1
|
Gao Y, Tillu VA, Wu Y, Rae J, Hall TE, Chen KE, Weeratunga S, Guo Q, Livingstone E, Tham WH, Parton RG, Collins BM. Nanobodies against Cavin1 reveal structural flexibility and regulated interactions of its N-terminal coiled-coil domain. J Cell Sci 2025; 138:jcs263756. [PMID: 40260863 DOI: 10.1242/jcs.263756] [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: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Caveolae are abundant plasma membrane structures that regulate signalling, membrane homeostasis and mechanoprotection. Their formation is driven by caveolins and cavins and their coordinated interactions with lipids. Here, we developed nanobodies against the trimeric HR1 coiled-coil domain of Cavin1. We identified specific nanobodies that do not perturb Cavin1 membrane binding and localise to caveolae when expressed in cells. The crystal structure of a nanobody-Cavin 1 HR1 complex reveals a symmetric 3:3 architecture as validated by mutagenesis. In this structure, the C-terminal half of the HR1 domain is disordered, suggesting that the nanobody stabilises an open conformation of Cavin1, which has previously been identified as important for membrane interactions. A phosphomimic mutation in a threonine-serine pair proximal to this region reveals selective regulation of Cavin2 and Cavin3 association. These studies provide new insights into cavin domains required for assembly of multiprotein caveolar assemblies and describe new nanobody tools for structural and functional studies of caveolae.
Collapse
Affiliation(s)
- Ya Gao
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Vikas A Tillu
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Yeping Wu
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - James Rae
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Kai-En Chen
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Saroja Weeratunga
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Qian Guo
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Emma Livingstone
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Wai-Hong Tham
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| |
Collapse
|
2
|
Maltseva M, Rossotti MA, Tanha J, Langlois MA. Characterization of Nanobody Binding to Distinct Regions of the SARS-CoV-2 Spike Protein by Flow Virometry. Viruses 2025; 17:571. [PMID: 40285013 PMCID: PMC12030927 DOI: 10.3390/v17040571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/31/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Nanobodies, or single-domain antibodies (VHHs) from camelid heavy-chain-only antibodies, offer significant advantages in therapeutic and diagnostic applications due to their small size and ability to bind cryptic protein epitopes inaccessible to conventional antibodies. In this study, we examined nanobodies specific to regions of the SARS-CoV-2 spike glycoprotein, including the receptor-binding domain (RBD), N-terminal domain (NTD), and subunit 2 (S2). Using flow virometry, a high-throughput technique for viral quantification, we achieved the efficient detection of pseudotyped viruses expressing the spike glycoprotein. RBD-targeting nanobodies showed the most effective staining, followed by NTD-targeting ones, while S2-specific nanobodies exhibited limited resolution. The simple genetic structure of nanobodies enables the creation of multimeric formats, improving binding specificity and avidity. Bivalent VHH-Fc constructs (VHHs fused to the Fc region of human IgG) outperformed monovalent formats in resolving viral particles from background noise. However, S2-specific monovalent VHHs demonstrated improved staining efficiency, suggesting their smaller size better accesses restricted antigenic sites. Furthermore, direct staining of cell supernatants was possible without virus purification. This versatile nanobody platform, initially developed for antiviral therapy against SARS-CoV-2, can be readily adapted for flow virometry applications and other diagnostic assays.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.M.); (J.T.)
| | - Martin A. Rossotti
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, Ottawa, ON K1N 1J1, Canada;
| | - Jamshid Tanha
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.M.); (J.T.)
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, Ottawa, ON K1N 1J1, Canada;
- uOttawa Center for Infection, Immunity, and Inflammation (CI3), Ottawa, ON K1H 8L1, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.M.); (J.T.)
- uOttawa Center for Infection, Immunity, and Inflammation (CI3), Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
3
|
Ketaren NE, Fridy PC, Malashkevich V, Sanyal T, Brillantes M, Thompson MK, Oren DA, Bonanno JB, Šali A, Almo SC, Chait BT, Rout MP. Unique mechanisms to increase structural stability and enhance antigen binding in nanobodies. Structure 2025; 33:677-690.e5. [PMID: 39938509 PMCID: PMC12042621 DOI: 10.1016/j.str.2025.01.019] [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: 10/15/2024] [Revised: 12/04/2024] [Accepted: 01/16/2025] [Indexed: 02/14/2025]
Abstract
Nanobodies are single domain antibody variants proving themselves to be compelling tools for research, disease diagnostics, and as therapeutics targeting a myriad of disease agents. However, despite this potential, their mechanisms of paratope presentation and structural stabilization have not been fully explored. Here, we show that unlike monoclonal antibodies, a nanobody repertoire maximizes sampling of an antigen surface by binding a single antigen in at least three different orientations, which are correlated with their paratope composition. Structure-guided reengineering of several nanobodies reveals that a single point mutation within the paratope or a highly conserved region of a nanobody's framework 3 (FR3) can markedly improve antigen affinity, nanobody stability, or both. Conversely, we show the negative impact on antigen affinity when "over-stabilizing" nanobodies. Collectively our results provide a universal strategy to tune a nanobody's affinity by modifying specific residues that can readily be applied to guide nanobody optimization and functionalization.
Collapse
Affiliation(s)
- Natalia E Ketaren
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA.
| | - Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA
| | - Vladimir Malashkevich
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Tanmoy Sanyal
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marc Brillantes
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA
| | - Mary K Thompson
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA
| | - Deena A Oren
- Structural Biology Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey B Bonanno
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrej Šali
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY 10065, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
4
|
Lai Y, Xie B, Zhang W, He W. Pure drug nanomedicines - where we are? Chin J Nat Med 2025; 23:385-409. [PMID: 40274343 DOI: 10.1016/s1875-5364(25)60851-x] [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: 08/11/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 04/26/2025]
Abstract
Pure drug nanomedicines (PDNs) encompass active pharmaceutical ingredients (APIs), including macromolecules, biological compounds, and functional components. They overcome research barriers and conversion thresholds associated with nanocarriers, offering advantages such as high drug loading capacity, synergistic treatment effects, and environmentally friendly production methods. This review provides a comprehensive overview of the latest advancements in PDNs, focusing on their essential components, design theories, and manufacturing techniques. The physicochemical properties and in vivo behaviors of PDNs are thoroughly analyzed to gain an in-depth understanding of their systematic characteristics. The review introduces currently approved PDN products and further explores the opportunities and challenges in expanding their depth and breadth of application. Drug nanocrystals, drug-drug cocrystals (DDCs), antibody-drug conjugates (ADCs), and nanobodies represent the successful commercialization and widespread utilization of PDNs across various disease domains. Self-assembled pure drug nanoparticles (SAPDNPs), a next-generation product, still require extensive translational research. Challenges persist in transitioning from laboratory-scale production to mass manufacturing and overcoming the conversion threshold from laboratory findings to clinical applications.
Collapse
Affiliation(s)
- Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wanting Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
5
|
Wang Q, Hong D, Li E, Cheng Z, Zhang J, Jin T, Chiu S. Generation of nanobodies against the F protein of respiratory syncytial virus and establishment of an indirect immunofluorescence assay. Microbiol Spectr 2025:e0278424. [PMID: 40130866 DOI: 10.1128/spectrum.02784-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/11/2025] [Indexed: 03/26/2025] Open
Abstract
Respiratory syncytial virus (RSV) infection is a leading cause of acute respiratory infections and death in children and older adults. Currently, there is a lack of effective vaccines and antibody treatments on the market, and the clinical manifestations of RSV are indistinguishable from those of acute respiratory infections caused by other etiologies. Therefore, the prevention and treatment of RSV require not only effective vaccines but also simple, fast, cost-effective, and accurate detection and diagnostic methods. Given the small molecular weight, excellent antigen-binding specificity, and affinity of nanobodies, we successfully obtained a panel of nanobodies by expressing the RSV F protein, immunizing camel, and establishing a phage display library. Among them, F-E2 has been shown to bind specifically to the RSV F protein and can be used for Western blot, enzyme-linked immunosorbent assay, flow cytometry, immunohistochemistry, and immunofluorescence assays. Moreover, an RSV immunofluorescence detection method based on F-E2 has been established and proved to be highly specific, sensitive, cost-effective, and fast, with great application potential. IMPORTANCE A respiratory syncytial virus (RSV) detection nanobody, F-E2, was successfully screened by constructing a dromedary camel immune library. F-E2 binds to RSV F with ng affinities and can be used for Western blot, enzyme-linked immunosorbent assay, flow cytometry, immunohistochemistry, and immunofluorescence assays. Importantly, a high-throughput RSV immunofluorescence detection method based on F-E2 has been established and proved to be highly specific, sensitive, cost-effective, and fast, with great application potential.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China
| | - Dongxiang Hong
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Entao Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China
| | - Zekai Cheng
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China
| | - Jiachen Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China
| | - Tengchuan Jin
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China
| | - Sandra Chiu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China
| |
Collapse
|
6
|
Bernasconi-Bisio F, Molina E, Ibarra V, Ibáñez-Sala I, Rochira F, Jauregui P, Rodríguez-Diaz S, Martínez-Turrillas R, Azagra-Barber I, Gómez-Cebrián N, Lasarte JJ, Puchades-Carrasco L, Vanrell L, Rodríguez-Madoz JR, Prósper F, Pineda-Lucena A. Discovery and preclinical development of a SdAb-based CAR-T technology for targeting CD33 in AML. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200949. [PMID: 40084273 PMCID: PMC11904528 DOI: 10.1016/j.omton.2025.200949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapies have revolutionized cancer immunotherapy. Traditional single-chain variable fragments (ScFvs) used as CAR recognition moieties face challenges such as high tonic signaling, compromised binding epitopes, and suboptimal affinity. Single-domain antibodies (SdAbs) offer an attractive alternative due to their smaller size, stability, and reduced immunogenicity. In this work, we developed an SdAb-CAR-T cell discovery platform integrating generation, characterization, and selection of SdAbs based on various properties. This approach was demonstrated by developing CAR-T cells with SdAbs against CD33, a target for acute myeloid leukemia (AML). We identified diverse SdAbs against CD33, with affinities ranging from 3.9-115 nM, and characterized their binding kinetics and epitope recognition. Using SdAb-based second-generation CARs, we assessed tonic signaling, T cell phenotypes, cytotoxicity and cytokine release in vitro, resulting in reduced tonic signaling and increased cytokine production. In vivo, SdAb-based CAR-T cells exhibited enhanced efficacy at lower doses, in a xenograft AML mouse model, demonstrating advantages over ScFv-based CD33 CAR-T cells.
Collapse
Affiliation(s)
| | - Eva Molina
- Therapeutic Innovation Program, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Vianca Ibarra
- Therapeutic Innovation Program, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Inés Ibáñez-Sala
- Therapeutic Innovation Program, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Federica Rochira
- Therapeutic Innovation Program, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Patricia Jauregui
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
| | - Saray Rodríguez-Diaz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
| | - Rebeca Martínez-Turrillas
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
| | - Iñigo Azagra-Barber
- Therapeutic Innovation Program, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Nuria Gómez-Cebrián
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Juan José Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
| | - Leonor Puchades-Carrasco
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | | | - Juan Roberto Rodríguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
- Hematology and Cell Therapy Department, Clínica Universidad de Navarra, IdiSNA, 31008 Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 31008 Pamplona, Spain
| | - Antonio Pineda-Lucena
- Therapeutic Innovation Program, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Cancer Center Clínica Universidad de Navarra (CCUN), 31008 Pamplona, Spain
| |
Collapse
|
7
|
Maksymova L, Pilger YA, Nuhn L, Van Ginderachter JA. Nanobodies targeting the tumor microenvironment and their formulation as nanomedicines. Mol Cancer 2025; 24:65. [PMID: 40033293 PMCID: PMC11877942 DOI: 10.1186/s12943-025-02270-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Among the emerging strategies for cancer theranostics, nanomedicines offer significant promise in advancing both patients' diagnosis and treatment. In combination with nanobodies, nanomedicines can potentially enhance the precision and efficiency of drug or imaging agent delivery, addressing key limitations of current approaches, such as off-target toxicities. The development of nanomedicines will be further accelerated by the creation of smart nanoparticles, and their integration with immunotherapy. Obviously, the success of nano-immunotherapy will depend on a comprehensive understanding of the tumor microenvironment, including the complex interplay of mechanisms that drive cancer-mediated immunosuppression and immune escape. Hence, effective therapeutic targeting of the tumor microenvironment requires modulation of immune cell function, overcoming resistance mechanisms associated with stromal components or the extracellular matrix, and/or direct elimination of cancer cells. Identifying key molecules involved in cancer progression and drug resistance is, therefore, essential for developing effective therapies and diagnostic tools that can predict patient responses to treatment and monitor therapeutic outcomes. Current nanomedicines are being designed with careful consideration of factors such as the choice of carrier (e.g., biocompatibility, controlled cargo release) and targeting moiety. The unique properties of nanobodies make them an effective engineering tool to target biological molecules with high affinity and specificity. In this review, we focus on the latest applications of nanobodies for targeting various components of the tumor microenvironment for diagnostic and therapeutic purposes. We also explore the main types of nanoparticles used as a carrier for cancer immunotherapies, as well as the strategies for formulating nanoparticle-nanobody conjugates. Finally, we highlight how nanobody-nanoparticle formulations can enhance current nanomedicines.
