1
|
Panikulam S, Morgan H, Gutknecht M, Villiger TK, Lebesgue N, Karle AC. Host cell protein-mediated adjuvanticity and immunogenicity risks of biotherapeutics. Biotechnol Adv 2025; 81:108575. [PMID: 40180137 DOI: 10.1016/j.biotechadv.2025.108575] [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: 10/18/2024] [Revised: 02/18/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Host cell proteins (HCPs) are process-related impurities of biotherapeutic production that might affect product quality and/or patient safety. In a few cases, adverse events were attributed to HCPs present in the administered biotherapeutic. HCP-associated immune risks include adjuvanticity and immunogenicity with potential cross-reactivity. Based on the published data, some HCPs can act as adjuvants increasing the immunogenicity of the biotherapeutic as a bystander effect. HCPs may also induce immunogenicity against themselves, resulting in anti-HCP T cell responses and anti-HCP antibody formation. Depending on sequence similarities, these anti-HCP immune responses might theoretically be cross-reactive to the biotherapeutic or human endogenous proteins. In this review, we examine HCP-associated immune-related risks reported from non-clinical and clinical studies. We also discuss the potential and limitations of in vitro and in silico methods to evaluate the adjuvanticity and immunogenicity potential of HCPs. A risk-based assessment of the safety impact of HCPs may include the identity of the HCP and similarity to the biotherapeutic and human proteins, as well as product, treatment-, and patient-related factors.
Collapse
Affiliation(s)
- Sherin Panikulam
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Hannah Morgan
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Thomas K Villiger
- Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Muttenz, Switzerland
| | - Nicolas Lebesgue
- Technical Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Anette C Karle
- Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
2
|
Jumadila O, Fakhri MD, Darsono A, Puspasari F, Kurniasih SD, Masduki FF, Vidilaseris K, Ihsanawati I, Artarini A, Tan MI, Giri-Rachman EA, Natalia D. Development of a self-assembling multimeric Bann-RBD fusion protein in Pichia pastoris as a potential COVID-19 vaccine candidate. Sci Rep 2025; 15:18467. [PMID: 40425664 PMCID: PMC12117094 DOI: 10.1038/s41598-025-01242-5] [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: 12/01/2024] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
The development of an affordable and accessible vaccine platform is essential for achieving global and long-term protection against COVID-19 and other emerging viral diseases. In this study, we developed a multimeric fusion protein comprising the SARS-CoV-2 receptor-binding domain (RBD) and the β-annulus (Bann) from the tomato bushy stunt virus (TBSV) as a potential subunit vaccine candidate. Molecular modeling of Bann-RBD revealed a 60-mer structure with the RBD displayed on its outer surface. The Bann-RBD gene was constructed and overexpressed in Pichia pastoris X-33. SDS-PAGE analysis of the purified Bann-RBD showed a protein band at 45 kDa, corresponding to monomeric glycosylated Bann-RBD. Peptide mapping analysis using LC-MS/MS confirmed that the expressed Bann-RBD was consistent with the designed protein fusion. The Bann-RBD protein was observed to spontaneously self-assemble into spherical nanocapsids with a diameter of approximately 50 nm. Antigenicity studies demonstrated that the purified Bann-RBD was strongly recognised by monoclonal human anti-SARS-CoV-2 spike-S1 IgG antibodies. Immunogenicity studies revealed that Bann-RBD elicited a robust humoral immune response in BALB/c mice, generating potent neutralising antibodies. Collectively, these findings suggest that the recombinant Bann-RBD produced in Pichia pastoris X-33 is a promising candidate for the development of a COVID-19 vaccine.
