1
|
Hifumi E, Ito Y, Tsujita M, Taguchi H, Uda T. Enzymatization of mouse monoclonal antibodies to the corresponding catalytic antibodies. Sci Rep 2024; 14:12184. [PMID: 38806597 PMCID: PMC11133420 DOI: 10.1038/s41598-024-63116-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024] Open
Abstract
Catalytic antibodies possess a dual function that enables both antigen recognition and degradation. However, their time-consuming preparation is a significant drawback. This study developed a new method for quickly converting mice monoclonal antibodies into catalytic antibodies using site-directed mutagenesis. Three mice type monoclonal antibodies targeting hemagglutinin molecule of influenza A virus could be transformed into the catalytic antibodies by deleting Pro95 in CDR-3 of the light chain. No catalytic activity was observed for monoclonal antibodies and light chains. In contrast, the Pro95-deleted light chains exhibited a catalytic activity to cleave the antigenic peptide including the portion of conserved region of hemagglutinin molecule. The affinity of the Pro95-deleted light chains to the antigen increased approximately 100-fold compared to the wild-type light chains. In the mutants, three residues (Asp1, Ser92, and His93) come closer to the appropriate position to create the catalytic site and contributing to the enhancement of both catalytic function and immunoreactivity. Notably, the Pro95-deleted catalytic light chains could suppress influenza virus infection in vitro assay, whereas the parent antibody and the light chain did not. This strategy offers a rapid and efficient way to create catalytic antibodies from existing antibodies, accelerating the development for various applications in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Emi Hifumi
- Institute for Research Management, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan.
- Research Center for GLOBAL/LOCAL Infectious Diseases, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan.
| | - Yuina Ito
- Institute for Research Management, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan
- Graduate School of Engineering, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan
| | - Moe Tsujita
- Institute for Research Management, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan
- Graduate School of Engineering, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan
| | - Hiroaki Taguchi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki-cho, Suzuka, 510-0293, Japan
| | - Taizo Uda
- Institute for Research Management, Oita University, 700 Dannoharu, Oita-shi, Oita, 870-1192, Japan
- Materials Open Laboratory, Institute of Systems, Information Technologies and Nanotechnologies (ISIT), Fukuoka, 819-0388, Japan
| |
Collapse
|
2
|
Hoang PT, Luong QXT, Ayun RQ, Lee Y, Oh KJ, Kim T, Lee TK, Lee S. A synergistic therapy against influenza virus A/H1N1/PR8 by a HA1 specific neutralizing single-domain V L and an RNA hydrolyzing scFv. Front Microbiol 2024; 15:1355599. [PMID: 38706966 PMCID: PMC11066198 DOI: 10.3389/fmicb.2024.1355599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
The emergence of anti-influenza drug-resistant strains poses a challenge for influenza therapy due to mutations in the virus's surface protein. Recently, there has been increasing interest in combination therapy consisting of two or more drugs as a potential alternative approach, aiming to enhance therapeutic efficacy. In this study, we investigated a novel synergistic therapy with a vertical effect using a single-domain VL-HA1-specific antibody against H1N1/PR8 and a horizontal effect using an RNA catalytic antibody with broad-spectrum influenza antiviral drug. We isolated a single-domain VL-HA1-specific (NVLH8) antibody binding to the virus particles showing a neutralizing activity against influenza virus A, specifically H1N1/PR8, as determined by the reduction in plaque number and lower viral HA protein expression in vitro. The neutralizing antibody likely prevented the viral entry, specifically at the viral genome-releasing step. Additionally, the 3D8 scFv hydrolyzed viral RNAs in the cytoplasm, including mRNA, vRNA, and cRNA in MDCK cells. The combined treatment of neutralizing antibodies for a vertical effect and 3D8 scFv for a horizontal effect produced a synergistic effect providing a novel approach against viral diseases when compared with a single treatment. Our results indicated that combining treatment, in particular two proteins exhibiting different mechanisms of action increased the antiviral activity against the influenza virus.
Collapse
Affiliation(s)
- Phuong Thi Hoang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Novelgen Co., Ltd., R&D Center, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Quynh Xuan Thi Luong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ramadhani Qurrota Ayun
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yongjun Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Ji Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Novelgen Co., Ltd., R&D Center, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Taehyun Kim
- Novelgen Co., Ltd., R&D Center, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Taek-Kyun Lee
- Risk Assessment Research Center, Korea Institute of Ocean Science and Technology, Geoje, Republic of Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
3
|
Luong QXT, Hoang PT, Lee Y, Ayun RQ, Na K, Park S, Lin C, Ho PT, Lee TK, Lee S. An RNA-hydrolyzing recombinant minibody prevents both influenza A virus and coronavirus in co-infection models. Sci Rep 2024; 14:8472. [PMID: 38605110 PMCID: PMC11009316 DOI: 10.1038/s41598-024-52810-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/23/2024] [Indexed: 04/13/2024] Open
Abstract
With the lifting of COVID-19 non-pharmaceutical interventions, the resurgence of common viral respiratory infections was recorded in several countries worldwide. It facilitates viral co-infection, further burdens the already over-stretched healthcare systems. Racing to find co-infection-associated efficacy therapeutic agents need to be rapidly established. However, it has encountered numerous challenges that necessitate careful investigation. Here, we introduce a potential recombinant minibody-associated treatment, 3D8 single chain variable fragment (scFv), which has been developed as a broad-spectrum antiviral drug that acts via its nucleic acid catalytic and cell penetration abilities. In this research, we demonstrated that 3D8 scFv exerted antiviral activity simultaneously against both influenza A viruses (IAVs) and coronaviruses in three established co-infection models comprising two types of coronaviruses [beta coronavirus-human coronavirus OC43 (hCoV-OC43) and alpha coronavirus-porcine epidemic diarrhea virus (PEDV)] in Vero E6 cells, two IAVs [A/Puerto Rico/8/1934 H1N1 (H1N1/PR8) and A/X-31 (H3N2/X-31)] in MDCK cells, and a combination of coronavirus and IAV (hCoV-OC43 and adapted-H1N1) in Vero E6 cells by a statistically significant reduction in viral gene expression, proteins level, and approximately around 85%, 65%, and 80% of the progeny of 'hCoV-OC43-PEDV', 'H1N1/PR8-H3N2/X-31', and 'hCoV-OC43-adapted-H1N1', respectively, were decimated in the presence of 3D8 scFv. Taken together, we propose that 3D8 scFv is a promising broad-spectrum drug for treatment against RNA viruses in co-infection.
Collapse
Affiliation(s)
- Quynh Xuan Thi Luong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Phuong Thi Hoang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Yongjun Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | | | - Kyungho Na
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Seonhyeon Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Chengmin Lin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Phuong Thi Ho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea
| | - Taek-Kyun Lee
- Ecological Risk Research Department, Korea Institute of Ocean Science & Technology, Geoje, 53201, Korea.
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
4
|
Zavialova M, Kamaeva D, Kazieva L, Skvortsov VS, Smirnova L. Some structural features of the peptide profile of myelin basic protein-hydrolyzing antibodies in schizophrenic patients. PeerJ 2023; 11:e15584. [PMID: 37431466 PMCID: PMC10329820 DOI: 10.7717/peerj.15584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/26/2023] [Indexed: 07/12/2023] Open
Abstract
The antibodies of schizophrenic patients that hydrolyze myelin basic protein (MBP) have been actively studied recently, but the mechanism of the catalytic properties of immunoglobulin molecules remains unknown. Determination of specific immunoglobulin sequences associated with the high activity of MBP proteolysis will help to understand the mechanisms of abzyme catalysis. In the course of comparative mass spectrometric analysis of IgG peptides from the blood serum of patients with acute schizophrenia and healthy people, 12 sequences were identified, which were found only in antibodies that hydrolyze MBP. These sequences belong to IgG heavy chains and κ- and λ-type light chains, with eight of them belonging to variable domains. The content of peptides from the variable regions of the light chains does not correlate with the proteolytic activity of IgG to MBP in patients with schizophrenia, whereas for two sequences from the variable regions of the heavy chains (FQ(+0.98)GWVTMTR and *LYLQMN(+0.98)SLR), an increase in activity with increasing their concentration. The results suggest that these sequences may be involved in one way or another in MBP hydrolysis.
Collapse
Affiliation(s)
| | - Daria Kamaeva
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | | | | | - Liudmila Smirnova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
5
|
Zhou M, Abid M, Cao S, Zhu S. Progress of Research into Novel Drugs and Potential Drug Targets against Porcine Pseudorabies Virus. Viruses 2022; 14:v14081753. [PMID: 36016377 PMCID: PMC9416328 DOI: 10.3390/v14081753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/06/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
Pseudorabies virus (PRV) is the causative agent of pseudorabies (PR), infecting most mammals and some birds. It has been prevalent around the world and caused huge economic losses to the swine industry since its discovery. At present, the prevention of PRV is mainly through vaccination; there are few specific antivirals against PRV, but it is possible to treat PRV infection effectively with drugs. In recent years, some drugs have been reported to treat PR; however, the variety of anti-pseudorabies drugs is limited, and the underlying mechanism of the antiviral effect of some drugs is unclear. Therefore, it is necessary to explore new drug targets for PRV and develop economic and efficient drug resources for prevention and control of PRV. This review will focus on the research progress in drugs and drug targets against PRV in recent years, and discuss the future research prospects of anti-PRV drugs.
Collapse
Affiliation(s)
- Mo Zhou
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China
| | - Muhammad Abid
- Viral Oncogenesis Group, The Pirbright Institute, Ash Road Pirbright, Woking, Surrey GU24 0NF, UK
| | - Shinuo Cao
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China
- Correspondence: (S.C.); (S.Z.)
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China
- Correspondence: (S.C.); (S.Z.)
| |
Collapse
|
6
|
A Novel Approach of Antiviral Drugs Targeting Viral Genomes. Microorganisms 2022; 10:microorganisms10081552. [PMID: 36013970 PMCID: PMC9414836 DOI: 10.3390/microorganisms10081552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Outbreaks of viral diseases, which cause morbidity and mortality in animals and humans, are increasing annually worldwide. Vaccines, antiviral drugs, and antibody therapeutics are the most effective tools for combating viral infection. The ongoing coronavirus disease 2019 pandemic, in particular, raises an urgent need for the development of rapid and broad-spectrum therapeutics. Current antiviral drugs and antiviral antibodies, which are mostly specific at protein levels, have encountered difficulties because the rapid evolution of mutant viral strains resulted in drug resistance. Therefore, degrading viral genomes is considered a novel approach for developing antiviral drugs. The current article highlights all potent candidates that exhibit antiviral activity by digesting viral genomes such as RNases, RNA interference, interferon-stimulated genes 20, and CRISPR/Cas systems. Besides that, we introduce a potential single-chain variable fragment (scFv) that presents antiviral activity against various DNA and RNA viruses due to its unique nucleic acid hydrolyzing characteristic, promoting it as a promising candidate for broad-spectrum antiviral therapeutics.