Collapse
Affiliation(s)
- Liudmyla Maksymova
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Pleinlaan 2, Brussels, B-1050, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yannick A Pilger
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Faculty of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg, 97070, Germany
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Faculty of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg, 97070, Germany.
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Pleinlaan 2, Brussels, B-1050, Belgium.
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
8
|
Zheng L, Wang H, Zhong X, Jia L, Shi G, Bai C, Yang R, Huang Z, Jiang Y, Wei J, Dong Z, Li J, Long Y, Dai L, Li Z, Chen C, Wang J. Reprogramming tumor microenvironment with precise photothermal therapy by calreticulin nanobody-engineered probiotics. Biomaterials 2025; 314:122809. [PMID: 39303415 DOI: 10.1016/j.biomaterials.2024.122809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024]
Abstract
Targeted therapies have revolutionized traditional cancer treatments by precisely targeting tumor cells, enhancing efficacy and safety. Despite this advancement, the proportion of cancer patients eligible for such therapies remains low due to the absence of suitable targets. Here, we investigate whether the translocation of the immunogenic cell death (ICD) marker calreticulin (CALR) from the endoplasmic reticulum (ER) to the cell surface following ICD induction can serve as a target for targeted therapies. To target CALR, a nanobody Nb215 identified from a naïve VHH phage library with high binding affinity to both human and mouse CALR was employed to engineer probiotic EcN 1917. Our results demonstrated that CALR nanobody-modified EcN-215 coupled with the photothermal dye indocyanine green (ICG) was able to exert NIR-II imaging-guide photothermal therapy (PTT). Moreover, PTT with EcN-215/ICG can reshape the tumor microenvironment by enhancing the infiltration of CD45+CD3+ T cells and CD11b+F4/80+ macrophages. Furthermore, the antitumor activity of CALR-targeted EcN-215/ICG is synergistically enhanced by blocking CD47-SIRPα axis. Collectively, our study provides a proof of concept for CALR-targeted therapy. Given that CALR translocation can be induced by various anticancer therapies across numerous tumor cell lines, CALR-targeted therapies hold promise as a novel approach for treating multiple types of cancers.
Collapse
Affiliation(s)
- Liuhai Zheng
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Huifang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaoru Zhong
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Lin Jia
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Guangwei Shi
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528399, China
| | - Chongzhi Bai
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Runwei Yang
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528399, China
| | - Zhenhui Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Yuke Jiang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Jinxi Wei
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Zhiyu Dong
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Jiexuan Li
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Ying Long
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Lingyun Dai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Zhijie Li
- Department of Hyperbaric Oxygen Medicine, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China.
| | - Chunbo Chen
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Jigang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
9
|
Mateusiak Ł, Hakuno S, de Jonge-Muller ESM, Floru S, Sier CFM, Hawinkels LJAC, Hernot S. Fluorescent Nanobodies for enhanced guidance in digestive tumors and liver metastasis surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109537. [PMID: 39753054 DOI: 10.1016/j.ejso.2024.109537] [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: 09/19/2024] [Revised: 11/04/2024] [Accepted: 12/05/2024] [Indexed: 03/17/2025]
Abstract
BACKGROUND Fluorescence molecular imaging, a potent and non-invasive technique, has become indispensable in medicine for visualizing molecular processes. In surgical oncology, it aids treatment by allowing visualization of tumor cells during fluorescence-guided surgery (FGS). Targeting the urokinase plasminogen activator receptor (uPAR), overexpressed during tissue remodeling and inflammation, holds promise for advancing FGS by specifically highlighting tumors. This study explores the extended use of Nanobody-based (Nb) anti-uPAR tracers, evaluating their receptor binding, ability to visualize and demarcate colorectal (CRC) and gastric cancer (GC), and detect localized (PC) and metastatic (PC-M) pancreatic carcinoma. METHODS First, the receptor structure interactions of Nb15, which binds specifically to the human homologue of uPAR, were characterized in vitro to deepen our understanding of these interactions. Subsequently, Nbs 15 and 13-where Nb13 targets the murine uPAR homologue-were labeled with the s775z fluorescent dye and validated in a randomized study in mice (n = 4 per group) using orthotopic human CRC, GC, and PC models, as well as a mouse PC-M model. RESULTS Nb15, which binds to the D1 domain of uPAR and competes with urokinase's binding fragment, showed rapid and specific tumor accumulation. It exhibited higher tumor-to-background ratios in CRC (3.35 ± 0.75) and PC (3.41 ± 0.46), and effectively differentiated tumors in GC (mean fluorescence intensity: 0.084 ± 0.017), as compared to control Nbs. Nb13 successfully identified primary tumors and liver metastases in PC-M models. CONCLUSION The tested fluorescently-labeled anti-uPAR Nbs show significant preclinical and clinical potential for improving surgical precision and patient outcomes, with Nb15 demonstrating promise for real-time surgical guidance.
Collapse
Affiliation(s)
- Łukasz Mateusiak
- Vrije Universiteit Brussel (VUB), Molecular Imaging and Therapy Research Group, MITH, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Sarah Hakuno
- Leiden University Medical Center (LUMC), Department of Gastroenterology and Hepatology, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Eveline S M de Jonge-Muller
- Leiden University Medical Center (LUMC), Department of Gastroenterology and Hepatology, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Sam Floru
- Vrije Universiteit Brussel (VUB), Molecular Imaging and Therapy Research Group, MITH, Laarbeeklaan 103, 1090, Brussels, Belgium
| | | | - Lukas J A C Hawinkels
- Leiden University Medical Center (LUMC), Department of Gastroenterology and Hepatology, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Sophie Hernot
- Vrije Universiteit Brussel (VUB), Molecular Imaging and Therapy Research Group, MITH, Laarbeeklaan 103, 1090, Brussels, Belgium
| |
Collapse
|
10
|
Liang W, Zhang Y, Li J, Ji C, Jiang X. Her-2 nanobody modified cisplatin nanoparticles for precise chemotherapy of colon cancer. J Microencapsul 2025:1-9. [PMID: 39976420 DOI: 10.1080/02652048.2025.2467060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
Construct a Her-2 nanobody modified nanoplatform as a versatile carrier of cisplatin and evaluate its anti-tumour effects. Size, morphology, cellular uptake, in vitro release, cell viability, bio-distribution and antitumor efficacy were respectively measured by dynamic light scattering, transmission electron microscopy, confocal microscopy, HPLC, MTT assay, ICP-Mass and tumour volume. Nb-CDDP NPs was prepared with average diameter 60.4 ± 8.4 nm, PDI 0.2 ± 0.02, Zeta potential -35.74 mV, entrapment efficiency 89.5%±0.8% and drug loading 28.7%±1.3% (w/w). From which cisplatin could release more rapidly in acidic solution. NPs could be easily phagocytised and exhibited stronger cytotoxic effect in HCT-116 cells with IC50 1.46 ± 0.019 μg/mL. The concentration of Nb-CDDP NPs in tumour and its inhibition ratio on tumour volume were both higher than without Nb modification, with hardly any influence on body weight. This cisplatin nanoplatform exhibits exceptional properties and high targeting anti-tumour efficacy in colon cancer cells and mice, which maybe provide a promising strategy for precise chemotherapy.
Collapse
Affiliation(s)
- Weina Liang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Yan Zhang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Jianpeng Li
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Chenglin Ji
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| | - Xuexin Jiang
- Yantai Food and Drug Inspection and Testing Center, Yantai, China
| |
Collapse
|
11
|
Zhu N, Smallwood PM, Williams J, Wang Y, Nathans JH. Utility of binding protein fusions to immunoglobulin heavy chain constant regions from mammalian and avian species. J Biol Chem 2025; 301:108324. [PMID: 39971160 PMCID: PMC11964738 DOI: 10.1016/j.jbc.2025.108324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025] Open
Abstract
Antibodies are of central importance as reagents for the localization of proteins and other biomolecules in cells and tissues. To expand the repertoire of antibody-based reagents, we have constructed a series of plasmid vectors that permit expression of amino-terminal fusions to the hinge and Fc regions from goat, guinea pig, human, mouse, and rabbit immunoglobulin Gs, and chicken immunogloblin Y. The resulting fusion proteins can be produced in transfected mammalian cells and detected with commercially available and species-specific secondary antibody reagents. We demonstrate the utility of this platform by constructing and testing Fc fusions with DARPin, single-chain Fv, nanobody, toxin, and chemokine partners. The resulting fusion proteins were used to detect their targets in tissue sections or on the surface of transfected cells by immunofluorescent staining or on the surface of immune cells by flow cytometry. By expanding the range of Fc sequences available for fusion protein production, this platform will expand the repertoire of primary antibody reagents for multiplexed immunostaining and fluorescence-activated cell sorting analyses.
Collapse
Affiliation(s)
- Ningyu Zhu
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John Williams
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy H Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
12
|
Galindo G, Maejima D, DeRoo J, Burlingham SR, Fixen G, Morisaki T, Febvre HP, Hasbrook R, Zhao N, Ghosh S, Mayton EH, Snow CD, Geiss BJ, Ohkawa Y, Sato Y, Kimura H, Stasevich TJ. AI-assisted protein design to rapidly convert antibody sequences to intrabodies targeting diverse peptides and histone modifications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636921. [PMID: 39975170 PMCID: PMC11839053 DOI: 10.1101/2025.02.06.636921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Intrabodies are engineered antibodies that function inside living cells, enabling therapeutic, diagnostic, and imaging applications. While powerful, their development has been hindered by challenges associated with their folding, solubility, and stability in the reduced intracellular environment. Here, we present an AI-driven pipeline integrating AlphaFold2, ProteinMPNN, and live-cell screening to optimize antibody framework regions while preserving epitope-binding complementarity-determining regions. Using this approach, we successfully converted 19 out of 26 antibody sequences into functional single-chain variable fragment (scFv) intrabodies, including a panel targeting diverse histone modifications for real-time imaging of chromatin dynamics and gene regulation. Notably, 18 of these 19 sequences had failed to convert using the standard approach, demonstrating the unique effectiveness of our method. As antibody sequence databases expand, our method will accelerate intrabody design, making their development easier, more cost-effective, and broadly accessible for biological research.
Collapse
Affiliation(s)
- Gabriel Galindo
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Daiki Maejima
- School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Jacob DeRoo
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Scott R Burlingham
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Gretchen Fixen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Tatsuya Morisaki
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Hallie P Febvre
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Ryan Hasbrook
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Ning Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - E Handly Mayton
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins, CO, USA
| | - Christopher D Snow
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Chemical & Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Brian J Geiss
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology, & Pathology, Colorado State University, Fort Collins, CO, USA
| | - Yasuyuki Ohkawa
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yuko Sato
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Kimura
- School of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama, Japan
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- Department of Chemical & Biological Engineering, Colorado State University, Fort Collins, CO, USA
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama, Japan
| |
Collapse
|
13
|
Al-Seragi M, Chen Y, Duong van Hoa F. Advances in nanobody multimerization and multispecificity: from in vivo assembly to in vitro production. Biochem Soc Trans 2025; 53:BST20241419. [PMID: 39927832 DOI: 10.1042/bst20241419] [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: 10/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/11/2025]
Abstract
NANOBODIES® (Nbs) have emerged as valuable tools across therapeutic, diagnostic, and industrial applications owing to their small size and consequent ability to bind unique epitopes inaccessible to conventional antibodies. While Nbs retrieved from immune libraries normally possess sufficient affinity and specificity for their cognate antigens in the practical use case, their multimerization will often increase functional affinity via avidity effects. Therefore, to rescue binding affinity and broaden targeting specificities, recent efforts have focused on conjugating multiple Nb clones - of identical or unique antigen cognates - together. In vivo and in vitro approaches, including flexible linkers, antibody domains, self-assembling coiled coils, chemical conjugation, and self-clustering hydrophobic sequences, have been employed to produce multivalent and multispecific Nb constructs. Examples of successful Nb multimerization are diverse, ranging from immunoassaying reagents to virus-neutralizing moieties. This review aims to recapitulate the in vivo and in vitro modalities to produce multivalent and multispecific Nbs while highlighting the applications, advantages, and drawbacks tied to each method.
