1
|
Cao G, Li Z, Wang Z, Yang Y, Li J, Qi H. High-throughput optimization of peptide-linker for fusing function protein with GFP. Protein Expr Purif 2025; 231:106718. [PMID: 40239936 DOI: 10.1016/j.pep.2025.106718] [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: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Fusion proteins are pivotal in bioengineering, with applications in purification, delivery, and imaging. However, the development of specialized peptide linkers tailored for target fusion proteins remains an unmet challenge. In this study, we demonstrate the optimization of fusing a functional protein with green fluorescent protein (GFP) through the screening of peptide linker sequences. Using seamless cloning methodology, a nanobody protein was fused to the N-terminus of GFP via a randomized 18-amino acid peptide linker library. Initial screening of fusion protein clones was conducted on solid plates to identify those expressing robust GFP fluorescence. A total of 153 clones with unique linker sequences were identified using Sanger sequencing. A wide range of normalized fluorescence signals was observed, revealing significant variability in linker performance. Among the screened linkers, one exhibited high fluorescence activity, outperforming commonly used flexible and rigid linkers. This finding underscores the necessity of optimize linker sequences for specific fusion proteins. Furthermore, the results demonstrated that the screened linker is compatible with diverse N-terminal proteins while maintaining GFP functionality. Additionally, to investigate the effect of linker on the function of target protein, we determined the reverse transcription efficiency of the murine leukemia virus reverse transcriptase (MLV-RT) in the fusion proteins by a two-step RT-qPCR method. In conclusion, this study presents an efficient optimization of peptide linkers, offering a novel methodology for the engineering and application of specialized linkers for fusion proteins.
Collapse
Affiliation(s)
- Gaili Cao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China; State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Zhong Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China; State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Zhaoguan Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China; State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Youhui Yang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China; State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Jiawei Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China; State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Hao Qi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China; State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China.
| |
Collapse
|
2
|
Sanchez C, Ramirez A, Hodgson L. Unravelling molecular dynamics in living cells: Fluorescent protein biosensors for cell biology. J Microsc 2025; 298:123-184. [PMID: 38357769 PMCID: PMC11324865 DOI: 10.1111/jmi.13270] [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: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.
Collapse
Affiliation(s)
- Colline Sanchez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrea Ramirez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Louis Hodgson
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
3
|
Koyano B, Shibuya T. Faster and More Accurate Estimation of Protein Hinges Based on Information Criteria. J Comput Biol 2025; 32:498-519. [PMID: 40293732 DOI: 10.1089/cmb.2024.0731] [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: 04/30/2025] Open
Abstract
Protein hinges are flexible parts connecting several rigid substructures of proteins that are crucial to determine protein function. Various methods have been developed for efficiently and accurately estimating protein hinge positions by comparing two different conformations of the same protein for a growing number of protein structures. However, few studies have focused on accurately estimating the number of hinges, and it is required to accurately estimate both the number and positions of hinges. We propose faster and more accurate algorithms for estimating the number and positions of hinges by utilizing information criteria that run in O(n2)-time, where n is the protein length. Our algorithms utilize Bayesian Information Criterion (BIC) or Akaike's Information Criterion based on a newly proposed k-hinge structure generation model that models the hinge motions between two protein conformations. Our exact algorithm based on BIC outperformed the most accurate previous method in terms of both hinge number and position accuracy on our simulation dataset. Our exact algorithm was approximately as fast as the previous fastest method, DynDom, on our simulation dataset. We evaluated the hinge number and position accuracy of our exact algorithm and previous methods on one hinge-annotated dataset. The hinge number and position accuracy of our exact algorithm were comparable to the most accurate previous method on the hinge-annotated dataset. We further propose even faster O(n)-time heuristic algorithms, where n is the protein length. Our heuristic algorithm achieved almost the same hinge number and position accuracy as our exact algorithm, and was over 18 times faster than our exact algorithm and DynDom.
Collapse
Affiliation(s)
- Bunsho Koyano
- Department of Computer Science, Graduate School of Information Science and Technology, The University of Tokyo, Japan
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Japan
| | - Tetsuo Shibuya
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Japan
| |
Collapse
|
4
|
Šulc J, Vondrášek J. Unraveling the Tether: Exploring Representative Protein Linkers and Their Structural and Thermodynamical Properties. J Phys Chem B 2025; 129:3720-3730. [PMID: 40188464 PMCID: PMC12010332 DOI: 10.1021/acs.jpcb.4c04194] [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: 07/02/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/08/2025]
Abstract
This study explores the thermodynamic and structural behaviors of linker peptides, short polypeptide segments that often bridge protein domains. We are focusing on three prototypical classes─glycine-serine (GS), glycine-glycine (GG), and alanine-proline (AP)─and exploring their conformational dynamics as isolated entities outside a multidomain protein context. Using extensive molecular dynamics (MD) simulations and free energy perturbation (FEP) analyses, we characterize the free energy landscapes, entropic properties, and solvation energetics of 20 representative linkers. Our results reveal a pronounced linear relationship between linker length and key thermodynamic contributions, including zero-point vibrational energy (ZPVE), potential energy, and entropy. Notably, vibrational entropy emerges as a dominant stabilizing term. We also found that AP linkers display more rigid, yet extended conformations compared to the highly flexible GS and moderately flexible GG linkers. These findings underscore the nuanced role of linker composition in contributing to multidomain protein architecture and dynamics, and highlight how thermodynamic forces shape linker conformational behavior. Collectively, our work enhances the mechanistic understanding of protein linkers, offering valuable insights for the rational design of peptide-based systems and informing future efforts to modulate interdomain flexibility and stability in multidomain proteins.
Collapse
Affiliation(s)
- Josef Šulc
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 6 166 10, Czech Republic
- Faculty
of Science, Charles University, Albertov 2038, Prague 128 00, Czech Republic
| | - Jiří Vondrášek
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 6 166 10, Czech Republic
| |
Collapse
|
5
|
Chatrdooz H, Sargolzaei J. An Overview of Property, Design, and Functionality of Linkers for Fusion Protein Construction. Proteins 2025. [PMID: 40099816 DOI: 10.1002/prot.26812] [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: 08/09/2024] [Revised: 11/03/2024] [Accepted: 02/06/2025] [Indexed: 03/20/2025]
Abstract
Linkers are naturally occurring short amino acid sequences that are used to separate domains within a protein. The advent of recombinant DNA technology has made it possible to combine two interacting partners by introducing artificial linkers that often, allow for the production of stable and functional proteins. Glycine-rich linkers are useful for transient interactions, especially where the interaction is weak, by covalently linking proteins and forming a stable protein-protein complex. These linkers have also been used to generate covalently stable dimers and to connect two independent domains that create a ligand binding site or recognition sequence. Various structures of covalently linked protein complexes have been described using nuclear magnetic resonance methods, cryo-electron microscopy techniques, and X-ray crystallography; in addition, several structures where linkers have been used to generate stable protein-protein complexes, improve protein solubility, and obtain protein dimers are investigated, and also the design and engineering of the linker in fusion proteins is discussed. Therefore, one of the main factors for linker design and optimization is their flexibility, which can directly contribute to the physical distance between the domains of a fusion protein and describe the tendency of a linker to maintain a stable conformation during expression. We summarize the research on design and bioinformatics can be used to predict the spatial structure of the fusion protein. To perform simulations of spatial structures and drug molecule design, future research will concentrate on various correlation models.
Collapse
Affiliation(s)
- Hadis Chatrdooz
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Javad Sargolzaei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| |
Collapse
|
6
|
Abstract
The use of enzymes to convert substrates into valuable products has been an integral part of biocatalysis. However, some reactions are energy-demanding that requires the use of NAD(P)H to proceed. This NAD(P)H can be costly impeding the progress of enzyme usage at a bigger scale. The rise of sophisticated cloning methods has allowed the possibility of constructing multi-enzyme complexes such as coupling NAD(P)H-requiring enzymes with NADH-regeneration systems such as formate dehydrogenases. This allows a more-efficient way to recycle co-factors or co-substrates with cheaper sacrificial substrate such as formate for formate dehydrogenases or glucose for glucose dehydrogenases. However, the design of fusion proteins requires careful attention especially on the peptide linker that will be used to connect two protein domains. The length and the property of the linker and even the orientation of the genes encoding for the proteins in the open reading frame can significantly affect the outcome of the fusion protein. In this chapter, we present a step-by-step procedure for the design of a fusion protein construct via Gibson assembly and how to design linker libraries from one construct using site-directed mutagenesis.
Collapse
Affiliation(s)
- Beyzanur Celebi
- Microbial Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Janina Lawniczek
- Microbial Biotechnology, Ruhr University Bochum, Bochum, Germany
| | | | | |
Collapse
|
7
|
Girardo B, Yue Y, Lockridge O, Bartling AM, Schopfer LM, Augusto L, Larson MA. Francisella tularensis universal stress protein contributes to persistence during growth arrest and paraquat-induced superoxide stress. J Bacteriol 2025; 207:e0037724. [PMID: 39846732 PMCID: PMC11841066 DOI: 10.1128/jb.00377-24] [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: 09/05/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025] Open
Abstract
Francisella tularensis is one of the most virulent bacterial pathogens known and causes the disease tularemia, which can be fatal if untreated. This zoonotic and intracellular pathogen is exposed to diverse environmental and host stress factors that require an appropriate response to survive. However, the stress tolerance mechanisms used by F. tularensis to persist are not fully understood. To address this aspect, we evaluated the highly conserved universal stress protein (Usp) that is encoded by a single-copy gene in F. tularensis, unlike the majority of other bacterial pathogens that produce several to many Usp homologs. We determined that the F. tularensis Usp transcript is unusually stable with a half-life of over 30 minutes, and that usp transcript and protein levels remained abundant when exposed to low pH, nutrient deprivation, hydrogen peroxide, and paraquat. Of these and other stress conditions evaluated, the F. tularensis Δusp mutant only exhibited reduced survival relative to the wild type during stationary phase and exposure to paraquat, a highly toxic compound that generates superoxide anions and other free radicals. Comparison of transcript levels in untreated and paraquat-treated F. tularensis wild type and Δusp indicated that Usp contributes to enhanced expression of antioxidant defense genes, oxyR and katG. In summary, the high abundance and stability of Usp provide prompt protection during extended periods of growth arrest and free radical exposure, promoting F. tularensis persistence. We propose that F. tularensis Usp contributes to an adaptive response that prolongs viability and increases the longevity of this zoonotic pathogen in the environment. IMPORTANCE Francisella tularensis is classified as a Tier 1 select agent due to the low infectious dose, ease of transmission, and potential use as a bioweapon. To better understand the stress defense mechanisms that contribute to the ability of this highly virulent pathogen to persist, we evaluated the conserved F. tularensis universal stress protein (Usp). We show that F. tularensis Usp is unusually stable and remains abundant, regardless of the stress conditions tested, differing from other bacterial Usp homologs. We also determined that F. tularensis Usp enhances the expression of several critical antioxidant defense genes and increases survival during paraquat exposure and growth arrest. Determining the factors that promote F. tularensis persistence in the environment is needed to prevent tularemia transmission.
Collapse
Affiliation(s)
- Benjamin Girardo
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yinshi Yue
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Oksana Lockridge
- Eppley Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Amanda M. Bartling
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | - Leonardo Augusto
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Marilynn A. Larson
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
8
|
Tripathy RK, Pande AH. Nanobody-Oligonucleotide Conjugates (NucleoBodies): The Next Frontier in Oligonucleotide Therapy. Pharm Res 2025; 42:219-236. [PMID: 39953265 DOI: 10.1007/s11095-025-03829-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: 10/08/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
As of now, more than 15 oligonucleotide drugs, primarily small interfering RNAs and antisense oligonucleotide classes, have been approved by the US FDA for therapeutic use, and many more are under clinical trials. However, safe and effective delivery of the oligonucleotide-based drugs to the target tissue still remains a major challenge. For enhanced plasma half-life, effective endosomal release, and other multiple functionalities, various carrier molecules have been used over the years. The successful therapeutic application of antibody-drug conjugates has made antibodies a popular choice for the delivery of oligonucleotide payloads into the target tissues. Single-chain variable domains of heavy chain antibodies (nanobodies) have proven a promising alternative to antibodies in recent years due to their small size, high affinity for the target, cell-penetrating potency, simple and easy production. The present review highlights the oligonucleotide drug types and their conjugation with nanobodies called NucleoBodies for effective targeted delivery, detection and diagnostics.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India.
| |
Collapse
|
9
|
Rahman MM, Masum MHU, Parvin R, Das SC, Talukder A. Designing of an mRNA vaccine against high-risk human papillomavirus targeting the E6 and E7 oncoproteins exploiting immunoinformatics and dynamic simulation. PLoS One 2025; 20:e0313559. [PMID: 39761277 PMCID: PMC11703113 DOI: 10.1371/journal.pone.0313559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/25/2024] [Indexed: 05/01/2025] Open
Abstract
Human papillomavirus 16 and human papillomavirus 18 have been associated with different life-threatening cancers, including cervical, lung, penal, vulval, vaginal, anal, and oropharyngeal cancers, while cervical cancer is the most prominent one. Several research studies have suggested that the oncoproteins E6 and E7 are the leading cause of cancers associated with the human papillomavirus infection. Therefore, we developed two mRNA vaccines (V1 and V2) targeting these oncoproteins. We used several bioinformatics tools to predict helper T lymphocyte, cytotoxic T lymphocyte, and B-cell epitopes derived from the proteins and assessed their antigenicity, allergenicity, and toxicity. Both vaccines were constructed using selected epitopes, linkers, and adjuvants. After that, the vaccines were applied for physicochemical properties, secondary and tertiary structure predictions, and subsequent docking and simulation analyses. Accordingly, vaccine 1 (V1) and vaccine 2 (V2) showed better hydrophilicity with the grand average hydropathicity score of -0.811 and -0.648, respectively. The secondary and tertiary structures of the vaccines were also deemed satisfactory, with high stability indicated by the Ramachandran plot (V1:94.5% and V2:87.1%) and Z scores (V1: -5.15 and V2: -4.1). Docking analysis revealed substantial affinity of the vaccines towards the toll-like receptor-2 (V1: -1159.3, V2: -1246.3) and toll-like receptor-4 (V1: -1109.3, V2: -1244.8) receptors. Molecular dynamic simulation validated structural integrity and indicated varying stability throughout the simulation. Codon optimization showed significant expression of the vaccines (V1:51.88% and V2:51.63%) in E. coli vectors. Furthermore, regarding immune stimulation, the vaccines elicited significant B-cell and T-cell responses, including sustained adaptive and innate immune responses. Finally, thermodynamic predictions indicated stable mRNA structures of the vaccines (V1: -502.60 kcal/mol and V2: -450.90 kcal/mol). The proposed vaccines designed effectively targeting human papillomavirus oncoproteins have demonstrated promising results via robust immune responses, suggesting their suitability for further clinical advancement, including in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, Australia
| | - Md. Habib Ullah Masum
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chittagong, Bangladesh
| | - Rehana Parvin
- Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Khulshi, Chittagong, Bangladesh
| | - Shuvo Chandra Das
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Asma Talukder
- Microbiology, Cancer and Bioinformatics Research Group, Noakhali Science and Technology University, Noakhali, Bangladesh
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, Australia
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
10
|
Senthilkumar S, Mahesh S, Jaisankar S, Yennamalli RM. Surface exposed and charged residues drive thermostability in fungi. Proteins 2025; 93:226-240. [PMID: 37909647 DOI: 10.1002/prot.26623] [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: 05/18/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
Fungi, though mesophilic, include thermophilic and thermostable species, as well. The thermostability of proteins observed in these fungi is most likely to be attributed to several molecular factors, such as the presence of salt bridges and hydrogen bond interactions between side chains. These factors cannot be generalized for all fungi. Factors impacting thermostability can guide how fungal thermophilic proteins gain thermostability. We curated a dataset of proteins for 14 thermophilic fungi and their evolutionarily closer mesophiles. Additionally, the proteome of Chaetomium thermophilum and its evolutionarily related mesophile Chaetomium globosum was analyzed. Using eggNOG, we categorized the proteomes into clusters of orthologous groups (COGs). While the individual count of proteins is over-represented in mesophiles (for COGs S, G, L, and Q), there are certain features that are significantly enriched in thermophiles (such as charged residues, exposed residues, polar residues, etc.). Since fungi are known to be cellulolytic and chitinolytic by nature, we selected 37 existing carbohydrate-active enzymes (CAZyme) families in Eurotiales, Mucorales, and Sordariales. We looked at closely similar sequences and their modeled structures for further comparison. Comparing solvent accessibilities of thermophilic and mesophilic proteins, exposed and intermediate residues are observed higher in thermophiles whereas buried residues are observed higher in mesophiles. For specific five CAZYme families (GH7, GH11, GH18, GH45, and CBM1) we looked at position-specific substitutions between thermophiles and mesophiles. We also found that there are relatively more intramolecular interactions in thermophiles compared to mesophiles. Thus, we found factors such as surface exposed residues and charged residues that are highly likely to impart thermostability in fungi, and this study sets the stage for further studies in the area of fungal thermostability.
Collapse
Affiliation(s)
- Shricharan Senthilkumar
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sankar Mahesh
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Subachandran Jaisankar
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Ragothaman M Yennamalli
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
11
|
Sidhanta SPD, Sowdhamini R, Srinivasan N. Comparative analysis of permanent and transient domain-domain interactions in multi-domain proteins. Proteins 2025; 93:197-208. [PMID: 37828826 DOI: 10.1002/prot.26581] [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: 05/31/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 10/14/2023]
Abstract
Protein domains are structural, functional, and evolutionary units. These domains bring out the diversity of functionality by means of interactions with other co-existing domains and provide stability. Hence, it is important to study intra-protein inter-domain interactions from the perspective of types of interactions. Domains within a chain could interact over short timeframes or permanently, rather like protein-protein interactions (PPIs). However, no systematic study has been carried out between two classes, namely permanent and transient domain-domain interactions. In this work, we studied 263 two-domain proteins, belonging to either of these classes and their interfaces on the basis of several factors, such as interface area and details of interactions (number, strength, and types of interactions). We also characterized them based on residue conservation at the interface, correlation of residue motions across domains, its involvement in repeat formation, and their involvement in particular molecular processes. Finally, we could analyze the interactions arising from domains in two-domain monomeric proteins, and we observed significant differences between these two classes of domain interactions and a few similarities. This study will help to obtain a better understanding of structure-function and folding principles of multi-domain proteins.
Collapse
Affiliation(s)
| | - Ramanathan Sowdhamini
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Computational Approaches to Protein Science, National Centre for Biological Sciences, Bangalore, India
- Computational Biology, Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| | | |
Collapse
|
12
|
Sohn M, Kim S, Jeong HJ, Ko IY, Moon JW, Lee D, Oh J. Strategic Optimization of the Middle Domain IIIA in RBP-Albumin IIIA-IB Fusion Protein to Enhance Productivity and Thermostability. Int J Mol Sci 2024; 26:137. [PMID: 39795995 PMCID: PMC11720212 DOI: 10.3390/ijms26010137] [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: 11/29/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
The protein therapeutics market, including antibody and fusion proteins, has experienced steady growth over the past decade, underscoring the importance of optimizing amino acid sequences. In our previous study, we developed a fusion protein, R31, which combines retinol-binding protein (RBP) with albumin domains IIIA and IB, linked by a sequence (AAAA), and includes an additional disulfide bond (N227C-V254C) in IIIA. This fusion protein effectively inhibited hepatic stellate cell activation. In this study, we further optimized the sequence. The G176K mutation at the C-terminus of RBP altered the initiation site of the first α-helix in domain IIIA, shifting it from P182 to K176, and promoted polar interactions between K176 and adjacent residues, enhancing the rigidity of the RBP/IIIA interface. The introduction of an additional disulfide bond (V231C/Y250C) connecting helices 3 and 4 in IIIA resulted in a three-fold increase in productivity and a 2 °C improvement in thermal stability compared to R31. Furthermore, combining the G176K mutation with V231C/Y250C further enhanced both productivity and anti-fibrotic activity. These findings suggest that the enhanced stability of domain IIIA, conferred by V231C/Y250C, along with the increased rigidity of the RBP/IIIA interface, optimizes interdomain distance and alignment, facilitating proper protein folding.
Collapse
Affiliation(s)
- Myungho Sohn
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; (M.S.); (S.K.); (H.J.J.); (I.Y.K.)
| | - Sanggil Kim
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; (M.S.); (S.K.); (H.J.J.); (I.Y.K.)
| | - Hyeon Ju Jeong
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; (M.S.); (S.K.); (H.J.J.); (I.Y.K.)
| | - In Young Ko
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; (M.S.); (S.K.); (H.J.J.); (I.Y.K.)
| | - Ji Wook Moon
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea; (J.W.M.); (D.L.)
| | - Dowon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea; (J.W.M.); (D.L.)
| | - Junseo Oh
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea; (J.W.M.); (D.L.)
| |
Collapse
|
13
|
Sharif E, Nezafat N, Ahmadi FM, Mohit E. In Silico Design of CT26 Polytope and its Surface Display by ClearColi™-Derived Outer Membrane Vesicles as a Cancer Vaccine Candidate Against Colon Carcinoma. Appl Biochem Biotechnol 2024; 196:8820-8847. [PMID: 38958886 DOI: 10.1007/s12010-024-04971-x] [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] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
Simultaneous targeting of several mutations can be useful in colorectal cancer (CRC) due to its heterogeneity and presence of somatic mutations. As CT26 mutations and expression profiles resemble those of human CRC, we focused on designing a polyepitope vaccine based on CT26 neoepitopes. Due to its low immunogenicity, outer membrane vesicles (rOMV) as an antigen delivery system and adjuvant was applied. Herein, based on previous experimental and our in silico studies four CT26 neoepitopes with the ability to bind MHC-I and MHC-II, TCR, and induce IFN-α production were selected. To increase their immunogenicity, the gp70 and PADRE epitopes were added. The order of the neoepitopes was determined through 3D structure analysis using ProSA, Verify 3D, ERRAT, and Ramachandran servers. The stable peptide-protein docking between the selected epitopes and MHC alleles strengthen our prediction. The CT26 polytope vaccine sequence was fused to the C-terminal of cytolysin A (ClyA) anchor protein and rOMVs were isolated from endotoxin-free ClearColi™ strain. The results of the C-ImmSim server showed that the ClyA-CT26 polytope vaccine could induce T and B cells immunity.The ClyA-CT26 polytope was characterized as a soluble, stable, immunogen, and non-allergen vaccine and optimized for expression in ClearColi™ 24 h after induction with 1 mM IPTG at 25 °C. Western blot analysis confirmed the expression of ClyA-CT26 polytope by ClearColi™ and also on ClearColi™-derived rOMVs. In conclusion, we found that ClearColi™-derived rOMVs with CT26 polytope can deliver CRC neoantigens and induce antitumor immunity, but in vivo immunological studies are needed to confirm vaccine efficacy.
Collapse
Affiliation(s)
- Elham Sharif
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, No. 2660, Vali-e-Asr Ave, Tehran, 1991953381, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, No. 2660, Vali-e-Asr Ave, Tehran, 1991953381, Iran.
| |
Collapse
|
14
|
Wei Y, Qiu T, Ai Y, Zhang Y, Xie J, Zhang D, Luo X, Sun X, Wang X, Qiu J. Advances of computational methods enhance the development of multi-epitope vaccines. Brief Bioinform 2024; 26:bbaf055. [PMID: 39951549 PMCID: PMC11827616 DOI: 10.1093/bib/bbaf055] [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: 09/12/2024] [Revised: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Vaccine development is one of the most promising fields, and multi-epitope vaccine, which does not need laborious culture processes, is an attractive alternative to classical vaccines with the advantage of safety, and efficiency. The rapid development of algorithms and the accumulation of immune data have facilitated the advancement of computer-aided vaccine design. Here we systemically reviewed the in silico data and algorithms resource, for different steps of computational vaccine design, including immunogen selection, epitope prediction, vaccine construction, optimization, and evaluation. The performance of different available tools on epitope prediction and immunogenicity evaluation was tested and compared on benchmark datasets. Finally, we discuss the future research direction for the construction of a multiepitope vaccine.
Collapse
Affiliation(s)
- Yiwen Wei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Tianyi Qiu
- Institute of Clinical Science, Zhongshan Hospital; Intelligent Medicine Institute; Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, No. 180, Fenglin Road, Xuhui Destrict, Shanghai 200032, China
| | - Yisi Ai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Yuxi Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Junting Xie
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Dong Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Xiaochuan Luo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Lihu Avenue 1800, Wuxi, Jiangsu 214122, China
| | - Xin Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| | - Jingxuan Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
- Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, No. 334, Jungong Road, Yangpu District, Shanghai 200093, China
| |
Collapse
|
15
|
Zhong C, Vyas A, Liu JDH, Oostenbrink C, Nidetzky B. Keeping the Distance: Activity Control in Solid-Supported Sucrose Phosphorylase by a Rigid α-Helical Linker of Tunable Spacer Length. ACS Catal 2024; 14:17090-17102. [PMID: 39569159 PMCID: PMC11574764 DOI: 10.1021/acscatal.4c05616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024]
Abstract
Enzyme immobilization into carrier materials has broad importance in biotechnology, yet understanding the catalysis of enzymes bound to solid surfaces remains challenging. Here, we explore surface effects on the catalysis of sucrose phosphorylase through a fusion protein approach. We immobilize the enzyme via a structurally rigid α-helical linker [EA3K] n of tunable spacer length due to the variable number of pentapeptide repeats used (n = 6, 14, 19). Molecular modeling and simulation approaches delineate the conformational space sampled by each linker relative to its His-tag cap used for surface tethering. The population distribution of linker conformers gets broader, with a consequent shift of the enzyme-to-surface distance to larger values (≤15 nm), as the spacer length increases. Based on temperature kinetic studies, we obtain an energetic description of catalysis by the enzyme-to-linker fusions in solution and immobilize on Ni2+-chelate agarose. The solid-supported enzymes involve distinct changes in enthalpy-entropy partitioning within the frame of invariant Gibbs free energy of activation (ΔG ‡ = ∼61 kJ/mol at 30 °C). The entropic contribution (-TΔS ‡) to ΔG ‡ increases with the spacer length, from -16.4 kJ/mol in the linker-free enzyme to +7.9 kJ/mol in the [EA3K]19 linked fusion. The immobilized [EA3K]19 fusion protein is indistinguishable in its catalytic properties from the enzymes in solution, which behave identically regardless of their linker. Enzymes positioned closer to the surface arguably experience a higher degree of molecular organization ("rigidification") that must relax for catalysis through the additional uptake of heat, compensated by a gain in entropy. Increased thermostability of these enzymes (up to 2.8-fold) is consistent with the proposed rigidification effect. Collectively, our study reveals surface effects on the activation parameters of sucrose phosphorylase catalysis and shows their consistent dependence on the length of the surface-tethering linker. The fundamental insight here obtained, together with the successful extension of the principle to a different enzyme (nigerose phosphorylase), suggests that rigid linker-based control of the protein-surface distance can be used as an engineering strategy to optimize the activity characteristics of immobilized enzymes.
Collapse
Affiliation(s)
- Chao Zhong
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, Graz 8010, Austria
| | - Anisha Vyas
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, Graz 8010, Austria
- Austrian Centre of Industrial Biotechnology (ACIB), Krenngasse 37, Graz 8010, Austria
| | - Jakob D H Liu
- Institute of Molecular Modeling and Simulation, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, Vienna 1190, Austria
| | - Chris Oostenbrink
- Institute of Molecular Modeling and Simulation, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, Vienna 1190, Austria
| | - Bernd Nidetzky
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, Graz 8010, Austria
- Austrian Centre of Industrial Biotechnology (ACIB), Krenngasse 37, Graz 8010, Austria
| |
Collapse
|
16
|
Munetomo S, Uchiyama J, Takemura-Uchiyama I, Wanganuttara T, Yamamoto Y, Tsukui T, Hagiya H, Kanamaru S, Kanda H, Matsushita O. Examination of yield, bacteriolytic activity and cold storage of linker deletion mutants based on endolysin S6_ORF93 derived from Staphylococcus giant bacteriophage S6. PLoS One 2024; 19:e0310962. [PMID: 39441843 PMCID: PMC11498662 DOI: 10.1371/journal.pone.0310962] [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: 04/28/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus spp. present challenges in clinical and veterinary settings because effective antimicrobial agents are limited. Phage-encoded peptidoglycan-degrading enzyme, endolysin, is expected to be a novel antimicrobial agent. The enzymatic activity has recently been shown to be influenced by the linker between functional domains in the enzyme. S6_ORF93 (ORF93) is one of the endolysins derived from previously isolated Staphylococcus giant phage S6. The ORF93 was speculated to have a catalytic and peptidoglycan-binding domain with a long linker. In this study, we examined the influence of linker shortening on the characteristics of ORF93. We produce wild-type ORF93 and the linker deletion mutants using an Escherichia coli expression system. These mutants were designated as ORF93-Δ05, ORF93-Δ10, ORF93-Δ15, and ORF93-Δ20, from which 5, 10, 15, and 20 amino acids were removed from the linker, respectively. Except for the ORF93-Δ20, ORF93 and its mutants were expressed as soluble proteins. Moreover, ORF93-Δ15 showed the highest yield and bacteriolytic activity, while the antimicrobial spectrum was homologous. The cold storage experiment showed a slight effect by the linker deletion. According to our results and other studies, linker investigations are crucial in endolysin development.
Collapse
Affiliation(s)
- Sosuke Munetomo
- Department of Public Health, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Jumpei Uchiyama
- Department of Bacteriology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Iyo Takemura-Uchiyama
- Department of Bacteriology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Thamonwan Wanganuttara
- Department of Bacteriology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yumiko Yamamoto
- Department of Bacteriology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | | | - Hideharu Hagiya
- Department of Infectious Diseases, Okayama University Hospital, Kita-ku, Okayama, Japan
| | - Shuji Kanamaru
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama Kanagawa, Japan
| | - Hideyuki Kanda
- Department of Public Health, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Osamu Matsushita
- Department of Bacteriology, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
17
|
Messaabi A, Merindol N, Bohnenblust L, Fantino E, Meddeb-Mouelhi F, Desgagné-Penix I. In vivo thrombin activity in the diatom Phaeodactylum tricornutum: biotechnological insights. Appl Microbiol Biotechnol 2024; 108:481. [PMID: 39377797 PMCID: PMC11461642 DOI: 10.1007/s00253-024-13322-z] [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: 07/02/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Diatoms are responsible for 20% of global carbon dioxide fixation and have significant potential in various biotechnological and industrial applications. Recently, the pennate diatom Phaeodactylum tricornutum has emerged as a prominent platform organism for metabolic engineering and synthetic biology. The availability of its genome sequence has facilitated the development of new bioengineering tools. In this study, we used in silico analyses to identify sequences potentially encoding thrombin-like proteins, which are involved in recognizing and cleaving the thrombin sequence LVPRGS in P. tricornutum. Protein structure prediction and docking studies indicated a similar active site and ligand positioning compared to characterized human and bovine thrombin. The evidence and efficiency of the cleavage were determined in vivo using two fusion-protein constructs that included YFP to measure expression, protein accumulation, and cleavage. Western blot analysis revealed 50-100% cleavage between YFP and N-terminal fusion proteins. Our findings suggest the existence of a novel thrombin-like protease in P. tricornutum. This study advances the application of diatoms for the synthesis and production of complex proteins and enhances our understanding of the functional role of these putative thrombin sequences in diatom physiology. KEY POINTS: • Protein structure predictions reveal thrombin-like active sites in P. tricornutum. • