loops of GFP of the ribosome display construct are selectable upon binding to the solid phase IgE antigen. An input of 1.0 microg of the dual site aptameric GFP library exhibiting a diversity of 7.5x10(11) was transcribed, translated and incubated with solid phase IgE. RT-PCR products were amplified from mRNA of the aptamer-ribosome-mRNA (ARM) complex captured on the solid phase IgE. Clones of aptameric GFP were prepared from RT-PCR product of ARM complex following repetitive selection. Recombinant aptameric GFP proteins from the selected clones bind IgE coated on the 96-well plate, and the binding was abrogated by incubation with soluble human IgE but not human IgG. Selected aptameric GFP proteins also exhibit binding to three different sources of human IgE (IgE PS, BED, and JW8) but not irrelevant proteins. These observations indicate that appropriately selected aptameric GFP on a solid phase ligand by ribosome display may serve as an affinity reagent for blocking reactivity of a biological ligand.
Collapse
Affiliation(s)
- Swey-Shen Chen
- The Institute of Genetics, Allergy and Immunology, 6370 Lusk Boulevard, F109-F110, San Diego, CA 92121, USA.
| | | | | |
Collapse
|
30
|
Fisher AC, DeLisa MP. Laboratory evolution of fast-folding green fluorescent protein using secretory pathway quality control. PLoS One 2008; 3:e2351. [PMID: 18545653 PMCID: PMC2396501 DOI: 10.1371/journal.pone.0002351] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2008] [Accepted: 04/28/2008] [Indexed: 11/19/2022] Open
Abstract
Green fluorescent protein (GFP) has undergone a long history of optimization to become one of the most popular proteins in all of cell biology. It is thermally and chemically robust and produces a pronounced fluorescent phenotype when expressed in cells of all types. Recently, a superfolder GFP was engineered with increased resistance to denaturation and improved folding kinetics. Here we report that unlike other well-folded variants of GFP (e.g., GFPmut2), superfolder GFP was spared from elimination when targeted for secretion via the SecYEG translocase. This prompted us to hypothesize that the folding quality control inherent to this secretory pathway could be used as a platform for engineering similar ‘superfolded’ proteins. To test this, we targeted a combinatorial library of GFPmut2 variants to the SecYEG translocase and isolated several superfolded variants that accumulated in the cytoplasm due to their enhanced folding properties. Each of these GFP variants exhibited much faster folding kinetics than the parental GFPmut2 protein and one of these, designated superfast GFP, folded at a rate that even exceeded superfolder GFP. Remarkably, these GFP variants exhibited little to no loss in specific fluorescence activity relative to GFPmut2, suggesting that the process of superfolding can be accomplished without altering the proteins' normal function. Overall, we demonstrate that laboratory evolution combined with secretory pathway quality control enables sampling of largely unexplored amino-acid sequences for the discovery of artificial, high-performance proteins with properties that are unparalleled in their naturally occurring analogues.
Collapse
Affiliation(s)
- Adam C. Fisher
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, United States of America
| | - Matthew P. DeLisa
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, United States of America
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
31
|
Dai M, Temirov J, Pesavento E, Kiss C, Velappan N, Pavlik P, Werner JH, Bradbury ARM. Using T7 phage display to select GFP-based binders. Protein Eng Des Sel 2008; 21:413-24. [PMID: 18469345 DOI: 10.1093/protein/gzn016] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Filamentous phage do not display cytoplasmic proteins very effectively. As T7 is a cytoplasmic phage, released by cell lysis, it has been prospected as being more efficient for the display of such proteins. Here we investigate this proposition, using a family of GFP-based cytoplasmic proteins that are poorly expressed by traditional phage display. Using two single-molecule detection techniques, fluorescence correlation spectroscopy and anti-bunching, we show that the number of displayed fluorescent proteins ranges from one to three. The GFP derivatives displayed on T7 contain binding loops able to recognize specific targets. By mixing these in a large background of non-binders, these derivatives were used to optimize selection conditions. Using the optimal selection conditions determined in these experiments, we then demonstrated the selection of specific binders from a library of GFP clones containing heavy chain CDR3 antibody binding loops derived from normal donors inserted at a single site. The selected GFP-based binders were successfully used to detect binding without the use of secondary reagents in flow cytometry, fluorescence-linked immunosorbant assays and immunoblotting. These results demonstrate that specific GFP-based affinity reagents, selected from T7-based libraries, can be used in applications in which only the intrinsic fluorescence is used for detection.