Collapse
|
7
|
A Therapeutically Active Minibody Exhibits an Antiviral Activity in Oseltamivir-Resistant Influenza-Infected Mice via Direct Hydrolysis of Viral RNAs. Viruses 2022; 14:v14051105. [PMID: 35632846 PMCID: PMC9146509 DOI: 10.3390/v14051105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Emerging Oseltamivir-resistant influenza strains pose a critical public health threat due to antigenic shifts and drifts. We report an innovative strategy for controlling influenza A infections by use of a novel minibody of the 3D8 single chain variable fragment (scFv) showing intrinsic viral RNA hydrolyzing activity, cell penetration activity, and epidermal cell penetration ability. In this study, we examined 3D8 scFv’s antiviral activity in vitro on three different H1N1 influenza strains, one Oseltamivir-resistant (A/Korea/2785/2009pdm) strain, and two Oseltamivir-sensitive (A/PuertoRico/8/1934 and A/X-31) strains. Interestingly, the 3D8 scFv directly digested viral RNAs in the ribonucleoprotein complex. scFv’s reduction of influenza viral RNA including viral genomic RNA, complementary RNA, and messenger RNA during influenza A infection cycles indicated that this minibody targets all types of viral RNAs during the early, intermediate, and late stages of the virus’s life cycle. Moreover, we further addressed the antiviral effects of 3D8 scFv to investigate in vivo clinical outcomes of influenza-infected mice. Using both prophylactic and therapeutic treatments of intranasal administered 3D8 scFv, we found that Oseltamivir-resistant H1N1-infected mice showed 90% (prophylactic effects) and 40% (therapeutic effects) increased survival rates, respectively, compared to the control group. The pathological signs of influenza A in the lung tissues, and quantitative analyses of the virus proliferations supported the antiviral activity of the 3D8 single chain variable fragment. Taken together, these results demonstrate that 3D8 scFv has antiviral therapeutic potentials against a wide range of influenza A viruses via the direct viral RNA hydrolyzing activity.
Collapse
|
8
|
Benn JA, Mukadam AS, McEwan WA. Targeted protein degradation using intracellular antibodies and its application to neurodegenerative disease. Semin Cell Dev Biol 2021; 126:138-149. [PMID: 34654628 DOI: 10.1016/j.semcdb.2021.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 01/10/2023]
Abstract
Antibodies mediate the majority of their effects in the extracellular domain, or in intracellular compartments isolated from the cytosol. Under a growing list of circumstances, however, antibodies are found to gain access to the cytoplasm. Cytosolic immune complexes are bound by the atypical antibody receptor TRIM21, which mediates the rapid degradation of the immune complexes at the proteasome. These discoveries have informed the development of TRIM-Away, a technique to selectively deplete proteins using delivery of antibodies into cells. A range of related approaches that elicit selective protein degradation using intracellular constructs linking antibody fragments to degradative effector functions have also been developed. These methods hold promise for inducing the degradation of proteins as both research tools and as a novel therapeutic approach. Protein aggregates are a pathophysiological feature of neurodegenerative diseases and are considered to have a causal role in pathology. Immunotherapy is emerging as a promising route towards their selective targeting, and a role of antibodies in the cytosol has been demonstrated in cell-based assays. This review will explore the mechanisms by which therapeutic antibodies engage and eliminate intracellularly aggregated proteins. We will discuss how future developments in intracellular antibody technology may enhance the therapeutic potential of such antibody-derived therapies.
Collapse
Affiliation(s)
- Jonathan A Benn
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK
| | - Aamir S Mukadam
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK.
| |
Collapse
|
9
|
Lee G, Budhathoki S, Lee GY, Oh KJ, Ham YK, Kim YJ, Lim YR, Hoang PT, Lee Y, Lim SW, Kim JM, Cho S, Kim TH, Song JW, Lee S, Kim WK. Broad-Spectrum Antiviral Activity of 3D8, a Nucleic Acid-Hydrolyzing Single-Chain Variable Fragment (scFv), Targeting SARS-CoV-2 and Multiple Coronaviruses In Vitro. Viruses 2021; 13:650. [PMID: 33918914 PMCID: PMC8068894 DOI: 10.3390/v13040650] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 11/25/2022] Open
Abstract
The virus behind the current pandemic, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the etiology of novel coronavirus disease (COVID-19) and poses a critical public health threat worldwide. Effective therapeutics and vaccines against multiple coronaviruses remain unavailable. Single-chain variable fragment (scFv), a recombinant antibody, exhibits broad-spectrum antiviral activity against DNA and RNA viruses owing to its nucleic acid-hydrolyzing property. The antiviral activity of 3D8 scFv against SARS-CoV-2 and other coronaviruses was evaluated in Vero E6 cell cultures. Viral growth was quantified with quantitative RT-qPCR and plaque assay. The nucleic acid-hydrolyzing activity of 3D8 was assessed through abzyme assays of in vitro viral transcripts and cell viability was determined by MTT assay. We found that 3D8 inhibited the replication of SARS-CoV-2, human coronavirus OC43 (HCoV-OC43), and porcine epidemic diarrhea virus (PEDV). Our results revealed the prophylactic and therapeutic effects of 3D8 scFv against SARS-CoV-2 in Vero E6 cells. Immunoblot and plaque assays showed the reduction of coronavirus nucleoproteins and infectious particles, respectively, in 3D8 scFv-treated cells. These data demonstrate the broad-spectrum antiviral activity of 3D8 against SARS-CoV-2 and other coronaviruses. Thus, it could be considered a potential antiviral countermeasure against SARS-CoV-2 and zoonotic coronaviruses.
Collapse
Affiliation(s)
- Gunsup Lee
- R&D Center, Novelgen Co., Ltd., 77, Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Korea; (G.L.); (K.-j.O.); (Y.K.H.); (Y.-J.K.); (Y.R.L.); (T.-H.K.)
| | - Shailesh Budhathoki
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Korea;
| | - Geum-Young Lee
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.-W.S.)
| | - Kwang-ji Oh
- R&D Center, Novelgen Co., Ltd., 77, Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Korea; (G.L.); (K.-j.O.); (Y.K.H.); (Y.-J.K.); (Y.R.L.); (T.-H.K.)
| | - Yeon Kyoung Ham
- R&D Center, Novelgen Co., Ltd., 77, Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Korea; (G.L.); (K.-j.O.); (Y.K.H.); (Y.-J.K.); (Y.R.L.); (T.-H.K.)
| | - Young-Jun Kim
- R&D Center, Novelgen Co., Ltd., 77, Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Korea; (G.L.); (K.-j.O.); (Y.K.H.); (Y.-J.K.); (Y.R.L.); (T.-H.K.)
| | - Ye Rin Lim
- R&D Center, Novelgen Co., Ltd., 77, Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Korea; (G.L.); (K.-j.O.); (Y.K.H.); (Y.-J.K.); (Y.R.L.); (T.-H.K.)
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Phuong Thi Hoang
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea; (P.T.H.); (Y.L.)
| | - Yongjun Lee
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea; (P.T.H.); (Y.L.)
| | - Seok-Won Lim
- Animal Functional Genomics & Bioinformatics Lab., Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea; (S.-W.L.); (J.-M.K.)
| | - Jun-Mo Kim
- Animal Functional Genomics & Bioinformatics Lab., Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea; (S.-W.L.); (J.-M.K.)
| | - Seungchan Cho
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.-W.S.)
| | - Tai-Hyun Kim
- R&D Center, Novelgen Co., Ltd., 77, Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Korea; (G.L.); (K.-j.O.); (Y.K.H.); (Y.-J.K.); (Y.R.L.); (T.-H.K.)
| | - Jin-Won Song
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea; (G.-Y.L.); (S.C.); (J.-W.S.)
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Sukchan Lee
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea; (P.T.H.); (Y.L.)
| | - Won-Keun Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Korea;
- Institute of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
10
|
Sureshkumar S, Jung SK, Kim D, Oh KB, Yang H, Lee HC, Jo YJ, Lee HS, Lee S, Byun SJ. Administration of L. salivarius expressing 3D8 scFv as a feed additive improved the growth performance, immune homeostasis, and gut microbiota of chickens. Anim Sci J 2020; 91:e13399. [PMID: 32512648 DOI: 10.1111/asj.13399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/02/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022]
Abstract
Probiotics have been defined as live microorganisms that are administered in an appropriate amount to provide health benefits to the host animal. In this study, we investigated the effect of L. salivarius DJ-sa-01 secreting the 3D8 single-chain variable fragment (3D8 scFv) on the growth performance, cytokine secretion, and intestinal microbial flora of chickens. The experiment was divided into the control group and L. salivarius expressing 3D8 scFv experimental group. Chicken was fed 109 colony-forming units (CFUs) of wild-type (WT) L. salivarius or 3D8 scFv-secreting L. salivarius daily for 35 days. The administration of L. salivarius expressing 3D8 scFv significantly improved the body weight of chickens compared with the administration of WT L. salivarius. A 16S ribosomal RNA metagenomic analysis showed that Firmicutes, Proteobacteria, Actinobacteria, and Bacteroidetes were the dominant phyla in both experimental groups. At the genus level, Lactobacillus was more abundant (22.82%) in the L. salivarius/3D8 group compared with the WT L. salivarius group. The serum levels of cytokines, such as IL-8, TNF-α, IL-1β, IFN-γ, IL-4, and IGF1, were significantly reduced in the L. salivarius/3D8-treated chickens. In summary, our results suggest that L. salivarius expressing 3D8 scFv could be considered a feed additive for improving the growth performance, immune function, and disease resistance of poultry.
Collapse
Affiliation(s)
- Shanmugam Sureshkumar
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea.,Department of Animal Resource and Science, Dankook University, Cheonan, Republic of Korea
| | - Sun Keun Jung
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Dongjun Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Gyeonggi-do, Republic of Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Hyeon Yang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Hwi Cheul Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Yong Jin Jo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Hae Sun Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Gyeonggi-do, Republic of Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| |
Collapse
|
11
|
Gordon RE, Nemeth JF, Singh S, Lingham RB, Grewal IS. Harnessing SLE Autoantibodies for Intracellular Delivery of Biologic Therapeutics. Trends Biotechnol 2020; 39:298-310. [PMID: 32807530 DOI: 10.1016/j.tibtech.2020.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Intracellular delivery of therapeutic antibodies is highly desirable but remains a challenge for biomedical research and the pharmaceutical industry. Approximately two-thirds of disease-associated targets are found inside the cell. Difficulty blocking these targets with available drugs creates a need for technology to deliver highly specific therapeutic antibodies intracellularly. Historically, antibodies have not been believed to traverse the cell membrane and neutralize intracellular targets. Emerging evidence has revealed that anti-DNA autoantibodies found in systemic lupus erythematosus (SLE) patients can penetrate inside the cell. Harnessing this technology has the potential to accelerate the development of drugs against intracellular targets. Here, we dissect the mechanisms of the intracellular localization of SLE antibodies and discuss how to apply these insights to engineer successful cell-penetrating antibody drugs.