Collapse
Affiliation(s)
- Ozi Jumadila
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Muhammad Dzul Fakhri
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Adam Darsono
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Fernita Puspasari
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Sari Dewi Kurniasih
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Fifi Fitriyah Masduki
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
- Biosciences and Biotechnology Research Centre, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Keni Vidilaseris
- The Molecular and Integrative Biosciences Research Programme (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, Biocenter 1/Viikinkaari 9, 00014, Helsinki, Finland
| | - Ihsanawati Ihsanawati
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Anita Artarini
- Laboratory of Pharmaceutical Biotechnology, School of Pharmacy, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
- Biosciences and Biotechnology Research Centre, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Marselina Irasonia Tan
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
- Biosciences and Biotechnology Research Centre, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Ernawati Arifin Giri-Rachman
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
- Biosciences and Biotechnology Research Centre, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia
| | - Dessy Natalia
- Biochemistry and Biomolecular Engineering Research Division, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia.
- Biosciences and Biotechnology Research Centre, Institut Teknologi Bandung, Jl. Ganesa No. 10, Bandung, 40132, Indonesia.
| |
Collapse
|
3
|
Islam MD, Islam MM, Inoue A, Yesmin S, Brindha S, Yoshizue T, Tsurui H, Kurosu T, Kuroda Y. Neutralizing antibodies against the Japanese encephalitis virus are produced by a 12 kDa E. coli- expressed envelope protein domain III (EDIII) tagged with a solubility-controlling peptide. Vaccine 2025; 56:127143. [PMID: 40267616 DOI: 10.1016/j.vaccine.2025.127143] [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: 01/08/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
Escherichia coli is a powerful and cost-effective platform for producing recombinant proteins. However, E. coli- produced proteins lack side-chain glycosylation and may be misfolded due to non-native disulfide bonds, often leading to poor immunogenicity. As a result, they are commonly perceived as unsuitable for use as antiviral vaccine antigens. This study addresses this challenge using the small 12 kDa envelope protein domain III of the Japanese encephalitis virus (JEV-EDIII) as a model. We demonstrate that the low immunogenicity of E. coli- produced proteins can be effectively overcome by employing a solubility-controlling peptide tag (SCP-tag) composed of five isoleucines (C5I). E. coli-produced JEV-EDIII oligomerized into 100 nm (Rh) soluble oligomers upon attachment of the C5I-tag, whereas the untagged JEV-EDIII remained monomeric (Rh ∼ 1.9 nm). The C5I-tag significantly enhanced anti-JEV EDIII IgG titers, as evidenced by ELISA, and increased the population of memory T cells in the spleen, as assessed by flow cytometry. Most notably, the C5I-tagged JEV-EDIII elicited neutralizing antibodies, confirmed by the FRNT50 neutralization assay using live JEV. These findings suggest that oligomerization via SCP-tagging offers a promising, adjuvant-free approach for producing neutralizing antibodies with long-term T cell memory, paving the way for developing E. coli- produced, protein domain-based vaccines.
Collapse
Affiliation(s)
- Md Din Islam
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - M Monirul Islam
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan; Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong 4331, Bangladesh
| | - Ayae Inoue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - Sanjida Yesmin
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - Subbaian Brindha
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan
| | - Takahiro Yoshizue
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan
| | - Hiromichi Tsurui
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan; Department of Immunological Diagnosis, Juntendo University School of Medicine, Hongo 2-1-1, Tokyo 113-8421, Japan
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Gakuen 4-7-1, Tokyo 208-0011, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Faculty of Engineering, Tokyo University of Agriculture and Technology, Nakamachi 2-24-16, Tokyo 184-8588, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1 Harumi-cho, Fuchu-shi, Tokyo 183-8538, Japan.
| |
Collapse
|
4
|
Li W, Liu Z. Advances in glycan-specific biomimetic molecular recognition and its biomedical applications. Chem Commun (Camb) 2025; 61:6739-6754. [PMID: 40243224 DOI: 10.1039/d5cc01003a] [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: 04/18/2025]
Abstract
Glycan-mediated recognition is critically involved in a variety of pathophysiological events, so strategies targeting unique glycosylation could offer opportunities for novel disease diagnostics and therapeutics. Herein, we survey the current progress in glycan-binding entities and their biomedical applications. Particularly focusing on biologically promising artificial receptors, including boronate affinity-based molecularly imprinted polymers (MIPs) and anti-glycan aptamers, we summarize significant efforts in the recognition of glycans by MIPs and aptamers with high affinity and exquisite specificity. Furthermore, we highlight successful examples in biomedical fields of antiviral treatment, cancer diagnostics and targeted therapeutics. Finally, we briefly sketch the remaining challenges and future perspectives. Collectively, this review provides significant insights for further exploration of glycan-specific biomimetic materials in the broad biomedical area.
