1
|
Zhang Y, Guo Y, Song L, Liu W, Nian R, Fan X. Streamlined on-column refolding and purification of nanobodies from inclusion bodies expressed as fusion proteins. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1246:124279. [PMID: 39186888 DOI: 10.1016/j.jchromb.2024.124279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
This study introduces an efficient on-column refolding and purification method for preparing nanobodies (Nbs) expressed as inclusion bodies and fusion proteins. The HisTrapTM FF system was successfully employed for the purification of the fusion protein FN1-ΔI-CM-2D5. The intein ΔI-CM cleavage activity was activated at 42 °C, followed by incubation for 4 h. Leveraging the remarkable thermal stability of Nbs, 2D5 was further purified through heat treatment at 80 °C for 1h. This method yielded up to 107.2 mg of pure 2D5 with a purity of 99.2 % from just 1L of bacterial culture grown in a shaker flask. Furthermore, this approach successfully restored native secondary structure and affinity of 2D5. Additionally, the platform was effectively applied to the refolding and purification of a polystyrene-binding nanobody (B2), which exhibited limited expression in the periplasmic and cytoplasmic spaces of E. coli. This endeavor resulted in the isolation of 53.2 mg of pure B2 Nb with a purity exceeding 99.5 % from the same volume of bacterial culture. Significantly, this approach restored the native secondary structure of the Nbs, highlighting its potential for addressing challenges associated with expressing complex Nbs in E. coli. Overall, this innovative platform provides a scientifically rigorous and reproducible method for the efficient preparation of Nbs, offering a valuable tool for antibody research and development.
Collapse
Affiliation(s)
- Yiwen Zhang
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Yang Guo
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Liang Song
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Wenshuai Liu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Rui Nian
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Xiying Fan
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China.
| |
Collapse
|
2
|
Lu Y, Li Q, Fan H, Liao C, Zhang J, Hu H, Yi H, Peng Y, Lu J, Chen Z. A Multivalent and Thermostable Nanobody Neutralizing SARS-CoV-2 Omicron (B.1.1.529). Int J Nanomedicine 2023; 18:353-367. [PMID: 36700149 PMCID: PMC9869787 DOI: 10.2147/ijn.s387160] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Background The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variants have risen to dominance, which contains far more mutations in the spike protein in comparison to previously reported variants, compromising the efficacy of most existing vaccines or therapeutic monoclonal antibodies. Nanobody screened from high-throughput naïve libraries is a potential candidate for developing preventive and therapeutic antibodies. Methods Four nanobodies specific to the SARS-CoV-2 wild-type receptor-binding domain (RBD) were screened from a naïve phage display library. Their affinity and neutralizing activity were evaluated by surface plasmon resonance assays, surrogate virus neutralization tests, and pseudovirus neutralization assays. Preliminary identification of the binding epitopes of nanobodies by peptide-based ELISA and competition assay. Then four multivalent nanobodies were engineered by attaching the monovalent nanobodies to an antibody-binding nanoplatform constructed based on the lumazine synthase protein cage nanoparticles isolated from the Aquifex aeolicus (AaLS). Finally, the differences in potency between the monovalent and multivalent nanobodies were compared using the same methods. Results Three of the four specific nanobodies could maintain substantial inhibitory activity against the Omicron (B.1.1.529), of them, B-B2 had the best neutralizing activity against the Omicron (B.1.1.529) pseudovirus (IC50 = 1.658 μg/mL). The antiviral ability of multivalent nanobody LS-B-B2 was improved in the Omicron (B.1.1.529) pseudovirus assays (IC50 = 0.653 μg/mL). The results of peptide-based ELISA indicated that LS-B-B2 might react with the linear epitopes in the SARS-CoV-2 RBD conserved regions, which would clarify the mechanisms for the maintenance of potent neutralization of Omicron (B.1.1.529) preliminary. Conclusion Our study indicated that the AaLS could be used as an antibody-binding nanoplatform to present nanobodies on its surface and improve the potency of nanobodies. The multivalent nanobody LS-B-B2 may serve as a potential agent for the neutralization of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yuying Lu
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Qianlin Li
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Conghui Liao
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Jingsong Zhang
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Huan Hu
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Huaimin Yi
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Yuanli Peng
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China
| | - Jiahai Lu
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, People’s Republic of China,Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, People’s Republic of China,Correspondence: Jiahai Lu; Zeliang Chen, One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China, Email ;
| | - Zeliang Chen
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-Sen University, Guangzhou, People’s Republic of China,National Medical Products Administration Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, People’s Republic of China,Key Laboratory of Tropical Diseases Control, Sun Yat-Sen University, Ministry of Education, Guangzhou, People’s Republic of China,Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, People’s Republic of China
| |
Collapse
|
3
|
Tian L, Jackson K, Chan M, Saif A, He L, Didar TF, Hosseinidoust Z. Phage display for the detection, analysis, disinfection, and prevention of Staphylococcus aureus. SMART MEDICINE 2022; 1:e20220015. [PMID: 39188734 PMCID: PMC11235639 DOI: 10.1002/smmd.20220015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/25/2022] [Indexed: 08/28/2024]
Abstract
The World Health Organization has designated Staphylococcus aureus as a global health concern. This designation stems from the emergence of multiple drug-resistant strains that already account for hundreds of thousands of deaths globally. The development of novel treatment strategies to eradicate S. aureus or mitigate its pathogenic potential is desperately needed. In the effort to develop emerging strategies to combat S. aureus, phage display is uniquely positioned to assist in this endeavor. Leveraging bacteriophages, phage display enables researchers to better understand interactions between proteins and their antagonists. In doing so, researchers have the capacity to design novel inhibitors, biosensors, disinfectants, and immune modulators that can target specific S. aureus strains. In this review, we highlight how phage display can be leveraged to design novel solutions to combat S. aureus. We further discuss existing uses of phage display as a detection, intervention, and prevention platform against S. aureus and provide outlooks on how this technology can be optimized for future applications.
Collapse
Affiliation(s)
- Lei Tian
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Kyle Jackson
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Michael Chan
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Ahmed Saif
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Leon He
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Tohid F. Didar
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioCanada
- Michael DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonOntarioCanada
- Department of Mechanical EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Zeinab Hosseinidoust
- Department of Chemical EngineeringMcMaster UniversityHamiltonOntarioCanada
- School of Biomedical EngineeringMcMaster UniversityHamiltonOntarioCanada
- Michael DeGroote Institute for Infectious Disease ResearchMcMaster UniversityHamiltonOntarioCanada
| |
Collapse
|
4
|
Sanaei M, Setayesh N, Sepehrizadeh Z, Mahdavi M, Yazdi MH. Nanobodies in Human Infections: Prevention, Detection, and Treatment. Immunol Invest 2019; 49:875-896. [PMID: 31856615 DOI: 10.1080/08820139.2019.1688828] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Despite the existence of vaccination, antibiotic therapy, and antibody therapies, infectious diseases still remain as one of the biggest challenges to human health all over the world. Among the different methods for treatment and prevention of infectious diseases, antibodies are well known but poorly developed. There is a new subclass of antibodies calledheavy-chain antibodies that belong to the IgG isotype. However, they are low in molecular weight and lost the first constant domain (CH1). Their single-domain antigen-binding fragments, identified as nanobodies, have unique characteristics, which make them superior in comparison with the conventional antibodies. Low molecular weight and small size, high stability and solubility, ease of expression, good tissue penetration, and low-cost production make nanobodies an appropriate alternative to use against infectious disease. In this research, we review the properties of nanobodies and their potential applications in controlling human infections and inflammations.
