1
|
Rizk SS, Moustafa DM, ElBanna SA, Nour El-Din HT, Attia AS. Nanobodies in the fight against infectious diseases: repurposing nature's tiny weapons. World J Microbiol Biotechnol 2024; 40:209. [PMID: 38771414 PMCID: PMC11108896 DOI: 10.1007/s11274-024-03990-4] [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: 02/20/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
Nanobodies are the smallest known antigen-binding molecules to date. Their small size, good tissue penetration, high stability and solubility, ease of expression, refolding ability, and negligible immunogenicity in the human body have granted them excellence over conventional antibodies. Those exceptional attributes of nanobodies make them promising candidates for various applications in biotechnology, medicine, protein engineering, structural biology, food, and agriculture. This review presents an overview of their structure, development methods, advantages, possible challenges, and applications with special emphasis on infectious diseases-related ones. A showcase of how nanobodies can be harnessed for applications including neutralization of viruses and combating antibiotic-resistant bacteria is detailed. Overall, the impact of nanobodies in vaccine design, rapid diagnostics, and targeted therapies, besides exploring their role in deciphering microbial structures and virulence mechanisms are highlighted. Indeed, nanobodies are reshaping the future of infectious disease prevention and treatment.
Collapse
Affiliation(s)
- Soha S Rizk
- Microbiology and Immunology Postgraduate Program, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Dina M Moustafa
- Department of Medical Sciences, Faculty of Dentistry, The British University in Egypt, El Sherouk City, Cairo, 11837, Egypt
| | - Shahira A ElBanna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hanzada T Nour El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed S Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
2
|
Salmen W, Hu L, Bok M, Chaimongkol N, Ettayebi K, Sosnovtsev SV, Soni K, Ayyar BV, Shanker S, Neill FH, Sankaran B, Atmar RL, Estes MK, Green KY, Parreño V, Prasad BVV. A single nanobody neutralizes multiple epochally evolving human noroviruses by modulating capsid plasticity. Nat Commun 2023; 14:6516. [PMID: 37845211 PMCID: PMC10579229 DOI: 10.1038/s41467-023-42146-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/28/2023] [Indexed: 10/18/2023] Open
Abstract
Acute gastroenteritis caused by human noroviruses (HuNoVs) is a significant global health and economic burden and is without licensed vaccines or antiviral drugs. The GII.4 HuNoV causes most epidemics worldwide. This virus undergoes epochal evolution with periodic emergence of variants with new antigenic profiles and altered specificity for histo-blood group antigens (HBGA), the determinants of cell attachment and susceptibility, hampering the development of immunotherapeutics. Here, we show that a llama-derived nanobody M4 neutralizes multiple GII.4 variants with high potency in human intestinal enteroids. The crystal structure of M4 complexed with the protruding domain of the GII.4 capsid protein VP1 revealed a conserved epitope, away from the HBGA binding site, fully accessible only when VP1 transitions to a "raised" conformation in the capsid. Together with dynamic light scattering and electron microscopy of the GII.4 VLPs, our studies suggest a mechanism in which M4 accesses the epitope by altering the conformational dynamics of the capsid and triggering its disassembly to neutralize GII.4 infection.
Collapse
Affiliation(s)
- Wilhelm Salmen
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Marina Bok
- Virology Institute and Technology Innovation, IVIT, CONICET-INTA, Hurlingham, Buenos Aires, Argentina
| | - Natthawan Chaimongkol
- Caliciviruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Khalil Ettayebi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Stanislav V Sosnovtsev
- Caliciviruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaundal Soni
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - B Vijayalakshmi Ayyar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sreejesh Shanker
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Frederick H Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Banumathi Sankaran
- Berkeley Center for Structural Biology, Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley Laboratory, Berkeley, CA, USA
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kim Y Green
- Caliciviruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Viviana Parreño
- Virology Institute and Technology Innovation, IVIT, CONICET-INTA, Hurlingham, Buenos Aires, Argentina
| | - B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Prado NDR, Brilhante-Da-Silva N, Sousa RMO, Morais MSDS, Roberto SA, Luiz MB, Assis LCD, Marinho ACM, Araujo LFLD, Pontes RDS, Stabeli RG, Fernandes CFC, Pereira SDS. Single-domain antibodies applied as antiviral immunotherapeutics. J Virol Methods 2023; 320:114787. [PMID: 37516366 DOI: 10.1016/j.jviromet.2023.114787] [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: 12/12/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Viral infections have been the cause of high mortality rates throughout different periods in history. Over the last two decades, outbreaks caused by zoonotic diseases and transmitted by arboviruses have had a significant impact on human health. The emergence of viral infections in different parts of the world encourages the search for new inputs to fight pathologies of viral origin. Antibodies represent the predominant class of new drugs developed in recent years and approved for the treatment of various human diseases, including cancer, autoimmune and infectious diseases. A promising group of antibodies are single-domain antibodies derived from camelid heavy chain immunoglobulins, or VHHs, are biomolecules with nanometric dimensions and unique pharmaceutical and biophysical properties that can be used in the diagnosis and immunotherapy of viral infections. For viral neutralization to occur, VHHs can act in different stages of the viral cycle, including the actual inhibition of infection, to hindering viral replication or assembly. This review article addresses advances involving the use of VHHs in therapeutic propositions aimed to battle different viruses that affect human health.
Collapse
Affiliation(s)
- Nidiane Dantas Reis Prado
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | - Nairo Brilhante-Da-Silva
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil
| | - Rosa Maria Oliveira Sousa
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil
| | | | - Sibele Andrade Roberto
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Marcos Barros Luiz
- Instituto Federal de Rondônia Campus Guajará-Mirim, IFRO, Guajará-Mirim, RO, Brazil
| | - Livia Coelho de Assis
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil
| | - Anna Carolina M Marinho
- Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Luiz Felipe Lemes de Araujo
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rafael de Souza Pontes
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade de São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Rodrigo Guerino Stabeli
- Plataforma Bi-institucional de Medicina Translacional, Fundação Oswaldo Cruz-USP, Ribeirão Preto, SP, Brazil
| | - Carla Freire Celedonio Fernandes
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Laboratório Multiusuário de Pesquisa e Desenvolvimento, Fundação Oswaldo Cruz, Fiocruz unidade Ceará, Eusebio, CE, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Soraya Dos Santos Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, FIOCRUZ, unidade Rondônia, Porto Velho, RO, Brazil; Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, RJ, Brazil; Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
4
|
Misson Mindrebo L, Liu H, Ozorowski G, Tran Q, Woehl J, Khalek I, Smith J, Barman S, Zhao F, Keating C, Limbo O, Verma M, Liu J, Stanfield R, Zhu X, Turner H, Sok D, Huang PS, Burton D, Ward A, Wilson I, Jardine J. Fully synthetic platform to rapidly generate tetravalent bispecific nanobody-based immunoglobulins. Proc Natl Acad Sci U S A 2023; 120:e2216612120. [PMID: 37276407 PMCID: PMC10268213 DOI: 10.1073/pnas.2216612120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
Nanobodies bind a target antigen with a kinetic profile similar to a conventional antibody, but exist as a single heavy chain domain that can be readily multimerized to engage antigen via multiple interactions. Presently, most nanobodies are produced by immunizing camelids; however, platforms for animal-free production are growing in popularity. Here, we describe the development of a fully synthetic nanobody library based on an engineered human VH3-23 variable gene and a multispecific antibody-like format designed for biparatopic target engagement. To validate our library, we selected nanobodies against the SARS-CoV-2 receptor-binding domain and employed an on-yeast epitope binning strategy to rapidly map the specificities of the selected nanobodies. We then generated antibody-like molecules by replacing the VH and VL domains of a conventional antibody with two different nanobodies, designed as a molecular clamp to engage the receptor-binding domain biparatopically. The resulting bispecific tetra-nanobody immunoglobulins neutralized diverse SARS-CoV-2 variants with potencies similar to antibodies isolated from convalescent donors. Subsequent biochemical analyses confirmed the accuracy of the on-yeast epitope binning and structures of both individual nanobodies, and a tetra-nanobody immunoglobulin revealed that the intended mode of interaction had been achieved. This overall workflow is applicable to nearly any protein target and provides a blueprint for a modular workflow for the development of multispecific molecules.
Collapse
Affiliation(s)
- Laetitia Misson Mindrebo
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Gabriel Ozorowski
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
| | - Quoc Tran
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Jordan Woehl
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Irene Khalek
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Jessica M. Smith
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Shawn Barman
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA92037
| | - Fangzhu Zhao
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA92037
| | - Celina Keating
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA92037
| | - Oliver Limbo
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Megan Verma
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| | - Jingjia Liu
- Department of Bioengineering, Stanford University, Stanford, CA94305
| | - Robyn L. Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Hannah L. Turner
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
| | - Devin Sok
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA92037
| | - Po-Ssu Huang
- Department of Bioengineering, Stanford University, Stanford, CA94305
| | - Dennis R. Burton
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA92037
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA02139
| | - Andrew B. Ward
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA92037
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA92037
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Joseph G. Jardine
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA92037
- International AIDS Vaccine Initiative, New York, NY10004
| |
Collapse
|
5
|
Bhattacharya M, Chatterjee S, Lee SS, Chakraborty C. Therapeutic applications of nanobodies against SARS-CoV-2 and other viral infections: Current update. Int J Biol Macromol 2023; 229:70-80. [PMID: 36586649 PMCID: PMC9797221 DOI: 10.1016/j.ijbiomac.2022.12.284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/15/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022]
Abstract
In the last two years, the world encountered the SARS-CoV-2 virus, which is still dominating the population due to the absence of a viable treatment. To eradicate the global pandemic, scientists, doctors, and researchers took an exceptionally significant initiative towards the development of effective therapeutics to save many lifes. This review discusses about the single-domain antibodies (sdAbs), also called nanobodies, their structure, and their types against the infections of dreadful SARS-CoV-2 virus. A precise description highlights the nanobodies and their therapeutic application against the other selected viruses. It aims to focus on the extraordinary features of these antibodies compared to the conventional therapeutics like mAbs, convalescent plasma therapy, and vaccines. The stable structure of these nanobodies along with the suitable mechanism of action also confers greater resistance to the evolving variants with numerous mutations. The nanobodies developed against SARS-CoV-2 and its mutant variants have shown the greater neutralization potential than the primitive ones. Engineering of these specialized antibodies by modern biotechnological approaches will surely be more beneficial in treating this COVID-19 pandemic along with certain other viral infections.
Collapse
Affiliation(s)
- Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore 756020, Odisha, India
| | - Srijan Chatterjee
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India.
| |
Collapse
|
6
|
Chen Y, Wang X, Zhang M, Li J, Gao X, Nan Y, Zhao Q, Zhou EM, Liu B. Identification of two novel neutralizing nanobodies against swine hepatitis E virus. Front Microbiol 2022; 13:1048180. [PMID: 36504801 PMCID: PMC9727072 DOI: 10.3389/fmicb.2022.1048180] [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: 09/19/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
Hepatitis E virus (HEV) is thought to be a zoonotic pathogen that causes serious economic loss and threatens human health. However, there is a lack of efficient antiviral strategies. As a more promising tool for antiviral therapy, nanobodies (also named single-domain antibodies, sdAbs) exhibit higher specificity and affinity than traditional antibodies. In this study, nanobody anti-genotype four HEV open reading frame 2 (ORF2) was screened using phage display technology, and two nanobodies (nb14 and nb53) with high affinity were prokaryotically expressed. They were identified to block HEV ORF2 virus like particle (VLP) sp239 (aa 368-606) absorbing HepG2 cells in vitro. With the previously built animal model, the detection indicators of fecal shedding, viremia, seroconversion, alanine aminotransferase (ALT) levels, and liver lesions showed that nb14 could completely protect rabbits from swine HEV infection, and nb53 partially blocked swine HEV infection in rabbits. Collectively, these results revealed that nb14, with its anti-HEV neutralizing activity, may be developed as an antiviral drug for HEV.