Collapse
Affiliation(s)
- Wei Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
5
|
Liu M, Xiao R, Li X, Zhao Y, Huang J. A comprehensive review of recombinant technology in the food industry: Exploring expression systems, application, and future challenges. Compr Rev Food Sci Food Saf 2025; 24:e70078. [PMID: 39970011 DOI: 10.1111/1541-4337.70078] [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: 09/21/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 02/21/2025]
Abstract
Biotechnology has significantly advanced the production of recombinant proteins (RPs). This review examines the latest advancements in protein production technologies, including CRISPR, genetic engineering, vector integration, and fermentation, and their implications for the food industry. This review delineates the merits and shortcomings of prevailing host systems for RP production, underscoring molecular and process strategies pivotal for amplifying yields and purity. It traverses the spectrum of RP applications, challenges, and burgeoning trends, highlighting the imperative of employing robust hosts and cutting-edge genetic engineering to secure high-quality, high-yield outputs while circumventing protein aggregation and ensuring correct folding for enhanced activity. Recombinant technology has paved the way for the food industry to produce alternative proteins like leghemoglobin and cytokines, along with enzyme preparations such as proteases and lipases, and to modify microbial pathways for synthesizing beneficial compounds, including pigments, terpenes, flavonoids, and functional sugars. However, scaling microbial production to industrial scales presents economic, efficiency, and environmental challenges that demand innovative solutions, including high-throughput screening and CRISPR/Cas9 systems, to bolster protein yield and quality. Although recombinant technology holds much promise, it must navigate high costs and scalability to satisfy the escalating global demand for RPs in therapeutics and food. The variability in ethical and regulatory hurdles across regions further complicates market acceptance, underscoring an urgent need for robust regulatory frameworks for genetically modified organisms. These frameworks are essential for safeguarding the production process, ensuring product safety, and upholding the efficacy of RPs in industrial applications.
Collapse
Affiliation(s)
- Ming Liu
- College of Grain and Food Science, Henan University of Technology, Zhengzhou, Henan, P. R. China
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou, Henan, P. R. China
| | - Ran Xiao
- College of Agriculture, Henan University, Kaifeng, Henan, P. R. China
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou, Henan, P. R. China
| | - Xiaolin Li
- College of Grain and Food Science, Henan University of Technology, Zhengzhou, Henan, P. R. China
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou, Henan, P. R. China
| | - Yingyu Zhao
- College of Grain and Food Science, Henan University of Technology, Zhengzhou, Henan, P. R. China
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou, Henan, P. R. China
| | - Jihong Huang
- College of Agriculture, Henan University, Kaifeng, Henan, P. R. China
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou, Henan, P. R. China
- School of Food and Pharmacy, Xuchang University, Xuchang, Henan, P. R. China
| |
Collapse
|
6
|
Xu X, Peng Q, Jiang X, Tan S, Yang W, Han Y, Oyang L, Lin J, Shen M, Wang J, Li H, Xia L, Peng M, Wu N, Tang Y, Wang H, Liao Q, Zhou Y. Altered glycosylation in cancer: molecular functions and therapeutic potential. Cancer Commun (Lond) 2024; 44:1316-1336. [PMID: 39305520 PMCID: PMC11570773 DOI: 10.1002/cac2.12610] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 11/19/2024] Open
Abstract
Glycosylation, a key mode of protein modification in living organisms, is critical in regulating various biological functions by influencing protein folding, transportation, and localization. Changes in glycosylation patterns are a significant feature of cancer, are associated with a range of pathological activities in cancer-related processes, and serve as critical biomarkers providing new targets for cancer diagnosis and treatment. Glycoproteins like human epidermal growth factor receptor 2 (HER2) for breast cancer, alpha-fetoprotein (AFP) for liver cancer, carcinoembryonic antigen (CEA) for colon cancer, and prostate-specific antigen (PSA) for prostate cancer are all tumor biomarkers approved for clinical use. Here, we introduce the diversity of glycosylation structures and newly discovered glycosylation substrate-glycosylated RNA (glycoRNA). This article focuses primarily on tumor metastasis, immune evasion, metabolic reprogramming, aberrant ferroptosis responses, and cellular senescence to illustrate the role of glycosylation in cancer. Additionally, we summarize the clinical applications of protein glycosylation in cancer diagnostics, treatment, and multidrug resistance. We envision a promising future for the clinical applications of protein glycosylation.