Collapse
Affiliation(s)
- Mohammed Al-Seragi
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yilun Chen
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Franck Duong van Hoa
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
14
|
Davies LJ, Ghosh P, Siryer S, Ullrich S, Nitsche C. Peptide-Bismuth Tricycles: Maximizing Stability by Constraint. Chemistry 2025; 31:e202500064. [PMID: 39803821 DOI: 10.1002/chem.202500064] [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: 01/08/2025] [Indexed: 01/23/2025]
Abstract
Constrained peptides possess excellent properties for identifying lead compounds in drug discovery. While it has become increasingly straightforward to discover selective high-affinity peptide ligands, especially through genetically encoded libraries, their stability and bioavailability remain significant challenges. By integrating macrocyclization chemistry with bismuth binding, we generated series of linear, cyclic, bicyclic, and tricyclic peptides with identical sequences. Utilizing bismuth to rigidify the peptide structure allows for a better comparison of different constraint levels, reducing confounding effects of interactions often seen with hydrophobic stapling reagents. Our study facilitated the identification of a peptide-bismuth tricycle that fully withstands cellular levels of glutathione, acts as a nanomolar protease inhibitor without being proteolytically digested by its target, and is fully stable in human plasma. Importantly, this multicyclic peptide does not possess any non-canonical amino acid modifications. Using oxime ligation, we conjugated an analogue of this tricycle to the N-terminus of two nanobodies to demonstrate potential applications in targeted therapy.
Collapse
Affiliation(s)
- Lani J Davies
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Pritha Ghosh
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Sauhta Siryer
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Sven Ullrich
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
15
|
Zhang J, Wang M, Wang D, Zhang X, Ma Y, Pardon E, Steyaert J, Abskharon R, Wang F, Ma J. Investigating the In Vivo Effects of Anti-Prion Protein Nanobodies on Prion Disease with AAV Vector. Pathogens 2025; 14:131. [PMID: 40005509 PMCID: PMC11858434 DOI: 10.3390/pathogens14020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Prion diseases are fatal neurodegenerative disorders affecting humans and animals, and the central pathogenic event is the conversion of normal prion protein (PrPC) into the pathogenic PrPSc isoform. Previous studies have identified nanobodies that specifically recognize PrPC and inhibit the PrPC to PrPSc conversion in vitro. In this study, we investigated the potential for in vivo expression of anti-PrPC nanobodies and evaluated their impact on prion disease. The coding sequences of three nanobodies were packaged into recombinant adeno-associated virus (rAAV) and were administered via intracerebroventricular (ICV) injection in newborn mice. We found that the expression of these nanobodies remained robust for over 180 days, with no observed detrimental effects. To assess their therapeutic potential, we performed ICV injections of nanobody-expressing rAAVs in newborn mice, followed by intracerebral prion inoculation at 5-6 weeks of age. One nanobody exhibited a small yet statistically significant therapeutic effect, extending survival time from 176 days to 184 days. Analyses of diseased brains revealed that the nanobodies did not alter the pathological changes. Our findings suggest that high levels of anti-PrPC nanobodies are necessary to delay disease progression. Further optimization of the nanobodies, AAV vectors, or delivery methods is essential to achieve a significant therapeutic effect.
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102206, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Mengfei Wang
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102206, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Dan Wang
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102206, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Xiangyi Zhang
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102206, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Yue Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium (R.A.)
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium (R.A.)
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium
| | - Romany Abskharon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium (R.A.)
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium
- National Institute of Oceanography and Fisheries (NIOF), Cairo 11516, Egypt
| | - Fei Wang
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jiyan Ma
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102206, China
- Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
16
|
Li Y, Duan Z, Li Z, Xue W. Data and AI-driven synthetic binding protein discovery. Trends Pharmacol Sci 2025; 46:132-144. [PMID: 39755458 DOI: 10.1016/j.tips.2024.12.002] [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: 10/12/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025]
Abstract
Synthetic binding proteins (SBPs) are a class of protein binders that are artificially created and do not exist naturally. Their broad applications in tackling challenges of research, diagnostics, and therapeutics have garnered significant interest. Traditional protein engineering is pivotal to the discovery of SBPs. Recently, this discovery has been significantly accelerated by computational approaches, such as molecular modeling and artificial intelligence (AI). Furthermore, while numerous bioinformatics databases offer a wealth of resources that fuel SBP discovery, the full potential of these data has not yet been fully exploited. In this review, we present a comprehensive overview of SBP data ecosystem and methodologies in SBP discovery, highlighting the critical role of high-quality data and AI technologies in accelerating the discovery of innovative SBPs with promising applications in pharmacological sciences.
Collapse
Affiliation(s)
- Yanlin Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zixin Duan
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zhenwen Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China; Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Park, Chongqing 401329, China.
| |
Collapse
|
17
|
Deng C, Ma J, Liu Y, Tong X, Wang L, Dong J, Shi P, Wang M, Zheng W, Ma X. Targeting intracellular cancer proteins with tumor-microenvironment-responsive bispecific nanobody-PROTACs for enhanced therapeutic efficacy. MedComm (Beijing) 2025; 6:e70068. [PMID: 39830023 PMCID: PMC11742431 DOI: 10.1002/mco2.70068] [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: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Proteolysis targeting chimeras (PROTACs) are pivotal in cancer therapy for their ability to degrade specific proteins. However, their non-specificity can lead to systemic toxicity due to protein degradation in normal cells. To address this, we have integrated a nanobody into the PROTACs framework and leveraged the tumor microenvironment to enhance drug specificity. In this study, we engineered BumPeD, a novel bispecific nanobody-targeted PROTACs-like platform, by fusing two nanobodies with a Furin protease cleavage site (RVRR) and a degron sequence (ALAPYIP or KIGLGRQKPPKATK), enabling the tumor microenvironment to direct the degradation of intracellular proteins. We utilized KN035 and Nb4A to target PD-L1 (programmed death ligand 1) on the cell surface and intracellular Survivin, respectively. In vitro experiments showed that BumPeD triggers Survivin degradation via the ubiquitin-proteasome pathway, inducing tumor apoptosis and suppressing bladder tumor cell proliferation and migration. In vivo experiments further confirmed BumPeD's robust anti-tumor efficacy, underscoring its potential as a precise protein degradation strategy for cancer therapy. Our platform provides a systematic approach to developing effective and practical protein degraders, offering a targeted theoretical basis and experimental support for the development of novel degradative drugs, as well as new directions for cancer therapy.
Collapse
Affiliation(s)
- Changping Deng
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
- Key Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Center for Systems BiomedicineShanghai Jiao Tong UniversityShanghaiP. R. China
| | - Jiacheng Ma
- Department of Information EngineeringThe Chinese University of Hong KongHong KongP. R. China
| | - Yuping Liu
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghaiP. R. China
| | - Xikui Tong
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Lei Wang
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Jiayi Dong
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Ping Shi
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| | - Meiyan Wang
- School of MedicineShanghai UniversityShanghaiP. R. China
| | - Wenyun Zheng
- Shanghai Key Laboratory of New Drug DesignSchool of PharmacyEast China University of Science and TechnologyShanghaiP. R. China
| | - Xingyuan Ma
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghaiP. R. China
| |
Collapse
|
18
|
Ghosh P, Davies LJ, Nitsche C. Engineered Nanobodies Bind Bismuth, Indium and Gallium for Applications in Theranostics. Angew Chem Int Ed Engl 2025; 64:e202419455. [PMID: 39481115 DOI: 10.1002/anie.202419455] [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: 10/09/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/02/2024]
Abstract
Targeted theranostics heavily rely on metal isotopes conjugated to antibodies. Single-domain antibodies, known as nanobodies, are much smaller in size without compromising specificity and affinity. The conventional way of conjugating metals to nanobodies involves non-specific modification of amino acid residues with bifunctional chelating agents. We demonstrate that mutagenesis of a single residue in a nanobody creates a triple cysteine motif that selectively binds bismuth which is, for example, used in targeted alpha therapy. Two mutations create a quadruple cysteine mutant specific for gallium and indium used in positron emission tomography and single-photon emission computed tomography, respectively. Labelling is quantitative within a few minutes. The metal nanobodies maintain structural integrity and stability over weeks, resist competition from endogenous metal binders like glutathione, and retain functionality.
Collapse
Affiliation(s)
- Pritha Ghosh
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Lani J Davies
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
19
|
Truong DT, Chwastyk M, Phung VBT, Nguyen MT. Presence of EGF ligand restricts the binding ability of EgB4 nanobody to EGFR extracellular domain. Sci Rep 2025; 15:2420. [PMID: 39827297 PMCID: PMC11742981 DOI: 10.1038/s41598-025-86646-z] [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: 09/26/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
EgB4 is a nanobody that could facilitate the development of drug-nanobody conjugates or drug delivery in cancer treatment due to its specific binding ability to the EGFR transmembrane protein. More significantly, EgB4 does not hamper the EGFR function and associates with EGFR in both the presence and absence of an EGF ligand. However, the difference in EgB4-EGFR interaction with and without EGF ligand is not clear. To explore this aspect, we carried out computations including classical molecular dynamics simulations, steered molecular dynamics simulations and the umbrella sampling method. The three main objectives of the study are: (i) how EgB4 interacts with EGFR, (ii) the role of an EGF molecule, and (iii) how to mutate EgB4 to efficiently construct a stronger nanobody. Our computed results showed that, in addition to the residues asp98 and asp110 that were previously reported, two other residues, arg105 and asp108, are also highly interactive with the EGFR extracellular domain. Notably, the absolute binding free energy of the EgB4-EGFR complex decreases from - 17.1 to -13.3 kcal/mol in going from an absence to a presence of EGF. With the EGF presence, the interacting space between both EgB4 and EGFR entities is also reduced. When EGF stabilizes the dimerization of EGFR, there is less opportunity for EgB4 to bind. This crucial aspect has not been reported before.
Collapse
Affiliation(s)
- Duc Toan Truong
- Laboratory for Chemical Computation and Modeling, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, 70000, Vietnam
- Faculty of Applied Technology, School of Technology, Van Lang University, Ho Chi Minh City, 70000, Vietnam
| | - Mateusz Chwastyk
- Institute of Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Viet Bac T Phung
- Center for Environmental Intelligence and College of Engineering and Computer Science, VinUniversity, Gia Lam District, Hanoi, 10000, Vietnam.
| | - Minh Tho Nguyen
- Laboratory for Chemical Computation and Modeling, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, 70000, Vietnam.