Collapse
|
7
|
Saied AA, Metwally AA, Alobo M, Shah J, Sharun K, Dhama K. Bovine-derived antibodies and camelid-derived nanobodies as biotherapeutic weapons against SARS-CoV-2 and its variants: A review article. Int J Surg 2022; 98:106233. [PMID: 35065260 PMCID: PMC8768012 DOI: 10.1016/j.ijsu.2022.106233] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected 305 million individuals worldwide and killed about 5.5 million people as of January 10, 2022. SARS-CoV-2 is the third major outbreak caused by a new coronavirus in the previous two decades, following SARS-CoV and MERS-CoV. Even though vaccination against SARS-CoV-2 is considered a critical strategy for preventing virus spread in the population and limiting COVID-19 clinical manifestations, new therapeutic drugs, and management strategies are urgently needed, particularly in light of the growing number of SARS-CoV-2 variants (such as Delta and Omicron variants). However, the use of conventional antibodies has faced many challenges, such as viral escape mutants, increased instability, weak binding, large sizes, the need for large amounts of plasma, and high-cost manufacturing. Furthermore, the emergence of new SARS-CoV-2 variants in the human population and recurrent coronavirus spillovers highlight the need for broadly neutralizing antibodies that are not affected by an antigenic drift that could limit future zoonotic infection. Bovine-derived antibodies and camelid-derived nanobodies are more potent and protective than conventional human antibodies, thanks to their inbuilt characteristics, and can be produced in large quantities. In addition, it was reported that these biotherapeutics are effective against a broad spectrum of epitopes, reducing the opportunity of viral pathogens to develop mutational escape. In this review, we focus on the potential benefits behind our rationale for using bovine-derived antibodies and camelid-derived nanobodies in countering SARS-CoV-2 and its emerging variants and mutants.
Collapse
Affiliation(s)
- AbdulRahman A. Saied
- Department of Food Establishments Licensing (Aswan Branch), National Food Safety Authority (NFSA), Aswan, 81511, Egypt,Touristic Activities and Interior Offices Sector (Aswan Office), Ministry of Tourism and Antiquities, Aswan, 81511, Egypt,Corresponding author. Department of Food Establishments Licensing (Aswan Branch), National Food Safety Authority (NFSA), Aswan, 81511, Egypt
| | - Asmaa A. Metwally
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81511, Egypt,Corresponding author. Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Moses Alobo
- Grand Challenges Africa, Science for Africa Foundation, Nairobi, Kenya
| | - Jaffer Shah
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
8
|
Gransagne M, Aymé G, Brier S, Chauveau-Le Friec G, Meriaux V, Nowakowski M, Dejardin F, Levallois S, Dias de Melo G, Donati F, Prot M, Brûlé S, Raynal B, Bellalou J, Goncalves P, Montagutelli X, Di Santo JP, Lazarini F, England P, Petres S, Escriou N, Lafaye P. Development of a highly specific and sensitive VHH-based sandwich immunoassay for the detection of the SARS-CoV-2 nucleoprotein. J Biol Chem 2021; 298:101290. [PMID: 34678315 PMCID: PMC8526496 DOI: 10.1016/j.jbc.2021.101290] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/15/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022] Open
Abstract
The current COVID-19 pandemic illustrates the importance of obtaining reliable methods for the rapid detection of SARS-CoV-2. A highly specific and sensitive diagnostic test able to differentiate the SARS-CoV-2 virus from common human coronaviruses is therefore needed. Coronavirus nucleoprotein (N) localizes to the cytoplasm and the nucleolus and is required for viral RNA synthesis. N is the most abundant coronavirus protein, so it is of utmost importance to develop specific antibodies for its detection. In this study, we developed a sandwich immunoassay to recognize the SARS-CoV-2 N protein. We immunized one alpaca with recombinant SARS-CoV-2 N and constructed a large single variable domain on heavy chain (VHH) antibody library. After phage display selection, seven VHHs recognizing the full N protein were identified by ELISA. These VHHs did not recognize the nucleoproteins of the four common human coronaviruses. Hydrogen Deuterium eXchange–Mass Spectrometry (HDX-MS) analysis also showed that these VHHs mainly targeted conformational epitopes in either the C-terminal or the N-terminal domains. All VHHs were able to recognize SARS-CoV-2 in infected cells or on infected hamster tissues. Moreover, the VHHs could detect the SARS variants B.1.17/alpha, B.1.351/beta, and P1/gamma. We propose that this sandwich immunoassay could be applied to specifically detect the SARS-CoV-2 N in human nasal swabs.
Collapse
Affiliation(s)
| | - Gabriel Aymé
- Plateforme d'Ingénierie des Anticorps, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Sébastien Brier
- Plateforme technologique de RMN biologique, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | | | - Véronique Meriaux
- Plateforme d'Ingénierie des Anticorps, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Mireille Nowakowski
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - François Dejardin
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Sylvain Levallois
- Biology of Infection Unit, Institut Pasteur, Inserm U1117, Paris, France
| | | | - Flora Donati
- Molecular Genetics of RNA viruses, UMR 3569 CNRS, University of Paris, Institut Pasteur, Paris, France; National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
| | - Matthieu Prot
- Evolutionary Genomics of RNA viruses, Institut Pasteur, Paris, France
| | - Sébastien Brûlé
- Plateforme de Biophysique Moléculaire, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Bertrand Raynal
- Plateforme de Biophysique Moléculaire, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Jacques Bellalou
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Pedro Goncalves
- Unité d'Immunité Innée, Institut Pasteur, Paris, France; INSERM U1223, Paris, France
| | | | - James P Di Santo
- Unité d'Immunité Innée, Institut Pasteur, Paris, France; INSERM U1223, Paris, France
| | - Françoise Lazarini
- Perception and Memory Unit, Institut Pasteur, CNRS UMR 3571, Paris, France
| | - Patrick England
- Plateforme de Biophysique Moléculaire, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Stéphane Petres
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Nicolas Escriou
- Département de Santé Globale, Institut Pasteur, Paris, France
| | - Pierre Lafaye
- Plateforme d'Ingénierie des Anticorps, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France.
| |
Collapse
|
9
|
Abstract
Tick-borne encephalitis virus (TBEV), of the genus Flavivirus, is a causative agent of severe encephalitis in endemic regions of northern Asia and central and northern Europe. Interferon induced transmembrane proteins (IFITMs) are restriction factors that inhibit the replication cycles of numerous viruses, including flaviviruses such as the West Nile virus, dengue virus, and Zika virus. Here, we demonstrate the role of IFITM1, IFITM2, and IFITM3 in the inhibition of TBEV infection and in protection against virus-induced cell death. We show the most significant role being that of IFITM3, including the dissection of its functional motifs by mutagenesis. Furthermore, through the use of CRISPR-Cas9-generated IFITM1/3-knockout monoclonal cell lines, we confirm the role and additive action of endogenous IFITMs in TBEV suppression. However, the results of co-culture assays suggest that TBEV might partially escape IFN- and IFITM-mediated suppression during high-density co-culture infection when the virus enters naïve cells directly from infected donor cells. Thus, cell-to-cell spread may constitute a strategy for virus escape from innate host defenses. Importance: TBEV infection may result in encephalitis, chronic illness or death. TBEV is endemic in northern Asia and Europe; however, due to climate change, new endemic centers arise. Although effective TBEV vaccines have been approved, vaccination coverage is low, and, due to the lack of specific therapeutics, infected individuals depend on their immune responses to control the infection. The IFITM proteins are components of the innate antiviral defenses that suppress cell entry of many viral pathogens. However, no studies regarding the role of IFITM proteins in the TBEV infection have been published so far. Understanding of antiviral innate immune responses is crucial for future development of antiviral strategies. Here, we show the important role of IFITM proteins in the inhibition of TBEV infection and virus-mediated cell death. However, our data suggest that TBEV cell-to-cell spread may be less prone to both IFN- and IFITM-mediated suppression, potentially facilitating escape from IFITM-mediated immunity.
Collapse
|
10
|
Ye Q, Lu S, Corbett KD. Structural Basis for SARS-CoV-2 Nucleocapsid Protein Recognition by Single-Domain Antibodies. Front Immunol 2021; 12:719037. [PMID: 34381460 PMCID: PMC8351461 DOI: 10.3389/fimmu.2021.719037] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
The COVID-19 pandemic, caused by the coronavirus SARS-CoV-2, is the most severe public health event of the twenty-first century. While effective vaccines against SARS-CoV-2 have been developed, there remains an urgent need for diagnostics to quickly and accurately detect infections. Antigen tests, particularly those that detect the abundant SARS-CoV-2 Nucleocapsid protein, are a proven method for detecting active SARS-CoV-2 infections. Here we report high-resolution crystal structures of three llama-derived single-domain antibodies that bind the SARS-CoV-2 Nucleocapsid protein with high affinity. Each antibody recognizes a specific folded domain of the protein, with two antibodies recognizing the N-terminal RNA binding domain and one recognizing the C-terminal dimerization domain. The two antibodies that recognize the RNA binding domain affect both RNA binding affinity and RNA-mediated phase separation of the Nucleocapsid protein. All three antibodies recognize highly conserved surfaces on the Nucleocapsid protein, suggesting that they could be used to develop affordable diagnostic tests to detect all circulating SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Qiaozhen Ye
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, United States
| | - Shan Lu
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, United States
| | - Kevin D Corbett
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, United States.,Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
11
|
Sokullu E, Gauthier MS, Coulombe B. Discovery of Antivirals Using Phage Display. Viruses 2021; 13:v13061120. [PMID: 34200959 PMCID: PMC8230593 DOI: 10.3390/v13061120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
The latest coronavirus disease outbreak, COVID-19, has brought attention to viral infections which have posed serious health threats to humankind throughout history. The rapid global spread of COVID-19 is attributed to the increased human mobility of today's world, yet the threat of viral infections to global public health is expected to increase continuously in part due to increasing human-animal interface. Development of antiviral agents is crucial to combat both existing and novel viral infections. Recently, there is a growing interest in peptide/protein-based drug molecules. Antibodies are becoming especially predominant in the drug market. Indeed, in a remarkably short period, four antibody therapeutics were authorized for emergency use in COVID-19 treatment in the US, Russia, and India as of November 2020. Phage display has been one of the most widely used screening methods for peptide/antibody drug discovery. Several phage display-derived biologics are already in the market, and the expiration of intellectual property rights of phage-display antibody discovery platforms suggests an increment in antibody drugs in the near future. This review summarizes the most common phage display libraries used in antiviral discovery, highlights the approaches employed to enhance the antiviral potency of selected peptides/antibody fragments, and finally provides a discussion about the present status of the developed antivirals in clinic.
Collapse
Affiliation(s)
- Esen Sokullu
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Correspondence: (E.S.); (B.C.)
| | - Marie-Soleil Gauthier
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
| | - Benoit Coulombe
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Correspondence: (E.S.); (B.C.)
| |
Collapse
|
12
|
Ye Q, Lu S, Corbett KD. Structural basis for SARS-CoV-2 Nucleocapsid protein recognition by single-domain antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.06.01.446591. [PMID: 34100017 PMCID: PMC8183014 DOI: 10.1101/2021.06.01.446591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic, caused by the coronavirus SARS-CoV-2, is the most severe public health event of the twenty-first century. While effective vaccines against SARS-CoV-2 have been developed, there remains an urgent need for diagnostics to quickly and accurately detect infections. Antigen tests, particularly those that detect the abundant SARS-CoV-2 Nucleocapsid protein, are a proven method for detecting active SARS-CoV-2 infections. Here we report high-resolution crystal structures of three llama-derived single-domain antibodies that bind the SARS-CoV-2 Nucleocapsid protein with high affinity. Each antibody recognizes a specific folded domain of the protein, with two antibodies recognizing the N-terminal RNA binding domain and one recognizing the C-terminal dimerization domain. The two antibodies that recognize the RNA binding domain affect both RNA binding affinity and RNA-mediated phase separation of the Nucleocapsid protein. All three antibodies recognize highly-conserved surfaces on the Nucleocapsid protein, suggesting that they could be used to develop affordable diagnostic tests to detect all circulating SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Qiaozhen Ye
- Department of Cellular & Molecular Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA
| | - Shan Lu
- Department of Cellular & Molecular Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA
| | - Kevin D. Corbett
- Department of Cellular & Molecular Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA
| |
Collapse
|
13
|
Structural and Biophysical Characterization of the HCV E1E2 Heterodimer for Vaccine Development. Viruses 2021; 13:v13061027. [PMID: 34072451 PMCID: PMC8227786 DOI: 10.3390/v13061027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
An effective vaccine for the hepatitis C virus (HCV) is a major unmet medical and public health need, and it requires an antigen that elicits immune responses to multiple key conserved epitopes. Decades of research have generated a number of vaccine candidates; based on these data and research through clinical development, a vaccine antigen based on the E1E2 glycoprotein complex appears to be the best choice. One bottleneck in the development of an E1E2-based vaccine is that the antigen is challenging to produce in large quantities and at high levels of purity and antigenic/functional integrity. This review describes the production and characterization of E1E2-based vaccine antigens, both membrane-associated and a novel secreted form of E1E2, with a particular emphasis on the major challenges facing the field and how those challenges can be addressed.