Collapse
Affiliation(s)
- Xuemeng Xu
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Qiu Peng
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Xianjie Jiang
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
| | - Wenjuan Yang
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
| | - Yaqian Han
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Mengzhou Shen
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Jiewen Wang
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Haofan Li
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Mingjing Peng
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Nayiyuan Wu
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Key Laboratory of Translational Radiation OncologyChangshaHunanP. R. China
| | - Qianjin Liao
- Department of OncologyHunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaHunanP. R. China
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer MetabolismChangshaHunanP. R. China
- Hunan Engineering Research Center of Tumor organoid Technology and application, Public Service Platform of Tumor organoids TechnologyChangshaHunanP. R. China
- Hunan Key Laboratory of Translational Radiation OncologyChangshaHunanP. R. China
| |
Collapse
|
7
|
Murphy JF, Lavelle M, Asciak L, Burdis R, Levis HJ, Ligorio C, McGuire J, Polleres M, Smith PO, Tullie L, Uribe-Gomez J, Chen B, Dawson JI, Gautrot JE, Hooper NM, Kelly DJ, Li VSW, Mata A, Pandit A, Phillips JB, Shu W, Stevens MM, Williams RL, Armstrong JPK, Huang YYS. Biofabrication and biomanufacturing in Ireland and the UK. Biodes Manuf 2024; 7:825-856. [PMID: 39650072 PMCID: PMC11618173 DOI: 10.1007/s42242-024-00316-z] [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: 04/12/2024] [Accepted: 08/11/2024] [Indexed: 12/11/2024]
Abstract
As we navigate the transition from the Fourth to the Fifth Industrial Revolution, the emerging fields of biomanufacturing and biofabrication are transforming life sciences and healthcare. These sectors are benefiting from a synergy of synthetic and engineering biology, sustainable manufacturing, and integrated design principles. Advanced techniques such as 3D bioprinting, tissue engineering, directed assembly, and self-assembly are instrumental in creating biomimetic scaffolds, tissues, organoids, medical devices, and biohybrid systems. The field of biofabrication in the United Kingdom and Ireland is emerging as a pivotal force in bioscience and healthcare, propelled by cutting-edge research and development. Concentrating on the production of biologically functional products for use in drug delivery, in vitro models, and tissue engineering, research institutions across these regions are dedicated to innovating healthcare solutions that adhere to ethical standards while prioritising sustainability, affordability, and healthcare system benefits. Graphic abstract
Collapse
Affiliation(s)
- Jack F. Murphy
- Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ UK
| | - Martha Lavelle
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY UK
| | - Lisa Asciak
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G4 0NW UK
| | - Ross Burdis
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Hannah J. Levis
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX UK
| | - Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD UK
| | - Jamie McGuire
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, S016 6YD UK
| | - Marlene Polleres
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
| | - Poppy O. Smith
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX UK
| | - Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, NW1 1AT UK
| | - Juan Uribe-Gomez
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, H91 W2TY Ireland
| | - Biqiong Chen
- School of Mechanical and Aerospace Engineering, Queen’s University Belfast, Belfast, BT9 5AH UK
| | - Jonathan I. Dawson
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, S016 6YD UK
| | - Julien E. Gautrot
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS UK
| | - Nigel M. Hooper
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, M13 9PL UK
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 H903 Ireland
| | - Vivian S. W. Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, NW1 1AT UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD UK
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, H91 W2TY Ireland
| | - James B. Phillips
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX UK
| | - Wenmiao Shu