Collapse
Affiliation(s)
- Marzieh Sanaei
- Biotechnology Research Center, Tehran University of Medical Sciences , Tehran, Iran.,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy & Biotechnology Research Center, Tehran University of Medical Sciences , Tehran, Iran
| | - Neda Setayesh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy & Biotechnology Research Center, Tehran University of Medical Sciences , Tehran, Iran
| | - Zargham Sepehrizadeh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy & Biotechnology Research Center, Tehran University of Medical Sciences , Tehran, Iran
| | - Mehdi Mahdavi
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences , Tehran, Iran
| | - Mohammad Hossein Yazdi
- Biotechnology Research Center, Tehran University of Medical Sciences , Tehran, Iran.,Recombinant Vaccine Research Center, Tehran University of Medical Sciences , Tehran, Iran
| |
Collapse
|
5
|
Anderson GP, Liu JL, Shriver-Lake LC, Zabetakis D, Sugiharto VA, Chen HW, Lee CR, Defang GN, Wu SJL, Venkateswaran N, Goldman ER. Oriented Immobilization of Single-Domain Antibodies Using SpyTag/SpyCatcher Yields Improved Limits of Detection. Anal Chem 2019; 91:9424-9429. [DOI: 10.1021/acs.analchem.9b02096] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- George P. Anderson
- Center for Biomolecular Science and Engineering, U.S. Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, D.C. 20375, United States
| | - Jinny L. Liu
- Center for Biomolecular Science and Engineering, U.S. Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, D.C. 20375, United States
| | - Lisa C. Shriver-Lake
- Center for Biomolecular Science and Engineering, U.S. Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, D.C. 20375, United States
| | - Dan Zabetakis
- Center for Biomolecular Science and Engineering, U.S. Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, D.C. 20375, United States
| | - Victor A. Sugiharto
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Hua-Wei Chen
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Cheng-Rei Lee
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Gabriel N. Defang
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Shuenn-Jue L. Wu
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Neeraja Venkateswaran
- Tetracore, Inc., 9901 Belward Campus Drive, Suite 300, Rockville, Maryland 20850, United States
| | - Ellen R. Goldman
- Center for Biomolecular Science and Engineering, U.S. Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, D.C. 20375, United States
| |
Collapse
|
6
|
Olson MA, Legler PM, Zabetakis D, Turner KB, Anderson GP, Goldman ER. Sequence Tolerance of a Single-Domain Antibody with a High Thermal Stability: Comparison of Computational and Experimental Fitness Profiles. ACS OMEGA 2019; 4:10444-10454. [PMID: 31460140 PMCID: PMC6648363 DOI: 10.1021/acsomega.9b00730] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/09/2019] [Indexed: 06/10/2023]
Abstract
The sequence fitness of a llama single-domain antibody with an unusually high thermal stability is explored by a combined computational and experimental study. Starting with the X-ray crystallographic structure, RosettaBackrub simulations were applied to model sequence-structure tolerance profiles and identify key substitution sites. From the model calculations, an experimental site-directed mutagenesis was used to produce a panel of mutants, and their melting temperatures were determined by thermal denaturation. The results reveal a sequence fitness of an excess stability of approximately 12 °C, a value taken from a decrease in the melting temperature of an electrostatic charge-reversal substitution in the CRD3 without a deleterious effect on the binding affinity to the antigen. The tolerance for the disruption of antigen recognition without loss in the thermal stability was demonstrated by the introduction of a proline in place of a tyrosine in the CDR2, producing a mutant that eliminated binding. To further assist the sequence design and the selection of engineered single-domain antibodies, an assessment of different computational strategies is provided of their accuracy in the detection of substitution "hot spots" in the sequence tolerance landscape.
Collapse
Affiliation(s)
- Mark A. Olson
- Systems
and Structural Biology Division, USAMRIID, Frederick, Maryland 21702, United States
| | - Patricia M. Legler
- Center
for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| | - Daniel Zabetakis
- Center
for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| | - Kendrick B. Turner
- Center
for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| | - George P. Anderson
- Center
for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| | - Ellen R. Goldman
- Center
for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, District of Columbia 20375, United States
| |
Collapse
|
7
|
Shriver-Lake LC, Liu JL, Zabetakis D, Sugiharto VA, Lee CR, Defang GN, Wu SJL, Anderson GP, Goldman ER. Selection and Characterization of Anti-Dengue NS1 Single Domain Antibodies. Sci Rep 2018; 8:18086. [PMID: 30591706 PMCID: PMC6308234 DOI: 10.1038/s41598-018-35923-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/13/2018] [Indexed: 01/13/2023] Open
Abstract
Reliable detection and diagnosis of dengue virus (DENV) is important for both patient care and epidemiological control. Starting with a llama immunized with a mixture of recombinant nonstructural protein 1 (NS1) antigen from the four DENV serotypes, a phage display immune library of single domain antibodies was constructed and binders selected which exhibited specificity and affinity for DENV NS1. Each of these single domain antibodies was evaluated for its binding affinity to NS1 from the four serotypes, and incorporated into a sandwich format for NS1 detection. An optimal pair was chosen that provided the best combination of sensitivity for all four DENV NS1 antigens spiked into 50% human serum while showing no cross reactivity to NS1 from Zika virus, yellow fever virus, tick-borne encephalitis virus, and minimal binding to NS1 from Japanese encephalitis virus and West Nile virus. These rugged and robust recombinant binding molecules offer attractive alternatives to conventional antibodies for implementation into immunoassays destined for resource limited locals.
Collapse
Affiliation(s)
- Lisa C Shriver-Lake
- Center for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Jinny L Liu
- Center for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Dan Zabetakis
- Center for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Victor A Sugiharto
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Cheng-Rei Lee
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Gabriel N Defang
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Shuenn-Jue L Wu
- Viral and Rickettsial Diseases Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - George P Anderson
- Center for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Ellen R Goldman
- Center for Biomolecular Science and Engineering, US Naval Research Laboratory, 4555 Overlook Ave SW, Washington, DC, 20375, USA.