Collapse
Affiliation(s)
- Renata E Gordon
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Jennifer F Nemeth
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Sanjaya Singh
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Russell B Lingham
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA
| | - Iqbal S Grewal
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, 1400 McKean Road, Spring House, PA 19477, USA.
| |
Collapse
|
12
|
Byun SJ, Choi H, Sureshkumar S, Yuk SS, Kwon JH, Noh JY, Jung SK, Kim JS, Oh KB, Yang H, Lee G, Lee HC, Woo JS, Song CS. The 3D8 single chain variable fragment protein suppresses Newcastle disease virus transmission in transgenic chickens. BMC Vet Res 2020; 16:273. [PMID: 32762754 PMCID: PMC7409462 DOI: 10.1186/s12917-020-02462-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/06/2020] [Indexed: 02/01/2023] Open
Abstract
Background The 3D8 single chain variable fragment (scFv) is a mini-antibody sequence that exhibits independent nuclease activity against all types of nucleic acids. In this research, crossing a 3D8 scFv G1 transgenic rooster with wild-type hens produced 3D8 scFv G2 transgenic chickens to evaluate suppression of viral transmission. Result The transgenic chickens were identified using genomic PCR and immunohistochemistry. To evaluate Newcastle disease virus (NDV) protection conferred by 3D8 scFv expression, transgenic, non-transgenic, and specific pathogen-free (SPF) chickens were challenged with virulent NDV by direct injection or aerosol exposure. The three groups of chickens showed no significant differences (p < 0.05) in mean death time after being directly challenged with NDV; however, in contrast to chickens in the non-transgenic and SPF groups, chickens in the transgenic group survived after aerosol exposure. Although the transgenic chickens did not survive after direct challenge, we found that the chickens expressing the 3D8 scFv survived aerosol exposure to NDV. Conclusions Our finding suggest that the 3D8 scFv could be a useful tool to prevent chickens from spreading NDV and control virus transmission.
Collapse
Affiliation(s)
- Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Hoonsung Choi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Shanmugam Sureshkumar
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Seong-Su Yuk
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, 05029, Seoul, Republic of Korea
| | - Jung-Hoon Kwon
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, 05029, Seoul, Republic of Korea
| | - Jin-Yong Noh
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, 05029, Seoul, Republic of Korea
| | - Sun Keun Jung
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Jeom Sun Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Hyeon Yang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Gunsup Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Hwi-Cheul Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Jae-Seok Woo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 55365, Wanju-gun, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, 05029, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Choi H, Lee SI, Sureshkumar S, Jeon MH, Kim JS, Park MR, Kim KW, Jeon IS, Lee S, Byun SJ. Avian influenza virus transmission is suppressed in chickens fed Lactobacillus paracasei expressing the 3D8 single-chain variable fragment protein. Acta Vet Hung 2019; 67:610-618. [PMID: 31842597 DOI: 10.1556/004.2019.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The 3D8 single-chain variable fragment (scFv) is a mini-antibody sequence with independent nuclease activity that shows antiviral effects against all types of viruses in chickens and mice. In this study, chickens were treated daily with an oral dose of 109 CFU Lactobacillus paracasei (L. paracasei) expressing either a secreted or anchored 3D8 scFv for three weeks. After L. paracasei administration, the chickens were challenged with avian influenza virus (AIV). From each experimental group, three chickens were directly infected with 100 µL of 107.5 EID50/mL H9N2 AIV and seven chickens were indirectly challenged through contact transmission. oropharyngeal and cloacal swab samples were collected at 3, 5, 7, and 9 days post-inoculation (dpi) from AIV-challenged chickens, AIV Shedding titres were measured by quantitative real-time PCR. Contact transmission in the chickens that were fed 3D8 scFv-secreting L. paracasei showed a significant reduction in viral shedding when compared with other groups. These results suggest that L. paracasei secreting 3D8 provides a basis for the development of ingestible antiviral probiotics with activity against AIV.
Collapse
Affiliation(s)
- Hoonsung Choi
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Sang In Lee
- 2Department of Animal Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do, Republic of Korea
| | - Shanmugam Sureshkumar
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Mi-Hyang Jeon
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Jeom Sun Kim
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Mi-Ryung Park
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Kyung-Woon Kim
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Ik-Soo Jeon
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Sukchan Lee
- 3Department of Genetic Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Republic of Korea
| | - Sung June Byun
- 1Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| |
Collapse
|
14
|
Atomic resolution cryo-EM structure of a native-like CENP-A nucleosome aided by an antibody fragment. Nat Commun 2019; 10:2301. [PMID: 31127102 PMCID: PMC6534667 DOI: 10.1038/s41467-019-10247-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/26/2019] [Indexed: 12/20/2022] Open
Abstract
Genomic DNA in eukaryotes is organized into chromatin through association with core histones to form nucleosomes, each distinguished by their DNA sequences and histone variants. Here, we used a single-chain antibody fragment (scFv) derived from the anti-nucleosome antibody mAb PL2-6 to stabilize human CENP-A nucleosome containing a native α-satellite DNA and solved its structure by the cryo-electron microscopy (cryo-EM) to 2.6 Å resolution. In comparison, the corresponding cryo-EM structure of the free CENP-A nucleosome could only reach 3.4 Å resolution. We find that scFv binds to a conserved acidic patch on the histone H2A-H2B dimer without perturbing the nucleosome structure. Our results provide an atomic resolution cryo-EM structure of a nucleosome and insight into the structure and function of the CENP-A nucleosome. The scFv approach is applicable to the structural determination of other native-like nucleosomes with distinct DNA sequences. CENP-A histone variants replace histones H3 at centromeres. Here the authors use a single-chain antibody fragment (scFv) to stabilize human CENP-A nucleosome containing a native α-satellite DNA and solved its structure by cryo-EM to 2.6 Å resolution, providing insight into the structure and function of the CENP-A nucleosome.
Collapse
|
15
|
Lee G, Choi H, Sureshkumar S, Jung SK, Kim JS, Oh KB, Kim KW, Yang H, Kim DH, Byun SJ. The 3D8 single chain variable fragment protein suppress infectious bronchitis virus transmission in the transgenic chickens. Res Vet Sci 2019; 123:293-297. [PMID: 30738233 PMCID: PMC7111750 DOI: 10.1016/j.rvsc.2019.01.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/28/2018] [Accepted: 01/22/2019] [Indexed: 11/29/2022]
Abstract
Infectious bronchitis (IB) generated by the infectious bronchitis virus (IBV) causes economic difficulties for livestock farmers. The 3D8 single chain variable fragment (scFv) protein is a recombinant antibody with nuclease activity that shows antiviral effects against various DNA and RNA viruses in mice and chickens. In this experiment, 3D8 scFv G2 transgenic chickens produced by crossing 3D8 scFv G1 transgenic rooster and wild type hens were screened by genomic PCR and immunohistochemistry analysis. 3D8 scFv transgenic chickens, wild type sibling chickens, and SPF chickens were directly infected with IBV (5 chickens per group) and indirectly infected by airborne propagation (15 chickens per group). The relative IBV shedding titers were measured by quantitative real-time PCR using oropharyngeal and cloacal swabs on days 3 and 5 after intraocular infection. The viral load was significantly decreased in the 3D8 scFv transgenic chickens from the contact transmission group. Additionally, blood was collected from each group on day 17 post-infection. The ELISA results showed a marked reduction of the antibody titer against IBV in the 3D8 scFv transgenic chickens from the contact transmission group. These results suggest that the 3D8 scFv protein potentially inhibits infectious bronchitis virus transmission in chickens. Produced G2 3D8 single chain variable fragment (scFv) transgenic chickens. 3D8 scFv transgenic chickens showed reduced infectious bronchitis viral shedding level in the contact transmission group. 3D8 scFv transgenic chickens were 40% lower than the response in the control groups in IBV serum antibody titer.
Collapse
Affiliation(s)
- Gunsup Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea; Gyeongbuk Institute for Marine Bio-Industry (GIMB), Uljin 36315, Gyeongbuk, Republic of Korea
| | - Hoonsung Choi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Shanmugam Sureshkumar
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Sun Keun Jung
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Jeom Sun Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Kyung-Woon Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Hyeon Yang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Dong-Hoon Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Wanju-gun 441-706, Republic of Korea.
| |
Collapse
|
16
|
Park H, Kim M, Seo Y, Ham Y, Cho MY, Kwon MH. Cytosolic Internalization of Anti-DNA Antibodies by Human Monocytes Induces Production of Pro-inflammatory Cytokines Independently of the Tripartite Motif-Containing 21 (TRIM21)-Mediated Pathway. Front Immunol 2018; 9:2019. [PMID: 30233598 PMCID: PMC6131520 DOI: 10.3389/fimmu.2018.02019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/16/2018] [Indexed: 01/05/2023] Open
Abstract
Anti-DNA autoantibodies are a hallmark of systemic lupus erythematosus (SLE). A subset of anti-DNA IgG autoantibodies is cell-internalizable; thus they can enter living cells in the form of free IgG. However, the contribution made by the Fc region of internalized free-form IgG to the cytokine response has not been studied, despite the recent discovery of tripartite motif-containing 21 (TRIM21), a cytosolic Fc receptor involved in immune signaling. This study used an internalizable IgG anti-DNA antibody (3D8) to examine the cytokine responses of human monocytes to the Fc region of cytosolic free IgG. Internalization of 3D8 IgG and a 3D8 single-chain variable fragment-Fc (scFv-Fc) induced production of IL-8 and TNF-α via activation of NF-κB. By contrast, a 3D8 scFv (comprising variable domains alone) did not. This suggests Fc-dependent cytokine signaling. A 3D8 IgG-N434D mutant that is not recognized by TRIM21 induced greater production of cytokines than 3D8 IgG. Moreover the amounts of cytokines induced by 3D8 IgG in TRIM21-knockdown THP-1 cells were higher than those in control cells, indicating that cytokine signaling is not mediated by TRIM21. The results suggest the existence of a novel Fc-dependent signaling pathway that is activated upon internalization of IgG antibodies by human monocytes.
Collapse
Affiliation(s)
- Hyunjoon Park
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Minjae Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Yeonkyoung Ham
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Mi-Young Cho
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Myung-Hee Kwon
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
17
|
Probiotic Lactobacillus Paracasei Expressing a Nucleic Acid-Hydrolyzing Minibody (3D8 Scfv) Enhances Probiotic Activities in Mice Intestine as Revealed by Metagenomic Analyses. Genes (Basel) 2018; 9:genes9060276. [PMID: 29844265 PMCID: PMC6027128 DOI: 10.3390/genes9060276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 01/13/2023] Open
Abstract
Probiotics are well known for their beneficial effects for animals, including humans and livestock. Here, we tested the probiotic activity of Lactobacillus paracasei expressing 3D8 scFv, a nucleic acid-hydrolyzing mini-antibody, in mice intestine. A total of 18 fecal samples derived from three different conditions at two different time points were subjected to high-throughput 16S ribosomal RNA (rRNA) metagenomic analyses. Bioinformatic analyses identified an average of 290 operational taxonomic units. After administration of L. paracasei, populations of the probiotics L. paracasei, Lactobacillus reuteri, and Pediococcus acidilactici increased, whereas the population of harmful bacteria such as Helicobacter species decreased. Furthermore, continuous administration of L. paracasei resulted in L. paracasei emerging as the dominant probiotic after competition with other existing probiotics. Expression of 3D8 scFv protein specifically increased the population of P. acidilactici, which is another probiotic. In summary, our results showed that L. paracasei expressing 3D8 scFv protein enhanced probiotic activity in mice intestine with no observable side effects. Thus, the system developed in this study may be a good tool for the expression of recombinant protein using probiotics.
Collapse
|
18
|
Park JH, Lee JW, Choi H, Jung SK, Kim JS, Kim KW, Oh KB, Yang H, Byun SJ. Survival of Escherichia coli harboring nucleic acid-hydrolyzing 3D8 scFv during RNA virus infection. Regul Toxicol Pharmacol 2018; 94:286-292. [PMID: 29486271 PMCID: PMC7115797 DOI: 10.1016/j.yrtph.2018.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/19/2018] [Accepted: 02/19/2018] [Indexed: 11/24/2022]
Abstract
Previously, Escherichia coli harboring the codon-optimized 3D8scFv gene (E. coli 3D8scFv) was developed as a feed additive for use in preventing norovirus infection. Here, we evaluated whether the 3D8scFv gene affects the colonization of E coli when E. coli 3D8scFv passes through the mouse gastrointestinal tract. To determine the colonization ability of E. coli 3D8scFv, E. coli cells with or without the 3D8scFv gene were fed to mice. Total DNA was extracted from the animals’ stools, stomach, small intestine and colon. All samples were amplified using 3D8scFv gene-specific primer sets. E. coli 3D8scFv begins to be excreted 1 h after feeding and that all E. coli 3D8scFv cells were excreted between 12 and 24 h after the last feeding of the cells. The previously measured gastrointestinal transit time of the mice was between 8 h and 22 h. The results of this study therefore show that E. coli 3D8scFv cannot colonize the gastrointestinal tracts of mice. In addition, if the purified 3D8 scFv protein is used as a feed additive, any associated E. coli 3D8scFv bacteria will not colonize the gastrointestinal tracts of the livestock. Thus, this feed additive meets the safety assessment criteria for the commercial use of bacteria. It is evaluated whether E. coli 3D8scFv colonizes in the gastrointestinal tracts of mice. Orally ingested E. coli 3D8scFv is excreted from mice without colonization of the gastrointestinal tract. Purified 3D8 scFv is suitable for a feed additive according to the concept of substantial equivalence.
Collapse
Affiliation(s)
- Jung-Ho Park
- Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk-do 28116, South Korea
| | - Jae-Woo Lee
- Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk-do 28116, South Korea; Department of Food Science and Technology, College of Agriculture and Life Sciences, Chungnam National University, Daejeon 34134, South Korea
| | - Hoonsung Choi
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Sun Keun Jung
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Jeom Sun Kim
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Kyung-Woon Kim
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Hyeon Yang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea.
| |
Collapse
|
19
|
Park H, Kim M, Kim HJ, Lee Y, Seo Y, Pham CD, Lee J, Byun SJ, Kwon MH. Heparan sulfate proteoglycans (HSPGs) and chondroitin sulfate proteoglycans (CSPGs) function as endocytic receptors for an internalizing anti-nucleic acid antibody. Sci Rep 2017; 7:14373. [PMID: 29085061 PMCID: PMC5662561 DOI: 10.1038/s41598-017-14793-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/16/2017] [Indexed: 02/02/2023] Open
Abstract
A subset of monoclonal anti-DNA autoantibodies enters a variety of living cells. Here, we aimed to identify the endocytic receptors recognized by an internalizing anti-nucleic acid autoantibody, the 3D8 single-chain variable fragment (scFv). We found that cell surface binding and internalization of 3D8 scFv were inhibited markedly in soluble heparan sulfate (HS)/chondroitin sulfate (CS)-deficient or -removed cells and in the presence of soluble HS and CS. 3D8 scFv colocalized intracellularly with either HS proteoglycans (HSPGs) or CSPGs in HeLa cells. 3D8 scFv was co-endocytosed and co-precipitated with representative individual HSPG and CSPG molecules: syndecan-2 (a transmembrane HSPG), glypican-3 (a glycosylphosphatidylinositol (GPI)-anchored HSPG); CD44 (a transmembrane CSPG); and brevican (a GPI-anchored CSPG). Collected data indicate that 3D8 scFv binds to the negatively charged sugar chains of both HSPGs and CSPGs and is then internalized along with these molecules, irrespective of how these proteoglycans are associated with the cell membrane. This is the first study to show that anti-DNA antibodies enter cells via both HSPGs and CSPGs simultaneously. The data may aid understanding of endocytic receptors that bind anti-DNA autoantibodies. The study also provides insight into potential cell membrane targets for macromolecular delivery.
Collapse
Affiliation(s)
- Hyunjoon Park
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Minjae Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Hye-Jin Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Yeonjin Lee
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Chuong D Pham
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Joungmin Lee
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 565-851, South Korea
| | - Myung-Hee Kwon
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea. .,Department of Microbiology, Ajou University School of Medicine, 206 World cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea.
| |
Collapse
|
20
|
Choi H, Jung SK, Kim JS, Kim KW, Oh KB, Lee PY, Byun SJ. Effects of dietary recombinant chlorella supplementation on growth performance, meat quality, blood characteristics, excreta microflora, and nutrient digestibility in broilers. Poult Sci 2017; 96:710-716. [PMID: 27697932 DOI: 10.3382/ps/pew345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/11/2016] [Indexed: 11/20/2022] Open
Abstract
The use of chlorella as an immune stimulant to enhance nonspecific host defense mechanisms or as an antimicrobial to inhibit bacterial growth has been reported. Thus, the aim of the present study was to clarify the effect of recombinant chlorella supplementation on growth performance, meat quality, and the blood profile, excreta microflora, and nutrient digestibility in broilers. A total of 375 one-day-old ROSS 308 broilers (male and female) were allotted to 5 dietary treatments using 5 cages with 15 chicks per cage. Treatments were: 1) NC, basal diet supplemented with 1.0% E. coli fermented liquor (EFL); 2) PC1, 0.2% EFL with chlorella; 3) PC2, 1.0% EFL with chlorella; 4) T1, 0.2% EFL with chlorella (anti-viral); and 5) T2, 1.0% EFL with chlorella (anti-viral). The broilers in the T2 treatment groups showed higher body weight gain (BGW) by 2.55% (P < 0.01) and lower feed conversion ratio (FCR) by 2.75% (P < 0.05) compared with those fed the control NC treatment group. Moreover, the blood contents of blood urea nitrogen (BUN), creatinine, and IgA in the broilers of the T2 treatment group were significantly increased by 28.12, 23.07, and 29.72%, respectively -more than those found in the broilers of the NC treatment group (P < 0.01). In contrast, the LDL/C in the blood from the animals in the T2 treatment group was significantly decreased by 23.23% - more than that in the blood from the NC broilers (P < 0.05). Based on these results, we suggest that the dietary supplementation of broilers with recombinant chlorella could improve their growth performance, increase the concentration of IgA and apparently metabolizable nitrogen in the blood, and decrease ammonia emissions. Therefore, our findings have important implications for the effect of recombinant chlorella supplementation through increasing the concentration of IgA and the level of metabolizable nitrogen.
Collapse
|
21
|
Lee J, Kim M, Seo Y, Lee Y, Park H, Byun SJ, Kwon MH. The catalytic activity of a recombinant single chain variable fragment nucleic acid-hydrolysing antibody varies with fusion tag and expression host. Arch Biochem Biophys 2017; 633:110-117. [PMID: 28888872 DOI: 10.1016/j.abb.2017.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022]
Abstract
The antigen-binding properties of single chain Fv antibodies (scFvs) can vary depending on the position and type of fusion tag used, as well as the host cells used for expression. The issue is even more complicated with a catalytic scFv antibody that binds and hydrolyses a specific antigen. Herein, we investigated the antigen-binding and -hydrolysing activities of the catalytic anti-nucleic acid antibody 3D8 scFv expressed in Escherichia coli or HEK293f cells with or without additional amino acid residues at the N- and C-termini. DNA-binding activity was retained in all recombinant forms. However, the DNA-hydrolysing activity varied drastically between forms. The DNA-hydrolysing activity of E. coli-derived 3D8 scFvs was not affected by the presence of a C-terminal human influenza haemagglutinin (HA) or His tag. By contrast, the activity of HEK293f-derived 3D8 scFvs was completely lost when additional residues were included at the N-terminus and/or when a His tag was incorporated at the C-terminus, whereas a HA tag at the C-terminus did not diminish activity. Thus, we demonstrate that the antigen-binding and catalytic activities of a catalytic antibody can be separately affected by the presence of additional residues at the N- and C-termini, and by the host cell type.
Collapse
Affiliation(s)
- Joungmin Lee
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea
| | - Minjae Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea
| | - Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea
| | - Yeonjin Lee
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea
| | - Hyunjoon Park
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, 55365, South Korea
| | - Myung-Hee Kwon
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea; Department of Microbiology, Ajou University School of Medicine, 206 World Cup-ro, Yeongtong-gu, Suwon 16499, South Korea.
| |
Collapse
|
22
|
June Byun S, Yuk SS, Jang YJ, Choi H, Jeon MH, Erdene-Ochir TO, Kwon JH, Noh JY, Sun Kim J, Gyu Yoo J, Song CS. Transgenic Chickens Expressing the 3D8 Single Chain Variable Fragment Protein Suppress Avian Influenza Transmission. Sci Rep 2017; 7:5938. [PMID: 28724948 PMCID: PMC5517518 DOI: 10.1038/s41598-017-05270-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 05/26/2017] [Indexed: 12/21/2022] Open
Abstract
The 3D8 single chain variable fragment (scFv) is a mini-antibody that causes unusual sequence-independent nuclease activity against all types of nucleic acids. We used recombinant lentiviruses to generate transgenic chickens expressing the 3D8 scFv gene under the control of the chicken β-actin promoter. From 420 injected embryos, 200 chicks (G0) hatched and were screened for the 3D8 scFv using PCR, and 15 chicks were identified as transgenic birds expressing the transgene in their semen. The G0 founder birds were mated with wild-type hens to produce seven transgenic chicks (G1). 3D8 scFv expression in the chicken embryonic fibroblasts (CEFs) was verified by RT-PCR and Western blot analysis. Immunofluorescence staining for 3D8 scFv in the CEFs revealed that the 3D8 scFv protein was primarily cytosolic. To identify 3D8 scFv anti-viral activity, wild-type and two transgenic CEF lines were infected with H9N2 avian influenza virus (AIV). We selected one line of transgenic chickens that exhibited the lowest number of plaque-forming units to be challenged with H9N2 virus. The challenge experiment revealed that contact exposed transgenic chickens expressing 3D8 scFv exhibited suppressed viral shedding. This results suggest that the transgenic chickens developed in this study could be useful for controlling potential within-flock AIV transmission.