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G4 0NW UK
| | - Molly M. Stevens
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU UK
- Department of Engineering Science, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU UK
| | - Rachel L. Williams
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX UK
| | - James P. K. Armstrong
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY UK
| | | |
Collapse
|
8
|
Wang Y, Lei K, Zhao L, Zhang Y. Clinical glycoproteomics: methods and diseases. MedComm (Beijing) 2024; 5:e760. [PMID: 39372389 PMCID: PMC11450256 DOI: 10.1002/mco2.760] [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: 06/06/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Glycoproteins, representing a significant proportion of posttranslational products, play pivotal roles in various biological processes, such as signal transduction and immune response. Abnormal glycosylation may lead to structural and functional changes of glycoprotein, which is closely related to the occurrence and development of various diseases. Consequently, exploring protein glycosylation can shed light on the mechanisms behind disease manifestation and pave the way for innovative diagnostic and therapeutic strategies. Nonetheless, the study of clinical glycoproteomics is fraught with challenges due to the low abundance and intricate structures of glycosylation. Recent advancements in mass spectrometry-based clinical glycoproteomics have improved our ability to identify abnormal glycoproteins in clinical samples. In this review, we aim to provide a comprehensive overview of the foundational principles and recent advancements in clinical glycoproteomic methodologies and applications. Furthermore, we discussed the typical characteristics, underlying functions, and mechanisms of glycoproteins in various diseases, such as brain diseases, cardiovascular diseases, cancers, kidney diseases, and metabolic diseases. Additionally, we highlighted potential avenues for future development in clinical glycoproteomics. These insights provided in this review will enhance the comprehension of clinical glycoproteomic methods and diseases and promote the elucidation of pathogenesis and the discovery of novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yujia Wang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Kaixin Lei
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Lijun Zhao
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Yong Zhang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
9
|
Walimbwa SI, Maly P, Kafkova LR, Raska M. Beyond glycan barriers: non-cognate ligands and protein mimicry approaches to elicit broadly neutralizing antibodies for HIV-1. J Biomed Sci 2024; 31:83. [PMID: 39169357 PMCID: PMC11337606 DOI: 10.1186/s12929-024-01073-y] [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: 05/02/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) vaccine immunogens capable of inducing broadly neutralizing antibodies (bNAbs) remain obscure. HIV-1 evades immune responses through enormous diversity and hides its conserved vulnerable epitopes on the envelope glycoprotein (Env) by displaying an extensive immunodominant glycan shield. In elite HIV-1 viremic controllers, glycan-dependent bNAbs targeting conserved Env epitopes have been isolated and are utilized as vaccine design templates. However, immunological tolerance mechanisms limit the development of these antibodies in the general population. The well characterized bNAbs monoclonal variants frequently exhibit extensive levels of somatic hypermutation, a long third heavy chain complementary determining region, or a short third light chain complementarity determining region, and some exhibit poly-reactivity to autoantigens. This review elaborates on the obstacles to engaging and manipulating the Env glycoprotein as an effective immunogen and describes an alternative reverse vaccinology approach to develop a novel category of bNAb-epitope-derived non-cognate immunogens for HIV-1 vaccine design.
Collapse
Affiliation(s)
- Stephen Ian Walimbwa
- Department of Immunology, University Hospital Olomouc, Zdravotníků 248/7, 77900, Olomouc, Czech Republic.
| | - Petr Maly
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50, Vestec, Czech Republic
| | - Leona Raskova Kafkova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 779 00, Olomouc, Czech Republic
| | - Milan Raska
- Department of Immunology, University Hospital Olomouc, Zdravotníků 248/7, 77900, Olomouc, Czech Republic.