| |
Collapse
|
8
|
Karau MJ, Tilahun ME, Krogman A, Osborne BA, Goldsby RA, David CS, Mandrekar JN, Patel R, Rajagopalan G. Passive therapy with humanized anti-staphylococcal enterotoxin B antibodies attenuates systemic inflammatory response and protects from lethal pneumonia caused by staphylococcal enterotoxin B-producing Staphylococcus aureus. Virulence 2017; 8:1148-1159. [PMID: 27925510 PMCID: PMC5711449 DOI: 10.1080/21505594.2016.1267894] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023] Open
Abstract
Drugs such as linezolid that inhibit bacterial protein synthesis may be beneficial in treating infections caused by toxigenic Staphylococcus aureus. As protein synthesis inhibitors have no effect on preformed toxins, neutralization of pathogenic exotoxins with anti-toxin antibodies may be beneficial in conjunction with antibacterial therapy. Herein, we evaluated the efficacy of human-mouse chimeric high-affinity neutralizing anti-staphylococcal enterotoxin B (SEB) antibodies in the treatment of experimental pneumonia caused by SEB-producing S. aureus. Since HLA class II transgenic mice mount a stronger systemic immune response following challenge with SEB and are more susceptible to SEB-induced lethal toxic shock than conventional mice strains, HLA-DR3 transgenic mice were used. Lethal pneumonia caused by SEB-producing S. aureus in HLA-DR3 transgenic mice was characterized by robust T cell activation and elevated systemic levels of several pro-inflammatory cytokines and chemokines. Prophylactic administration of a single dose of linezolid 30 min prior to the onset of infection attenuated the systemic inflammatory response and protected from mortality whereas linezolid administered 60 min after the onset of infection failed to confer significant protection. Human-mouse chimeric high-affinity neutralizing anti-SEB antibodies alone, but not polyclonal human IgG, mitigated this response and protected from death when administered immediately after initiation of infection. Further, anti-SEB antibodies as well as intact polyclonal human IgG, but not its Fab or Fc fragments, protected from lethal pneumonia when followed with linezolid therapy 60 min later. In conclusion, neutralization of superantigens with high-affinity antibodies may have beneficial effects in pneumonia.
Collapse
Affiliation(s)
- Melissa J. Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mulualem E. Tilahun
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
- Department of Biology, Amherst College, Amherst, MA, USA
| | - Ashton Krogman
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Barbara A. Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | | | - Chella S. David
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jayawant N. Mandrekar
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Govindarajan Rajagopalan
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
9
|
Shriver-Lake LC, Zabetakis D, Goldman ER, Anderson GP. Evaluation of anti-botulinum neurotoxin single domain antibodies with additional optimization for improved production and stability. Toxicon 2017; 135:51-58. [DOI: 10.1016/j.toxicon.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 01/27/2023]
|
10
|
Goldman ER, Liu JL, Zabetakis D, Anderson GP. Enhancing Stability of Camelid and Shark Single Domain Antibodies: An Overview. Front Immunol 2017; 8:865. [PMID: 28791022 PMCID: PMC5524736 DOI: 10.3389/fimmu.2017.00865] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 07/07/2017] [Indexed: 11/13/2022] Open
Abstract
Single domain antibodies (sdAbs) are gaining a reputation as superior recognition elements as they combine the advantages of the specificity and affinity found in conventional antibodies with high stability and solubility. Melting temperatures (Tms) of sdAbs cover a wide range from below 50 to over 80°C. Many sdAbs have been engineered to increase their Tm, making them stable until exposed to extreme temperatures. SdAbs derived from the variable heavy chains of camelid and shark heavy chain-only antibodies are termed VHH and VNAR, respectively, and generally exhibit some ability to refold and bind antigen after heat denaturation. This ability to refold varies from 0 to 100% and is a property dependent on both intrinsic factors of the sdAb and extrinsic conditions such as the sample buffer ionic strength, pH, and sdAb concentration. SdAbs have also been engineered to increase their solubility and refolding ability, which enable them to function even after exposure to temperatures that exceed their melting point. In addition, efforts to improve their stability at extreme pH and in the presence of chemical denaturants or proteases have been undertaken. Multiple routes have been employed to engineer sdAbs with these enhanced stabilities. The methods utilized to achieve these goals include grafting complementarity-determining regions onto stable frameworks, introduction of non-canonical disulfide bonds, random mutagenesis combined with stringent selection, point mutations such as inclusion of negative charges, and genetic fusions. Increases of up to 20°C have been realized, pushing the Tm of some sdAbs to over 90°C. Herein, we present an overview of the work done to stabilize sdAbs derived from camelids and sharks. Utilizing these various strategies sdAbs have been stabilized without significantly compromising their affinity, thereby providing superior reagents for detection, diagnostic, and therapeutic applications.
Collapse
Affiliation(s)
- Ellen R Goldman
- Center for BioMolecular Science and Engineering, US Naval Research Laboratory, Washington, DC, United States
| | - Jinny L Liu
- Center for BioMolecular Science and Engineering, US Naval Research Laboratory, Washington, DC, United States
| | - Dan Zabetakis
- Center for BioMolecular Science and Engineering, US Naval Research Laboratory, Washington, DC, United States
| | - George P Anderson
- Center for BioMolecular Science and Engineering, US Naval Research Laboratory, Washington, DC, United States
| |
Collapse
|
11
|
Anderson GP, Liu JH, Zabetakis D, Liu JL, Goldman ER. Thermal stabilization of anti-α-cobratoxin single domain antibodies. Toxicon 2017; 129:68-73. [PMID: 28209480 DOI: 10.1016/j.toxicon.2017.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/09/2017] [Accepted: 02/11/2017] [Indexed: 10/20/2022]
Abstract
There is an unmet need for snake antivenoms that can be stored ready to use near the point of care. To address that need we have taken two anti-α-cobratoxin single domain antibodies and increased their thermal stability to improve their ambient temperature shelf-life. The anti-α-cobratoxin single domain antibodies C2 and C20 were first isolated, and demonstrated to be toxin neutralizing by Richard et al., 2013 (Richard, G., Meyers, A.J., McLean, M.D., Arbabi-Ghahroudi, M., MacKenzie, R., Hall, J.C., 2013. In vivo neutralization of alpha-cobratoxin with high-affinity llama single-domain antibodies (VHHs) and a VHH-Fc antibody. PLoS One 8, e69495). To thermal stabilize C2 and C20, we first made changes to their frame work 1 region that we had previously identified to be stabilizing, as well as reverted to the hallmark amino acids highly conserved in VHH domains; these changes improved their melting temperature (Tm) by 2 and 6 °C respectively. The further addition of a non-canonical disulfide bond raised the Tm an additional 13 and 9 °C respectively; giving final Tm values of 86 and 75 °C. Testing these mutants at 1 mg/mL at a range of elevated temperatures for an hour; we found that at 65 °C the wild type C2 and C20 had lost 35 and 95% of their binding activity respectively, while the mutants with the added disulfide bond retained nearly 100% of their initial binding activity. While significant work remains to formulate and field a shelf-stable antivenom, our results indicate such a product should be attainable in the near future.