- Faculty of Applied Technology, School of Technology, Van Lang University, Ho Chi Minh City, 70000, Vietnam.
| |
Collapse
|
20
|
Zhao S, Zeng W, Yu F, Xu P, Chen CY, Chen W, Dong Y, Wang F, Ma L. Visual and High-Efficiency Secretion of SARS-CoV-2 Nanobodies with Escherichia coli. Biomolecules 2025; 15:111. [PMID: 39858505 PMCID: PMC11762740 DOI: 10.3390/biom15010111] [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: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Nanobodies have gained attention as potential therapeutic and diagnostic agents for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) due to their ability to bind and neutralize the virus. However, rapid, scalable, and robust production of nanobodies for SARS-CoV-2 remains a crucial challenge. In this study, we developed a visual and high-efficiency biomanufacturing method for nanobodies with Escherichia coli by fusing the super-folder green fluorescent protein (sfGFP) to the N-terminus or C-terminus of the nanobody. Several receptor-binding domain (RBD)-specific nanobodies of the SARS-CoV-2 spike protein (S) were secreted onto the surface of E. coli cells and even into the culture medium, including Fu2, ANTE, mNb6, MR3-MR3, and n3113.1. The nanobodies secreted by E. coli retained equal activity as prior research, regardless of whether sfGFP was removed. Since some of the nanobodies bound to different regions of the RBD, we combined two nanobodies to improve the affinity. Fu2-sfGFP-ANTE was constructed to be bispecific for the RBD, and the bispecific nanobody exhibited significantly higher affinity than Fu2 (35.0-fold), ANTE (7.3-fold), and the combination of the two nanobodies (3.3-fold). Notably, Fu2-sfGFP-ANTE can be normally secreted into the culture medium and outer membrane. The novel nanobody production system enhances the efficiency of nanobody expression and streamlines the downstream purification process, enabling large-scale, cost-effective nanobody production. In addition, E. coli cells secreting the nanobodies on their surface facilitates screening and characterization of antigen-binding clones.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanming Dong
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China; (S.Z.); (W.Z.); (F.Y.); (P.X.); (C.-Y.C.); (W.C.); (F.W.)
| | | | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China; (S.Z.); (W.Z.); (F.Y.); (P.X.); (C.-Y.C.); (W.C.); (F.W.)
| |
Collapse
|
21
|
Knight K, Park JB, Oot RA, Khan MM, Roh SH, Wilkens S. Monoclonal nanobodies alter the activity and assembly of the yeast vacuolar H +-ATPase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632502. [PMID: 39829782 PMCID: PMC11741422 DOI: 10.1101/2025.01.10.632502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The vacuolar ATPase (V-ATPase; V1Vo) is a multi-subunit rotary nanomotor proton pump that acidifies organelles in virtually all eukaryotic cells, and extracellular spaces in some specialized tissues of higher organisms. Evidence suggests that metastatic breast cancers mislocalize V-ATPase to the plasma membrane to promote cell survival and facilitate metastasis, making the V-ATPase a potential drug target. We have generated a library of camelid single-domain antibodies (Nanobodies; Nbs) against lipid-nanodisc reconstituted yeast V-ATPase Vo proton channel subcomplex. Here, we present an in-depth characterization of three anti-Vo Nbs using biochemical and biophysical in vitro experiments. We find that the Nbs bind Vo with high affinity, with one Nb inhibiting holoenzyme activity and another one preventing enzyme assembly. Using cryoEM, we find that two of the Nbs bind the c subunit ring of the Vo on the lumen side of the complex. Additionally, we show that one of the Nbs raised against yeast Vo can pull down human V-ATPase (HsV1Vo). Our research demonstrates Nb versatility to target and modulate the activity of the V-ATPase, and highlights the potential for future therapeutic Nb development.
Collapse
Affiliation(s)
- Kassidy Knight
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Jun Bae Park
- Department of Biological Sciences, Seoul National University, Seoul, Korea
- Present address: Department of Cancer Biology, Lerner research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rebecca A. Oot
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Md. Murad Khan
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Present address: Howard Hughes Medical Institute, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Soung-Hun Roh
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
22
|
Gordon G, Greenshields-Watson A, Agarwal P, Wong A, Boyles F, Hummer A, Lujan Hernandez A, Deane C. PLAbDab-nano: a database of camelid and shark nanobodies from patents and literature. Nucleic Acids Res 2025; 53:D535-D542. [PMID: 39385626 PMCID: PMC11701533 DOI: 10.1093/nar/gkae881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/13/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
Nanobodies are essential proteins of the adaptive immune systems of camelid and shark species, complementing conventional antibodies. Properties such as their relatively small size, solubility and high thermostability make VHH (variable heavy domain of the heavy chain) and VNAR (variable new antigen receptor) modalities a promising therapeutic format and a valuable resource for a wide range of biological applications. The volume of academic literature and patents related to nanobodies has risen significantly over the past decade. Here, we present PLAbDab-nano, a nanobody complement to the Patent and Literature Antibody Database (PLAbDab). PLAbDab-nano is a self-updating, searchable repository containing ∼5000 annotated VHH and VNAR sequences. We describe the methods used to curate the entries in PLAbDab-nano, and highlight how PLAbDab-nano could be used to design diverse libraries, as well as find sequences similar to known patented or therapeutic entries. PLAbDab-nano is freely available as a searchable web server (https://opig.stats.ox.ac.uk/webapps/plabdab-nano/).
Collapse
Affiliation(s)
- Gemma L Gordon
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| | | | - Parth Agarwal
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| | - Ashley Wong
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| | - Fergus Boyles
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| | - Alissa Hummer
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| | | | - Charlotte M Deane
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| |
Collapse
|
23
|
Yi L, Liu H, Liu Y, Aiyisi, He J, Ming L, Jirimutu. Nanobody-based indirect competitive ELISA for the detection of aflatoxin M1 in dairy products. Sci Rep 2025; 15:785. [PMID: 39755714 PMCID: PMC11700155 DOI: 10.1038/s41598-024-83869-4] [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: 09/03/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
Aflatoxin M1 (AFM1) is known to be carcinogenic, mutagenic, and teratogenic and poses a serious threat to food safety and human health, which makes its surveillance critical. In this study, an indirect competitive ELISA (icELISA) based on a nanobody (Nb M4) was developed for the sensitive and rapid detection of AFM1 in dairy products. In our previous work, Nb M4 was screened from a Bactrian-camel-immunized phage-displayed library. It exhibits VH-like features, possesses higher thermal stability than monoclonal antibody (mAb 1E6) and tightly binds to AFM1-BSA with a KD value of 2.5 nM. Under the optimal conditions, its half-maximal inhibitory concentration was 0.338 ng/mL, the limit of detection was 0.051 ng/mL, and linearity was noted in the range of 0.168-0.679 ng/mL. Nb M4 displayed almost no cross-reactivity with other mycotoxins. No matrix effect was observed in milk and milk powder samples, and the matrix effect in yogurt samples could be weakened by 2-fold dilution. Furthermore, validation studies in spiked samples (milk, yogurt, and milk powder) resulted in good recoveries of 95.40-111.33%, with a low coefficient of variation (2.89-6.78%). High-performance liquid chromatography was used to evaluate the accuracy and reliability of the developed icELISA, which indicated a satisfactory consistent correlation (R2 = 0.9722). This study highlights the potential of Nb M4 as a promising component for detecting AFM1 in dairy products.
Collapse
Affiliation(s)
- Li Yi
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Haiyuan Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yingda Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Aiyisi
- Inner Mongolia Autonomous Region International Mongolian Hospital, Hohhot, 010018, China
| | - Jing He
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Liang Ming
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Jirimutu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
24
|
Tang AY, Jung S, Carrasco-López C, Avalos JL. Light-Induced Nanobody-Mediated Targeted Protein Degradation for Metabolic Flux Control. ACS Synth Biol 2024; 13:4110-4118. [PMID: 39527810 DOI: 10.1021/acssynbio.4c00552] [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] [Indexed: 11/16/2024]
Abstract
In metabolic engineering, increasing chemical production usually involves manipulating the expression levels of key enzymes. However, limited synthetic tools exist for modulating enzyme activity beyond the transcription level. Inspired by natural post-translational mechanisms, we present targeted enzyme degradation mediated by optically controlled nanobodies. We applied this method to a branched biosynthetic pathway, deoxyviolacein, and observed enhanced product specificity and yield. We then extend the biosynthesis pathway to violacein and show how simultaneous degradation of two target enzymes can further shift production profiles. Through the redirection of metabolic flux, we demonstrate how targeted enzyme degradation can be used to minimize unwanted intermediates and boost the formation of desired products.
Collapse
Affiliation(s)
- Allison Y Tang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Seyi Jung
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - César Carrasco-López
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - José L Avalos
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, New Jersey 08544, United States
- Andlinger Center for Energy and the Environment, Princeton University, Princeton, New Jersey 08544, United States
- High Meadows Environmental Institute, Princeton University, Princeton, New Jersey 08544, United States
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
25
|
Duan Z, Li D, Li N, Lin S, Ren H, Hong J, Hinrichs CS, Ho M. CAR-T cells based on a TCR mimic nanobody targeting HPV16 E6 exhibit antitumor activity against cervical cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200892. [PMID: 39524212 PMCID: PMC11546159 DOI: 10.1016/j.omton.2024.200892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/10/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
The E6 and E7 oncoproteins of human papillomavirus (HPV) are considered promising targets for HPV-related cancers. In this study, we evaluated novel T cell receptor mimic (TCRm) nanobodies targeting the E629-38 peptide complexed with human leukocyte antigen (HLA)-A∗02:01 in the chimeric antigen receptor (CAR) format. We isolated two dromedary camel nanobodies, F5 and G9, through phage display screening. F5 bound more efficiently to the complex expressed on cells, including peptide-pulsed T2, overexpressed 293E6, and cervical cancer lines CaSki and SS4050, compared to G9. CAR-T cells based on the F5 nanobody specifically killed target cells, including 293E6, CaSki, and SS4050 in vitro, through activation of nuclear factor of activated T cells (NFAT) and nuclear factor κB (NF-κB) signaling. Importantly, F5 CAR-T cells inhibited the growth of CaSki and SS4050 tumor xenografts in mice. These findings demonstrate that HPV-16+ cervical cancer can be targeted by F5 nanobody-based CAR-T cells, offering a valuable alternative strategy for treating HPV-16+ malignancies.
Collapse
Affiliation(s)
- Zhijian Duan
- Antibody Engineering Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shaoli Lin
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hua Ren
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jessica Hong
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Mitchell Ho
- Antibody Engineering Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Feng H, Sun X, Li N, Xu Q, Li Q, Zhang S, Xing G, Zhang G, Wang F. Machine Learning-Driven Methods for Nanobody Affinity Prediction. ACS OMEGA 2024; 9:47893-47902. [PMID: 39651108 PMCID: PMC11618429 DOI: 10.1021/acsomega.4c09718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024]
Abstract
Because of their high affinity, specificity, and environmental stability, nanobodies (Nbs) have continuously received attention from the field of biological research. However, it is tough work to obtain high-affinity Nbs using experimental methods. In the current study, 12 machine learning algorithms were compared in parallel to explore the potential patterns between Nb-ligand affinity and eight noncovalent interactions. After model comparison and optimization, four optimized models (SVMrB, RotFB, RFB, and C50B) and two stacked models (StackKNN and StackRF) based on nine uncorrelated (correlation coefficient <0.65) optimized models were selected. All the models showed an accuracy of around 0.70 and high specificity. Compared to the other models, RotFB and RFB were not capable of predicting nonaffinitive Nbs with lower precision (<0.44) but showed higher sensitivity at 0.6761 and 0.3521 and good model robustness (F1 score and MCC values). On the contrary, SVMrB, C50B, and StackKNN were able to effectively predict the future nonaffinitive Nbs (specificity >0.92) and reduce the number of true affinitive Nbs (precision >0.5). On the other hand, StackRF showed intermediate model performance. Furthermore, an in-depth feature analysis indicated that hydrogen bonding and aromatic-associated interactions were the key noncovalent interactions in determining Nb-ligand binding affinity. In summary, the current study provides, for the first time, a tool that can effectively predict whether there is an affinity between nanobodies and their intended ligands and explores the key factors that influence their affinity, which could improve the screening and design process of Nbs and accelerate the development of Nb drugs and applications.