Collapse
Affiliation(s)
- Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, 441-706, Republic of Korea
| | - Seong-Su Yuk
- Department of Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Ye-Jin Jang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, 441-706, Republic of Korea
| | - Hoonsung Choi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, 441-706, Republic of Korea
| | - Mi-Hyang Jeon
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, 441-706, Republic of Korea
| | - T O Erdene-Ochir
- Department of Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Jung-Hoon Kwon
- Department of Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Jin-Yong Noh
- Department of Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Jeom Sun Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, 441-706, Republic of Korea
| | - Jae Gyu Yoo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, 441-706, Republic of Korea
| | - Chang-Seon Song
- Department of Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Korea.
| |
Collapse
|
23
|
Fan CY, Huang SY, Chou MY, Lyu PC. De novo protein sequencing, humanization and in vitro effects of an antihuman CD34 mouse monoclonal antibody. Biochem Biophys Rep 2016; 9:51-60. [PMID: 28955989 PMCID: PMC5614552 DOI: 10.1016/j.bbrep.2016.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/25/2016] [Accepted: 11/15/2016] [Indexed: 01/08/2023] Open
Abstract
QBEND/10 is a mouse immunoglobulin lambda-chain monoclonal antibody with strict specificity against human hematopoietic progenitor cell antigen CD34. Our in vitro study showed that QBEND/10 impairs the tube formation of human umbilical vein endothelial cells (HUVECs), suggesting that the antibody may be of potential benefit in blocking tumor angiogenesis. We provided a de novo protein sequencing method through tandem mass spectrometry to identify the amino acid sequences in the variable heavy and light chains of QBEND/10. To reduce immunogenicity for clinical applications, QBEND/10 was further humanized using the resurfacing approach. We demonstrate that the de novo sequenced and humanized QBEND/10 retains the biological functions of the parental mouse counterpart, including the binding kinetics to CD34 and blockage of the tube formation of the HUVECs.
Collapse
Affiliation(s)
- Chia-Yu Fan
- Institute of Bioinformatics and Structural Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.,Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | | | - Min-Yuan Chou
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ping-Chiang Lyu
- Institute of Bioinformatics and Structural Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
24
|
Ermakov EA, Smirnova LP, Parkhomenko TA, Dmitrenok PS, Krotenko NM, Fattakhov NS, Bokhan NA, Semke AV, Ivanova SA, Buneva VN, Nevinsky GA. DNA-hydrolysing activity of IgG antibodies from the sera of patients with schizophrenia. Open Biol 2016; 5:150064. [PMID: 26382278 PMCID: PMC4593665 DOI: 10.1098/rsob.150064] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is believed that damage to the membranes of brain cells of schizophrenia (SCZ) patients induces the formation of autoantigens and autoantibodies. Nevertheless, the importance of immunological changes leading to the loss of tolerance to self-antigens in the genesis of SCZ has not been established. The MALDI mass spectra of the IgG light chains of 20 healthy donors were relatively homogeneous and characterized by one peak with only one maximum. In contrast to the healthy donors, the MALDI mass spectra of IgG light chains corresponding to 20 SCZ patients demonstrated, similarly to 20 autoimmune systemic lupus erythematosus (SLE) patients, two maxima of a comparable intensity. In addition, the MALDI spectra of the IgG light chains of five SLE and four SCZ patients contained a small additional brightly pronounced peak with remarkably lower molecular mass compared with the main one. DNase autoantibodies (abzymes) can be found in the blood of patients with several autoimmune diseases, while the blood of healthy donors or patients with diseases without a significant disturbance of the immune status does not contain DNase abzymes. Here, we present the first analysis of anti-DNA antibodies and DNase abzymes in the sera of SCZ patients. Several strict criteria have been applied to show that the DNase activity is an intrinsic property of IgGs from the sera of SCZ patients. The sera of approximately 30% of SCZ patients displayed a higher content of antibodies (compared with 37% of SLE) interacting with single- and double-stranded DNA compared with healthy donors. Antibodies with DNase activity were revealed in 80% of the patients. These data indicate that some SCZ patients may show signs of typical autoimmune processes to a certain extent.
Collapse
Affiliation(s)
- Evgeny A Ermakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk 630090, Russia Novosibirsk State University, 2 Pirogova Street, Novosibirsk 630090, Russia
| | - Ludmila P Smirnova
- Mental Health Research Institute, Russian Academy of Medical Sciences, 4 Aleutskaya Avenue, Tomsk 634014, Russia
| | - Taisiya A Parkhomenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk 630090, Russia
| | - Pavel S Dmitrenok
- Pacific Institute of Bioorganic Chemistry, Far East Division, Russian Academy of Sciences, Vladivostok 690022, Russia
| | - Nina M Krotenko
- Mental Health Research Institute, Russian Academy of Medical Sciences, 4 Aleutskaya Avenue, Tomsk 634014, Russia
| | - Nikolai S Fattakhov
- Mental Health Research Institute, Russian Academy of Medical Sciences, 4 Aleutskaya Avenue, Tomsk 634014, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Russian Academy of Medical Sciences, 4 Aleutskaya Avenue, Tomsk 634014, Russia
| | - Arkadiy V Semke
- Mental Health Research Institute, Russian Academy of Medical Sciences, 4 Aleutskaya Avenue, Tomsk 634014, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Russian Academy of Medical Sciences, 4 Aleutskaya Avenue, Tomsk 634014, Russia
| | - Valentina N Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk 630090, Russia Novosibirsk State University, 2 Pirogova Street, Novosibirsk 630090, Russia
| | - Georgy A Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk 630090, Russia Novosibirsk State University, 2 Pirogova Street, Novosibirsk 630090, Russia
| |
Collapse
|
25
|
Lee J, Park H, Kim M, Seo Y, Lee Y, Byun SJ, Lee S, Kwon MH. Functional stability of 3D8 scFv, a nucleic acid-hydrolyzing single chain antibody, under different biochemical and physical conditions. Int J Pharm 2015; 496:561-70. [PMID: 26536531 DOI: 10.1016/j.ijpharm.2015.10.076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/07/2015] [Accepted: 10/29/2015] [Indexed: 12/13/2022]
Abstract
3D8 single-chain Fv (scFv) is a catalytic nucleic acid antibody with anti-viral activity against a broad spectrum of viruses. Here we investigated the functional stability of 3D8 scFv to provide a basis for engineering a 3D8 scFv derivative and for developing stable formulations with improved stability and potential use as an anti-viral agent. The stability of 3D8 scFv was assessed by measuring its DNA-hydrolyzing activity under different biochemical and physical conditions using a fluorescence resonance energy transfer (FRET)-based method. In addition, the anti-influenza (H9N2) effect of 3D8 scFv was evaluated in A549 cells. 3D8 scFv was stable at 50°C for 6h at pH 7.2, for 3 days at pH 4-10 at 37°C and 30 days at pH 4-8 at 37°C. The stability was not affected by a reducing condition, freeze-thawing for up to 30 cycles, or lyophilization. Evaluation of the anti-virus effect showed that cells treated with 32-128 units of 3D8 scFv showed a 50% decrease in influenza replication compared to untreated cells. Based on its enzymatic stability in various biochemical and physical environments, 3D8 scFv holds good potential for development as an anti-viral therapeutic.
Collapse
Affiliation(s)
- Joungmin Lee
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea
| | - Hyunjoon Park
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea
| | - Minjae Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea
| | - Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea
| | - Yeonjin Lee
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon 441-706, Republic of Korea
| | - Sukchan Lee
- Department of Genetic Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Republic of Korea
| | - Myung-Hee Kwon
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea; Department of Microbiology, Ajou University School of Medicine, 206 World Cup-ro, Yeongtong-gu, Suwon 443-749, Republic of Korea.
| |
Collapse
|
26
|
Im SR, Im SW, Chung HY, Pravinsagar P, Jang YJ. Cell- and nuclear-penetrating anti-dsDNA autoantibodies have multiple arginines in CDR3 of VH and increase cellular level of pERK and Bcl-2 in mesangial cells. Mol Immunol 2015; 67:377-87. [DOI: 10.1016/j.molimm.2015.06.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/19/2015] [Accepted: 06/24/2015] [Indexed: 01/27/2023]
|
27
|
Preventive Activity against Influenza (H1N1) Virus by Intranasally Delivered RNA-Hydrolyzing Antibody in Respiratory Epithelial Cells of Mice. Viruses 2015; 7:5133-44. [PMID: 26402693 PMCID: PMC4584307 DOI: 10.3390/v7092863] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/10/2015] [Accepted: 09/14/2015] [Indexed: 01/22/2023] Open
Abstract
The antiviral effect of a catalytic RNA-hydrolyzing antibody, 3D8 scFv, for intranasal administration against avian influenza virus (H1N1) was described. The recombinant 3D8 scFv protein prevented BALB/c mice against H1N1 influenza virus infection by degradation of the viral RNA genome through its intrinsic RNA-hydrolyzing activity. Intranasal administration of 3D8 scFv (50 μg/day) for five days prior to infection demonstrated an antiviral activity (70% survival) against H1N1 infection. The antiviral ability of 3D8 scFv to penetrate into epithelial cells from bronchial cavity via the respiratory mucosal layer was confirmed by immunohistochemistry, qRT-PCR, and histopathological examination. The antiviral activity of 3D8 scFv against H1N1 virus infection was not due to host immune cytokines or chemokines, but rather to direct antiviral RNA-hydrolyzing activity of 3D8 scFv against the viral RNA genome. Taken together, our results suggest that the RNase activity of 3D8 scFv, coupled with its ability to penetrate epithelial cells through the respiratory mucosal layer, directly prevents H1N1 virus infection in a mouse model system.
Collapse
|
28
|
Lee G, Cho S, Hoang PM, Kim D, Lee Y, Kil EJ, Byun SJ, Lee TK, Kim DH, Kim S, Lee S. Therapeutic Strategy for the Prevention of Pseudorabies Virus Infection in C57BL/6 Mice by 3D8 scFv with Intrinsic Nuclease Activity. Mol Cells 2015; 38:773-80. [PMID: 26255831 PMCID: PMC4588720 DOI: 10.14348/molcells.2015.0073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/09/2015] [Accepted: 06/22/2015] [Indexed: 11/27/2022] Open
Abstract
3D8 single chain variable fragment (scFv) is a recombinant monoclonal antibody with nuclease activity that was originally isolated from autoimmune-prone MRL mice. In a previous study, we analyzed the nuclease activity of 3D8 scFv and determined that a HeLa cell line expressing 3D8 scFv conferred resistance to herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV). In this study, we demonstrate that 3D8 scFv could be delivered to target tissues and cells where it exerted a therapeutic effect against PRV. PRV was inoculated via intramuscular injection, and 3D8 scFv was injected intraperitoneally. The observed therapeutic effect of 3D8 scFv against PRV was also supported by results from quantitative reverse transcription polymerase chain reaction, southern hybridization, and immunohistochemical assays. Intraperitoneal injection of 5 and 10 μg 3D8 scFv resulted in no detectable toxicity. The survival rate in C57BL/6 mice was 9% after intramuscular injection of 10 LD50 PRV. In contrast, the 3D8 scFv-injected C57BL/6 mice showed survival rates of 57% (5 μg) and 47% (10 μg). The results indicate that 3D8 scFv could be utilized as an effective antiviral agent in several animal models.