Collapse
Affiliation(s)
- George P Anderson
- US Naval Research Laboratory, Center for Biomolecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA.
| | - Jessica H Liu
- Science and Engineering Apprenticeship Program, US Naval Research Laboratory, 4555, Overlook Ave SW, Washington, DC, USA
| | - Dan Zabetakis
- US Naval Research Laboratory, Center for Biomolecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Jinny L Liu
- US Naval Research Laboratory, Center for Biomolecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| | - Ellen R Goldman
- US Naval Research Laboratory, Center for Biomolecular Science and Engineering, 4555 Overlook Ave SW, Washington, DC, 20375, USA
| |
Collapse
|
12
|
Legler PM, Compton JR, Hale ML, Anderson GP, Olson MA, Millard CB, Goldman ER. Stability of isolated antibody-antigen complexes as a predictive tool for selecting toxin neutralizing antibodies. MAbs 2016; 9:43-57. [PMID: 27660893 PMCID: PMC5240650 DOI: 10.1080/19420862.2016.1236882] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ricin is an A-B ribosome inactivating protein (RIP) toxin composed of an A-chain subunit (RTA) that contains a catalytic N-glycosidase and a B-chain (RTB) lectin domain that binds cell surface glycans. Ricin exploits retrograde transport to enter into the Golgi and the endoplasmic reticulum, and then dislocates into the cytoplasm where it can reach its substrate, the rRNA. A subset of isolated antibodies (Abs) raised against the RTA subunit protect against ricin intoxication, and RTA-based vaccine immunogens have been shown to provide long-lasting protective immunity against the holotoxin. Anti-RTA Abs are unlikely to cross a membrane and reach the cytoplasm to inhibit the enzymatic activity of the A-chain. Moreover, there is not a strict correlation between the apparent binding affinity (Ka) of anti-RTA Abs and their ability to successfully neutralize ricin toxicity. Some anti-RTA antibodies are toxin-neutralizing, whereas others are not. We hypothesize that neutralizing anti-RTA Abs may interfere selectively with conformational change(s) or partial unfolding required for toxin internalization. To test this hypothesis, we measured the melting temperatures (Tm) of neutralizing single-domain Ab (sdAb)-antigen (Ag) complexes relative to the Tm of the free antigen (Tm-shift = Tmcomplex – TmAg), and observed increases in the Tmcomplex of 9–20 degrees. In contrast, non-neutralizing sdAb-Ag complexes shifted the TmComplex by only 6–7 degrees. A strong linear correlation (r2 = 0.992) was observed between the magnitude of the Tm-shift and the viability of living cells treated with the sdAb and ricin holotoxin. The Tm-shift of the sdAb-Ag complex provided a quantitative biophysical parameter that could be used to predict and rank-order the toxin-neutralizing activities of Abs. We determined the first structure of an sdAb-RTA1-33/44-198 complex, and examined other sdAb-RTA complexes. We found that neutralizing sdAb bound to regions involved in the early stages of unfolding. These Abs likely interfere with steps preceding or following endocytosis that require conformational changes. This method may have utility for the characterization or rapid screening of other Ab that act to prevent conformational changes or unfolding as part of their mechanism of action.
Collapse
Affiliation(s)
| | | | - Martha L Hale
- c US Army Medical Research Institute of Infectious Diseases , Frederick , MD , USA
| | | | - Mark A Olson
- c US Army Medical Research Institute of Infectious Diseases , Frederick , MD , USA
| | - Charles B Millard
- c US Army Medical Research Institute of Infectious Diseases , Frederick , MD , USA
| | | |
Collapse
|
13
|
Single-domain antibody based thermally stable electrochemical immunosensor. Biosens Bioelectron 2016; 83:162-8. [PMID: 27125838 DOI: 10.1016/j.bios.2016.04.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 02/07/2023]
Abstract
Conventional monoclonal and polyclonal antibodies are sensitive to changes in environmental factors such as temperature, pH, humidity, etc. This limits the current cost-effective and portable electrochemical immunosensors in harsh environments. Using Ricin Chain-A, a naturally occurring toxin, as a model analyte we report fabrication of a thermally stable electrochemical immunosensor. Single-domain antibodies (sdAb) or nanobodies have been employed as recognition elements for direct detection of Ricin at temperatures great than 4°C. Immunosensor fabricated using the conventional Ricin monoclonal and polyclonal antibodies have also been demonstrated for comparison. In the case of sdAb immunosensor, Ricin was detected in a linear range of 1log(fg/mL)-1log(μg/mL) with a sensitivity of 0.07μA/log(g/mL)/cm(2) using cyclic voltammetry. The fabricated miniaturized sensors have demonstrated higher shelf life and stability at temperatures up to 40°C. Therefore these electrochemical sensors can be integrated as a part of a portable device for point-of-care immunosensing.
Collapse
|
14
|
Vuong C, Yeh AJ, Cheung GYC, Otto M. Investigational drugs to treat methicillin-resistant Staphylococcus aureus. Expert Opin Investig Drugs 2015; 25:73-93. [PMID: 26536498 DOI: 10.1517/13543784.2016.1109077] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Staphylococcus aureus remains one of the leading causes of morbidity and mortality worldwide. This is to a large extent due to antibiotic-resistant strains, in particular methicillin-resistant S. aureus (MRSA). While the toll of invasive MRSA infections appears to decrease in U.S. hospitals, the rate of community-associated MRSA infections remains constant and there is a surge of MRSA in many other countries, a situation that calls for continuing if not increased efforts to find novel strategies to combat MRSA infections. AREAS COVERED This review provides an overview of current investigational drugs and therapeutic antibodies against S. aureus in early clinical development (up to phase II clinical development). It includes a short description of the mechanism of action and a presentation of microbiological and clinical data. EXPERT OPINION Increased recent antibiotic development efforts and results from pathogenesis research have led to several new antibiotics and therapies, such as anti-virulence drugs, as well as a more informed selection of targets for vaccination efforts against MRSA. This developing portfolio of novel anti-staphylococcal drugs will hopefully provide us with additional and more efficient ways to combat MRSA infections in the near future and prevent us from running out of treatment options, even if new resistances arise.
Collapse
Affiliation(s)
- Cuong Vuong
- a Principal Scientist/Laboratory Head, Bacteriology , AiCuris GmbH & Co. KG, Friedrich-Ebert-Str. 475/Geb. 302, 42117 Wuppertal , Germany
| | - Anthony J Yeh
- b Post-baccalaureate IRTA, Laboratory of Bacteriology , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| | - Gordon Y C Cheung
- c Staff Scientist, National Institute of Allergy and Infectious Diseases , National Institutes of Health, Laboratory of Bacteriology , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| | - Michael Otto
- d Senior Investigator, National Institute of Allergy and Infectious Diseases , National Institutes of Health, Laboratory of Bacteriology , Bldg. 33, 1W10, 9000 Rockville Pike, Bethesda , MD 20892 , USA
| |
Collapse
|
15
|
Liu JL, Goldman ER, Zabetakis D, Walper SA, Turner KB, Shriver-Lake LC, Anderson GP. Enhanced production of a single domain antibody with an engineered stabilizing extra disulfide bond. Microb Cell Fact 2015; 14:158. [PMID: 26449768 PMCID: PMC4599338 DOI: 10.1186/s12934-015-0340-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Single domain antibodies derived from the variable region of the unique heavy chain antibodies found in camelids yield high affinity and regenerable recognition elements. Adding an additional disulfide bond that bridges framework regions is a proven method to increase their melting temperature, however often at the expense of protein production. To fulfill their full potential it is essential to achieve robust protein production of these stable binding elements. In this work, we tested the hypothesis that decreasing the isoelectric point of single domain antibody extra disulfide bond mutants whose production fell due to the incorporation of the extra disulfide bond would lead to recovery of the protein yield, while maintaining the favorable melting temperature and affinity. RESULTS Introduction of negative charges into a disulfide bond mutant of a single domain antibody specific for the L1 antigen of the vaccinia virus led to approximately 3.5-fold increase of protein production to 14 mg/L, while affinity and melting temperature was maintained. In addition, refolding following heat denaturation improved from 15 to 70 %. It also maintained nearly 100 % of its binding function after heating to 85 °C for an hour at 1 mg/mL. Disappointingly, the replacement of neutral or positively charged amino acids with negatively charged ones to lower the isoelectric point of two anti-toxin single domain antibodies stabilized with a second disulfide bond yielded only slight increases in protein production. Nonetheless, for one of these binders the charge change itself stabilized the structure equivalent to disulfide bond addition, thus providing an alternative route to stabilization which is not accompanied by loss in production. CONCLUSION The ability to produce high affinity, stable single domain antibodies is critical for their utility. While the addition of a second disulfide bond is a proven method for enhancing stability of single domain antibodies, it frequently comes at the cost of reduced yields. While decreasing the isoelectric point of double disulfide mutants of single domain antibodies may improve protein production, charge addition appears to consistently improve refolding and some charge changes can also improve thermal stability, thus providing a number of benefits making the examination of such mutations worth consideration.