Collapse
Affiliation(s)
- M Dai
- Biosciences Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Velappan N, Clements J, Kiss C, Valero-Aracama R, Pavlik P, Bradbury ARM. Fluorescence linked immunosorbant assays using microtiter plates. J Immunol Methods 2008; 336:135-41. [PMID: 18514691 DOI: 10.1016/j.jim.2008.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2007] [Revised: 04/07/2008] [Accepted: 04/08/2008] [Indexed: 11/28/2022]
Abstract
Fluorescence methods are widely used in the detection of antibodies and other binding events. However, as a general screening and detection tool in microtiter plates, enzyme linked immunosorbant (ELISA) methods predominate. In this paper we explore all parameters for effective use of fluorescence as a plate based detection method, including which microtiter plates can be used, the most effective means of immobilization, and the use of different fluorescent dyes or fluorescent proteins. These studies indicate that fluorescent immunosorbant assays (FLISA) can be used as effectively as enzymatic method in microtiter plate based screening methods, including the screening of phage antibody selections.
Collapse
Affiliation(s)
- N Velappan
- B division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | | | | | | | | | | |
Collapse
|
33
|
Chen SS, Barankiewicz T, Yang YM, Zanetti M, Hill P. Protection of IgE-mediated allergic sensitization by active immunization with IgE loops constrained in GFP protein scaffold. J Immunol Methods 2007; 333:10-23. [PMID: 18281056 DOI: 10.1016/j.jim.2007.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Accepted: 10/04/2007] [Indexed: 10/22/2022]
Abstract
Green fluorescent protein (GFP) exhibits a rigid central beta-barrel, formed by eleven beta-strands with floppy loops spanning between the stands. Herein, we evaluate whether the rigid beta-barrel may serve as a scaffold that can constrain the loops of a foreign protein, and thus its antigenicity. The spanning loops, site 6 of GFP, were engineered with RE cloning sites for inserting oligonucleotides corresponding to FcepsilonRI-binding sequence of human IgE. In a high-throughput format, shortened oligonucleotides encoding eight amino acid residues of the receptor-binding regions were inserted into site 6 of GFP by PCR, followed by enabling sequences for in vitro transcription and translation at the 5' end. Antigenized C2-3 linker (C2-3L) was shown by immuno-blots with polyclonal anti-IgE under native gel electrophoresis and transfer. Recombinant antigenized GFP was expressed and purified to homogeneity by metal affinity column, followed by Sephacryl S-200 high resolution gel filtration. Hyperimmune sera from mice immunized with C2-3L antigenized GFP contain anti-IgE reactive with JW8 murine/human chimeric IgE. Further, elevated serum anti-C2-3L and affinity pure antibodies effectively inhibits binding of JW8 IgE to recombinant FcepsilonRIalpha, and desensitizes JW8 to rat RBL-2H3 transfected with human FcepsilonRIalpha. This observation raised the possibility that active IgE vaccine may be employed in raising active protective anti-IgE in allergic patients as an alternative to passive immunization with MAb-E25 anti-IgE. Taken together, GFP appears suitable protein scaffold for spanning/constraining the C2-3L of human IgE as active vaccine; and this technique may be generally employed for eliciting antibodies to specific B-cell epitopes of other proteins.
Collapse
Affiliation(s)
- Swey-Shen Chen
- Department of Allergy and Immunology, The Institute of Genetics, San Diego, CA 92121, USA.
| | | | | | | | | |
Collapse
|
34
|
Medhekar B, Miller JF. Diversity-generating retroelements. Curr Opin Microbiol 2007; 10:388-95. [PMID: 17703991 PMCID: PMC2703298 DOI: 10.1016/j.mib.2007.06.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 06/01/2007] [Accepted: 06/01/2007] [Indexed: 01/09/2023]
Abstract
Parasite adaptation to dynamic host characteristics is a recurrent theme in biology. Diversity-generating retroelements (DGRs) are a newly discovered family of genetic elements that function to diversify DNA sequences and the proteins they encode. The prototype DGR was identified in a temperate bacteriophage, BPP-1, on the basis of its ability to generate variability in a gene that specifies tropism for receptor molecules on host Bordetella species. Tropism switching is a template-dependent, reverse transcriptase mediated process that introduces nucleotide substitutions at defined locations within a target gene. This cassette-based mechanism is theoretically capable of generating trillions of different amino acid sequences in a distal tail fiber protein, providing a vast repertoire of potential ligand-receptor interactions. Variable residues are displayed in the context of a specialized C-type lectin fold, which has evolved a unique solution for balancing protein diversity against structural stability. Homologous DGRs have been identified in the chromosomes of diverse bacterial species. These unique genetic elements have the potential to confer powerful selective advantages to their hosts, and their ability to generate novel binding specificities and dynamic antimicrobial agents suggests numerous applications.
Collapse
Affiliation(s)
- Bob Medhekar
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jeff F Miller
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- The Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- The California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
- Correspondence E-mail ; Tel. (+1) 310 206 7926; Fax (+1) 310 206 3865
| |
Collapse
|
35
|
Rich RL, Myszka DG. Survey of the year 2006 commercial optical biosensor literature. J Mol Recognit 2007; 20:300-66. [DOI: 10.1002/jmr.862] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|