Collapse
Affiliation(s)
- Hua Feng
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
- Longhu Laboratory, 218 Ping AN Avenue, Zhengzhou 450002, China
| | - Xuefeng Sun
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
| | - Ning Li
- College of
Food Science and Technology, Henan Agricultural
University, 218 Ping AN Avenue, Zhengzhou 450002, China
| | - Qian Xu
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
| | - Qin Li
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
| | - Shenli Zhang
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
| | - Guangxu Xing
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
| | - Gaiping Zhang
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
- School of
Advanced Agricultural Sciences, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
- Jiangsu Co-Innovation
Center for the Prevention and Control of Important Animal Infectious
Diseases and Zoonoses, Yangzhou University, 88 South Daxue Road, Yangzhou 225009, China
| | - Fangyu Wang
- Institute
for Animal Health, Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, 116 Huayuan Road, Zhengzhou 450002, China
- Longhu Laboratory, 218 Ping AN Avenue, Zhengzhou 450002, China
| |
Collapse
|
27
|
Minne RL, Luo NY, Mork CM, Wopat MR, Esbona K, Javeri S, Nickel KP, Hernandez R, LeBeau AM, Kimple RJ, Baschnagel AM. Evaluation of a Novel MET-Targeting Camelid-Derived Antibody in Head and Neck Cancer. Mol Pharm 2024; 21:6376-6384. [PMID: 39513517 PMCID: PMC11987585 DOI: 10.1021/acs.molpharmaceut.4c00938] [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] [Indexed: 11/15/2024]
Abstract
In head and neck squamous cell carcinoma (HNSCC), the mesenchymal epithelial transition (MET) receptor drives cancer growth, proliferation, and metastasis. MET is known to be overexpressed in HNSCC and, therefore, is an appealing therapeutic target. In this study, we evaluated MET expression in patients with HNSCC and investigated the potential imaging application of a novel MET-binding single-domain camelid antibody using positron emission tomography/computed tomography (PET/CT) in a preclinical MET-expressing HNSCC model. Multiplex immunostaining for MET protein performed on a tissue microarray from 203 patients with HNSCC found 86% of patients to have MET expression, with 14% having high expression and 53% having low MET expression. Using The Cancer Genome Atlas (TCGA) database, high MET RNA expression was associated with worse progression-free survival and overall survival in patients with HPV-negative HSNCC. Utilizing flow cytometry and immunofluorescence, our novel camelid antibody fused to a human IgG Fc chain (1E7-Fc) showed high binding affinity and specificity to high MET-expressing Detroit 562 cells but not to low MET-expressing HNSCC cells. The efficacy and biodistribution of [89Zr]Zr-1E7-Fc as a PET imaging agent was then investigated in a MET-expressing head and neck xenograft model. [89Zr]Zr-1E7-Fc rapidly localized and showed high tumor uptake in Detroit 562 xenografts (8.4% ID/g at 72 h postinjection), with rapid clearance from the circulatory system (2.7 tumor-to-blood radioactivity ratio at 72 h postinjection). Our preclinical data suggest that the camelid antibody 1E7-Fc could be a potential theranostic agent for HNSCC. Further investigations are warranted to confirm these findings in patients and to evaluate 1E7-Fc as an imaging agent and platform to deliver radionuclide or drug therapy to MET-driven cancers.
Collapse
Affiliation(s)
- Rachel L Minne
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Natalie Y Luo
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Caroline M Mork
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Madalynn R Wopat
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Karla Esbona
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Saahil Javeri
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Kwangok P Nickel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Reinier Hernandez
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Aaron M LeBeau
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| | - Andrew M Baschnagel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, United States
| |
Collapse
|
28
|
Kang L, Dong Y, Wang W, Li Z, Wang Y, Yan L, Yin C, Zhang X, Dai H, Wu B, Zhao H, Wang J. Identification of a novel anti-ROR1 nanobody through phage display and its biochemical characterization. Biotechnol Appl Biochem 2024; 71:1226-1234. [PMID: 39645564 DOI: 10.1002/bab.2623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/25/2024] [Indexed: 12/09/2024]
Abstract
In this study, we aimed to develop nanobodies targeting receptor tyrosine kinase-like orphan receptor 1 (ROR1) for cancer diagnosis and therapy. We immunized alpacas with ROR1, extracted RNA from their blood, and converted it to complementary DNA (cDNA) to amplify the VHH (variable domain of heavy-chain antibodies) sequence. This sequence was used to construct a phage library with a capacity of 8 ×108. Screening identified a high-affinity nanobody, HCAbs1, which binds effectively to ROR1. ELISA and surface plasmon resonance analyses revealed HCAbs1's binding affinities to ROR1 at 4.42 and 12.9 nM, respectively. Functional tests showed HCAbs1 could reduce extracellular signal-regulated kinase (ERK) phosphorylation levels induced by Wnt5a in ROR1-transfected cells. Our findings highlight the potential of HCAbs1 nanobodies in diagnosing and treating cancers through targeting ROR1.
Collapse
Affiliation(s)
- Li Kang
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Yingkui Dong
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Wanxue Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zehua Li
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Yizhuo Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- University of Science and Technology of China, Hefei, Anhui, China
| | - Li Yan
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Cunlong Yin
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - XiaoHui Zhang
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - Han Dai
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Bo Wu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Hongxin Zhao
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
- Hefei China Science Longwood Biological Technology Co., Ltd, Hefei, Anhui, China
| | - Junfeng Wang
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| |
Collapse
|
29
|
Liu Y, Wang H, Chiu CKC, Wu Y, Bi Y. 1H, 13C and 15N resonance assignments of a shark variable new antigen receptor against hyaluronan synthase. BIOMOLECULAR NMR ASSIGNMENTS 2024; 18:213-217. [PMID: 39141198 DOI: 10.1007/s12104-024-10190-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
Single domain antibody (sdAb) is only composed of a variable domain of the heavy-chain-only antibody, which is devoid of light chain and naturally occurring in camelids and cartilaginous fishes. Variable New Antigen Receptor (VNAR), a type of single domain antibody present in cartilaginous fishes such as sharks, is the smallest functional antigen-binding fragment found in nature. The unique features, including flexible paratope, high solubility and outstanding stability make VNAR a promising prospect in antibody drug development and structural biology research. However, VNAR's research has lagged behind camelid-derived sdAb, especially in the field of structural research. Here we report the 1H,15N,13C resonance assignments of a VNAR derived from the immune library of Chiloscyllium plagiosum, termed B2-3, which recognizes the hyaluronan synthase. Analysis of the backbone chemical shifts demonstrates that the secondary structure of VNAR is predominately composed of β-sheets corresponding to around 40% of the B2-3 backbone. The Cβ chemical shift values of cysteine residues, combined with mass spectrometry data, clearly shows that B2-3 contains two pairs of disulfide bonds, which is import for protein stability. The assignments will be essential for determining the high resolution solution structure of B2-3 by NMR spectroscopy.
Collapse
Affiliation(s)
- Yuxin Liu
- College of Life Sciences, Qingdao University, Qingdao, China
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Hao Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | | | - Yujie Wu
- Shenzhen Bay Laboratory, Shenzhen, China.
| | - Yunchen Bi
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
30
|
Yi L, Guo X, Liu Y, Jirimutu, Wang Z. Single-cell 5' RNA sequencing of camelid peripheral B cells provides insights into cellular basis of heavy-chain antibody production. Comput Struct Biotechnol J 2024; 23:1705-1714. [PMID: 38689719 PMCID: PMC11059136 DOI: 10.1016/j.csbj.2024.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Camelids produce both conventional tetrameric antibodies (Abs) and dimeric heavy-chain antibodies (HCAbs). Although B cells that generate these two types of Abs exhibit distinct B cell receptors (BCRs), whether these two B cell populations differ in their phenotypes and developmental processes remains unclear. Here, we performed single-cell 5' RNA profiling of peripheral blood mononuclear cell samples from Bactrian camels before and after immunization. We characterized the functional subtypes and differentiation trajectories of circulating B cells in camels, and reconstructed single-cell BCR sequences. We found that in contrast to humans, the proportion of T-bet+ B cells was high among camelid peripheral B cells. Several marker genes of human B cell subtypes, including CD27 and IGHD, were expressed at low levels in the corresponding camel B cell subtypes. Camelid B cells expressing variable genes of HACbs (VHH) were widely present in various functional subtypes and showed highly overlapping differentiation trajectories with B cells expressing variable genes of conventional Abs (VH). After immunization, the transcriptional changes in VHH+ and VH+ B cells were largely consistent. Through structure modeling, we identified a variety of scaffold types among the reconstructed VHH sequences. Our study provides insights into the cellular context of HCAb production in camels and lays the foundation for developing single-B cell-based camelid single-domain Ab screening.
Collapse
Affiliation(s)
- Li Yi
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot 010018, China
| | - Xin Guo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuexing Liu
- Guangzhou Laboratory, Guangzhou 510005, China
| | - Jirimutu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot 010018, China
- Inner Mongolia China-Kazakhstan Camel Research Institute, Alxa 750306, China
| | - Zhen Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
31
|
Chen Y, Huang Y, Yang R, Cui Y, Wang Y, Wang X, Wang J. Preparation of divalent camelid single-domain antibody and its application in immunoassays for Salmonella detection in food. Anal Bioanal Chem 2024; 416:7063-7072. [PMID: 39460771 DOI: 10.1007/s00216-024-05610-4] [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: 07/11/2024] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Salmonella-related foodborne infections are commonly caused by the serovars of S. Typhimurium, which can be detected using antibody-based immunoassays. The monovalent variable domain of the camelid heavy chain antibody (VHH) performs excellently in constructing multivalent VHH variants, which generally exhibit higher affinities with antigens and consequently enhance the assay sensitivity. In this study, the divalent variants of VHHs (diVHHs) targeting S. Typhimurium were generated by encoding the monovalent VHH genes assembled in tandem with a flexible linker peptide (G4S)2. Soluble diVHHs were produced in a prokaryotic expression system and purified with a yield of 4.22 mg/L. Benefiting from their stability and antigen-binding abilities towards tested Salmonella serovars, diVHH-based immunoassays were developed. The diVHH-based sandwich immunoassay, using diVHH as capture antibody, exhibited a detection limit of 1.04×102 CFU/mL and enabled as low as 10 CFU/mL S. Typhimurium to be detected after 6 h of enrichment in lettuce. Furthermore, this assay can be applied to spiked lettuce, chicken, and pork samples, showing acceptable recoveries ranging from 83 to 106%. This study presented feasible strategies for VHH multivalence and established a superior sensitivity VHH-based immunoassay for monitoring and analyzing Salmonella contamination in food.
Collapse
Affiliation(s)
- Yanhong Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yingjie Huang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Ruo Yang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yongliang Cui
- Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Yanru Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaohong Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jia Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
32
|
Fridy PC, Rout MP, Ketaren NE. Nanobodies: From High-Throughput Identification to Therapeutic Development. Mol Cell Proteomics 2024; 23:100865. [PMID: 39433212 PMCID: PMC11609455 DOI: 10.1016/j.mcpro.2024.100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024] Open
Abstract
The camelid single-domain antibody fragment, commonly referred to as a nanobody, achieves the targeting power of conventional monoclonal antibodies (mAbs) at only a fraction of their size. Isolated from camelid species (including llamas, alpacas, and camels), their small size at ∼15 kDa, low structural complexity, and high stability compared with conventional antibodies have propelled nanobody technology into the limelight of biologic development. Nanobodies are proving themselves to be a potent complement to traditional mAb therapies, showing success in the treatment of, for example, autoimmune diseases and cancer, and more recently as therapeutic options to treat infectious diseases caused by rapidly evolving biological targets such as the SARS-CoV-2 virus. This review highlights the benefits of applying a proteomic approach to identify diverse nanobody sequences against a single antigen. This proteomic approach coupled with conventional yeast/phage display methods enables the production of highly diverse repertoires of nanobodies able to bind the vast epitope landscape of an antigen, with epitope sampling surpassing that of mAbs. Additionally, we aim to highlight recent findings illuminating the structural attributes of nanobodies that make them particularly amenable to comprehensive antigen sampling and to synergistic activity-underscoring the powerful advantage of acquiring a large, diverse nanobody repertoire against a single antigen. Lastly, we highlight the efforts being made in the clinical development of nanobodies, which have great potential as powerful diagnostic reagents and treatment options, especially when targeting infectious disease agents.
Collapse
Affiliation(s)
- Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Natalia E Ketaren
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
33
|
Jiang F, Li M, Dong J, Yu Y, Sun X, Wu B, Huang J, Kang L, Pei Y, Zhang L, Wang S, Xu W, Xin J, Ouyang W, Fan G, Zheng L, Tan Y, Hu Z, Xiong Y, Feng Y, Yang G, Liu Q, Song J, Liu J, Hong L, Tan P. A general temperature-guided language model to design proteins of enhanced stability and activity. SCIENCE ADVANCES 2024; 10:eadr2641. [PMID: 39602544 PMCID: PMC11601203 DOI: 10.1126/sciadv.adr2641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024]
Abstract
Designing protein mutants with both high stability and activity is a critical yet challenging task in protein engineering. Here, we introduce PRIME, a deep learning model, which can suggest protein mutants with improved stability and activity without any prior experimental mutagenesis data for the specified protein. Leveraging temperature-aware language modeling, PRIME demonstrated superior predictive ability compared to current state-of-the-art models on the public mutagenesis dataset across 283 protein assays. Furthermore, we validated PRIME's predictions on five proteins, examining the impact of the top 30 to 45 single-site mutations on various protein properties, including thermal stability, antigen-antibody binding affinity, and the ability to polymerize nonnatural nucleic acid or resilience to extreme alkaline conditions. More than 30% of PRIME-recommended mutants exhibited superior performance compared to their premutation counterparts across all proteins and desired properties. We developed an efficient and effective method based on PRIME to rapidly obtain multisite mutants with enhanced activity and stability. Hence, PRIME demonstrates broad applicability in protein engineering.