Collapse
Affiliation(s)
- Jinny L Liu
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| | - Ellen R Goldman
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| | - Dan Zabetakis
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| | - Scott A Walper
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| | - Kendrick B Turner
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| | - Lisa C Shriver-Lake
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| | - George P Anderson
- Naval Research Laboratory, Center for Bio/Molecular Science and Engineering, Washington, DC, 20375, USA.
| |
Collapse
|
16
|
Olson MA, Zabetakis D, Legler PM, Turner KB, Anderson GP, Goldman ER. Can template-based protein models guide the design of sequence fitness for enhanced thermal stability of single domain antibodies? Protein Eng Des Sel 2015; 28:395-402. [PMID: 26374895 DOI: 10.1093/protein/gzv047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/14/2015] [Indexed: 12/18/2022] Open
Abstract
We investigate the practical use of comparative (template-based) protein models in replica-exchange simulations of single-domain antibody (sdAb) chains to evaluate if the models can correctly predict in rank order the thermal susceptibility to unfold relative to experimental melting temperatures. The baseline model system is the recently determined crystallographic structure of a llama sdAb (denoted as A3), which exhibits an unusually high thermal stability. An evaluation of the simulation results for the A3 comparative model and crystal structure shows that, despite the overall low Cα root-mean-square deviation between the two structures, the model contains misfolded regions that yields a thermal profile of unraveling at a lower temperature. Yet comparison of the simulations of four different comparative models for sdAb A3, C8, A3C8 and E9, where A3C8 is a design of swapping the sequence of the complementarity determining regions of C8 onto the A3 framework, discriminated among the sequences to detect the highest and lowest experimental melting transition temperatures. Further structural analysis of A3 for selected alanine substitutions by a combined computational and experimental study found unexpectedly that the comparative model performed admirably in recognizing substitution 'hot spots' when using a support-vector machine algorithm.
Collapse
Affiliation(s)
- Mark A Olson
- Department of Cell Biology and Biochemistry, Molecular and Translational Sciences Division, USAMRIID, Frederick, MD, USA
| | - Dan Zabetakis
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, DC, USA
| | - Patricia M Legler
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, DC, USA
| | - Kendrick B Turner
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, DC, USA
| | - George P Anderson
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, DC, USA
| | - Ellen R Goldman
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Avenue SW, Washington, DC, USA
| |
Collapse
|
17
|
Li J, Yang J, Lu YW, Wu S, Wang MR, Zhu JM. Possible Role of Staphylococcal Enterotoxin B in the Pathogenesis of Autoimmune Diseases. Viral Immunol 2015; 28:354-9. [PMID: 26086678 DOI: 10.1089/vim.2015.0017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
As a member of superantigens (SAgs) produced by Staphylococcus aureus, staphylococcal enterotoxin B (SEB) is a exotoxin superantigen that can regulate the activity of immunomodulatory and pro-inflammatory cell types. In addition, SEB plays a critical role in the pathogenesis of autoimmune disorders either by initiating the autoimmune process or by inducing a relapse in an individual in clinical remission from an autoimmune disorder. SEB can directly activate T lymphocytes, leading to the release of cytokines, superoxides, or other mediators of inflammation either directly or indirectly, because of its unique ability to cross-link human major histocompatibility complex (MHC) class II and T cell receptors (TCR), forming a trimolecular complex. This review discusses the potential effects of SEB in the pathogenesis of autoimmune diseases such as multiple sclerosis, systemic lupus erythematosus, and rheumatoid arthritis, and explores some updated therapeutic medications to neutralize SEB.
Collapse
Affiliation(s)
- Jing Li
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Jie Yang
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Yu-wei Lu
- 2 Department of Information, The Second Hospital of Anhui Medical University , Hefei, China
| | - Song Wu
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Ming-rui Wang
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Ji-min Zhu
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| |
Collapse
|
18
|
Rousserie G, Grinevich R, Brazhnik K, Even-Desrumeaux K, Reveil B, Tabary T, Chames P, Baty D, Cohen JH, Nabiev I, Sukhanova A. Detection of carcinoembryonic antigen using single-domain or full-size antibodies stained with quantum dot conjugates. Anal Biochem 2015; 478:26-32. [DOI: 10.1016/j.ab.2015.02.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 02/23/2015] [Accepted: 02/28/2015] [Indexed: 12/18/2022]
|
19
|
Geoghegan EM, Zhang H, Desai PJ, Biragyn A, Markham RB. Antiviral activity of a single-domain antibody immunotoxin binding to glycoprotein D of herpes simplex virus 2. Antimicrob Agents Chemother 2015; 59:527-35. [PMID: 25385102 PMCID: PMC4291438 DOI: 10.1128/aac.03818-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/31/2014] [Indexed: 01/30/2023] Open
Abstract
Despite years of research dedicated to preventing the sexual transmission of herpes simplex virus 2 (HSV-2), there is still no protective vaccine or microbicide against one of the most common sexually transmitted infections in the world. Using a phage display library constructed from a llama immunized with recombinant HSV-2 glycoprotein D, we identified a single-domain antibody VHH, R33, which binds to the viral surface glycoprotein D. Although R33 does not demonstrate any HSV-2 neutralization activity in vitro, when expressed with the cytotoxic domain of exotoxin A, the resulting immunotoxin (R33ExoA) specifically and potently kills HSV-2-infected cells, with a 50% neutralizing dilution (IC50) of 6.7 nM. We propose that R33ExoA could be used clinically to prevent transmission of HSV-2 through killing of virus-producing epithelial cells during virus reactivation. R33 could also potentially be used to deliver other cytotoxic effectors to HSV-2-infected cells.