Collapse
Affiliation(s)
- Gunsup Lee
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
- Fruit Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Suwon 440-706,
Korea
| | - SeungChan Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
| | - Phuong Mai Hoang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
| | - Dongjun Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
| | - Yongjun Lee
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
| | - Eui-Joon Kil
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
| | - Sung-June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon 441-706,
Korea
| | - Taek-Kyun Lee
- South Sea Environment Research Department, Korea Institute of Ocean Science and Technology, Geoje 656-834,
Korea
| | - Dae-Hyun Kim
- Fruit Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Suwon 440-706,
Korea
| | - Sunghan Kim
- Department of Plant Science, Research Institute of Agriculture and Life Sciences, and Plant Genomics and Breeding Institute, Seoul National University, Seoul 151-921,
Korea
| | - Sukchan Lee
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746,
Korea
| |
Collapse
|
29
|
Timofeeva AM, Ivanisenko NV, Buneva VN, Nevinsky GA. Systemic lupus erythematosus: molecular cloning and analysis of recombinant monoclonal kappa light chain NGTA2-Me-pro-ChTr possessing two different activities-trypsin-like and metalloprotease. Int Immunol 2015; 27:633-45. [PMID: 26174315 DOI: 10.1093/intimm/dxv042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/09/2015] [Indexed: 11/14/2022] Open
Abstract
Polyclonal antibodies hydrolyzing myelin basic protein (MBP) can play an important role in the pathogenesis of multiple sclerosis and systemic lupus erythematosus (SLE). An immunoglobulin light chain phagemid library derived from peripheral blood lymphocytes of patients with SLE was used. The small pools of phage particles displaying light chains with different affinity for MBP were isolated by affinity chromatography on MBP-Sepharose. The fraction eluted with 0.5M NaCl was used for preparation of individual monoclonal light chains (MLChs, 26-27kDa). The clones were expressed in Escherichia coli in a soluble form; MLChs were purified by metal-chelating chromatography followed by gel filtration. In mammalians, there are serine proteases and metalloproteases. These and many other enzymes usually have only one active site and catalyze only one chemical reaction. In contrast to canonical proteases, one MLCh (NGTA2-Me-pro-ChTr) efficiently hydrolyzed MBP (but not other proteins) and four different oligopeptides corresponding to four immunodominant sequences containing cleavage sites of MBP. The proteolytic activity of MLCh was efficiently inhibited only by specific inhibitors of serine-like (phenylmethanesulfonylfluoride, PMSF) and metalloproteases (EDTA). It was shown that MLCh possess independent serine-like and metal-dependent activities. The principal existence of monoclonal antibodies with two different proteolytic activities is unexpected but very important for the further understanding of at present unknown biological functions of human antibodies.
Collapse
Affiliation(s)
- Anna M Timofeeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nikita V Ivanisenko
- Institute of Cytology and Genetics, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Valentina N Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia Novosibirsk State University, Novosibirsk 630090, Russia
| | - Georgy A Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
30
|
Abstract
Transgenic resistance to plant viruses is an important technology for control of plant virus infection, which has been demonstrated for many model systems, as well as for the most important plant viruses, in terms of the costs of crop losses to disease, and also for many other plant viruses infecting various fruits and vegetables. Different approaches have been used over the last 28 years to confer resistance, to ascertain whether particular genes or RNAs are more efficient at generating resistance, and to take advantage of advances in the biology of RNA interference to generate more efficient and environmentally safer, novel "resistance genes." The approaches used have been based on expression of various viral proteins (mostly capsid protein but also replicase proteins, movement proteins, and to a much lesser extent, other viral proteins), RNAs [sense RNAs (translatable or not), antisense RNAs, satellite RNAs, defective-interfering RNAs, hairpin RNAs, and artificial microRNAs], nonviral genes (nucleases, antiviral inhibitors, and plantibodies), and host-derived resistance genes (dominant resistance genes and recessive resistance genes), and various factors involved in host defense responses. This review examines the above range of approaches used, the viruses that were tested, and the host species that have been examined for resistance, in many cases describing differences in results that were obtained for various systems developed in the last 20 years. We hope this compilation of experiences will aid those who are seeking to use this technology to provide resistance in yet other crops, where nature has not provided such.
Collapse
Affiliation(s)
| | - Peter Palukaitis
- Department of Horticultural Sciences, Seoul Women's University, Seoul, Republic of Korea.
| |
Collapse
|
31
|
Timofeeva AM, Buneva VN, Nevinsky GA. Systemic lupus erythematosus: molecular cloning and analysis of 22 individual recombinant monoclonal kappa light chains specifically hydrolyzing human myelin basic protein. J Mol Recognit 2015; 28:614-27. [DOI: 10.1002/jmr.2476] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/25/2015] [Accepted: 03/06/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Anna M. Timofeeva
- SB RAS; Institute of Chemical Biology and Fundamental Medicine; 8 Lavrentiev Ave. 630090 Novosibirsk Russia
| | - Valentina N. Buneva
- SB RAS; Institute of Chemical Biology and Fundamental Medicine; 8 Lavrentiev Ave. 630090 Novosibirsk Russia
- Novosibirsk State University; 2 Pirogova St. 630090 Novosibirsk Russia
| | - Georgy A. Nevinsky
- SB RAS; Institute of Chemical Biology and Fundamental Medicine; 8 Lavrentiev Ave. 630090 Novosibirsk Russia
- Novosibirsk State University; 2 Pirogova St. 630090 Novosibirsk Russia
| |
Collapse
|
32
|
Scior T, Paiz-Candia B, Islas ÁA, Sánchez-Solano A, Millan-Perez Peña L, Mancilla-Simbro C, Salinas-Stefanon EM. Predicting a double mutant in the twilight zone of low homology modeling for the skeletal muscle voltage-gated sodium channel subunit beta-1 (Nav1.4 β1). Comput Struct Biotechnol J 2015; 13:229-40. [PMID: 25904995 PMCID: PMC4402383 DOI: 10.1016/j.csbj.2015.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/18/2015] [Accepted: 03/21/2015] [Indexed: 12/18/2022] Open
Abstract
The molecular structure modeling of the β1 subunit of the skeletal muscle voltage-gated sodium channel (Nav1.4) was carried out in the twilight zone of very low homology. Structural significance can per se be confounded with random sequence similarities. Hence, we combined (i) not automated computational modeling of weakly homologous 3D templates, some with interfaces to analogous structures to the pore-bearing Nav1.4 α subunit with (ii) site-directed mutagenesis (SDM), as well as (iii) electrophysiological experiments to study the structure and function of the β1 subunit. Despite the distant phylogenic relationships, we found a 3D-template to identify two adjacent amino acids leading to the long-awaited loss of function (inactivation) of Nav1.4 channels. This mutant type (T109A, N110A, herein called TANA) was expressed and tested on cells of hamster ovary (CHO). The present electrophysiological results showed that the double alanine substitution TANA disrupted channel inactivation as if the β1 subunit would not be in complex with the α subunit. Exhaustive and unbiased sampling of “all β proteins” (Ig-like, Ig) resulted in a plethora of 3D templates which were compared to the target secondary structure prediction. The location of TANA was made possible thanks to another “all β protein” structure in complex with an irreversible bound protein as well as a reversible protein–protein interface (our “Rosetta Stone” effect). This finding coincides with our electrophysiological data (disrupted β1-like voltage dependence) and it is safe to utter that the Nav1.4 α/β1 interface is likely to be of reversible nature.
Collapse
Affiliation(s)
- Thomas Scior
- Facultad de Ciencias Químicas, Universidad Autónoma de Puebla, Puebla, Mexico
| | - Bertin Paiz-Candia
- Facultad de Ciencias Químicas, Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ángel A Islas
- Laboratorio de Biofísica, Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alfredo Sánchez-Solano
- Laboratorio de Biofísica, Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla, Mexico
| | | | - Claudia Mancilla-Simbro
- Laboratorio de Biofísica, Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla, Mexico
| | | |
Collapse
|
33
|
Roh J, Byun SJ, Seo Y, KIm M, Lee JH, Kim S, Lee Y, Lee KW, Kim JK, Kwon MH. Generation of a chickenized catalytic anti-nucleic acid antibody by complementarity-determining region grafting. Mol Immunol 2015; 63:513-20. [PMID: 25458312 DOI: 10.1016/j.molimm.2014.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/07/2014] [Accepted: 10/11/2014] [Indexed: 12/24/2022]
Abstract
In contrast to a number of studies on the humanization of non-human antibodies, the reshaping of a non-human antibody into a chicken antibody has never been attempted. Therefore, nothing is known about the animal species-dependent compatibility of the framework regions (FRs) that sustain the appropriate conformation of the complementarity-determining regions (CDRs). In this study, we attempted the reshaping of the variable domains of the mouse catalytic anti-nucleic acid antibody 3D8 (m3D8) into the FRs of a chicken antibody (“chickenization”) by CDR grafting, which is a common method for the humanization of antibodies. CDRs of the acceptor chicken antibody that showed a high homology to the FRs of m3D8 were replaced with those of m3D8, resulting in the chickenized antibody (ck3D8). ck3D8 retained the biochemical properties (DNA binding, DNA hydrolysis, and cellular internalizing activities) and three-dimensional structure of m3D8 and showed reduced immunogenicity in chickens. Our study demonstrates that CDR grafting can be applied to the chickenization of a mouse antibody, probably due to the interspecies compatibility of the FRs.
Collapse
|
34
|
Seo Y, Jun HR, Lee J, Park H, Kim M, Lee Y, Kwon MH. In-Cell RNA Hydrolysis Assay: A Method for the Determination of the RNase Activity of Potential RNases. Mol Biotechnol 2015; 57:506-12. [PMID: 25632893 PMCID: PMC4432088 DOI: 10.1007/s12033-015-9844-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Conventional procedures to assay RNA degradation by a protein with ribonuclease (RNase) activity require a step to isolate intact RNA molecules, which are used as a substrate. Here, we established a novel “In-cell RNA hydrolysis assay” in which RNAs within cells are used as a substrate for the RNA-hydrolyzing protein, thereby avoiding the need to prepare intact RNA molecules. In this method, the degree of RNA degradation is indicated by the fluorescence intensity of RNA molecules released from fixed and permeabilized cells following treatment with the potential RNase. A catalytic 3D8 antibody capable of degrading RNAs and pancreatic RNase A were used as model RNases. Our data demonstrate that the novel In-cell RNA hydrolysis assay is a reliable and sensitive method to analyze the activities of potential RNA-hydrolyzing proteins such as catalytic antibodies.
Collapse
Affiliation(s)
- Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon, 443-749, South Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
Hoang PM, Cho S, Kim KE, Byun SJ, Lee TK, Lee S. Development of Lactobacillus paracasei harboring nucleic acid-hydrolyzing 3D8 scFv as a preventive probiotic against murine norovirus infection. Appl Microbiol Biotechnol 2014; 99:2793-803. [PMID: 25487889 DOI: 10.1007/s00253-014-6257-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/18/2014] [Accepted: 11/20/2014] [Indexed: 01/15/2023]
Abstract
The protein 3D8 single-chain variable fragment (3D8 scFv) has potential anti-viral activity due to its ability to penetrate into cells and hydrolyze nucleic acids. Probiotic Lactobacillus paracasei engineered to secrete 3D8 scFv for oral administration was used to test the anti-viral effects of 3D8 scFv against gastrointestinal virus infections. We found that injection of 3D8 scFv into the intestinal lumen resulted in the penetration of 3D8 scFv into the intestinal villi and lamina propria. 3D8 scFv secreted from engineered L. paracasei retained its cell-penetrating and nucleic acid-hydrolyzing activities, which were previously shown with 3D8 scFv expressed in Escherichia coli. Pretreatment of RAW264.7 cells with 3D8 scFv purified from L. paracasei prevented apoptosis induction by murine norovirus infection and decreased messenger RNA (mRNA) expression of the viral capsid protein VP1. In a mouse model, oral administration of the engineered L. paracasei prior to murine norovirus infection reduced the expression level of mRNA encoding viral polymerase. Taken together, these results suggest that L. paracasei secreting 3D8 scFv provides a basis for the development of ingestible anti-viral probiotics active against gastrointestinal viral infection.
Collapse
Affiliation(s)
- Phuong Mai Hoang
- Department of Genetic Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 440-746, Korea
| | | | | | | | | | | |
Collapse
|
36
|
Lee G, Yu J, Cho S, Byun SJ, Kim DH, Lee TK, Kwon MH, Lee S. A nucleic-acid hydrolyzing single chain antibody confers resistance to DNA virus infection in hela cells and C57BL/6 mice. PLoS Pathog 2014; 10:e1004208. [PMID: 24968358 PMCID: PMC4072776 DOI: 10.1371/journal.ppat.1004208] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 05/12/2014] [Indexed: 01/19/2023] Open
Abstract
Viral protein neutralizing antibodies have been developed but they are limited only to the targeted virus and are often susceptible to antigenic drift. Here, we present an alternative strategy for creating virus-resistant cells and animals by ectopic expression of a nucleic acid hydrolyzing catalytic 3D8 single chain variable fragment (scFv), which has both DNase and RNase activities. HeLa cells (SCH7072) expressing 3D8 scFv acquired significant resistance to DNA viruses. Virus challenging with Herpes simplex virus (HSV) in 3D8 scFv transgenic cells and fluorescence resonance energy transfer (FRET) assay based on direct DNA cleavage analysis revealed that the induced resistance in HeLa cells was acquired by the nucleic acid hydrolyzing catalytic activity of 3D8 scFv. In addition, pseudorabies virus (PRV) infection in WT C57BL/6 mice was lethal, whereas transgenic mice (STG90) that expressed high levels of 3D8 scFv mRNA in liver, muscle, and brain showed a 56% survival rate 5 days after PRV intramuscular infection. The antiviral effects against DNA viruses conferred by 3D8 scFv expression in HeLa cells as well as an in vivo mouse system can be attributed to the nuclease activity that inhibits viral genome DNA replication in the nucleus and/or viral mRNA translation in the cytoplasm. Our results demonstrate that the nucleic-acid hydrolyzing activity of 3D8 scFv confers viral resistance to DNA viruses in vitro in HeLa cells and in an in vivo mouse system. Most strategies for developing virus-resistant transgenic cells and animals are based on the concept of virus-derived resistance, in which dysfunctional virus-derived products are expressed to interfere with the pathogenic process of the virus in transgenic cells or animals. However, these viral protein targeting approaches are limited because they only target specific viruses and are susceptible to viral mutations. We describe a novel strategy that targets the viral genome itself, rather than viral gene products, to generate virus-resistant transgenic cells and animals. We functionally expressed 3D8 scFv which has both DNase and RNase activities, in HeLa cells and transgenic mice. We found that the transgenic cells and mice acquired complete resistance to two DNA viruses (HSV and PRV) without accumulating the virus, and showed delayed onset of disease symptoms. The antiviral effects against DNA viruses demonstrated in this study were caused by (1) DNase activity of 3D8 scFv in the nucleus, which inhibited DNA replication or RNA transcription and (2) 3D8 scFv RNase activity in the cytoplasm, which blocked protein translation. This strategy may facilitate control of a broad spectrum of viruses, including viruses uncharacterized at the molecular level, regardless of their genome type or variations in gene products.
Collapse
Affiliation(s)
- Gunsup Lee
- Department of Genetic Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Korea
- Fruit Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Suwon, Korea
| | - Jaelim Yu
- Department of Genetic Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Korea
| | - Seungchan Cho
- Department of Genetic Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Korea
| | - Sung-June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Suwon, Korea
| | - Dae Hyun Kim
- Fruit Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Suwon, Korea
| | - Taek-Kyun Lee
- South Sea Environment Research Department, Korea Institute of Ocean Science and Technology, Geoje, Korea
| | - Myung-Hee Kwon
- Department of Microbiology, Ajou University School of Medicine, San 5, Woncheon-dong, Yeongtong-gu, Suwon, Korea
| | - Sukchan Lee
- Department of Genetic Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Korea
- * E-mail: ,
| |
Collapse
|
37
|
Systemic lupus erythematosus: Molecular cloning of fourteen recombinant DNase monoclonal kappa light chains with different catalytic properties. Biochim Biophys Acta Gen Subj 2014; 1840:1725-37. [DOI: 10.1016/j.bbagen.2014.01.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 11/15/2022]
|
38
|
Kostrikina IA, Odintsova ES, Buneva VN, Nevinsky GA. Systemic lupus erythematosus: molecular cloning and analysis of recombinant DNase monoclonal κ light chain NGK-1. Int Immunol 2014; 26:439-50. [PMID: 24919596 DOI: 10.1093/intimm/dxu047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Because DNase antibodies are cytotoxic, enter the nucleus and cause DNA fragmentation inducing cell death by apoptosis, they can play an important role in the pathogenesis of different autoimmune pathologies and especially systemic lupus erythematosus (SLE). The interesting goal of catalytic antibodies research is not only to study a possible biological role of such antibodies, but also to develop in future new human and animal therapies that use the advantages offered by abzymes. An immunoglobulin κ light chain library from SLE patients was cloned into a phagemid vector. Phage particles displaying recombinant monoclonal antibody light chains (MLChs) capable of binding DNA were isolated by affinity chromatography on DNA-cellulose. Sixteen of the 46 MLChs efficiently hydrolyzed DNA; one MLCh (approximately 27-28kDa) was expressed in Escherichia coli and purified by metal chelating and gel filtration. MLCh NGK-1 was electrophoretically homogeneous and demonstrated a positive answer with mouse IgGs against light chains of human antibodies after western blotting. SDS-PAGE in a gel containing DNA demonstrated that the MLCh hydrolyzes DNA and is not contaminated by canonical DNases. The DNase MLCh was activated by several metal ions. The protein sequence of the DNase MLCh has homology with mammalian DNases I and shares with them several identical or similar (with the same side chain functionality) important amino acid residues, which are necessary for DNA hydrolysis and binding of Mg(2+) and Ca(2+) ions. The affinity of DNA for this first example of a MLCh (K(M) = 0.3 microM) was 150- to 200-fold higher than for human DNase I.
Collapse
Affiliation(s)
- Irina A Kostrikina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Elena S Odintsova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Valentina N Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia Novosibirsk State University, Novosibirsk 630090, Russia
| | - Georgy A Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, Novosibirsk 630090, Russia Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
39
|
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
|
40
|
Hifumi E, Fujimoto N, Arakawa M, Saito E, Matsumoto S, Kobayashi N, Uda T. Biochemical features of a catalytic antibody light chain, 22F6, prepared from human lymphocytes. J Biol Chem 2013; 288:19558-68. [PMID: 23677996 PMCID: PMC3707657 DOI: 10.1074/jbc.m113.454579] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Human antibody light chains belonging to subgroup II of germ line genes were amplified by a seminested PCR technique using B-lymphocytes taken from a human adult infected with influenza virus. Each gene of the human light chains was transferred into the Escherichia coli system. The recovered light chain was highly purified using a two-step purification system. Light chain 22F6 showed interesting catalytic features. The light chain cleaved a peptide bond of synthetic peptidyl-4-methyl-coumaryl-7-amide (MCA) substrates, such as QAR-MCA and EAR-MCA, indicating amidase activity. It also hydrolyzed a phosphodiester bond of both DNA and RNA. From the analysis of amino acid sequences and molecular modeling, the 22F6 light chain possesses two kinds of active sites as amidase and nuclease in close distances. The 22F6 catalytic light chain could suppress the infection of influenza virus type A (H1N1) of Madin-Darby canine kidney cells in an in vitro assay. In addition, the catalytic light chain clearly inhibited the infection of the influenza virus of BALB/c mice via nasal administration in an in vivo assay. In the experiment, the titer in the serum of the mice coinfected with the 22F6 light chain and H1N1 virus became considerably lowered compared with that of 22F6-non-coinfected mice. Note that the catalytic light chain was prepared from human peripheral lymphocyte and plays an important role in preventing infection by influenza virus. Considering the fact that the human light chain did not show any acute toxicity for mice, our procedure developed in this study must be unique and noteworthy for developing new drugs.
Collapse
Affiliation(s)
- Emi Hifumi
- Research Center for Applied Medical Engineering, Oita University, Dan-noharu 700, Oita-shi, Oita 870-1192, Japan.
| | | | | | | | | | | | | |
Collapse
|
41
|
Buneva VN, Krasnorutskii MA, Nevinsky GA. Natural antibodies to nucleic acids. BIOCHEMISTRY (MOSCOW) 2013; 78:127-143. [DOI: 10.1134/s0006297913020028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
42
|
Kim A, Shin TH, Shin SM, Pham CD, Choi DK, Kwon MH, Kim YS. Cellular internalization mechanism and intracellular trafficking of filamentous M13 phages displaying a cell-penetrating transbody and TAT peptide. PLoS One 2012; 7:e51813. [PMID: 23251631 PMCID: PMC3522607 DOI: 10.1371/journal.pone.0051813] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 11/06/2012] [Indexed: 11/19/2022] Open
Abstract
Cellular internalization of bacteriophage by surface-displayed cell penetrating peptides has been reported, though the underlying mechanism remains elusive. Here we describe in detail the internalization mechanism and intracellular trafficking and stability of filamentous M13 phages, the cellular entry of which is mediated by surface-displayed cell-penetrating light chain variable domain 3D8 VL transbody (3D8 VL-M13) or TAT peptide (TAT-M13). Recombinant 3D8 VL-M13 and TAT-M13 phages were efficiently internalized into living mammalian cells via physiologically relevant, energy-dependent endocytosis and were recovered from the cells in their infective form with the yield of 3D8 VL-M13 being higher (0.005∼0.01%) than that of TAT-M13 (0.001∼0.005%). Biochemical and genetic studies revealed that 3D8 VL-M13 was internalized principally by caveolae-mediated endocytosis via interaction with heparan sulfate proteoglycans as cell surface receptors, whereas TAT-M13 was internalized by clathrin- and caveolae-mediated endocytosis utilizing chondroitin sulfate proteoglycans as cell surface receptors, suggesting that phage internalization occurs by physiological endocytotic mechanism through specific cell surface receptors rather than non-specific transcytotic pathways. Internalized 3D8 VL-M13 phages routed to the cytosol and remained stable for more than 18 h without further trafficking to other subcellular compartments, whereas TAT-M13 phages routed to several subcellular compartments before being degraded in lysosomes even after 2 h of internalization. Our results suggest that the internalizing mechanism and intracellular trafficking of filamentous M13 bacteriophages largely follow the attributes of the displayed cell-penetrating moiety. Efficient internalization and cytosolic localization of 3D8 VL transbody-displayed phages will provide a useful tool for intracellular delivery of polar macromolecules such as proteins, peptides, and siRNAs.