Collapse
Affiliation(s)
- Fan Jiang
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingchen Li
- Shanghai Artificial Intelligence Laboratory, Shanghai 200030, China
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai 200240, China
| | - Jiajun Dong
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, China
| | - Yuanxi Yu
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinyu Sun
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230001, China
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Banghao Wu
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Life Sciences and Biotechnology, & State Key Laboratory of Microbial Metabolism, & Joint International Research Laboratory of Metabolic, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jin Huang
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Life Sciences and Biotechnology, & State Key Laboratory of Microbial Metabolism, & Joint International Research Laboratory of Metabolic, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liqi Kang
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yufeng Pei
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Liang Zhang
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shaojie Wang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wenxue Xu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jingyao Xin
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wanli Ouyang
- Shanghai Artificial Intelligence Laboratory, Shanghai 200030, China
| | - Guisheng Fan
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai 200240, China
| | - Lirong Zheng
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yang Tan
- Shanghai Artificial Intelligence Laboratory, Shanghai 200030, China
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai 200240, China
| | | | - Yi Xiong
- School of Life Sciences and Biotechnology, & State Key Laboratory of Microbial Metabolism, & Joint International Research Laboratory of Metabolic, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan Feng
- School of Life Sciences and Biotechnology, & State Key Laboratory of Microbial Metabolism, & Joint International Research Laboratory of Metabolic, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guangyu Yang
- School of Life Sciences and Biotechnology, & State Key Laboratory of Microbial Metabolism, & Joint International Research Laboratory of Metabolic, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Key Biological Raw Material, Shanghai Academy of Experimental Medicine, Shanghai 201401, China
- Hzymes Biotechnology Co. Ltd, Wuhan, Hubei 430075, China
| | - Qian Liu
- School of Life Sciences and Biotechnology, & State Key Laboratory of Microbial Metabolism, & Joint International Research Laboratory of Metabolic, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Song
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Liang Hong
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200030, China
- Zhanjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Pan Tan
- School of Physics and Astronomy, & Shanghai National Center for Applied Mathematics (SJTU Center), & Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200030, China
| |
Collapse
|
34
|
Zhang Y, Tian C, Yu X, Yu G, Han X, Wang Y, Zhou H, Zhang S, Li M, Yang T, Sun Y, Tai W, Yin Q, Zhao G. Lung-Selective Delivery of mRNA-Encoding Anti-MERS-CoV Nanobody Exhibits Neutralizing Activity Both In Vitro and In Vivo. Vaccines (Basel) 2024; 12:1315. [PMID: 39771977 PMCID: PMC11680347 DOI: 10.3390/vaccines12121315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a highly pathogenic virus causing severe respiratory illness, with limited treatment options that are mostly supportive. The success of mRNA technology in COVID-19 vaccines has opened avenues for antibody development against MERS-CoV. mRNA-based antibodies, expressed in vivo, offer rapid adaptability to viral mutations while minimizing long-term side effects. This study aimed to develop a lung-targeted lipid nanoparticle (LNP) system for mRNA-encoding neutralizing nanobodies against MERS-CoV, proposing a novel therapeutic strategy. Methods: An mRNA-encoding nanobody NbMS10 (mRNA-NbMS10) was engineered for enhanced stability and reduced immunogenicity. This mRNA was encapsulated in lung-selective LNPs using microfluidics to form the LNP-mRNA-NbMS10 system. Efficacy was assessed through in vitro assays and in vivo mouse studies, focusing on antigen-binding, neutralization, and sustained nanobody expression in lung tissues. Results: The LNP-mRNA-NbMS10 system expressed the nanobody in vitro, showing strong antigen-binding and significant MERS-CoV pseudovirus neutralization. In vivo studies confirmed selective lung mRNA delivery, with high nanobody expression sustained for up to 24 h, confirming lung specificity and prolonged antiviral activity. Conclusions: Extensive in vitro and in vivo evaluations demonstrate the LNP-mRNA-NbMS10 system's potential as a scalable, cost-effective, and adaptable alternative to current MERS-CoV therapies. This innovative platform offers a promising solution for preventing and treating respiratory infections, and countering emerging viral threats.
Collapse
Affiliation(s)
- Yuhang Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Y.Z.); (H.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
| | - Chongyu Tian
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China;
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030031, China
| | - Xinyang Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China; (X.Y.); (G.Y.)
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China; (X.Y.); (G.Y.)
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yuan Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Haisheng Zhou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Y.Z.); (H.Z.)
| | - Shuai Zhang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China;
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Tiantian Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Yali Sun
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Public Health School, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, China;
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Guangyu Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Y.Z.); (H.Z.)
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing 100071, China; (X.H.); (Y.W.); (M.L.); (T.Y.); (Y.S.)
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
35
|
Meng Q, Li H, Zhao W, Song M, Zhang W, Li X, Chen J, Wang L. Overcoming Debye screening effect in field-effect transistors for enhanced biomarker detection sensitivity. NANOSCALE 2024; 16:20864-20884. [PMID: 39452895 DOI: 10.1039/d4nr03481c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Field-effect transistor (FET)-based biosensors not only enable label-free detection by measuring the intrinsic charges of biomolecules, but also offer advantages such as high sensitivity, rapid response, and ease of integration. This enables them to play a significant role in disease diagnosis, point-of-care detection, and drug screening, among other applications. However, when FET sensors detect biomolecules in physiological solutions (such as whole blood, serum, etc.), the charged molecules will be surrounded by oppositely charged ions in the solution. This causes the effective charge carried by the biomolecules to be shielded, thereby significantly weakening their ability to induce charge rearrangement at the sensing interface. Such shielding hinders the change of carriers inside the sensing material, reduces the variation of current between the source and drain electrodes of the FET, and seriously limits the sensitivity and reliability of the device. In this article, we summarize the research progress in overcoming the Debye screening effect in FET-based biosensors over the past decade. Here, we first elucidate the working principles of FET sensors for detecting biomarkers and the mechanism of the Debye screening. Subsequently, we emphasize optimization strategies to overcome the Debye screening effect. Finally, we summarize and provide an outlook on the research on FET biosensors in overcoming the Debye screening effect, hoping to help the development of FET electronic devices with high sensitivity, specificity, and stability. This work is expected to provide new ideas for next-generation biosensing technology.
Collapse
Affiliation(s)
- Qi Meng
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Huimin Li
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Weilong Zhao
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Ming Song
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
36
|
Zhou L, Zhou H, Wang P, Xu H, Wu J, Zhou Y, Feng J, Zheng W. Construction of engineered probiotic that adhere and display nanobody to neutralize porcine reproductive and respiratory syndrome virus. Arch Microbiol 2024; 206:466. [PMID: 39540965 DOI: 10.1007/s00203-024-04198-8] [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: 09/11/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Pathogenic blue ear disease caused by porcine reproductive and respiratory syndrome virus (PRRSV) bring severe loss to breeding industry due to high infectivity and mortality. L. plantarum serves as the probiotic host strain, known for its beneficial properties in the gut microbiota. E. coli is used as a cloning host for the initial genetic engineering steps, facilitating the construction and amplification of the desired genetic constructs. In this study, using synthetic biology technology, we constructed engineered probiotics which could adhere and display nanobody on the surface to neutralize virus. Firstly, we screen an optimal nanobody to effectively bind with PRRSV by building library, expression and purification. Then, the integration of adhesion protein and nanobody into the genome of probiotics significantly improved its adhesion to IPEC-J2 cells. In addition, this engineered probiotic is almost non-toxic to cells with good safety, which can be used as a daily probiotics to prevent virus fecal transmission. Our study proposed this novel construction strategy of engineering probiotics with both adhesion and neutralization effects, which provided a new therapeutic view for intestinal virus clearance.
Collapse
Affiliation(s)
- Li Zhou
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Hanlin Zhou
- College of Life Science and Technology, Mudanjiang Normal University, 191 Wenhua Street, Mudanjiang, 157011, China
- College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luo Yu Road, Wuhan, 430074, China
- Liangzhun (Wuhan) Life Science & Technology Co. Ltd., 666 Gaoxin Avenue, Wuhan, 430073, China
| | - Panying Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Hang Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Affiliated to Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, P. R. China
| | - Jiayi Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Yuanzhuo Zhou
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Jiaying Feng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Weiyi Zheng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China.
| |
Collapse
|
37
|
Chen P, Yang Q, Li S, Liu X, Li B, Zhang J, Wang J, Yue X, Wang Y. Nanobody-Induced Aggregation of Gold Nanoparticles: A Mix-and-Read Strategy for the Rapid Detection of Cronobacter sakazakii. Anal Chem 2024; 96:17602-17611. [PMID: 39445484 DOI: 10.1021/acs.analchem.4c03463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Protein-nanoparticle interactions play a crucial role in both biomedical applications and the biosafety assessment of nanomaterials. Here, we found that nanobodies can induce citrate-capped gold nanoparticles (AuNPs) to aggregate into large clusters. Subsequently, we explored the mechanism behind this aggregation and proposed the "gold nucleation mechanism" to explain this phenomenon. Building on this observation, we developed a one-step label-free colorimetric method based on nanobody-induced AuNP aggregation. When nanobodies bind to target bacteria, spatial hindrance occurs, preventing further AuNPs aggregation. This alteration in surface plasmon resonance properties results in visible color changes. As an example, we present a simple and sensitive "mix-and-read" chromogenic immunosensor for Cronobacter sakazakii (C. sakazakii). The experiment can be completed within 20 min, with a visual detection limit of 103 CFU/mL and a quantitative detection limit of 136 CFU/mL. Importantly, our method exhibits no cross-reactivity with other bacterial species. This strategy harnesses the excellent properties of nanobodies and the optical characteristics of AuNPs for direct and rapid detection of foodborne pathogen.
Collapse
Affiliation(s)
- Pengyu Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qinkai Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shurong Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xuan Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Bingzhi Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jiahao Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaoyue Yue
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, Henan Province 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control (Zhengzhou University of Light Industry), Ministry of Education, Zhengzhou 450001, China
| | - Yanru Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
38
|
Culkins C, Adomanis R, Phan N, Robinson B, Slaton E, Lothrop E, Chen Y, Kimmel BR. Unlocking the Gates: Therapeutic Agents for Noninvasive Drug Delivery Across the Blood-Brain Barrier. Mol Pharm 2024; 21:5430-5454. [PMID: 39324552 DOI: 10.1021/acs.molpharmaceut.4c00604] [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] [Indexed: 09/27/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective network of various cell types that acts as a filter between the blood and the brain parenchyma. Because of this, the BBB remains a major obstacle for drug delivery to the central nervous system (CNS). In recent years, there has been a focus on developing various modifiable platforms, such as monoclonal antibodies (mAbs), nanobodies (Nbs), peptides, and nanoparticles, as both therapeutic agents and carriers for targeted drug delivery to treat brain cancers and diseases. Methods for bypassing the BBB can be invasive or noninvasive. Invasive techniques, such as transient disruption of the BBB using low pulse electrical fields and intracerebroventricular infusion, lack specificity and have numerous safety concerns. In this review, we will focus on noninvasive transport mechanisms that offer high levels of biocompatibility, personalization, specificity and are regarded as generally safer than their invasive counterparts. Modifiable platforms can be designed to noninvasively traverse the BBB through one or more of the following pathways: passive diffusion through a physio-pathologically disrupted BBB, adsorptive-mediated transcytosis, receptor-mediated transcytosis, shuttle-mediated transcytosis, and somatic gene transfer. Through understanding the noninvasive pathways, new applications, including Chimeric Antigen Receptors T-cell (CAR-T) therapy, and approaches for drug delivery across the BBB are emerging.