Collapse
Affiliation(s)
- Eileen M Geoghegan
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hong Zhang
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Prashant J Desai
- Viral Oncology Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, Maryland, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland, USA
| | - Richard B Markham
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
20
|
Evaluation of disulfide bond position to enhance the thermal stability of a highly stable single domain antibody. PLoS One 2014; 9:e115405. [PMID: 25526640 PMCID: PMC4272287 DOI: 10.1371/journal.pone.0115405] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 11/21/2014] [Indexed: 11/21/2022] Open
Abstract
Single domain antibodies are the small recombinant variable domains derived from camelid heavy-chain-only antibodies. They are renowned for their stability, in large part due to their ability to refold following thermal or chemical denaturation. In addition to refolding after heat denaturation, A3, a high affinity anti-Staphylococcal Enterotoxin B single domain antibody, possesses a melting temperature of ∼84°C, among the highest reported for a single domain antibody. In this work we utilized the recently described crystal structure of A3 to select locations for the insertion of a second disulfide bond and evaluated the impact that the addition of this second bond had on the melting temperature. Four double-disulfide versions of A3 were constructed and each was found to improve the melting temperature relative to the native structure without reducing affinity. Placement of the disulfide bond at a previously published position between framework regions 2 and 3 yielded the largest improvement (>6°C), suggesting this location is optimal, and seemingly provides a universal route to raise the melting temperature of single domain antibodies. This study further demonstrates that even single domain antibodies with extremely high melting points can be further stabilized by addition of disulfide bonds.
Collapse
|
21
|
George J, Compton JR, Leary DH, Olson MA, Legler PM. Structural and mutational analysis of a monomeric and dimeric form of a single domain antibody with implications for protein misfolding. Proteins 2014; 82:3101-16. [PMID: 25136772 DOI: 10.1002/prot.24671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/01/2014] [Accepted: 08/11/2014] [Indexed: 11/10/2022]
Abstract
Camelid single domain antibodies (sdAb) are known for their thermal stability and reversible refolding. We have characterized an unusually stable sdAb recognizing Staphylococcal enterotoxin B with one of the highest reported melting temperatures (T(m) = 85°C). Unexpectedly, ∼10-20% of the protein formed a dimer in solution. Three other cases where <20% of the sdAb dimerized have been reported; however, this is the first report of both the monomeric and dimeric X-ray crystal structures. Concentration of the monomer did not lead to the formation of new dimer suggesting a stable conformationally distinct species in a fraction of the cytoplasmically expressed protein. Comparison of periplasmic and cytoplasmic expression showed that the dimer was associated with cytoplasmic expression. The disulfide bond was partially reduced in the WT protein purified from the cytoplasm and the protein irreversibly unfolded. Periplasmic expression produced monomeric protein with a fully formed disulfide bond and mostly reversible refolding. Crystallization of a disulfide-bond free variant, C22A/C99V, purified from the periplasm yielded a structure of a monomeric form, while crystallization of C22A/C99V from the cytoplasm produced an asymmetric dimer. In the dimer, a significant conformational asymmetry was found in the loop residues of the edge β-strands (S50-Y60) containing the highly variable complementarity determining region, CDR2. Two dimeric assemblies were predicted from the crystal packing. Mutation of a residue at one of the interfaces, Y98A, disrupted the dimer in solution. The pleomorphic homodimer may yield insight into the stability of misfolded states and the importance of the conserved disulfide bond in preventing their formation.
Collapse
Affiliation(s)
- Jade George
- Bowie State University, Bowie, 14000 Jericho Park Road, Maryland, 20715-9465
| | | | | | | | | |
Collapse
|
22
|
Bever CRS, Majkova Z, Radhakrishnan R, Suni I, McCoy M, Wang Y, Dechant J, Gee S, Hammock BD. Development and utilization of camelid VHH antibodies from alpaca for 2,2',4,4'-tetrabrominated diphenyl ether detection. Anal Chem 2014; 86:7875-82. [PMID: 25005746 PMCID: PMC4144723 DOI: 10.1021/ac501807j] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
An
antibody-based analytical method for the detection of a chemical
flame retardant using antibody fragments isolated from an alpaca has
been developed. One specific chemical flame retardant congener, 2,2′,4,4′-tetrabrominated
diphenyl ether (BDE-47), is often the major poly-BDE (PBDE) congener
present in human and environmental samples and that which is the most
frequently detected. An alpaca was immunized with a surrogate of BDE-47
covalently attached to a carrier protein. The resulting mRNA coding
for the variable domain of heavy-chain antibodies (VHH) were isolated,
transcribed to cDNA, and cloned into a phagemid vector for phage display
library construction. Selection of VHHs recognizing BDE-47 was achieved
by panning under carefully modified conditions. The assay sensitivity
for detecting BDE-47 was down to the part-per-billion (microgram per
liter) level. Cross-reactivity analyses confirmed that this method
was highly selective for BDE-47 and selected hydroxylated metabolites.
When exposed to elevated temperatures, the camelid VHH antibodies
retained more reactivity than a polyclonal antibody developed to the
same target analyte. The use of this VHH antibody reagent immobilized
onto a Au electrode for impedance biosensing demonstrates the increased
versatility of VHH antibodies.
Collapse
Affiliation(s)
- Candace R S Bever
- Department of Entomology and Nematology, University of California-Davis , Davis, California 95616, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Turner KB, Zabetakis D, Legler P, Goldman ER, Anderson GP. Isolation and epitope mapping of staphylococcal enterotoxin B single-domain antibodies. SENSORS (BASEL, SWITZERLAND) 2014; 14:10846-63. [PMID: 24949641 PMCID: PMC4118376 DOI: 10.3390/s140610846] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 12/22/2022]
Abstract
Single-domain antibodies (sdAbs), derived from the heavy chain only antibodies found in camelids such as llamas have the potential to provide rugged detection reagents with high affinities, and the ability to refold after denaturation. We have isolated and characterized sdAbs specific to staphylococcal enterotoxin B (SEB) which bind to two distinct epitopes and are able to function in a sandwich immunoassay for toxin detection. Characterization of these sdAbs revealed that each exhibited nanomolar binding affinities or better. Melting temperatures for the sdAbs ranged from approximately 60 °C to over 70 °C, with each demonstrating at least partial refolding after denaturation and several were able to completely refold. A first set of sdAbs was isolated by panning the library using adsorbed antigen, all of which recognized the same epitope on SEB. Epitope mapping suggested that these sdAbs bind to a particular fragment of SEB (VKSIDQFLYFDLIYSI) containing position L45 (underlined), which is involved in binding to the major histocompatibility complex (MHC). Differences in the binding affinities of the sdAbs to SEB and a less-toxic vaccine immunogen, SEBv (L45R/Y89A/Y94A) were also consistent with binding to this epitope. A sandwich panning strategy was utilized to isolate sdAbs which bind a second epitope. This epitope differed from the initial one obtained or from that recognized by previously isolated anti-SEB sdAb A3. Using SEB-toxin spiked milk we demonstrated that these newly isolated sdAbs could be utilized in sandwich-assays with each other, A3, and with various monoclonal antibodies.
Collapse
Affiliation(s)
- Kendrick B Turner
- American Society for Engineering Education, Postdoctoral Fellow at the Naval Research Laboratory, Washington, DC 20375, USA.
| | - Dan Zabetakis
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA.
| | - Patricia Legler
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA.
| | - Ellen R Goldman
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA.
| | - George P Anderson
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA.
| |
Collapse
|
24
|
Pan LY, Salas-Solano O, Valliere-Douglass JF. Conformation and dynamics of interchain cysteine-linked antibody-drug conjugates as revealed by hydrogen/deuterium exchange mass spectrometry. Anal Chem 2014; 86:2657-64. [PMID: 24512515 DOI: 10.1021/ac404003q] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Antibody-drug conjugates (ADCs) are protein therapeutics in which a target specific monoclonal antibody (mAb) is conjugated with drug molecules. The manufacturing of ADCs involves additional conjugation steps, which are carried out on the parent mAbs, and it is important to evaluate how the drug conjugation process impacts the conformation and dynamics of the mAb. Here, we present a comparative study of interchain cysteine linked IgG1 ADCs and the corresponding mAb by hydrogen/deuterium exchange mass spectrometry (HDX-MS). We found that ∼90% of the primary sequence of the ADC conjugated with either monomethyl auristatin E or F (vcMMAE/mcMMAF) displayed the same HDX kinetics as the mAb, indicating the ADCs and mAbs share very similar conformation and dynamics in solution. Minor increases in HDX kinetic rates were observed in two Fc regions in the ADCs relative to the mAb which indicated that both regions become more structurally dynamic and/or more solvent-accessible in the ADCs. The findings led to a subsequent inquiry into whether the local conformational changes were due to the presence of drugs on the interchain cysteine residues or the absence of intact interchain disulfides or both. To address this question, a side-by-side HDX comparison of ADCs, mAbs, reduced mAbs (containing 8 reduced interchain cysteine thiols), and partially reduced mAbs (conjugation process intermediate) was performed. Our results indicated that the slight increase in conformational dynamics detected at the two regions in the ADCs was due to the absence of intact interchain disulfide bonds and not the presence of vcMMAE or mcMMAF on the alkylated interchain cysteine residues. These results highlight the utility of HDX-MS for interrogating the higher-order structure of ADCs and other protein therapeutics.
Collapse
Affiliation(s)
- Lucy Yan Pan
- Seattle Genetics, Inc., 21823 30th Drive SE, Bothell, Washington 98021, United States
| | | | | |
Collapse
|
25
|
Turner KB, Zabetakis D, Goldman ER, Anderson GP. Enhanced stabilization of a stable single domain antibody for SEB toxin by random mutagenesis and stringent selection. Protein Eng Des Sel 2014; 27:89-95. [PMID: 24488977 DOI: 10.1093/protein/gzu001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Single domain antibodies, recombinant variable heavy domains derived from the unique heavy-chain only antibodies found in camelids and sharks, are exceptionally rugged due to their ability to refold following heat or chemical denaturation. In addition, a number of single domain antibodies have been found to possess high melting points which provide an even greater degree of stability; one of these, llama-derived A3, is a binder of Staphylococcal enterotoxin B and has a Tm of 83.5 °C. In this work, we utilized random mutagenesis and stringent selection in an effort to obtain variants of A3 with even higher melting points. This effort resulted in the selection of a double mutant, A3-T28I-S72I, which has a melting point of 90.0 °C and near wild-type affinity for the target antigen. We further characterized the mutations individually to determine that while both contributed to the thermal stabilization, the T28I mutation accounted for ∼ 4.1 °C of the 6.5 °C increase. This work demonstrates that by the addition of relatively subtle changes it is possible to further improve the melting temperature of single domain antibodies that are already remarkably stable.
Collapse
Affiliation(s)
- Kendrick B Turner
- American Society for Engineering Education Postdoctoral Fellow, Naval Research Laboratory, Washington, DC 20375, USA
| | | | | | | |
Collapse
|
26
|
Goldman ER, Brozozog-Lee PA, Zabetakis D, Turner KB, Walper SA, Liu JL, Anderson GP. Negative tail fusions can improve ruggedness of single domain antibodies. Protein Expr Purif 2014; 95:226-32. [PMID: 24440507 DOI: 10.1016/j.pep.2014.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 01/06/2014] [Accepted: 01/08/2014] [Indexed: 12/31/2022]
Abstract
Single-domain antibodies (sdAbs), the recombinantly expressed binding domains derived from the heavy-chain-only antibodies found in camelids and sharks, are valued for their ability to refold after heat denaturation. However, some sdAbs are prone to aggregation on extended heating at high concentration. Additionally, sdAbs prepared cytoplasmically often lack the conserved disulfide bond found in variable heavy domains, which both decreases their melting point and can decrease their ability to refold. Genetic fusions of sdAbs with the acid tail of α-synuclein (ATS) resulted in constructs that had enhanced ability to resist aggregation. In addition, almost complete refolding was observed even in the absence of the disulfide bond. These sdAb-ATS fusions expand the utility of sdAbs. They provide sdAbs that are resistant to aggregation, and enable the production of re-foldable sdAbs in the reducing environment of the cytoplasm.
Collapse
Affiliation(s)
- Ellen R Goldman
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave. SW, Washington, DC 20375, USA
| | | | - Dan Zabetakis
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave. SW, Washington, DC 20375, USA
| | - Kendrick B Turner
- Science and Engineering Apprenticeship Program, American Society for Engineering Education, 4555 Overlook Ave. SW, Washington, DC 20375, USA
| | - Scott A Walper
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave. SW, Washington, DC 20375, USA
| | - Jinny L Liu
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave. SW, Washington, DC 20375, USA
| | - George P Anderson
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, 4555 Overlook Ave. SW, Washington, DC 20375, USA.
| |
Collapse
|
27
|
Contributions of the complementarity determining regions to the thermal stability of a single-domain antibody. PLoS One 2013; 8:e77678. [PMID: 24143255 PMCID: PMC3797041 DOI: 10.1371/journal.pone.0077678] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/03/2013] [Indexed: 11/19/2022] Open
Abstract
Single domain antibodies (sdAbs) are the recombinantly-expressed variable domain from camelid (or shark) heavy chain only antibodies and provide rugged recognition elements. Many sdAbs possess excellent affinity and specificity; most refold and are able to bind antigen after thermal denaturation. The sdAb A3, specific for the toxin Staphylococcal enterotoxin B (SEB), shows both sub-nanomolar affinity for its cognate antigen (0.14 nM) and an unusually high melting point of 85°C. Understanding the source of sdAb A3's high melting temperature could provide a route for engineering improved melting temperatures into other sdAbs. The goal of this work was to determine how much of sdAb A3's stability is derived from its complementarity determining regions (CDRs) versus its framework. Towards answering this question we constructed a series of CDR swap mutants in which the CDRs from unrelated sdAbs were integrated into A3's framework and where A3's CDRs were integrated into the framework of the other sdAbs. All three CDRs from A3 were moved to the frameworks of sdAb D1 (a ricin binder that melts at 50°C) and the anti-ricin sdAb C8 (melting point of 60°C). Similarly, the CDRs from sdAb D1 and sdAb C8 were moved to the sdAb A3 framework. In addition individual CDRs of sdAb A3 and sdAb D1 were swapped. Melting temperature and binding ability were assessed for each of the CDR-exchange mutants. This work showed that CDR2 plays a critical role in sdAb A3's binding and stability. Overall, results from the CDR swaps indicate CDR interactions play a major role in the protein stability.
Collapse
|
28
|
Arezumand R, Mahdian R, Behdani M, Khanahmad H, Langari J, Namvarasl N, Hassanzadeh-Ghasabeh R, Zeinali S. Recombinant expression and purification of human placental growth factor 1 and specific camel heavy chain polyclonal antibody preparation. Saudi J Biol Sci 2013; 21:35-9. [PMID: 24596498 DOI: 10.1016/j.sjbs.2013.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/18/2013] [Accepted: 04/24/2013] [Indexed: 01/28/2023] Open
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family. Unlike VEGF, PlGF is dispensable for normal cell development as well as playing various roles in pathological angiogenesis which occurs in tissue ischemia, inflammation, and malignancy. The PlGF-1 has been considered as a potential candidate for the diagnosis and targeting of pathological angiogenesis. Camelidae serum contains an important fraction of functional antibodies, called heavy-chain antibodies (HcAbs) that are naturally devoid of light chains. Camelid HcAbs recognize their cognate antigens by a single variable-domain, referred to as VHH or Nanobody. Here, we describe the expression and purification of recombinant human PlGF-1 (rhPlGF-1). This protein was subsequently used for the preparation of camel heavy chain polyclonal antibody against rhPlGF-1. The recombinant expression plasmid pET-26b-hPlGF-1 was introduced into Escherichia coli BL21 cells to express the rhPlGF-1 protein. Purified rhPlGF-1 was used to immunize camel, the specific reactivity of HcAb was determined with ELISA and western blot. Western blot analysis indicated that the antiserum specifically reacted to the recombinant protein. The rhPlGF-1 protein and its antibody may be used for the development of detection assays needed for clinical research.
Collapse
Affiliation(s)
- Roghaye Arezumand
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Mahdian
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Venom & Biotherapeutics Molecules Lab., Pasteur Institute of Iran, Tehran, Iran
| | - Hossein Khanahmad
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran ; Department of Genetics and Molecular Biology, Medical School of Isfahan University of Medical Science, Isfahan, Iran
| | - Jahangir Langari
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Nabiollah Namvarasl
- Department of Laboratory Animal Science, Research and Production Complex, Pasteur Institute of Iran, Karaj, Iran
| | - Reza Hassanzadeh-Ghasabeh
- Laboratory of Cellular and Molecular Immunology, Vrije University Brussel, Brussels, Belgium ; Department of Structural Biology, NSF, VIB, Brussels, Belgium
| | - Sirous Zeinali
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
29
|
Abstract
Staphylococcus aureus plays an important role in numerous human cases of food poisoning, soft tissue, and bone infections, as well as potentially lethal toxic shock. This common bacterium synthesizes various virulence factors that include staphylococcal enterotoxins (SEs). These protein toxins bind directly to major histocompatibility complex class II on antigen-presenting cells and specific Vβ regions of T-cell receptors, resulting in potentially life-threatening stimulation of the immune system. Picomolar concentrations of SEs ultimately elicit proinflammatory cytokines that can induce fever, hypotension, multi-organ failure, and lethal shock. Various in vitro and in vivo models have provided important tools for studying the biological effects of, as well as potential vaccines/therapeutics against, the SEs. This review succinctly presents known physical and biological properties of the SEs, including various intervention strategies. In particular, SEB will often be portrayed as per biodefense concerns dating back to the 1960s.
Collapse
Affiliation(s)
- Teresa Krakauer
- Integrated Toxicology Division; United States Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD USA
| | | |
Collapse
|
30
|
Legler PM, Zabetakis D, Anderson GP, Lam A, Hol WGJ, Goldman ER. Structure of a low-melting-temperature anti-cholera toxin: llama V(H)H domain. Acta Crystallogr Sect F Struct Biol Cryst Commun 2013; 69:90-3. [PMID: 23385744 DOI: 10.1107/s1744309112050750] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 12/13/2012] [Indexed: 01/29/2023]
Abstract
Variable heavy domains derived from the heavy-chain-only antibodies found in camelids (V(H)H domains) are known for their thermal stability. Here, the structure of A9, an anti-cholera toxin V(H)H domain (K(d) = 77 ± 5 nM) that has an unusually low melting temperature of 319.9 ± 1.6 K, is reported. The CDR3 residues of A9 form a β-hairpin that is directed away from the former V(H)-V(L) interfacial surface, exposing hydrophobic residues to the solvent. A DALI structural similarity search showed that this CDR3 conformation is uncommon.
Collapse
Affiliation(s)
- Patricia M Legler
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Selection and evaluation of single domain antibodies toward MS2 phage and coat protein. Mol Immunol 2013; 53:118-25. [DOI: 10.1016/j.molimm.2012.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 07/16/2012] [Accepted: 07/24/2012] [Indexed: 11/23/2022]
|
32
|
Comparison of single domain antibody immobilization strategies evaluated by surface plasmon resonance. J Immunol Methods 2012; 388:68-77. [PMID: 23261918 DOI: 10.1016/j.jim.2012.11.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 11/23/2012] [Accepted: 11/26/2012] [Indexed: 11/22/2022]
Abstract
The use of single domain antibodies (sdAbs) in place of conventional antibodies for both therapeutic and diagnostic applications continues to grow. SdAbs offer a number of advantages when compared to conventional antibodies such as their small size and low structural complexity which allows them to readily be produced as fusions in a variety formats. In this work we compared the utility of various C-terminal fusions and immobilization strategies for two sdAbs; one which recognizes ricin and the other EA1, an S-layer protein, of Bacillus anthracis. Comparisons were made between direct covalent attachment and affinity immobilization using a biotin-streptavidin interaction for the standard sdAb monomers, randomly and site-specifically biotinylated monomers, and fusion constructs of alkaline phosphatase dimers and streptavidin core tetramers. The sdAb binding and regeneration was evaluated by surface plasmon resonance in a multiplexed format. The construct that provided the highest density of active molecules by at least a factor of two was the sdAb-streptavidin core tetramer, followed by the sdAb-alkaline phosphatase and then the site-specifically biotinylated monomer. The poorest performing immobilization methods were the two most common, direct covalent attachment and the randomly biotinylated sdAb attached via NeutrAvidin. These improvements directly correlated to antigen capture in SPR assays. Similarly, the oriented immobilization method also translated to improvements in limit of detection assays using a bead-based system. The sdAb-streptavidin core provided more than a 100-fold improvement in the limit of detection of EA1, from ~200 ng/mL to to 1.6 ng/mL, while improvement for ricin detection was less but still a significant 5-fold decrease, going from 1.6 ng/mL down to 0.32 ng/mL. This demonstrated improvement in limits of detection is an advantage that should be transferable to most assay formats.
Collapse
|