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 779 00, Olomouc, Czech Republic.
| |
Collapse
|
10
|
Singh BN, Tabatabaei M, Pandit A, Elling L, Gupta VK. Emerging advances in glycoengineering of carbohydrates/glycans and their industrial applications. Biotechnol Adv 2024; 72:108324. [PMID: 38360155 DOI: 10.1016/j.biotechadv.2024.108324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Affiliation(s)
- Brahma N Singh
- Herbal Nanobiotechnology Lab, Pharmacology Division, CSIR-National Botanical Research Institute, Lucknow 226001, India
| | - Meisam Tabatabaei
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries (AKUATROP), Universiti Malaysia Terengganu, Kuala Nerus, Terengganu 21030, Malaysia
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Ireland
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, D-52074 Aachen, Germany
| | - Vijai Kumar Gupta
- School of Biotechnology, Dublin City University, Glasnevin, Dublin D09 K20V, Ireland; Biodesign Europe, Dublin City University, Glasnevin, Dublin D09 K20V, Ireland.
| |
Collapse
|
11
|
Hills FR, Eruera AR, Hodgkinson-Bean J, Jorge F, Easingwood R, Brown SHJ, Bouwer JC, Li YP, Burga LN, Bostina M. Variation in structural motifs within SARS-related coronavirus spike proteins. PLoS Pathog 2024; 20:e1012158. [PMID: 38805567 PMCID: PMC11236199 DOI: 10.1371/journal.ppat.1012158] [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: 02/05/2024] [Revised: 07/10/2024] [Accepted: 03/28/2024] [Indexed: 05/30/2024] Open
Abstract
SARS-CoV-2 is the third known coronavirus (CoV) that has crossed the animal-human barrier in the last two decades. However, little structural information exists related to the close genetic species within the SARS-related coronaviruses. Here, we present three novel SARS-related CoV spike protein structures solved by single particle cryo-electron microscopy analysis derived from bat (bat SL-CoV WIV1) and civet (cCoV-SZ3, cCoV-007) hosts. We report complex glycan trees that decorate the glycoproteins and density for water molecules which facilitated modeling of the water molecule coordination networks within structurally important regions. We note structural conservation of the fatty acid binding pocket and presence of a linoleic acid molecule which are associated with stabilization of the receptor binding domains in the "down" conformation. Additionally, the N-terminal biliverdin binding pocket is occupied by a density in all the structures. Finally, we analyzed structural differences in a loop of the receptor binding motif between coronaviruses known to infect humans and the animal coronaviruses described in this study, which regulate binding to the human angiotensin converting enzyme 2 receptor. This study offers a structural framework to evaluate the close relatives of SARS-CoV-2, the ability to inform pandemic prevention, and aid in the development of pan-neutralizing treatments.
Collapse
Affiliation(s)
- Francesca R. Hills
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alice-Roza Eruera
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - James Hodgkinson-Bean
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Fátima Jorge
- Otago Microscopy and Nano Imaging Unit, University of Otago, Dunedin, New Zealand
| | - Richard Easingwood
- Otago Microscopy and Nano Imaging Unit, University of Otago, Dunedin, New Zealand
| | - Simon H. J. Brown
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, University of Wollongong, Wollongong, New South Wales, Australia
| | - James C. Bouwer
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, University of Wollongong, Wollongong, New South Wales, Australia
| | - Yi-Ping Li
- Institute of Human Virology and Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Laura N. Burga
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Otago Microscopy and Nano Imaging Unit, University of Otago, Dunedin, New Zealand
| |
Collapse
|
12
|
Rocamora F, Schoffelen S, Arnsdorf J, Toth EA, Abdul Y, Cleveland TE, Bjørn SP, Wu MYM, McElvaney NG, Voldborg BGR, Fuerst TR, Lewis NE. Glycoengineered recombinant alpha1-antitrypsin results in comparable in vitro and in vivo activities to human plasma-derived protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587088. [PMID: 38585818 PMCID: PMC10996670 DOI: 10.1101/2024.03.27.587088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Alpha-1-antitrypsin (A1AT) is a multifunctional, clinically important, high value therapeutic glycoprotein that can be used for the treatment of many diseases such as alpha-1-antitrypsin deficiency, diabetes, graft-versus-host-disease, cystic fibrosis and various viral infections. Currently, the only FDA-approved treatment for A1AT disorders is intravenous augmentation therapy with human plasma-derived