Collapse
Affiliation(s)
- Aeyung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Tae-Hwan Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Seung-Min Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Chuong D. Pham
- Department of Microbiology, Ajou University School of Medicine, Suwon, Korea
| | - Dong-Ki Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Myung-Hee Kwon
- Department of Microbiology, Ajou University School of Medicine, Suwon, Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- * E-mail:
| |
Collapse
|
43
|
Pham CD, Woo MY, Kim YS, Park S, Kwon MH. An anti-nucleic acid antibody delivers antigen to the cross-presentation pathway in dendritic cells and potentiates therapeutic antitumor effects. THE JOURNAL OF IMMUNOLOGY 2012; 189:5755-63. [PMID: 23152565 DOI: 10.4049/jimmunol.1200804] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cross-presentation is important for initiating CTL responses against tumors. Delivery of exogenous Ags to the cross-presentation pathway in dendritic cells (DCs), using a number of different carriers, has been attempted to further understand the mechanisms underlying cross-presentation and to develop therapeutic tumor vaccines. The present study reports a new antigenic carrier molecule: a single-chain V region fragment (scFv) of a nucleic acid-hydrolyzing Ab, 3D8. A fusion protein comprising 3D8 scFv and the CTL epitope OVA(250-264) (chicken OVA aa 250-264) was internalized by DC2.4 DCs and processed via a proteasome-dependent, brefeldin- and cycloheximide-sensitive, chloroquine- and primaquine-insensitive pathway, resulting in loading of the CTL epitope onto H-2K(b). In vivo cross-presentation and cross-priming were efficient, even without adjuvant; injection of mice with 3D8 scFv-OVA(250-264) induced cross-presentation of the CTL epitope by draining lymph node CD11c(+) B7.1(+) MHC class II(high) DCs, elicited a CTL response, and suppressed the growth of tumors expressing the OVA epitope. This report shows that an anti-nucleic acid Ab is used to deliver exogenous Ag to the cross-presentation pathway and inhibit in vivo tumor growth.
Collapse
Affiliation(s)
- Chuong D Pham
- Department of Microbiology, Ajou University School of Medicine, Suwon 443-749, South Korea
| | | | | | | | | |
Collapse
|
44
|
Role of Structure-Based Changes due to Somatic Mutation in Highly Homologous DNA-Binding and DNA-Hydrolyzing Autoantibodies Exemplified by A23P Substitution in the VH Domain. Autoimmune Dis 2012. [PMID: 23193442 PMCID: PMC3502752 DOI: 10.1155/2012/683829] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Anti-DNA autoantibodies are responsible for tissue injury in lupus. A subset of DNA-specific antibodies capable of DNA cleavage can be even more harmful after entering the living cells by destroying nuclear DNA. Origins of anti-DNA autoantibodies are not fully understood, and the mechanism of induction of DNA-cleaving activity remains speculative. The autoantibody BV04-01 derived from lupus-prone mouse is the only DNA-hydrolyzing immunoglobulin with known 3D structure. Identification and analysis of antibodies homologous to BV04-01 may help to understand molecular bases and origins of DNA-cleaving activity of autoantibodies. BLAST search identified murine anti-DNA autoantibody MRL-4 with sequences of variable region genes highly homologous to those of autoantibody BV04-01. Despite significant homology to BV04-01, not only MRL-4 had no DNA-cleaving activity, but also reversion of its unusual P23 mutation to the germline alanine resulted in a dramatic loss of affinity to DNA. Contrary to this effect, transfer of the P23 mutation to the BV04-01 has resulted in a significant drop in DNA binding and almost complete loss of catalytic activity. In the present paper we analyzed the properties of two homologous autoantibodies and mutants thereof and discussed the implications of unusual somatic mutations for the development of autoantibodies with DNA-binding and DNA-hydrolyzing activity.
Collapse
|
45
|
Baranova SV, Buneva VN, Kharitonova MA, Sizyakina LP, Zakharova OD, Nevinsky GA. Diversity of integrase-hydrolyzing IgGs and IgMs from sera of HIV-infected patients. BIOCHEMISTRY (MOSCOW) 2012; 76:1300-11. [PMID: 22150275 DOI: 10.1134/s0006297911120030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
It was previously shown that small fractions of IgGs and IgMs from the sera of AIDS patients specifically hydrolyze only HIV integrase (IN) but not many other tested proteins. Here we present evidence showing that these IgGs and IgMs are extreme catalytically heterogeneous. Affinity chromatography on IN-Sepharose using elution of IgGs (or IgMs) with different concentration of NaCl and acidic buffer separated catalytic antibodies (ABs) into many AB subfractions demonstrating different values of K(m) for IN and k(cat). Nonfractionated IgGs and IgMs possess serine-, thiol-, acidic-like, and metal-dependent proteolytic activity. Metal-dependent activity of abzymes increases in the presence of ions of different metals. In contrast to canonical proteases having one pH optimum, initial nonfractionated IgGs and IgMs demonstrate several optima at pH from 3 to 10. The data obtained show that IN-hydrolyzing polyclonal IgG and IgM of HIV-infected patients are cocktails of anti-IN ABs with different structure of the active centers possessing various affinity to IN, pH optima, and relative rates of the specific substrate hydrolysis.
Collapse
Affiliation(s)
- S V Baranova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of the Russian Academy of Sciences, pr. Lavrentieva 8, 630090 Novosibirsk, Russia
| | | | | | | | | | | |
Collapse
|
46
|
Interfering transbody-mediated Her2 gene silencing induces apoptosis by G0/G1 cell cycle arrest in Her2-overexpressing SK-BR-3 breast cancer cells. BIOTECHNOL BIOPROC E 2012. [DOI: 10.1007/s12257-011-0609-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
47
|
Kim A, Lee JY, Byun SJ, Kwon MH, Kim YS. Viral genome RNA degradation by sequence-selective, nucleic-acid hydrolyzing antibody inhibits the replication of influenza H9N2 virus without significant cytotoxicity to host cells. Antiviral Res 2012; 94:157-67. [PMID: 22484663 DOI: 10.1016/j.antiviral.2012.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/29/2012] [Accepted: 03/23/2012] [Indexed: 11/26/2022]
Abstract
Influenza A virus infection is a great threat to avian species and humans. Targeting viral proteins by antibody has a limited success due to the antigen drift and shift. Here we present a novel antibody-based antiviral strategy of targeting viral genomic RNA (vRNA) for degradation rather than neutralizing viral proteins. Based on the template of a sequence-nonspecific nucleic acid-hydrolyzing, single domain antibody of the light chain variable domain, 3D8 VL, we generated a synthetic library on the yeast surface by randomizing putative nucleic acid interacting residues. To target nucleocapsid protein (NP)-encoding viral genomic RNA (NP-vRNA) of H9N2 influenza virus, the library was screened against a 18-nucleotide single stranded nucleic acid substrate, dubbed asNP(18), the sequence of which is unique to the NP-vRNA. We isolated a 3D8 VL variant, NP25, that had ∼15-fold higher affinity (∼54nM) and ∼3-fold greater selective hydrolyzing activity for the target substrate than for off targets. In contrast to 3D8 VL WT, asNP(18)-selective NP25 constitutively expressed in the cytosol of human lung carcinoma A549 cells does not exhibit any significant cytotoxicity and selectively degrades a reporter mRNA carrying the target asNP(18) sequence in the stable cell lines. NP25 more potently inhibits the replication of H9N2 influenza virus than 3D8 VL WT in the stable cell lines. NP25 more selectively reduces the amount of the targeted NP-vRNA than 3D8 VL WT from the early stage of virus infection in the stable cell lines, without noticeable harmful effects on the endogenous mRNA, suggesting that NP25 indeed more specifically recognizes to hydrolyze the target NP-vRNA of H9N2 virus than off-targets. Our results provide a new strategy of targeting viral genomic RNA for degradation by antibody for the prevention of influenza virus infection in humans and animals.
Collapse
Affiliation(s)
- Aeyung Kim
- Dept. of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | | | | | | | | |
Collapse
|
48
|
Li JW, Xia L, Su Y, Liu H, Xia X, Lu Q, Yang C, Reheman K. Molecular imprint of enzyme active site by camel nanobodies: rapid and efficient approach to produce abzymes with alliinase activity. J Biol Chem 2012; 287:13713-21. [PMID: 22374998 DOI: 10.1074/jbc.m111.336370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Screening of inhibitory Ab1 antibodies is a critical step for producing catalytic antibodies in the anti-idiotypic approach. However, the incompatible surface of the active site of the enzyme and the antigen-binding site of heterotetrameric conventional antibodies become the limiting step. Because camelid-derived nanobodies possess the potential to preferentially bind to the active site of enzymes due to their small size and long CDR3, we have developed a novel approach to produce antibodies with alliinase activities by exploiting the molecular mimicry of camel nanobodies. By screening the camelid-derived variable region of the heavy chain cDNA phage display library with alliinase, we obtained an inhibitory nanobody VHHA4 that recognizes the active site. Further screening with VHHA4 from the same variable domain of the heavy chain of a heavy-chain antibody library led to a higher incidence of anti-idiotypic Ab2 abzymes with alliinase activities. One of the abzymes, VHHC10, showed the highest activity that can be inhibited by Ab1 VHHA4 and alliinase competitive inhibitor penicillamine and significantly suppressed the B16 tumor cell growth in the presence of alliin in vitro. The results highlight the feasibility of producing abzymes via anti-idiotypic nanobody approach.
Collapse
Affiliation(s)
- Jiang-Wei Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
A novel molecular analysis of genes encoding catalytic antibodies. Mol Immunol 2012; 50:160-8. [PMID: 22325472 DOI: 10.1016/j.molimm.2012.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 01/09/2012] [Accepted: 01/12/2012] [Indexed: 10/14/2022]
Abstract
Among the numerous questions remaining opened about catalytic antibodies (abzymes), the understanding of the origin of the genes encoding them is of vital significance. An original statistical analysis of genes encoding abzymes is described in the present report. Results suggested that these genes display a high conservation degree with their germline counterpart and a limited number of amino acid changes. Hence, on the contrary with high-affinity antibodies, maturation process by accumulation of somatic hypermutations is not required for the catalytic function. We demonstrated that despite a weak somatic mutation rate, the physicochemical properties of mutated amino acid (AA) are predominantly dissimilar with that of the germline AA. Further, we developed a novel approach in order to analyze the nature of genes encoding catalytic antibodies. For the first time, an unexpected and significant high level expression of rare gene subgroups was noticed and emphasized. The data described in this paper would lay the foundation for future studies about origin of genes encoding catalytic antibodies.
Collapse
|
50
|
Sustained Cytoplasmic Delivery and Anti-viral Effect of PLGA Nanoparticles Carrying a Nucleic Acid-Hydrolyzing Monoclonal Antibody. Pharm Res 2011; 29:932-42. [DOI: 10.1007/s11095-011-0633-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 11/21/2011] [Indexed: 12/17/2022]
|