Collapse
Affiliation(s)
- Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Roman Adomanis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Slaton
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
39
|
Yu T, Zheng F, He W, Muyldermans S, Wen Y. Single domain antibody: Development and application in biotechnology and biopharma. Immunol Rev 2024; 328:98-112. [PMID: 39166870 PMCID: PMC11659936 DOI: 10.1111/imr.13381] [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] [Indexed: 08/23/2024]
Abstract
Heavy-chain antibodies (HCAbs) are a unique type of antibodies devoid of light chains, and comprised of two heavy chains-only that recognize their cognate antigen by virtue of a single variable domain also referred to as VHH, single domain antibody (sdAb), or nanobody (Nb). These functional HCAbs, serendipitous discovered about three decades ago, are exclusively found in camelids, comprising dromedaries, camels, llamas, and vicugnas. Nanobodies have become an essential tool in biomedical research and medicine, both in diagnostics and therapeutics due to their beneficial properties: small size, high stability, strong antigen-binding affinity, low immunogenicity, low production cost, and straightforward engineering into more potent affinity reagents. The occurrence of HCAbs in camelids remains intriguing. It is believed to be an evolutionary adaptation, equipping camelids with a robust adaptive immune defense suitable to respond to the pressure from a pathogenic invasion necessitating a more profound antigen recognition and neutralization. This evolutionary innovation led to a simplified HCAb structure, possibly supported by genetic mutations and drift, allowing adaptive mutation and diversification in the heavy chain variable gene and constant gene regions. Beyond understanding their origins, the application of nanobodies has significantly advanced over the past 30 years. Alongside expanding laboratory research, there has been a rapid increase in patent application for nanobodies. The introduction of commercial nanobody drugs such as Cablivi, Nanozora, Envafolimab, and Carvykti has boosted confidence among in their potential. This review explores the evolutionary history of HCAbs, their ontogeny, and applications in biotechnology and pharmaceuticals, focusing on approved and ongoing medical research pipelines.
Collapse
Affiliation(s)
- Ting Yu
- Center for Microbiome Research of Med‐X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated HospitalXi'an Jiaotong UniversityXi'anChina
| | - Fang Zheng
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science CenterXi'an Jiaotong UniversityXi'anChina
| | - Wenbo He
- Center for Microbiome Research of Med‐X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated HospitalXi'an Jiaotong UniversityXi'anChina
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Yurong Wen
- Center for Microbiome Research of Med‐X Institute, Shaanxi Provincial Key Laboratory of Sepsis in Critical Care Medicine, The First Affiliated HospitalXi'an Jiaotong UniversityXi'anChina
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science CenterXi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
40
|
Hu Y, Huo L, Chen W, Shen J, Wang W. Resistance-based directed evolution of nanobodies for higher affinity in prokaryotes. Biochim Biophys Acta Gen Subj 2024; 1868:130710. [PMID: 39245149 DOI: 10.1016/j.bbagen.2024.130710] [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: 08/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
A prokaryotic resistance-based directed evolution system leveraging protein-fragment complementation assay (PCA) was devised, and its proficiency in detecting protein-protein interactions and discriminating varying degrees of binding affinity was demonstrated by two well-characterized protein pairs. Furthermore, we constructed a random mutant library based on the GBPR36K/E45K mutant, characterized by almost no affinity towards EGFP. This library was subjected to PCA-based prokaryotic directed evolution, resulting in the isolation of back-mutated variants. In summary, we have established an expedited, cost-effective, and structural information-independent PCA-based prokaryotic directed evolution platform for nanobody affinity maturation, featuring tunable screening stringency via modulation of antibiotic concentrations.
Collapse
Affiliation(s)
- Yue Hu
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China & Key Laboratory for Biotechnology of the State Ethnic Affairs Commission, College of Life Sciences, South-Central Minzu University, Wuhan, Hubei 430074, PR China
| | - Li Huo
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China & Key Laboratory for Biotechnology of the State Ethnic Affairs Commission, College of Life Sciences, South-Central Minzu University, Wuhan, Hubei 430074, PR China
| | - Weiwei Chen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China & Key Laboratory for Biotechnology of the State Ethnic Affairs Commission, College of Life Sciences, South-Central Minzu University, Wuhan, Hubei 430074, PR China
| | - Jinhua Shen
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China & Key Laboratory for Biotechnology of the State Ethnic Affairs Commission, College of Life Sciences, South-Central Minzu University, Wuhan, Hubei 430074, PR China.
| | - Wenyi Wang
- Institute for Medical Biology & Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China & Key Laboratory for Biotechnology of the State Ethnic Affairs Commission, College of Life Sciences, South-Central Minzu University, Wuhan, Hubei 430074, PR China.
| |
Collapse
|
41
|
Sun B, Li G, Wu Y, Gai J, Zhu M, Ji W, Wang X, Zhang F, Li W, Hu J, Lou Y, Feng G, Han X, Dong J, Peng J, Pei J, Wan Y, Li Y, Ma L. Ce-MOF@Au-Based Electrochemical Immunosensor for Apolipoprotein A1 Detection Using Nanobody Technology. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58405-58416. [PMID: 39413767 DOI: 10.1021/acsami.4c14027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Apolipoprotein A1 (Apo-A1) is a well-recognized biomarker in tissues, closely associated with cardiovascular diseases such as atherosclerosis, coronary artery disease, and heart failure. However, existing methods for Apo-A1 determination are limited by costly equipment and intricate operational procedures. Given the distinct advantages of electrochemical immunosensors, including affordability and high sensitivity, along with the unique attributes of nanobodies (Nbs), such as enhanced specificity and better tissue permeability, we developed an electrochemical immunosensor for Apo-A1 detection utilizing Nb technology. In our study, Ce-MOF@AuNPs nanocomposites were synthesized by using ultrasonic methods and applied to modify a glassy carbon electrode. The Nb6, screened from an Apo-A1 immunized phage library, was immobilized onto the nanocomposite material, establishing a robust binding interaction with Apo-A1. The recorded peak current values demonstrated a logarithmic increase corresponding to Apo-A1 concentrations ranging from 1 to 100,000 pg/mL, with a detection limit of 36 fg/mL. Additionally, the developed immunosensors demonstrated high selectivity, good stability, and reproducibility. Our methodology was also effectively utilized for serum sample analysis, showing good performance in clinical assessments. This electrochemical immunosensor represents a promising tool for Apo-A1 detection, with significant potential for advancing cardiovascular disease diagnostics.
Collapse
Affiliation(s)
- Baihe Sun
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Guanghui Li
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai 201318, China
| | - Yue Wu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Junwei Gai
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai 201318, China
| | - Min Zhu
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai 201318, China
| | - Weiwei Ji
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai 201318, China
| | - Xiaoying Wang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Fenghua Zhang
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Wanting Li
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jingjin Hu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Yuxin Lou
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Gusheng Feng
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Xijun Han
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jinwen Dong
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jiayuan Peng
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jiawei Pei
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai 201318, China
| | - Yanfei Li
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Linlin Ma
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| |
Collapse
|
42
|
Alexander E, Leong KW. Discovery of nanobodies: a comprehensive review of their applications and potential over the past five years. J Nanobiotechnology 2024; 22:661. [PMID: 39455963 PMCID: PMC11515141 DOI: 10.1186/s12951-024-02900-y] [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: 06/19/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Nanobodies (Nbs) are antibody fragments derived from heavy-chain-only IgG antibodies found in the Camelidae family as well as cartilaginous fish. Their unique structural and functional properties, such as their small size, the ability to be engineered for high antigen-binding affinity, stability under extreme conditions, and ease of production, have made them promising tools for diagnostics and therapeutics. This potential was realized in 2018 with the approval of caplacizumab, the world's first Nb-based drug. Currently, Nbs are being investigated in clinical trials for a broad range of treatments, including targeted therapies against PDL1 and Epidermal Growth Factor Receptor (EGFR), cardiovascular diseases, inflammatory conditions, and neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. They are also being studied for their potential for detecting and imaging autoimmune conditions and infectious diseases such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A variety of methods are now available to generate target-specific Nbs quickly and efficiently at low costs, increasing their accessibility. This article examines these diverse applications of Nbs and their promising roles. Only the most recent articles published in the last five years have been used to summarize the most advanced developments in the field.
Collapse
Affiliation(s)
- Elena Alexander
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| |
Collapse
|
43
|
Zhu M, Ma L, Zhong P, Huang J, Gai J, Li G, Li Y, Qiao P, Gu H, Li X, Yin Y, Zhang L, Deng Z, Sun B, Chen Z, Ding Y, Wan Y. A novel inhalable nanobody targeting IL-4Rα for the treatment of asthma. J Allergy Clin Immunol 2024; 154:1008-1021. [PMID: 38871183 DOI: 10.1016/j.jaci.2024.05.027] [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: 11/20/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Inhalable biologics represent a promising approach to improve the efficacy and safety of asthma treatment. Although several mAbs targeting IL-4 receptor α chain (IL-4Rα) have been approved or are undergoing clinical trials, the development of inhalable mAbs targeting IL-4Rα presents significant challenges. OBJECTIVE Capitalizing on the distinctive advantages of nanobodies (Nbs) in maintaining efficacy during storage and administration, we sought to develop a novel inhalable IL-4Rα Nb for effectively treating asthma. METHODS Three IL-4Rα immunized Nb libraries were used to generate specific and functional IL-4Rα Nbs. LQ036, a bivalent Nb comprising 2 HuNb103 units, was constructed with a high affinity and specificity for human IL-4Rα. The efficacy, pharmacokinetics, and safety of inhaled LQ036 were evaluated in B-hIL4/hIL4RA humanized mice. RESULTS LQ036 inhibited secreted embryonic alkaline phosphatase reporter activity, inhibited TF-1 cell proliferation, and suppressed phosphorylated signal transducer and activator of transduction 6 in T cells from patients with asthma. Crystal structure analysis revealed a binding region similar to dupilumab but with higher affinity, leading to better efficacy in blocking the signaling pathway. HuNb103 competed with IL-4 and IL-13 for IL-4Rα binding. Additionally, LQ036 significantly inhibited ovalbumin-specific IgE levels in serum, CCL17 levels in bronchoalveolar lavage fluid, bronchial mucous cell hyperplasia, and airway goblet cell hyperplasia in B-hIL4/hIL4RA humanized mice. Inhaled LQ036 exhibited favorable pharmacokinetics, safety, and tissue distribution, with higher concentrations observed in the lungs and bronchi. CONCLUSIONS These findings from preclinical studies establish the safety and efficacy of inhaled LQ036, underscoring its potential as a pioneering inhalable biologic therapy for asthma.
Collapse
Affiliation(s)
- Min Zhu
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Linlin Ma
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Peiyu Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Huang
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Junwei Gai
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Guanghui Li
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Yanfei Li
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Peng Qiao
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Huaiyu Gu
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Xiaofei Li
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Children's Medical Center Pediatric Medical Complex (Pudong), Shanghai, China; Pediatric AI Clinical Application and Research Center, Shanghai Children's Medical Center, Shanghai, China
| | - Lei Zhang
- Department of Respiratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenzhen Deng
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China
| | - Baihe Sun
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine of Zhongshan Hospital, Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China.
| | - Yu Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yakun Wan
- Shanghai Novamab Biopharmaceuticals Co, Ltd, Shanghai, China.
| |
Collapse
|
44
|
Schnider ST, Vigano MA, Affolter M, Aguilar G. Functionalized Protein Binders in Developmental Biology. Annu Rev Cell Dev Biol 2024; 40:119-142. [PMID: 39038471 DOI: 10.1146/annurev-cellbio-112122-025214] [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] [Indexed: 07/24/2024]
Abstract
Developmental biology has greatly profited from genetic and reverse genetic approaches to indirectly studying protein function. More recently, nanobodies and other protein binders derived from different synthetic scaffolds have been used to directly dissect protein function. Protein binders have been fused to functional domains, such as to lead to protein degradation, relocalization, visualization, or posttranslational modification of the target protein upon binding. The use of such functionalized protein binders has allowed the study of the proteome during development in an unprecedented manner. In the coming years, the advent of the computational design of protein binders, together with further advances in scaffold engineering and synthetic biology, will fuel the development of novel protein binder-based technologies. Studying the proteome with increased precision will contribute to a better understanding of the immense molecular complexities hidden in each step along the way to generate form and function during development.
Collapse
Affiliation(s)
| | | | | | - Gustavo Aguilar
- Current affiliation: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
- Biozentrum, Universität Basel, Basel, Switzerland;
| |
Collapse
|
45
|
Jiang D, Chen R, Wang L, Xu G. Screening, expression and anti-tumor functional identification of anti-LAG-3 nanobodies. Protein Expr Purif 2024; 222:106522. [PMID: 38851552 DOI: 10.1016/j.pep.2024.106522] [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: 04/22/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE To screen and obtain specific anti-lymphocyte activation gene-3 (LAG3) nanobody sequences, purify and express recombinant anti-LAG3 nanobody, and verify its effect on promoting T cells to kill tumor cells. METHODS Based on the camel derived natural nanobody phage display library constructed by the research group, the biotinylated LAG3 antigen was used as the target, and the anti-LAG3 nanobody sequences were screened by biotin-streptavidin liquid phase screening, phage-ELISA and sequencing. The sequence-conjμgated human IgG1 Fc fragment was obtained, the recombinant anti-LAG3 nanobody expression vector was constructed, the expression of the recombinant anti-LAG3 nanobody was induced by IPTG and purified, and the characteristics and functions of the recombinant anti-LAG3 nanobody were verified by SDS-PAGE, Western blot, cytotoxicity assay, etc. RESULTS: One anti-LAG3 nanobody sequence was successfully screened, and the corresponding recombinant anti-LAG3 nanobody-expressing bacteria were constructed. The results of SDS-PAGE, Western blot and cytotoxicity assay showed that the recombinant anti-LAG3 nanobody was successfully expressed, which was specific, and it could promote the killing ability of T cells against tumor cells, and the optimal concentration was 200 μg/mL. CONCLUSION The recombinant anti-LAG3 nanobody screened and expressed has specific and auxiliary anti-tumor cell effects, which lays a foundation for its subsequent application.