A1AT. In addition to its limited supply, this approach poses a risk of infection transmission, since it uses therapeutic A1AT harvested from donors. To address these issues, we sought to generate recombinant human A1AT (rhA1AT) that is chemically and biologically indistinguishable from its plasma-derived counterpart using glycoengineered Chinese Hamster Ovary (geCHO-L) cells. By deleting nine key genes that are part of the CHO glycosylation machinery and expressing the human ST6GAL1 and A1AT genes, we obtained stable, high producing geCHO-L lines that produced rhA1AT having an identical glycoprofile to plasma-derived A1AT (pdA1AT). Additionally, the rhA1AT demonstrated in vitro activity and in vivo half-life comparable to commercial pdA1AT. Thus, we anticipate that this platform will help produce human-like recombinant plasma proteins, thereby providing a more sustainable and reliable source of therapeutics that are cost-effective and better-controlled with regard to purity, clinical safety and quality.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| | - Sanne Schoffelen
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Johnny Arnsdorf
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, United States
| | - Yunus Abdul
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, United States
| | - Thomas E Cleveland
- National Institute of Standards and Technology, Rockville, MD, 20850, USA
| | - Sara Petersen Bjørn
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
| | - Noel G McElvaney
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, United States
- Department of Medicine, Irish Center for Genetic Lung Disease, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bjørn Gunnar Rude Voldborg
- National Biologics Facility, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, United States
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, United States
- NeuImmune, Inc., Sykesville, MD, United States
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, United States
- NeuImmune, Inc., Sykesville, MD, United States
| |
Collapse
|
13
|
Zimmer K, Chmielewska AM, Jackowiak P, Figlerowicz M, Bienkowska-Szewczyk K. Alterations in N-glycosylation of HCV E2 Protein in Children Patients with IFN-RBV Therapy Failure. Pathogens 2024; 13:256. [PMID: 38535599 PMCID: PMC10974529 DOI: 10.3390/pathogens13030256] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 02/11/2025] Open
Abstract
The glycosylation of viral envelope proteins plays an important role in virus biology and the immune response of the host to infection. Hepatitis C virus (HCV) envelope proteins E1 and E2, key players in virus entry and spread, are highly N-glycosylated and possess 4 (5 in certain genotypes) to 11 conserved glycosylation sites, respectively. Many published results based on recombinant proteins indicate that the glycan shield can mask the epitopes targeted by neutralizing antibodies. Glycan shifting within the conserved linear E2 region (412-423) could be one of the escape strategies used by HCV. In the present report, we isolated E2 genes from samples (collected before the IFN-RBV therapy) originating from pediatric patients infected with HCV gt 1a. We analyzed the biochemical properties of cloned E2 glycoprotein variants and investigated their glycosylation status. The sequencing of E2 genes isolated from patients who did not respond to therapy revealed mutations at N-glycosylation sites, thus leading to a lower molecular weight and a low affinity to both linear and conformational neutralizing antibodies. The loss of the glycosylation site within the conserved epitope (amino acid 417) impaired the binding with AP33, an antibody that potently neutralizes all genotypes of HCV. Our findings, based on clinical samples, confirm the influence of N-glycosylation aberrations on the antigenic and conformational properties of HCV E1/E2, which may possibly correlate with the outcome of therapy in patients.
Collapse
Affiliation(s)
- Karolina Zimmer
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology of UG and MUG, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (K.Z.); (A.M.C.)
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biala, Poland
| | - Alicja M. Chmielewska
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology of UG and MUG, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (K.Z.); (A.M.C.)
| | - Paulina Jackowiak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznań, Poland (M.F.)
| | - Marek Figlerowicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznań, Poland (M.F.)
| | - Krystyna Bienkowska-Szewczyk
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology of UG and MUG, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (K.Z.); (A.M.C.)
| |
Collapse
|