Collapse
|
14
|
Chen F, Liu Z, Jiang F. Prospects of Neutralizing Nanobodies Against SARS-CoV-2. Front Immunol 2021; 12:690742. [PMID: 34122456 PMCID: PMC8194341 DOI: 10.3389/fimmu.2021.690742] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/04/2021] [Indexed: 01/14/2023] Open
Abstract
Since December 2019, the SARS-CoV-2 has erupted on a large scale worldwide and spread rapidly. Passive immunization of antibody-related molecules provides opportunities for prevention and treatment of high-risk patients and children. Nanobodies (Nbs) have many strong physical and chemical properties. They can be atomized, administered by inhalation, and can be directly applied to the infected site, with fast onset, high local drug concentration/high bioavailability, and high patient compliance (no needles). It has very attractive potential in the treatment of respiratory viruses. Rapid and low-cost development of Nbs targeting SARS-CoV-2 can quickly be achieved. Nbs against SARS-CoV-2 mutant strains also can be utilized quickly to prevent the virus from escaping. It provides important technical supports for the treatment of the SARS-CoV-2 and has the potential to become an essential medicine in the toolbox against the SARS-CoV-2.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhihong Liu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Fan Jiang
- NanoAI Biotech Co., Ltd., Huahan Technology Industrial Park, Shenzhen, China
| |
Collapse
|
15
|
Zare H, Aghamollaei H, Hosseindokht M, Heiat M, Razei A, Bakherad H. Nanobodies, the potent agents to detect and treat the Coronavirus infections: A systematic review. Mol Cell Probes 2021; 55:101692. [PMID: 33358936 PMCID: PMC7832525 DOI: 10.1016/j.mcp.2020.101692] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
The newly emerged coronavirus (SARS-CoV-2) continues to infect humans, and no effective treatment has yet been found. Antibody therapy is one way to control infection caused by COVID-19. However, the use of classical antibodies raises complex issues. Heavy chain antibodies (HCAbs) are single-domain antibodies derived from the Camelidae family. The variable part of these antibodies (Nanobodies or VHH) has interesting properties such as small size, cost-effective production, and good tissue permeability, causing VHH to be regarded as an antiviral therapeutics. However, the small size of nanobodies may lead to low antigen binding affinity and rapid renal clearance. In this systematic review, the application of nanobodies in the treatment of COVID-19 infection and other similar infections (MERS and SARS) was reviewed.
Collapse
Affiliation(s)
- Hamed Zare
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Hosseindokht
- Department of Molecular Medicine, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Razei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Science, Tehran, Iran
| | - Hamid Bakherad
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
16
|
Samrat SK, Tharappel AM, Li Z, Li H. Prospect of SARS-CoV-2 spike protein: Potential role in vaccine and therapeutic development. Virus Res 2020; 288:198141. [PMID: 32846196 PMCID: PMC7443330 DOI: 10.1016/j.virusres.2020.198141] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/08/2023]
Abstract
The recent outbreak of the betacoronavirus SARS-CoV-2 has become a significant concern to public health care worldwide. As of August 19, 2020, more than 22,140,472 people are infected, and over 781,135 people have died due to this deadly virus. In the USA alone, over 5,482,602 people are currently infected, and more than 171,823 people have died. SARS-CoV-2 has shown a higher infectivity rate and a more extended incubation period as compared to previous coronaviruses. SARS-CoV-2 binds much more strongly than SARS-CoV to the same host receptor, angiotensin-converting enzyme 2 (ACE2). Previously, several methods to develop a vaccine against SARS-CoV or MERS-CoV have been tried with limited success. Since SARS-CoV-2 uses the spike (S) protein for entry to the host cell, it is one of the most preferred targets for making vaccines or therapeutics against SARS-CoV-2. In this review, we have summarised the characteristics of the S protein, as well as the different approaches being used for the development of vaccines and/or therapeutics based on the S protein.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2
- Antibodies, Viral/biosynthesis
- Antibody-Dependent Enhancement/drug effects
- Betacoronavirus/drug effects
- Betacoronavirus/immunology
- Betacoronavirus/pathogenicity
- COVID-19
- COVID-19 Vaccines
- Clinical Trials as Topic
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Genetic Vectors/chemistry
- Genetic Vectors/immunology
- Humans
- Immunogenicity, Vaccine
- Pandemics/prevention & control
- Patient Safety
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/immunology
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- Receptors, Virus/genetics
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Vaccines, Attenuated
- Vaccines, DNA
- Vaccines, Subunit
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/biosynthesis
- Vaccines, Virus-Like Particle/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Anil M Tharappel
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Zhong Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA; Department of Biomedical Sciences, School of Public Health, University at Albany, 1 University Place, Rensselaer, NY 12144, USA.
| |
Collapse
|
17
|
Qian X, Xu C, Wu B, Tang H, Zhao P, Qi Z. SNORD126 Promotes Hepatitis C Virus Infection by Upregulating Claudin-1 via Activation of PI3K-AKT Signaling Pathway. Front Microbiol 2020; 11:565590. [PMID: 33042070 PMCID: PMC7522514 DOI: 10.3389/fmicb.2020.565590] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Hepatitis C virus (HCV) infection involves a variety of viral and host factors, some of which promote the infection process. A small nucleolar RNA, C/D box 126 (SNORD126), was previously shown to be associated with hepatocellular carcinoma (HCC). However, the role of SNORD126 in HCV infection, which is one of the primary reasons for HCC development, has not been elucidated. In the present study, using small nucleolar RNA profiling, we observed that SNORD126 was significantly downregulated during HCV infection in both Huh7 and Huh7.5.1 cells. In addition, overexpression of SNORD126 enhanced HCV entry into host cells, whereas SNORD126 knockdown showed the opposite effect, suggesting that SNORD126 promotes HCV infection, especially through viral entry. Further functional analysis revealed that SNORD126 could enhance the expression level of claudin-1 (CLDN1), a key HCV entry factor, by increasing the levels of phosphorylated AKT. Additionally, the function of SNORD126 in HCV infection was associated with ribonucleoprotein (RNP) complexes. In summary, our findings demonstrate that oncogenic SNORD126 levels are decreased during HCV infection probably due to the host defense reaction, and SNORD126 may be important to promote viral entry by increasing CLDN1 expression through activation of the PI3K-AKT pathway, the mechanism of which is partly associated with SNORD126-mediated snoRNA RNP (snoRNP) function. Our work here provides initial evidence that endogenous snoRNA takes part in HCV infection and shows potential as a diagnostic or antiviral agent.
Collapse
Affiliation(s)
- Xijing Qian
- Department of Microbiology, Second Military Medical University, Shanghai, China
| | - Chen Xu
- Spine Center, Department of Orthopedics, Changzheng Hospital Affiliated to Second Military Medical University, Shanghai, China
| | - Bingan Wu
- Department of Microbiology, Second Military Medical University, Shanghai, China
| | - Hailin Tang
- Department of Microbiology, Second Military Medical University, Shanghai, China
| | - Ping Zhao
- Department of Microbiology, Second Military Medical University, Shanghai, China
| | - Zhongtian Qi
- Department of Microbiology, Second Military Medical University, Shanghai, China
| |
Collapse
|
18
|
CD81 extracted in SMALP nanodiscs comprises two distinct protein populations within a lipid environment enriched with negatively charged headgroups. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183419. [PMID: 32735789 PMCID: PMC7456796 DOI: 10.1016/j.bbamem.2020.183419] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/05/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
Tetraspanins exert a wide range of cellular functions of broad medical importance. Despite this, their biophysical characteristics are incompletely understood. Only two high-resolution structures of full-length tetraspanins have been solved. One is that of human CD81, which is involved in the infectivity of human pathogens including influenza, HIV, the malarial Plasmodium parasite and hepatitis C virus (HCV). The CD81 crystal structure identifies a cholesterol-binding pocket, which has been suggested to be important in the regulation of tetraspanin function. Here we investigate the use of styrene-maleic anhydride co-polymers (SMA) for the solubilisation and purification of CD81 within a lipid environment. When CD81 was expressed in the yeast Pichia pastoris, it could be solubilised and purified using SMA2000. This SMALP-encapsulated CD81 retained its native folded structure, as determined by the binding of two conformation-sensitive anti-CD81 antibodies. Analysis by size exclusion chromatography revealed two distinct populations of CD81, only one of which bound the HCV glycoprotein, E2. Optimization of expression and buffer conditions increased the proportion of E2-binding competent CD81 protein. Mass spectrometry analysis indicated that the lipid environment surrounding CD81 is enriched with negatively charged lipids. These results establish a platform to study the influence of protein-lipid interactions in tetraspanin biology. CD81 expressed in Pichia pastoris can be solubilised and purified using SMA polymer. SMALP-encapsulated CD81 retains native folded structure. Expression and buffer conditions can be optimized to improve protein quality. The lipid environment surrounding CD81 is enriched with negatively charged lipids.
Collapse
|
19
|
Konwarh R. Nanobodies: Prospects of Expanding the Gamut of Neutralizing Antibodies Against the Novel Coronavirus, SARS-CoV-2. Front Immunol 2020; 11:1531. [PMID: 32655584 PMCID: PMC7324746 DOI: 10.3389/fimmu.2020.01531] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/10/2020] [Indexed: 11/25/2022] Open
Affiliation(s)
- Rocktotpal Konwarh
- Department of Biotechnology, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
- Centre of Excellence-Nanotechnology, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| |
Collapse
|
20
|
Antiviral Activity of a Llama-Derived Single-Domain Antibody against Enterovirus A71. Antimicrob Agents Chemother 2020; 64:AAC.01922-19. [PMID: 32152074 DOI: 10.1128/aac.01922-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/19/2020] [Indexed: 12/28/2022] Open
Abstract
In the past few decades, enterovirus A71 (EVA71) has caused devastating outbreaks in the Asia-Pacific region, resulting in serious sequelae in infected young children. No preventive or therapeutic interventions are currently available for curing EVA71 infection, highlighting a great unmet medical need for this disease. Here, we showed that one novel single-domain antibody (sdAb), F1, isolated from an immunized llama, could alleviate EVA71 infection both in vitro and in vivo We also confirmed that the sdAb clone F1 recognizes EVA71 through a novel conformational epitope comprising the highly conserved region of VP3 capsid protein by using competitive-binding and overlapping-peptide enzyme-linked immunosorbent assays (ELISAs). Because of the virion's icosahedral structure, we reasoned that adjacent epitopes must be clustered within molecular ranges that may be simultaneously bound by an engineered antibody with multiple valency. Therefore, two single-domain binding modules (F1) were fused to generate an sdAb-in-tandem design so that the capture of viral antigens could be further increased by valency effects. We showed that the tetravalent construct F1×F1-hFc, containing two sdAb-in-tandem on a fragment crystallizable (Fc) scaffold, exhibits more potent neutralization activity against EVA71 than does the bivalent sdAb F1-hFc by at least 5.8-fold. We also demonstrated that, using a human scavenger receptor class B member 2 (hSCARB2) transgenic mouse model, a half dose of the F1×F1-hFc provided better protection against EVA71 infection than did the F1-hFc. Thus, our study furnishes important insights into multivalent sdAb engineering against viral infection and provides a novel strategic deployment approach for preparedness of emerging infectious diseases such as EVA71.
Collapse
|
21
|
Melarkode Vattekatte A, Shinada NK, Narwani TJ, Noël F, Bertrand O, Meyniel JP, Malpertuy A, Gelly JC, Cadet F, de Brevern AG. Discrete analysis of camelid variable domains: sequences, structures, and in-silico structure prediction. PeerJ 2020; 8:e8408. [PMID: 32185102 PMCID: PMC7061911 DOI: 10.7717/peerj.8408] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/16/2019] [Indexed: 12/28/2022] Open
Abstract
Antigen binding by antibodies requires precise orientation of the complementarity- determining region (CDR) loops in the variable domain to establish the correct contact surface. Members of the family Camelidae have a modified form of immunoglobulin gamma (IgG) with only heavy chains, called Heavy Chain only Antibodies (HCAb). Antigen binding in HCAbs is mediated by only three CDR loops from the single variable domain (VHH) at the N-terminus of each heavy chain. This feature of the VHH, along with their other important features, e.g., easy expression, small size, thermo-stability and hydrophilicity, made them promising candidates for therapeutics and diagnostics. Thus, to design better VHH domains, it is important to thoroughly understand their sequence and structure characteristics and relationship. In this study, sequence characteristics of VHH domains have been analysed in depth, along with their structural features using innovative approaches, namely a structural alphabet. An elaborate summary of various studies proposing structural models of VHH domains showed diversity in the algorithms used. Finally, a case study to elucidate the differences in structural models from single and multiple templates is presented. In this case study, along with the above-mentioned aspects of VHH, an exciting view of various factors in structure prediction of VHH, like template framework selection, is also discussed.
Collapse
Affiliation(s)
- Akhila Melarkode Vattekatte
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Faculté des Sciences et Technologies, Saint Denis, La Réunion, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France
| | - Nicolas Ken Shinada
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France.,Discngine SAS, Paris, France
| | - Tarun J Narwani
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France
| | - Floriane Noël
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France.,PSL Research University, INSERM, UMR 932, Institut Curie, Paris, France.,Université Paris Sud, Université Paris-Saclay, Orsay, France
| | - Olivier Bertrand
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France
| | | | | | - Jean-Christophe Gelly
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France.,IBL, Paris, France
| | - Frédéric Cadet
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Faculté des Sciences et Technologies, Saint Denis, La Réunion, France.,Peaccel, Protein Engineering Accelerator, Paris, France
| | - Alexandre G de Brevern
- Biologie Intégrée du Globule Rouge UMR_S1134, Inserm, Univ. Paris, Univ. de la Réunion, Univ. des Antilles, Paris, France.,Laboratoire d'Excellence GR-Ex, Paris, France.,Faculté des Sciences et Technologies, Saint Denis, La Réunion, France.,Institut National de la Transfusion Sanguine (INTS), Paris, France.,IBL, Paris, France
| |
Collapse
|
22
|
Sroga P, Safronetz D, Stein DR. Nanobodies: a new approach for the diagnosis and treatment of viral infectious diseases. Future Virol 2020. [DOI: 10.2217/fvl-2019-0167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
With the rise of viral infections and antibiotic resistance, there is a constant need for the development of more sensitive and effective treatment and diagnostic tools. Since their discovery in the early 1990s, Camelidae antibodies have been investigated as potential tools due to their unique structure and favorable characteristics. Members of this family produce conventional IgG antibodies as well as heavy-chain only IgG antibodies that do not possess light chains. The variable domain (VHH), or nanobody, demonstrates unique antigen-binding capabilities, enhanced stability, and its small size allows for delivery into the body using a nebulizer, thereby eliminating the unfavorable use of injections. In addition, the cost-effective and easy in vitro production of these antibodies are an attractive quality in terms of mass production. This review covers the past and current nanobody treatment and diagnostic developments aimed at viral infectious diseases, including a brief overview of protozoal, bacterial, and veterinary viral approaches.
Collapse
Affiliation(s)
- Patrycja Sroga
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - David Safronetz
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
- Zoonotic Diseases & Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | | |
Collapse
|
23
|
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: 16] [Impact Index Per Article: 2.7] [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
|
24
|
Merat SJ, Bru C, van de Berg D, Molenkamp R, Tarr AW, Koekkoek S, Kootstra NA, Prins M, Ball JK, Bakker AQ, de Jong MD, Spits H, Beaumont T, Schinkel J. Cross-genotype AR3-specific neutralizing antibodies confer long-term protection in injecting drug users after HCV clearance. J Hepatol 2019; 71:14-24. [PMID: 30797052 DOI: 10.1016/j.jhep.2019.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 01/29/2019] [Accepted: 02/12/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS In order to design an effective vaccine against hepatitis C virus (HCV) infection, it is necessary to understand immune protection. A number of broadly reactive neutralizing antibodies have been isolated from B cells of HCV-infected patients. However, it remains unclear whether B cells producing such antibodies contribute to HCV clearance and long-term immune protection against HCV. METHODS We analysed the B cell repertoire of 13 injecting drug users from the Amsterdam Cohort Study, who were followed up for a median of 17.5 years after primary infection. Individuals were classified into 2 groups based on the outcome of HCV infection: 5 who became chronically infected either after primary infection or after reinfection, and 8 who were HCV RNA negative following spontaneous clearance of ≥1 HCV infection(s). From each individual, 10,000 CD27+IgG+B cells, collected 0.75 year after HCV infection, were cultured to characterize the antibody repertoire. RESULTS Using a multiplex flow cytometry-based assay to study the antibody binding to E1E2 from genotype 1 to 6, we found that a high frequency of cross-genotype antibodies was associated with spontaneous clearance of 1 or multiple infections (p = 0.03). Epitope specificity of these cross-genotype antibodies was determined by alanine mutant scanning in 4 individuals who were HCV RNA negative following spontaneous clearance of 1 or multiple infections. Interestingly, the cross-genotype antibodies were mainly antigenic region 3 (AR3)-specific and showed cross-neutralizing activity against HCV. In addition to AR3 antibodies, 3 individuals developed antibodies recognizing antigenic region 4, of which 1 monoclonal antibody showed cross-neutralizing capacity. CONCLUSIONS Together, these data suggest that a strong B cell response producing cross-genotype and neutralizing antibodies, especially targeting AR3, contributes to HCV clearance and long-term immune protection against HCV. LAY SUMMARY Although effective treatments against hepatitis C virus (HCV) are available, 500,000 people die from liver disease caused by HCV each year and approximately 1.75 million people are newly infected. This could be prevented by a vaccine. To design a vaccine against HCV, more insight into the role of antibodies in the protection against HCV infection is needed. In a cohort of injecting drug users, we found that antibodies interfering with virus cell entry, and recognizing multiple HCV genotypes, conferred long-term protection against chronic HCV infection.
Collapse
Affiliation(s)
| | - Camille Bru
- AIMM Therapeutics, Amsterdam, the Netherlands
| | | | - Richard Molenkamp
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Alexander W Tarr
- School of Life Sciences, The University of Nottingham, Nottingham, UK; NIHR Nottingham BRC, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Sylvie Koekkoek
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Maria Prins
- Public Health Service of Amsterdam, Amsterdam, the Netherlands; Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jonathan K Ball
- School of Life Sciences, The University of Nottingham, Nottingham, UK; NIHR Nottingham BRC, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | | | - Menno D de Jong
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | - Janke Schinkel
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Keck ZY, Pierce BG, Lau P, Lu J, Wang Y, Underwood A, Bull RA, Prentoe J, Velázquez-Moctezuma R, Walker MR, Luciani F, Guest JD, Fauvelle C, Baumert TF, Bukh J, Lloyd AR, Foung SKH. Broadly neutralizing antibodies from an individual that naturally cleared multiple hepatitis C virus infections uncover molecular determinants for E2 targeting and vaccine design. PLoS Pathog 2019; 15:e1007772. [PMID: 31100098 PMCID: PMC6542541 DOI: 10.1371/journal.ppat.1007772] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/30/2019] [Accepted: 04/20/2019] [Indexed: 12/17/2022] Open
Abstract
Cumulative evidence supports a role for neutralizing antibodies contributing to spontaneous viral clearance during acute hepatitis C virus (HCV) infection. Information on the timing and specificity of the B cell response associated with clearance is crucial to inform vaccine design. From an individual who cleared three sequential HCV infections with genotypes 1b, 1a and 3a strains, respectively, we employed peripheral B cells to isolate and characterize neutralizing human monoclonal antibodies (HMAbs) to HCV after the genotype 1 infections. The majority of isolated antibodies, designated as HMAbs 212, target conformational epitopes on the envelope glycoprotein E2 and bound broadly to genotype 1–6 E1E2 proteins. Further, some of these antibodies showed neutralization potential against cultured genotype 1–6 viruses. Competition studies with defined broadly neutralizing HCV HMAbs to epitopes in distinct clusters, designated antigenic domains B, C, D and E, revealed that the selected HMAbs compete with B, C and D HMAbs, previously isolated from subjects with chronic HCV infections. Epitope mapping studies revealed domain B and C specificity of these HMAbs 212. Sequential serum samples from the studied subject inhibited the binding of HMAbs 212 to autologous E2 and blocked a representative domain D HMAb. The specificity of this antibody response appears similar to that observed during chronic infection, suggesting that the timing and affinity maturation of the antibody response are the critical determinants in successful and repeated viral clearance. While additional studies should be performed for individuals with clearance or persistence of HCV, our results define epitope determinants for antibody E2 targeting with important implications for the development of a B cell vaccine. Studies of hepatitis C virus (HCV) infected individuals spontaneously clearing acute infections provide an opportunity to characterize the specificities of associated protective antibody responses. In an individual who resolved three separate HCV infections with different HCV genotypes, the antibodies induced during these acute infection episodes were similar to those induced during chronic infection. Surprisingly, the earliest detected antibodies were directed against conformational HCV epitopes on the envelope glycoprotein E2 (including polyprotein residues 434–446) known to be targeted by broadly neutralizing antibodies. Taken together, the key B-cell determinants in spontaneous clearance are the timing and affinity maturation of broadly neutralizing antibody responses.
Collapse
Affiliation(s)
- Zhen-Yong Keck
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Brian G. Pierce
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, United States of America
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Patrick Lau
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Janine Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yong Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Alexander Underwood
- Viral Immunology Systems Program, The Kirby Institute and School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Rowena A. Bull
- Viral Immunology Systems Program, The Kirby Institute and School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jannick Prentoe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rodrigo Velázquez-Moctezuma
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melanie R. Walker
- Viral Immunology Systems Program, The Kirby Institute and School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Fabio Luciani
- Viral Immunology Systems Program, The Kirby Institute and School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Johnathan D. Guest
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, United States of America
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Catherine Fauvelle
- Inserm U1110, Institut de Recherche sur les Maladies et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm U1110, Institut de Recherche sur les Maladies et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Pole Hépato-digestif, Institut Hospitalo-Universitaire, Hopitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrew R. Lloyd
- Viral Immunology Systems Program, The Kirby Institute and School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Steven K. H. Foung
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Single-Domain Antibodies Represent Novel Alternatives to Monoclonal Antibodies as Targeting Agents against the Human Papillomavirus 16 E6 Protein. Int J Mol Sci 2019; 20:ijms20092088. [PMID: 31035322 PMCID: PMC6539864 DOI: 10.3390/ijms20092088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 01/18/2023] Open
Abstract
Approximately one fifth of all malignancies worldwide are etiologically associated with a persistent viral or bacterial infection. Thus, there is a particular interest in therapeutic molecules which use components of a natural immune response to specifically inhibit oncogenic microbial proteins, as it is anticipated they will elicit fewer off-target effects than conventional treatments. This concept has been explored in the context of human papillomavirus 16 (HPV16)-related cancers, through the development of monoclonal antibodies and fragments thereof against the viral E6 oncoprotein. Challenges related to the biology of E6 as well as the functional properties of the antibodies themselves appear to have precluded their clinical translation. Here, we addressed these issues by exploring the utility of the variable domains of camelid heavy-chain-only antibodies (denoted as VHHs). Through construction and panning of two llama, immune VHH phage display libraries, a pool of potential VHHs was isolated. The interactions of these with recombinant E6 were further characterized using an enzyme-linked immunosorbent assay (ELISA), Western blotting under denaturing and native conditions, and surface plasmon resonance. Three VHHs were identified that bound recombinant E6 with nanomolar affinities. Our results lead the way for subsequent studies into the ability of these novel molecules to inhibit HPV16-infected cells in vitro and in vivo.
Collapse
|
27
|
Single-Domain Antibodies and Their Formatting to Combat Viral Infections. Antibodies (Basel) 2018; 8:antib8010001. [PMID: 31544807 PMCID: PMC6640686 DOI: 10.3390/antib8010001] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Since their discovery in the 1990s, single-domain antibodies (VHHs), also known as Nanobodies®, have changed the landscape of affinity reagents. The outstanding solubility, stability, and specificity of VHHs, as well as their small size, ease of production and formatting flexibility favor VHHs over conventional antibody formats for many applications. The exceptional ease by which it is possible to fuse VHHs with different molecular modules has been particularly explored in the context of viral infections. In this review, we focus on VHH formats that have been developed to combat viruses including influenza viruses, human immunodeficiency virus-1 (HIV-1), and human respiratory syncytial virus (RSV). Such formats may significantly increase the affinity, half-life, breadth of protection of an antiviral VHH and reduce the risk of viral escape. In addition, VHHs can be equipped with effector functions, for example to guide components of the immune system with high precision to sites of viral infection.
Collapse
|
28
|
Lafaye P, Li T. Use of camel single-domain antibodies for the diagnosis and treatment of zoonotic diseases. Comp Immunol Microbiol Infect Dis 2018; 60:17-22. [PMID: 30396425 PMCID: PMC7112682 DOI: 10.1016/j.cimid.2018.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/01/2018] [Accepted: 09/17/2018] [Indexed: 12/22/2022]
Abstract
VHHs provide many advantages over complete IgG in diagnostics and therapy. Toxins and viruses are more efficiently neutralized by multivalent VHHs. Camelids could be a source of broadly neutralizing antibodies (bNAbs) to treat zoonotic diseases.
Camelids produce both conventional heterotetrameric antibodies and homodimeric heavy-chain only antibodies. The antigen-binding region of such homodimeric heavy-chain only antibodies consists of one single domain, called VHH. VHHs provide many advantages over conventional full-sized antibodies and currently used antibody-based fragments (Fab, scFv), including high specificity, stability and solubility, and small size, allowing them to recognize unusual antigenic sites and deeply penetrate tissues. Since their discovery, VHHs have been used extensively in diagnostics and therapy. In recent decades, the number of outbreaks of diseases transmissible from animals to humans has been on the rise. In this review, we evaluate the status of VHHs as diagnostic and therapeutic biomolecular agents for the detection and treatment of zoonotic diseases, such as bacterial, parasitic, and viral zoonosis. VHHs show great adaptability to inhibit or neutralize pathogenic agents for the creation of multifunctional VHH-based diagnostic and therapeutic molecules against zoonotic diseases.
Collapse
Affiliation(s)
- Pierre Lafaye
- Institut Pasteur, Plate forme d'Ingénierie des Anticorps, C2RT, Paris, France.
| | - Tengfei Li
- Université Paris Diderot, Paris 7, France
| |
Collapse
|
29
|
Tarr AW, Backx M, Hamed MR, Urbanowicz RA, McClure CP, Brown RJP, Ball JK. Immunization with a synthetic consensus hepatitis C virus E2 glycoprotein ectodomain elicits virus-neutralizing antibodies. Antiviral Res 2018; 160:25-37. [PMID: 30217650 DOI: 10.1016/j.antiviral.2018.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/06/2018] [Accepted: 09/10/2018] [Indexed: 01/06/2023]
Abstract
Global eradication of hepatitis C virus (HCV) infection will require an efficacious vaccine capable of eliciting protective immunity against genetically diverse HCV strains. Natural spontaneous resolution of HCV infection is associated with production of broadly-neutralizing antibodies targeting the HCV glycoproteins E1 and E2. As such, production of cross-neutralizing antibodies is an important endpoint for experimental vaccine trials. Varying success generating cross-neutralizing antibodies has been achieved with immunogens derived from naturally-occurring HCV strains. In this study the challenge of minimising the genetic diversity between the vaccine strain and circulating HCV isolates was addressed. Two novel synthetic E2 glycoprotein immunogens (NotC1 and NotC2) were derived from consensus nucleotide sequences deduced from samples of circulating genotype 1 HCV strains. These two synthetic sequences differed in their relative positions in the overall genotype 1a/1b phylogeny. Expression of these constructs in Drosophila melanogaster S2 cells resulted in high yields of correctly-folded, monomeric E2 protein, which were recognised by broadly neutralizing monoclonal antibodies. Immunization of guinea pigs with either of these consensus immunogens, or a comparable protein representing a circulating genotype 1a strain resulted in high titres of cross-reactive anti-E2 antibodies. All immunogens generated antibodies capable of neutralizing the H77 strain, but NotC1 elicited antibodies that more potently neutralized virus entry. These vaccine-induced antibodies neutralized some viruses representing genotype 1, but not strains representing genotype 2 or genotype 3. Thus, while this approach to vaccine design resulted in correctly folded, immunogenic protein, cross-neutralizing epitopes were not preferentially targeted by the host immune response generated by this immunogen. Greater immunofocussing of vaccines to common epitopes is necessary to successfully elicit broadly neutralizing antibodies.
Collapse
Affiliation(s)
- Alexander W Tarr
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Matthijs Backx
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Mohamed R Hamed
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK; Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Richard A Urbanowicz
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - C Patrick McClure
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Richard J P Brown
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Jonathan K Ball
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
30
|
Guest JD, Pierce BG. Computational Modeling of Hepatitis C Virus Envelope Glycoprotein Structure and Recognition. Front Immunol 2018; 9:1117. [PMID: 29892287 PMCID: PMC5985375 DOI: 10.3389/fimmu.2018.01117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) is a major global health concern, and though therapeutic options have improved, no vaccine is available despite decades of research. As HCV can rapidly mutate to evade the immune response, an effective HCV vaccine must rely on identification and characterization of sites critical for broad immune protection and viral neutralization. This knowledge depends on structural and mechanistic insights of the E1 and E2 envelope glycoproteins, which assemble as a heterodimer on the surface of the virion, engage coreceptors during host cell entry, and are the primary targets of antibodies. Due to the challenges in determining experimental structures, structural information on E1 and E2 and their interaction is relatively limited, providing opportunities to model the structures, interactions, and dynamics of these proteins. This review highlights efforts to model the E2 glycoprotein structure, the assembly of the functional E1E2 heterodimer, the structure and binding of human coreceptors, and recognition by key neutralizing antibodies. We also discuss a comparison of recently described models of full E1E2 heterodimer structures, a simulation of the dynamics of key epitope sites, and modeling glycosylation. These modeling efforts provide useful mechanistic hypotheses for further experimental studies of HCV envelope assembly, recognition, and viral fitness, and underscore the benefit of combining experimental and computational modeling approaches to reveal new insights. Additionally, computational design approaches have produced promising candidates for epitope-based vaccine immunogens that specifically target key epitopes, providing a possible avenue to optimize HCV vaccines versus using native glycoproteins. Advancing knowledge of HCV envelope structure and immune recognition is highly applicable toward the development of an effective vaccine for HCV and can provide lessons and insights relevant to modeling and characterizing other viruses.
Collapse
Affiliation(s)
- Johnathan D Guest
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| | - Brian G Pierce
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, United States.,Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| |
Collapse
|
31
|
Dustin LB. Innate and Adaptive Immune Responses in Chronic HCV Infection. Curr Drug Targets 2018; 18:826-843. [PMID: 26302811 DOI: 10.2174/1389450116666150825110532] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/25/2015] [Accepted: 07/27/2015] [Indexed: 12/14/2022]
Abstract
Hepatitis C virus (HCV) remains a public health problem of global importance, even in the era of potent directly-acting antiviral drugs. In this chapter, I discuss immune responses to acute and chronic HCV infection. The outcome of HCV infection is influenced by viral strategies that limit or delay the initiation of innate antiviral responses. This delay may enable HCV to establish widespread infection long before the host mounts effective T and B cell responses. HCV's genetic agility, resulting from its high rate of replication and its error prone replication mechanism, enables it to evade immune recognition. Adaptive immune responses fail to keep up with changing viral epitopes. Neutralizing antibody epitopes may be hidden by decoy structures, glycans, and lipoproteins. T cell responses fail due to changing epitope sequences and due to exhaustion, a phenomenon that may have evolved to limit immune-mediated pathology. Despite these difficulties, innate and adaptive immune mechanisms do impact HCV replication. Immune-mediated clearance of infection is possible, occurring in 20-50% of people who contract the disease. New developments raise hopes for effective immunological interventions to prevent or treat HCV infection.
Collapse
Affiliation(s)
- Lynn B Dustin
- University of Oxford, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, United Kingdom
| |
Collapse
|
32
|
Du T, Liang J, Dong N, Lu J, Fu Y, Fang L, Xiao S, Han H. Glutathione-Capped Ag 2S Nanoclusters Inhibit Coronavirus Proliferation through Blockage of Viral RNA Synthesis and Budding. ACS APPLIED MATERIALS & INTERFACES 2018; 10:4369-4378. [PMID: 29337529 DOI: 10.1021/acsami.7b13811] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Development of novel antiviral reagents is of great importance for the control of virus spread. Here, Ag2S nanoclusters (NCs) were proved for the first time to possess highly efficient antiviral activity by using porcine epidemic diarrhea virus (PEDV) as a model of coronavirus. Analyses of virus titers showed that Ag2S NCs significantly suppressed the infection of PEDV by about 3 orders of magnitude at the noncytotoxic concentration at 12 h postinfection, which was further confirmed by the expression of viral proteins. Mechanism investigations indicated that Ag2S NCs treatment inhibits the synthesis of viral negative-strand RNA and viral budding. Ag2S NCs treatment was also found to positively regulate the generation of IFN-stimulating genes (ISGs) and the expression of proinflammation cytokines, which might prevent PEDV infection. This study suggest the novel underlying of Ag2S NCs as a promising therapeutic drug for coronavirus.
Collapse
Affiliation(s)
- Ting Du
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Jiangong Liang
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Nan Dong
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Jian Lu
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Yiying Fu
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Liurong Fang
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| | - Heyou Han
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, College of Science, and ‡State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan 430070, P. R. China
| |
Collapse
|
33
|
Wu Y, Jiang S, Ying T. Single-Domain Antibodies As Therapeutics against Human Viral Diseases. Front Immunol 2017; 8:1802. [PMID: 29326699 PMCID: PMC5733491 DOI: 10.3389/fimmu.2017.01802] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022] Open
Abstract
In full-size formats, monoclonal antibodies have been highly successful as therapeutics against cancer and immune diseases. However, their large size leads to inaccessibility of some epitopes and relatively high production costs. As an alternative, single-domain antibodies (sdAbs) offer special advantages compared to full-size antibodies, including smaller size, larger number of accessible epitopes, relatively low production costs and improved robustness. Currently, sdAbs are being developed against a number of viruses, including human immunodeficiency virus-1 (HIV-1), influenza viruses, hepatitis C virus (HCV), respiratory syncytial virus (RSV), and enteric viruses. Although sdAbs are very potent inhibitors of viral infections, no sdAbs have been approved for clinical use against virial infection or any other diseases. In this review, we discuss the current state of research on sdAbs against viruses and their potential as therapeutics against human viral diseases.
Collapse
Affiliation(s)
- Yanling Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Discovery of a novel conformational equilibrium in urokinase-type plasminogen activator. Sci Rep 2017; 7:3385. [PMID: 28611361 PMCID: PMC5469797 DOI: 10.1038/s41598-017-03457-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/27/2017] [Indexed: 01/15/2023] Open
Abstract
Although trypsin-like serine proteases have flexible surface-exposed loops and are known to adopt higher and lower activity conformations, structural determinants for the different conformations have remained largely obscure. The trypsin-like serine protease, urokinase-type plasminogen activator (uPA), is central in tissue remodeling processes and also strongly implicated in tumor metastasis. We solved five X-ray crystal structures of murine uPA (muPA) in the absence and presence of allosteric molecules and/or substrate-like molecules. The structure of unbound muPA revealed an unsuspected non-chymotrypsin-like protease conformation in which two β-strands in the core of the protease domain undergoes a major antiparallel-to-parallel conformational transition. We next isolated two anti-muPA nanobodies; an active-site binding nanobody and an allosteric nanobody. Crystal structures of the muPA:nanobody complexes and hydrogen-deuterium exchange mass spectrometry revealed molecular insights about molecular factors controlling the antiparallel-to-parallel equilibrium in muPA. Together with muPA activity assays, the data provide valuable insights into regulatory mechanisms and conformational flexibility of uPA and trypsin-like serine proteases in general.
Collapse
|
35
|
Vasiliauskaite I, Owsianka A, England P, Khan AG, Cole S, Bankwitz D, Foung SKH, Pietschmann T, Marcotrigiano J, Rey FA, Patel AH, Krey T. Conformational Flexibility in the Immunoglobulin-Like Domain of the Hepatitis C Virus Glycoprotein E2. mBio 2017; 8:e00382-17. [PMID: 28512091 PMCID: PMC5433095 DOI: 10.1128/mbio.00382-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/24/2017] [Indexed: 12/16/2022] Open
Abstract
The hepatitis C virus (HCV) glycoprotein E2 is the major target of neutralizing antibodies and is therefore highly relevant for vaccine design. Its structure features a central immunoglobulin (Ig)-like β-sandwich that contributes to the binding site for the cellular receptor CD81. We show that a synthetic peptide corresponding to a β-strand of this Ig-like domain forms an α-helix in complex with the anti-E2 antibody DAO5, demonstrating an inside-out flip of hydrophobic residues and a secondary structure change in the composite CD81 binding site. A detailed interaction analysis of DAO5 and cross-competing neutralizing antibodies with soluble E2 revealed that the Ig-like domain is trapped by different antibodies in at least two distinct conformations. DAO5 specifically captures retrovirus particles bearing HCV glycoproteins (HCVpp) and infectious cell culture-derived HCV particles (HCVcc). Infection of cells by DAO5-captured HCVpp can be blocked by a cross-competing neutralizing antibody, indicating that a single virus particle simultaneously displays E2 molecules in more than one conformation on its surface. Such conformational plasticity of the HCV E2 receptor binding site has important implications for immunogen design.IMPORTANCE Recent advances in the treatment of hepatitis C virus (HCV) infection with direct-acting antiviral drugs have enabled the control of this major human pathogen. However, due to their high costs and limited accessibility in combination with the lack of awareness of the mostly asymptomatic infection, there is an unchanged urgent need for an effective vaccine. The viral glycoprotein E2 contains regions that are crucial for virus entry into the host cell, and antibodies that bind to these regions can neutralize infection. One of the major targets of neutralizing antibodies is the central immunoglobulin (Ig)-like domain within E2. We show here that this Ig-like domain is conformationally flexible at the surface of infectious HCV particles and pseudoparticles. Our study provides novel insights into the interactions of HCV E2 with the humoral immune system that should aid future vaccine development.
Collapse
Affiliation(s)
- Ieva Vasiliauskaite
- Unité de Virologie Structurale, Department Virologie, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Ania Owsianka
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Patrick England
- Plate-Forme de Biophysique Moléculaire, Institut Pasteur, Paris, France
- CNRS UMR 3528, Paris, France
| | - Abdul Ghafoor Khan
- Center for Advanced Biotechnology and Medicine, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, USA
| | - Sarah Cole
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Dorothea Bankwitz
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
| | - Steven K H Foung
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Thomas Pietschmann
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Germany
| | - Joseph Marcotrigiano
- Center for Advanced Biotechnology and Medicine, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, USA
| | - Felix A Rey
- Unité de Virologie Structurale, Department Virologie, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Arvind H Patel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Thomas Krey
- Unité de Virologie Structurale, Department Virologie, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Hannover-Braunschweig Site, Germany
| |
Collapse
|
36
|
Rodrigo C, Walker MR, Leung P, Eltahla AA, Grebely J, Dore GJ, Applegate T, Page K, Dwivedi S, Bruneau J, Morris MD, Cox AL, Osburn W, Kim AY, Schinkel J, Shoukry NH, Lauer GM, Maher L, Hellard M, Prins M, Luciani F, Lloyd AR, Bull RA. Limited naturally occurring escape in broadly neutralizing antibody epitopes in hepatitis C glycoprotein E2 and constrained sequence usage in acute infection. INFECTION GENETICS AND EVOLUTION 2017; 49:88-96. [PMID: 28065804 DOI: 10.1016/j.meegid.2017.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/07/2016] [Accepted: 01/03/2017] [Indexed: 12/12/2022]
Abstract
Broadly neutralizing antibodies have been associated with spontaneous clearance of the hepatitis C infection as well as viral persistence by immune escape. Further study of neutralizing antibody epitopes is needed to unravel pathways of resistance to virus neutralization, and to identify conserved regions for vaccine design. All reported broadly neutralizing antibody (BNAb) epitopes in the HCV Envelope (E2) glycoprotein were identified. The critical contact residues of these epitopes were mapped onto the linear E2 sequence. All publicly available E2 sequences were then downloaded and the contact residues within the BNAb epitopes were assessed for the level of conservation, as well as the frequency of occurrence of experimentally-proven resistance mutations. Epitopes were also compared between two sequence datasets obtained from samples collected at well-defined time points from acute (<180days) and chronic (>180days) infections, to identify any significant differences in residue usage. The contact residues for all BNAbs were contained within 3 linear regions of the E2 protein sequence. An analysis of 1749 full length E2 sequences from public databases showed that only 10 out of 29 experimentally-proven resistance mutations were present at a frequency >5%. Comparison of subtype 1a viral sequences obtained from samples collected during acute or chronic infection revealed significant differences at positions 610 and 655 with changes in residue (p<0.05), and at position 422 (p<0.001) with a significant difference in variability (entropy). The majority of experimentally-described escape variants do not occur frequently in nature. The observed differences between acute and chronically isolated sequences suggest constraints on residue usage early in infection.
Collapse
Affiliation(s)
- Chaturaka Rodrigo
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.
| | - Melanie R Walker
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Preston Leung
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Auda A Eltahla
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Jason Grebely
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Gregory J Dore
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Tanya Applegate
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Kimberly Page
- Department of Epidemiology and Biostatistics, University of New Mexico, Albuquerque, NM, USA
| | - Sunita Dwivedi
- Department of Epidemiology and Biostatistics, University of New Mexico, Albuquerque, NM, USA
| | - Julie Bruneau
- CRCHUM, Université de Montréal, Montreal, QC, Canada
| | - Meghan D Morris
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - William Osburn
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | - Janke Schinkel
- Department of Internal Medicine, Division of Infectious Diseases, Tropical Medicine and AIDS, Center for Infection and Immunity Amsterdam, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands
| | | | | | - Lisa Maher
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Margaret Hellard
- Burnet Institute, Melbourne, VIC, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Maria Prins
- Department of Internal Medicine, Division of Infectious Diseases, Tropical Medicine and AIDS, Center for Infection and Immunity Amsterdam, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands; GGD Public Health Service of Amsterdam, Amsterdam, The Netherlands
| | - Fabio Luciani
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Andrew R Lloyd
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Rowena A Bull
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
37
|
Tawar RG, Heydmann L, Bach C, Schüttrumpf J, Chavan S, King BJ, McClure CP, Ball JK, Pessaux P, Habersetzer F, Bartenschlager R, Zeisel MB, Baumert TF. Broad neutralization of hepatitis C virus-resistant variants by Civacir hepatitis C immunoglobulin. Hepatology 2016; 64:1495-1506. [PMID: 27531416 PMCID: PMC7615276 DOI: 10.1002/hep.28767] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/19/2016] [Indexed: 12/15/2022]
Abstract
Hepatitis C virus (HCV)-induced end-stage liver disease is the major indication for liver transplantation (LT). However, reinfection of the liver graft is still common, especially in patients with detectable viral load at the time of LT. Limited data are available on direct-acting antivirals in the transplant setting for prevention of graft infection. The human hepatitis C immunoglobulin (HCIG) Civacir is an investigational drug that is currently being developed in an ongoing phase 3 clinical trial assessing its safety and efficacy at preventing HCV recurrence after liver transplantation (LT) in the United States. Using well-characterized patient-derived HCV variants selected during LT, we studied the molecular mechanism of action of Civacir. Inhibition of HCV infection was studied using infectious HCV models including HCV pseudoparticles (HCVpp) and cell culture-derived HCV (HCVcc) containing patient-derived viral envelope glycoproteins from 22 HCV variants isolated from patients before and after LT. The human hepatitis C immune globulin Civacir is an investigational drug that is currently being developed in an ongoing phase 3 clinical trial assessing safety and efficacy to prevent HCV recurrence after LT in the United States. Using well-characterized patient-derived HCV variants selected during LT, we studied the molecular mechanism of action of Civacir. Inhibition of HCV infection was studied using infectious HCV models including HCV pseudoparticles and cell culture-derived HCV containing patient-derived viral envelope glycoproteins from 22 HCV variants isolated from patients before and after liver transplantation. Additionally, we studied neutralization of different HCV genotypes and of direct-acting antiviral-resistant viruses. Our results indicate that Civacir potently, broadly, and dose-dependently neutralizes all tested patient variants in HCV pseudoparticles and cell culture-derived HCV assays including variants displaying resistance to host neutralizing antibodies and antiviral monoclonal antibodies. The half-maximal inhibitory concentrations were independent of the phenotype of the viral variant, indicating that virus neutralization by Civacir is not affected by viral selection. Furthermore, Civacir is equally active against tested direct-acting antiviral-resistant HCV isolates in cell culture. CONCLUSION Collectively, these results demonstrate broad neutralizing activity of Civacir against resistant viruses, likely due to synergy between anti-HCV antibodies derived from different plasma donors, and support its further clinical development for prevention of liver graft infection. (Hepatology 2016;64:1495-1506).
Collapse
Affiliation(s)
- Rajiv G Tawar
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Laura Heydmann
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Charlotte Bach
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | | | | | - Barnabas J King
- School of Life Sciences and the NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - C Patrick McClure
- School of Life Sciences and the NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Jonathan K Ball
- School of Life Sciences and the NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Patrick Pessaux
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- University of Strasbourg, Strasbourg, France
- Institut Hospitalo-universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - François Habersetzer
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- University of Strasbourg, Strasbourg, France
- Institut Hospitalo-universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research, Heidelberg University, Heidelberg, Germany
| | - Mirjam B Zeisel
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Thomas F Baumert
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.
- University of Strasbourg, Strasbourg, France.
- Institut Hospitalo-universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
38
|
Viral evasion and challenges of hepatitis C virus vaccine development. Curr Opin Virol 2016; 20:55-63. [PMID: 27657659 DOI: 10.1016/j.coviro.2016.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/24/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
Hepatitis C virus (HCV) is a major global disease burden, often leading to chronic liver diseases, cirrhosis, cancer, and death in those infected. Despite the recent approval of antiviral therapeutics, a preventative vaccine is recognized as the most effective means to control HCV globally, particularly in at-risk and developing country populations. Here we describe the efforts and challenges related to the development of an HCV vaccine, which after decades of research have not been successful. Viral sequence variability poses a major challenge, yet recent research has provided unprecedented views of the atomic structure of HCV epitopes and immune recognition by antibodies and T cell receptors. This, coupled with insights from deep sequencing, robust neutralization assays, and other technological advances, is spurring research toward rationally HCV designed vaccines that preferentially elicit responses toward conserved epitopes of interest that are associated with viral neutralization and clearance.
Collapse
|
39
|
Steeland S, Vandenbroucke RE, Libert C. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discov Today 2016; 21:1076-113. [DOI: 10.1016/j.drudis.2016.04.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/26/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022]
|
40
|
McClure CP, Urbanowicz RA, King BJ, Cano-Crespo S, Tarr AW, Ball JK. Flexible and rapid construction of viral chimeras applied to hepatitis C virus. J Gen Virol 2016; 97:2187-2193. [PMID: 27329374 PMCID: PMC5042125 DOI: 10.1099/jgv.0.000530] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
A novel and broadly applicable strategy combining site-directed mutagenesis and DNA assembly for constructing seamless viral chimeras is described using hepatitis C virus (HCV) as an exemplar. Full-length HCV genomic cloning cassettes, which contained flexibly situated restriction endonuclease sites, were prepared via a single, site-directed mutagenesis reaction and digested to receive PCR-amplified virus envelope genes by In-Fusion cloning. Using this method, we were able to construct gene-shuttle cassettes for generation of cell culture-infectious JFH-1-based chimeras containing genotype 1–3 E1E2 genes. Importantly, using this method we also show that E1E2 clones that were not able to support cell entry in the HCV pseudoparticle assay did confer entry when shuttled into the chimeric cell culture chimera system. This method can be easily applied to other genes of study and other viruses and, as such, will greatly simplify reverse genetics studies of variable viruses.
Collapse
Affiliation(s)
- C Patrick McClure
- School of Life Sciences and NIHR Nottingham Digestive Diseases Biomedical Research Unit, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Richard A Urbanowicz
- School of Life Sciences and NIHR Nottingham Digestive Diseases Biomedical Research Unit, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Barnabas J King
- School of Life Sciences and NIHR Nottingham Digestive Diseases Biomedical Research Unit, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Sara Cano-Crespo
- School of Life Sciences and NIHR Nottingham Digestive Diseases Biomedical Research Unit, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Alexander W Tarr
- School of Life Sciences and NIHR Nottingham Digestive Diseases Biomedical Research Unit, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Jonathan K Ball
- School of Life Sciences and NIHR Nottingham Digestive Diseases Biomedical Research Unit, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, UK
| |
Collapse
|
41
|
Kromann-Hansen T, Oldenburg E, Yung KWY, Ghassabeh GH, Muyldermans S, Declerck PJ, Huang M, Andreasen PA, Ngo JCK. A Camelid-derived Antibody Fragment Targeting the Active Site of a Serine Protease Balances between Inhibitor and Substrate Behavior. J Biol Chem 2016; 291:15156-68. [PMID: 27226628 DOI: 10.1074/jbc.m116.732503] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 01/15/2023] Open
Abstract
A peptide segment that binds the active site of a serine protease in a substrate-like manner may behave like an inhibitor or a substrate. However, there is sparse information on which factors determine the behavior a particular peptide segment will exhibit. Here, we describe the first x-ray crystal structure of a nanobody in complex with a serine protease. The nanobody displays a new type of interaction between an antibody and a serine protease as it inserts its complementary determining region-H3 loop into the active site of the protease in a substrate-like manner. The unique binding mechanism causes the nanobody to behave as a strong inhibitor as well as a poor substrate. Intriguingly, its substrate behavior is incomplete, as 30-40% of the nanobody remained intact and inhibitory after prolonged incubation with the protease. Biochemical analysis reveals that an intra-loop interaction network within the complementary determining region-H3 of the nanobody balances its inhibitor versus substrate behavior. Collectively, our results unveil molecular factors, which may be a general mechanism to determine the substrate versus inhibitor behavior of other protease inhibitors.
Collapse
Affiliation(s)
- Tobias Kromann-Hansen
- From the Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark,
| | - Emil Oldenburg
- From the Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Kristen Wing Yu Yung
- the School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Gholamreza H Ghassabeh
- the Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, 1050 Brussels, Belgium, Nanobody Service Facility, Flanders Institute for Biotechnology, 1050 Brussels, Belgium
| | - Serge Muyldermans
- the Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, 1050 Brussels, Belgium
| | - Paul J Declerck
- the Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit, 3000 Leuven, Belgium, and
| | - Mingdong Huang
- the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Science, Fuzhou, Fujian 350002, China
| | - Peter A Andreasen
- From the Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Jacky Chi Ki Ngo
- the School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| |
Collapse
|
42
|
Desombere I, Fafi-Kremer S, Van Houtte F, Pessaux P, Farhoudi A, Heydmann L, Verhoye L, Cole S, McKeating JA, Leroux-Roels G, Baumert TF, Patel AH, Meuleman P. Monoclonal anti-envelope antibody AP33 protects humanized mice against a patient-derived hepatitis C virus challenge. Hepatology 2016; 63:1120-34. [PMID: 26710081 PMCID: PMC7613414 DOI: 10.1002/hep.28428] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/04/2015] [Accepted: 12/23/2015] [Indexed: 01/01/2023]
Abstract
UNLABELLED End-stage liver disease (ESLD) caused by hepatitis C virus (HCV) infection is a major indication for liver transplantation. However, immediately after transplantation, the liver graft of viremic patients universally becomes infected by circulating virus, resulting in accelerated liver disease progression. Currently available direct-acting antiviral therapies have reduced efficacy in patients with ESLD and prophylactic strategies to prevent HCV recurrence are still highly needed. In this study, we compared the ability of two broadly reactive monoclonal antibodies (mAbs), designated 3/11 and AP33, recognizing a distinct, but overlapping, epitope in the viral E2 glycoprotein to protect humanized mice from a patient-derived HCV challenge. Their neutralizing activity was assessed using the HCV pseudoparticles and cell-culture-derived HCV systems expressing multiple patient-derived envelopes and a human-liver chimeric mouse model. HCV RNA was readily detected in all control mice challenged with a patient-derived HCV genotype 1b isolate, whereas 3 of 4 AP33-treated mice were completely protected. In contrast, only one of four 3/11-treated mice remained HCV-RNA negative throughout the observation period, whereas the other 3 had a viral load that was indistinguishable from that in the control group. The increased in vivo efficacy of AP33 was in line with its higher affinity and neutralizing capacity observed in vitro. CONCLUSIONS Although mAbs AP33 and 3/11 target the same region in E2, only mAb AP33 can efficiently protect from challenge with a heterologous HCV population in vivo. Given that mAb AP33 efficiently neutralizes viral variants that escaped the humoral immune response and reinfected the liver graft of transplant patients, it may be a valuable candidate to prevent HCV recurrence. In addition, our data are valuable for the design of a prophylactic vaccine.
Collapse
Affiliation(s)
| | - Samira Fafi-Kremer
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques,Université de Strasbourg, Strasbourg, France,Laboratoire de Virologie
| | | | - Patrick Pessaux
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques,Université de Strasbourg, Strasbourg, France,Laboratoire de Virologie
| | - Ali Farhoudi
- Center for Vaccinology, Ghent University, Ghent, Belgium
| | - Laura Heydmann
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques,Université de Strasbourg, Strasbourg, France
| | - Lieven Verhoye
- Center for Vaccinology, Ghent University, Ghent, Belgium
| | - Sarah Cole
- MRC – University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Jane A. McKeating
- Viral Hepatitis Research Group, Centre for Human Virology, University of Birmingham, Birmingham, UK
| | | | - Thomas F. Baumert
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques,Université de Strasbourg, Strasbourg, France,Laboratoire de Virologie,corresponding authors; Contact information: Prof. Philip Meuleman, PhD, Center for Vaccinology - Ghent University, University Hospital Ghent, Building A, 1st floor, De Pintelaan 185, B-9000 Gent, Belgium, Phone: +32 (0)9 332 02 05 (direct); Phone: +32 (0)9 332 36 58 (office administrator), Fax: +32 (0)9 332 63 11; , Thomas F. Baumert: , Arvind H. Patel:
| | - Arvind H. Patel
- MRC – University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK,corresponding authors; Contact information: Prof. Philip Meuleman, PhD, Center for Vaccinology - Ghent University, University Hospital Ghent, Building A, 1st floor, De Pintelaan 185, B-9000 Gent, Belgium, Phone: +32 (0)9 332 02 05 (direct); Phone: +32 (0)9 332 36 58 (office administrator), Fax: +32 (0)9 332 63 11; , Thomas F. Baumert: , Arvind H. Patel:
| | - Philip Meuleman
- Center for Vaccinology, Ghent University, Ghent, Belgium,corresponding authors; Contact information: Prof. Philip Meuleman, PhD, Center for Vaccinology - Ghent University, University Hospital Ghent, Building A, 1st floor, De Pintelaan 185, B-9000 Gent, Belgium, Phone: +32 (0)9 332 02 05 (direct); Phone: +32 (0)9 332 36 58 (office administrator), Fax: +32 (0)9 332 63 11; , Thomas F. Baumert: , Arvind H. Patel:
| |
Collapse
|
43
|
Sautto GA, Wisskirchen K, Clementi N, Castelli M, Diotti RA, Graf J, Clementi M, Burioni R, Protzer U, Mancini N. Chimeric antigen receptor (CAR)-engineered T cells redirected against hepatitis C virus (HCV) E2 glycoprotein. Gut 2016; 65:512-23. [PMID: 25661083 PMCID: PMC4789830 DOI: 10.1136/gutjnl-2014-308316] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/18/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The recent availability of novel antiviral drugs has raised new hope for a more effective treatment of hepatitis C virus (HCV) infection and its severe sequelae. However, in the case of non-responding or relapsing patients, alternative strategies are needed. To this end we have used chimeric antigen receptors (CARs), a very promising approach recently used in several clinical trials to redirect primary human T cells against different tumours. In particular, we designed the first CARs against HCV targeting the HCV/E2 glycoprotein (HCV/E2). DESIGN Anti-HCV/E2 CARs were composed of single-chain variable fragments (scFvs) obtained from a broadly cross-reactive and cross-neutralising human monoclonal antibody (mAb), e137, fused to the intracellular signalling motif of the costimulatory CD28 molecule and the CD3ζ domain. Activity of CAR-grafted T cells was evaluated in vitro against HCV/E2-transfected cells as well as hepatocytes infected with cell culture-derived HCV (HCVcc). RESULTS In this proof-of-concept study, retrovirus-transduced human T cells expressing anti-HCV/E2 CARs were endowed with specific antigen recognition accompanied by degranulation and secretion of proinflammatory and antiviral cytokines, such as interferon γ, interleukin 2 and tumour necrosis factor α. Moreover, CAR-grafted T cells were capable of lysing target cells of both hepatic and non-hepatic origin expressing on their surface the HCV/E2 glycoproteins of the most clinically relevant genotypes, including 1a, 1b, 2a, 3a, 4 and 5. Finally, and more importantly, they were capable of lysing HCVcc-infected hepatocytes. CONCLUSIONS Clearance of HCV-infected cells is a major therapeutic goal in chronic HCV infection, and adoptive transfer of anti-HCV/E2 CARs-grafted T cells represents a promising new therapeutic tool.
Collapse
Affiliation(s)
- Giuseppe A Sautto
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| | - Karin Wisskirchen
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Nicola Clementi
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| | - Matteo Castelli
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| | - Roberta A Diotti
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| | - Julia Graf
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Massimo Clementi
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| | - Roberto Burioni
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ulrike Protzer
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Nicasio Mancini
- Laboratorio di Microbiologia e Virologia, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
44
|
A Diverse Panel of Hepatitis C Virus Glycoproteins for Use in Vaccine Research Reveals Extremes of Monoclonal Antibody Neutralization Resistance. J Virol 2015; 90:3288-301. [PMID: 26699643 PMCID: PMC4794667 DOI: 10.1128/jvi.02700-15] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023] Open
Abstract
Despite significant advances in the treatment of hepatitis C virus (HCV) infection, the need to develop preventative vaccines remains. Identification of the best vaccine candidates and evaluation of their performance in preclinical and clinical development will require appropriate neutralization assays utilizing diverse HCV isolates. We aimed to generate and characterize a panel of HCV E1E2 glycoproteins suitable for subsequent use in vaccine and therapeutic antibody testing. Full-length E1E2 clones were PCR amplified from patient-derived serum samples, cloned into an expression vector, and used to generate viral pseudoparticles (HCVpp). In addition, some of these clones were used to generate cell culture infectious (HCVcc) clones. The infectivity and neutralization sensitivity of these viruses were then determined. Bioinformatic and HCVpp infectivity screening of approximately 900 E1E2 clones resulted in the assembly of a panel of 78 functional E1E2 proteins representing distinct HCV genotypes and different stages of infection. These HCV glycoproteins differed markedly in their sensitivity to neutralizing antibodies. We used this panel to predict antibody efficacy against circulating HCV strains, highlighting the likely reason why some monoclonal antibodies failed in previous clinical trials. This study provides the first objective categorization of cross-genotype patient-derived HCV E1E2 clones according to their sensitivity to antibody neutralization. It has shown that HCV isolates have clearly distinguishable neutralization-sensitive, -resistant, or -intermediate phenotypes, which are independent of genotype. The panel provides a systematic means for characterization of the neutralizing response elicited by candidate vaccines and for defining the therapeutic potential of monoclonal antibodies. IMPORTANCE Hepatitis C virus (HCV) has a global burden of more than 170 million people, many of whom cannot attain the new, expensive, direct-acting antiviral therapies. A safe and effective vaccine that generates both T cell responses and neutralizing antibodies is required to eradicate the disease. Regions within the HCV surface glycoproteins E1 and E2 are essential for virus entry and are targets for neutralizing antibodies. Screening of vaccine candidates requires suitable panels of glycoproteins that represent the breadth of neutralization resistance. Use of a standard reference panel for vaccine studies will ensure comparability of data sets, as has become routine for HIV-1. Here, we describe a large panel of patient-derived HCV glycoproteins with an assessment of their neutralization sensitivity to defined monoclonal antibodies, which has enabled us to predict their likely efficacy in the wider HCV-infected population. The panel could also be important for future selection of additional therapeutic antibodies and for vaccine design.
Collapse
|
45
|
Hakami AR, Ball JK, Tarr AW. Non-ionic detergents facilitate non-specific binding of M13 bacteriophage to polystyrene surfaces. J Virol Methods 2015; 221:1-8. [DOI: 10.1016/j.jviromet.2015.04.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 01/13/2023]
|
46
|
Apolipoprotein E, but Not Apolipoprotein B, Is Essential for Efficient Cell-to-Cell Transmission of Hepatitis C Virus. J Virol 2015. [PMID: 26202245 DOI: 10.1128/jvi.00577-15] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Hepatitis C virus (HCV) infects hepatocytes through two different routes: (i) cell-free particle diffusion followed by engagement with specific cellular receptors and (ii) cell-to-cell direct transmission mediated by mechanisms not well defined yet. HCV exits host cells in association with very-low-density lipoprotein (VLDL) components. VLDL particles contain apolipoproteins B (ApoB) and E (ApoE), which are required for viral assembly and/or infectivity. Based on these precedents, we decided to study whether these VLDL components participate in HCV cell-to-cell transmission in vitro. We observed that cell-to-cell viral spread was compromised after ApoE interference in donor but not in acceptor cells. In contrast, ApoB knockdown in either donor or acceptor cells did not impair cell-to-cell viral transmission. Interestingly, ApoB participated in the assembly of cell-free infective virions, suggesting a differential regulation of cell-to-cell and cell-free HCV infection. This study identifies host-specific factors involved in these distinct routes of infection that may unveil new therapeutic targets and advance our understanding of HCV pathogenesis. IMPORTANCE This work demonstrates that cell-to-cell transmission of HCV depends on ApoE but not ApoB. The data also indicate that ApoB is required for the assembly of cell-free infective particles, strongly suggesting the existence of mechanisms involving VLDL components that differentially regulate cell-free and cell-to-cell HCV transmission. These data clarify some of the questions regarding the role of VLDL in HCV pathogenesis and the transmission of the virus cell to cell as a possible mechanism of immune evasion and open the door to therapeutic intervention.
Collapse
|
47
|
Tarr AW, Khera T, Hueging K, Sheldon J, Steinmann E, Pietschmann T, Brown RJP. Genetic Diversity Underlying the Envelope Glycoproteins of Hepatitis C Virus: Structural and Functional Consequences and the Implications for Vaccine Design. Viruses 2015; 7:3995-4046. [PMID: 26193307 PMCID: PMC4517138 DOI: 10.3390/v7072809] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/19/2015] [Accepted: 07/08/2015] [Indexed: 12/13/2022] Open
Abstract
In the 26 years since the discovery of Hepatitis C virus (HCV) a major global research effort has illuminated many aspects of the viral life cycle, facilitating the development of targeted antivirals. Recently, effective direct-acting antiviral (DAA) regimens with >90% cure rates have become available for treatment of chronic HCV infection in developed nations, representing a significant advance towards global eradication. However, the high cost of these treatments results in highly restricted access in developing nations, where the disease burden is greatest. Additionally, the largely asymptomatic nature of infection facilitates continued transmission in at risk groups and resource constrained settings due to limited surveillance. Consequently a prophylactic vaccine is much needed. The HCV envelope glycoproteins E1 and E2 are located on the surface of viral lipid envelope, facilitate viral entry and are the targets for host immunity, in addition to other functions. Unfortunately, the extreme global genetic and antigenic diversity exhibited by the HCV glycoproteins represents a significant obstacle to vaccine development. Here we review current knowledge of HCV envelope protein structure, integrating knowledge of genetic, antigenic and functional diversity to inform rational immunogen design.
Collapse
Affiliation(s)
- Alexander W Tarr
- School of Life Sciences, Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Tanvi Khera
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centrefor Infection Research (HZI), Hannover D-30625, Germany.
| | - Kathrin Hueging
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centrefor Infection Research (HZI), Hannover D-30625, Germany.
| | - Julie Sheldon
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centrefor Infection Research (HZI), Hannover D-30625, Germany.
| | - Eike Steinmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centrefor Infection Research (HZI), Hannover D-30625, Germany.
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centrefor Infection Research (HZI), Hannover D-30625, Germany.
- German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig 38124, Germany.
| | - Richard J P Brown
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centrefor Infection Research (HZI), Hannover D-30625, Germany.
| |
Collapse
|
48
|
Drummer HE. Editorial on "Broadly neutralizing antibodies abrogate established hepatitis C virus infection" published in Science Translational Medicine on 17th September 2014. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:S6. [PMID: 26046093 DOI: 10.3978/j.issn.2305-5839.2015.03.55] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/06/2015] [Indexed: 11/14/2022]
Abstract
Hepatitis C virus (HCV) is a blood borne pathogen that causes chronic liver disease and afflicts 170 million people world-wide. While direct acting antivirals now provide a highly effective means to cure those infected with HCV, there is no vaccine to prevent infection. Published in Science Translational Medicine, de Jong et al. [2014] show that highly potent neutralizing antibodies (NAbs) directed to one of the surface glycoproteins of HCV, E2, can not only prevent infection but can also eliminate established infection in experimental animal models of HCV. They provide compelling evidence that for HCV to maintain a chronic infection, it must infect new hepatocytes; infection cannot be sustained in reservoirs of infected cells alone and that E2-specific NAbs are sufficient to cure an infection. In addition, the manuscript further supports the importance of NAbs in preventing, controlling and possibly curing HCV. Thus NAbs are not only essential to the development of prophylactic vaccines but may yet have a role in therapeutic approaches to HCV treatment.
Collapse
Affiliation(s)
- Heidi E Drummer
- Viral Fusion Laboratory, Centre for Biomedical Research, Burnet Institute, Melbourne, Australia
| |
Collapse
|
49
|
Structural flexibility of a conserved antigenic region in hepatitis C virus glycoprotein E2 recognized by broadly neutralizing antibodies. J Virol 2014; 89:2170-81. [PMID: 25473061 DOI: 10.1128/jvi.02190-14] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Neutralizing antibodies (NAbs) targeting glycoprotein E2 are important for the control of hepatitis C virus (HCV) infection. One conserved antigenic site (amino acids 412 to 423) is disordered in the reported E2 structure, but a synthetic peptide mimicking this site forms a β-hairpin in complex with three independent NAbs. Our structure of the same peptide in complex with NAb 3/11 demonstrates a strikingly different extended conformation. We also show that residues 412 to 423 are essential for virus entry but not for E2 folding. Together with the neutralizing capacity of the 3/11 Fab fragment, this indicates an unexpected structural flexibility within this epitope. NAbs 3/11 and AP33 (recognizing the extended and β-hairpin conformations, respectively) display similar neutralizing activities despite converse binding kinetics. Our results suggest that HCV utilizes conformational flexibility as an immune evasion strategy, contributing to the limited immunogenicity of this epitope in patients, similar to the conformational flexibility described for other enveloped and nonenveloped viruses. IMPORTANCE Approximately 180 million people worldwide are infected with hepatitis C virus (HCV), and neutralizing antibodies play an important role in controlling the replication of this major human pathogen. We show here that one of the most conserved antigenic sites within the major glycoprotein E2 (amino acids 412 to 423), which is disordered in the recently reported crystal structure of an E2 core fragment, can adopt different conformations in the context of the infectious virus particle. Recombinant Fab fragments recognizing different conformations of this antigenic site have similar neutralization activities in spite of converse kinetic binding parameters. Of note, an antibody response targeting this antigenic region is less frequent than those targeting other more immunogenic regions in E2. Our results suggest that the observed conformational flexibility in this conserved antigenic region contributes to the evasion of the humoral host immune response, facilitating chronicity and the viral spread of HCV within an infected individual.
Collapse
|
50
|
Hamed MR, Brown RJ, Zothner C, Urbanowicz RA, Mason CP, Krarup A, McClure CP, Irving WL, Ball JK, Harris M, Hickling TP, Tarr AW. Recombinant human L-ficolin directly neutralizes hepatitis C virus entry. J Innate Immun 2014; 6:676-84. [PMID: 24854201 PMCID: PMC6741592 DOI: 10.1159/000362209] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 03/16/2014] [Accepted: 03/16/2014] [Indexed: 12/25/2022] Open
Abstract
L-ficolin is a soluble pattern recognition molecule expressed by the liver that contributes to innate immune defense against microorganisms. It is well described that binding of L-ficolin to specific pathogen-associated molecular patterns activates the lectin complement pathway, resulting in opsonization and lysis of pathogens. In this study, we demonstrated that in addition to this indirect effect, L-ficolin has a direct neutralizing effect against hepatitis C virus (HCV) entry. Specific, dose-dependent binding of recombinant L-ficolin to HCV glycoproteins E1 and E2 was observed. This interaction was inhibited by soluble L-ficolin ligands. Interaction of L-ficolin with E1 and E2 potently inhibited entry of retroviral pseudoparticles bearing these glycoproteins. L-ficolin also inhibited entry of cell-cultured HCV in a calcium-dependent manner. Neutralizing concentrations of L-ficolin were found to be circulating in the serum of HCV-infected individuals. This is the first description of direct neutralization of HCV entry by a ficolin and highlights a novel role for L-ficolin as a virus entry inhibitor.
Collapse
Affiliation(s)
- Mohamed R. Hamed
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Richard J.P. Brown
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Carsten Zothner
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Richard A. Urbanowicz
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Christopher P. Mason
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Anders Krarup
- Biochemistry Department, University of Oxford, Oxford, UK
| | - C. Patrick McClure
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - William L. Irving
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Jonathan K. Ball
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Timothy P. Hickling
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | - Alexander W. Tarr
- School of Life Sciences, and Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| |
Collapse
|