Collapse
Affiliation(s)
- Dan Jiang
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Rui Chen
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China
| | - Liyan Wang
- Ningxia Chinese Medicine Research Centre, Yinchuan, Ningxia, 750000, China
| | - Guangxian Xu
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, 523808, China.
| |
Collapse
|
46
|
Zhang Y, Guo Y, Song L, Liu W, Nian R, Fan X. Streamlined on-column refolding and purification of nanobodies from inclusion bodies expressed as fusion proteins. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1246:124279. [PMID: 39186888 DOI: 10.1016/j.jchromb.2024.124279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
This study introduces an efficient on-column refolding and purification method for preparing nanobodies (Nbs) expressed as inclusion bodies and fusion proteins. The HisTrapTM FF system was successfully employed for the purification of the fusion protein FN1-ΔI-CM-2D5. The intein ΔI-CM cleavage activity was activated at 42 °C, followed by incubation for 4 h. Leveraging the remarkable thermal stability of Nbs, 2D5 was further purified through heat treatment at 80 °C for 1h. This method yielded up to 107.2 mg of pure 2D5 with a purity of 99.2 % from just 1L of bacterial culture grown in a shaker flask. Furthermore, this approach successfully restored native secondary structure and affinity of 2D5. Additionally, the platform was effectively applied to the refolding and purification of a polystyrene-binding nanobody (B2), which exhibited limited expression in the periplasmic and cytoplasmic spaces of E. coli. This endeavor resulted in the isolation of 53.2 mg of pure B2 Nb with a purity exceeding 99.5 % from the same volume of bacterial culture. Significantly, this approach restored the native secondary structure of the Nbs, highlighting its potential for addressing challenges associated with expressing complex Nbs in E. coli. Overall, this innovative platform provides a scientifically rigorous and reproducible method for the efficient preparation of Nbs, offering a valuable tool for antibody research and development.
Collapse
Affiliation(s)
- Yiwen Zhang
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Yang Guo
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Liang Song
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Wenshuai Liu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Rui Nian
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Xiying Fan
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China.
| |
Collapse
|
47
|
Gong X, Zhang J, Zhu L, Bai S, Yu L, Sun Y. Fabrication of a Heptapeptide-Modified Poly(glycidyl Methac-Rylate) Nanosphere for Oriented Antibody Immobilization and Immunoassay. Molecules 2024; 29:4635. [PMID: 39407565 PMCID: PMC11477792 DOI: 10.3390/molecules29194635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Oriented antibody immobilization has been widely employed in immunoassays and immunodiagnoses due to its efficacy in identifying target antigens. Herein, a heptapeptide ligand, HWRGWVC (HC7), was coupled to poly(glycidyl methacrylate) (PGMA) nanospheres (PGMA-HC7). The antibody immobilization behavior and antigen recognition performance were investigated and compared with those on PGMA nanospheres by nonspecific adsorption and covalent coupling via carbodiimide chemistry. The antibodies tested included bovine, rabbit, and human immunoglobulin G (IgG), while the antigens included horseradish peroxidase (HRP) and β-2-Microglobulin (β2-MG). The nanospheres were characterized using zeta potential and particle size analyzers, scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, and reversed-phase chromatography, proving each synthesis step was succeeded. Isothermal titration calorimetry assay demonstrated the strong affinity interaction between IgG and PGMA-HC7. Notably, PGMA-HC7 achieved rapid and extremely high IgG adsorption capacity (~3 mg/mg) within 5 min via a specific recognition via HC7 without nonspecific interactions. Moreover, the activities of immobilized anti-HRP and anti-β2-MG antibodies obtained via affinity binding were 1.5-fold and 2-fold higher than those of their covalent coupling counterparts. Further, the oriented-immobilized anti-β2-MG antibody on PGMA-HC7 exhibited excellent performance in antigen recognition with a linear detection range of 0-5.3 μg/mL, proving its great potential in immunoassay applications.
Collapse
Affiliation(s)
- Xiaoxing Gong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (X.G.); (J.Z.); (L.Z.); (S.B.)
| | - Jie Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (X.G.); (J.Z.); (L.Z.); (S.B.)
| | - Liyan Zhu
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (X.G.); (J.Z.); (L.Z.); (S.B.)
| | - Shu Bai
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (X.G.); (J.Z.); (L.Z.); (S.B.)
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Linling Yu
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (X.G.); (J.Z.); (L.Z.); (S.B.)
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; (X.G.); (J.Z.); (L.Z.); (S.B.)
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
48
|
Alfadhli A, Bates TA, Barklis RL, Romanaggi C, Tafesse FG, Barklis E. A nanobody interaction with SARS-COV-2 Spike allows the versatile targeting of lentivirus vectors. J Virol 2024; 98:e0079524. [PMID: 39207135 PMCID: PMC11406891 DOI: 10.1128/jvi.00795-24] [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: 05/06/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
While investigating methods to target gene delivery vectors to specific cell types, we examined the potential of using a nanobody against the SARS-CoV-2 Spike protein receptor-binding domain to direct lentivirus infection of Spike-expressing cells. Using four different approaches, we found that lentiviruses with surface-exposed nanobody domains selectively infect Spike-expressing cells. Targeting is dependent on the fusion function of the Spike protein, and conforms to a model in which nanobody binding to the Spike protein triggers the Spike fusion machinery. The nanobody-Spike interaction also is capable of directing cell-cell fusion and the selective infection of nanobody-expressing cells by Spike-pseudotyped lentivirus vectors. Significantly, cells infected with SARS-CoV-2 are efficiently and selectively infected by lentivirus vectors pseudotyped with a chimeric nanobody protein. Our results suggest that cells infected by any virus that forms syncytia may be targeted for gene delivery by using an appropriate nanobody or virus receptor mimic. Vectors modified in this fashion may prove useful in the delivery of immunomodulators to infected foci to mitigate the effects of viral infections.IMPORTANCEWe have discovered that lentiviruses decorated on their surfaces with a nanobody against the SARS-CoV-2 Spike protein selectively infect Spike-expressing cells. Infection is dependent on the specificity of the nanobody and the fusion function of the Spike protein and conforms to a reverse fusion model, in which nanobody binding to Spike triggers the Spike fusion machinery. The nanobody-Spike interaction also can drive cell-cell fusion and infection of nanobody-expressing cells with viruses carrying the Spike protein. Importantly, cells infected with SARS-CoV-2 are selectively infected with nanobody-decorated lentiviruses. These results suggest that cells infected by any virus that expresses an active receptor-binding fusion protein may be targeted by vectors for delivery of cargoes to mitigate infections.
Collapse
Affiliation(s)
- Ayna Alfadhli
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Timothy A. Bates
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Robin Lid Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, Oregon, USA
| | - CeAnn Romanaggi
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Fikadu G. Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Eric Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, Oregon, USA
| |
Collapse
|
49
|
Saleem MZ, Jahangir GZ, Saleem A, Zulfiqar A, Khan KA, Ercisli S, Ali B, Saleem MH, Saleem A. Production Technologies for Recombinant Antibodies: Insights into Eukaryotic, Prokaryotic, and Transgenic Expression Systems. Biochem Genet 2024:10.1007/s10528-024-10911-5. [PMID: 39287779 DOI: 10.1007/s10528-024-10911-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Recombinant antibodies, a prominent class of recombinant proteins, are witnessing substantial growth in research and diagnostics. Recombinant antibodies are being produced employing diverse hosts ranging from highly complex eukaryotes, for instance, mammalian cell lines (and insects, fungi, yeast, etc.) to unicellular prokaryotic models like gram-positive and gram-negative bacteria. This review delves into these production methods, highlighting approaches like antibody phage display that employs bacteriophages for gene library creation. Recent studies emphasize monoclonal antibody generation through hybridoma technology, utilizing hybridoma cells from myeloma and B-lymphocytes. Transgenic plants and animals have emerged as sources for polyclonal and monoclonal antibodies, with transgenic animals preferred due to their human-like post-translational modifications and reduced immunogenicity risk. Chloroplast expression offers environmental safety by preventing transgene contamination in pollen. Diverse production technologies, such as stable cell pools and clonal cell lines, are available, followed by purification via techniques like affinity chromatography. The burgeoning applications of recombinant antibodies in medicine have led to their large-scale industrial production.
Collapse
Affiliation(s)
| | | | - Ammara Saleem
- Institute of Botany, University of the Punjab, Lahore, Pakistan.
| | - Asma Zulfiqar
- Institute of Botany, University of the Punjab, Lahore, Pakistan
| | - Khalid Ali Khan
- Applied College, Center of Bee Research and its Products, Unit of Bee Research and Honey Production, and Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, 61413, Abha, Saudi Arabia
| | - Sezai Ercisli
- Department of Horticulture, Agricultural Faculty, Ataturk University, 25240, Erzurum, Türkiye
- HGF Agro, Ata Teknokent, 25240, Erzurum, Türkiye
| | - Baber Ali
- Department of Plant Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- School of Science, Western Sydney University, Penrith, 2751, Australia
| | - Muhammad Hamzah Saleem
- Office of Academic Research, Office of VP for Research & Graduate Studies, Qatar University, 2713, Doha, Qatar
| | - Aroona Saleem
- Applied College, Center of Bee Research and its Products, Unit of Bee Research and Honey Production, and Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, 61413, Abha, Saudi Arabia.
- Department of Microbiology, Dr. Ikram-Ul-Haq Institute of Industrial Biotechnology (IIIB), Government College University, Lahore, 54000, Pakistan.
| |
Collapse
|
50
|
Xie X, He Z, Sun Z, Zhang S, Cao H, Hammock BD, Liu X. Shark anti-idiotypic variable new antigen receptor specific for an alpaca nanobody: Exploration of a nontoxic substitute to ochratoxin A in immunoassay. JOURNAL OF HAZARDOUS MATERIALS 2024; 477:135264. [PMID: 39032175 DOI: 10.1016/j.jhazmat.2024.135264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 07/22/2024]
Abstract
Nontoxic substitutes to mycotoxins can facilitate the development of eco-friendly immunoassays. To explore a novel nontoxic substitute to ochratoxin A (OTA), this study screened shark anti-idiotypic variable new antigen receptors (VNARs) against the alpaca anti-OTA nanobody Nb28 through phage display. After four rounds of biopanning of a naïve VNAR phage display library derived from six adult Chiloscyllium plagiosum sharks, one positive clone, namely, P-3, was validated through a phage enzyme-linked immunosorbent assay (phage ELISA). The recombinant anti-idiotypic VNAR AId-V3 was obtained by prokaryotic expression, and the interactions between Nb28 and AId-V3 were investigated via computer-assisted simulation. The affinity of AId-V3 for Nb28 and its heptamer Nb28-C4bpα was measured using Biacore assay. Combining Nb28-C4bpα with AId-V3, a novel direct competitive ELISA (dcELISA) was developed for OTA analysis, with a limit of detection of 0.44 ng/mL and a linear range of 1.77-32.25 ng/mL. The good selectivity, reliability, and precision of dcELISA were confirmed via cross-reaction analysis and recovery experiments. Seven commercial pepper powder samples were tested using dcELISA and validated using high-performance liquid chromatography. Overall, the shark anti-idiotypic VNAR was demonstrated as a promising nontoxic substitute to OTA, and the proposed method was confirmed as a reliable tool for detecting OTA in food.
Collapse
Affiliation(s)
- Xiaoxia Xie
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Zhenyun He
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Zhichang Sun
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Sihang Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Hongmei Cao
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Bruce D Hammock
- Department of Entomology and Nematology, and UC Davis Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| | - Xing Liu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|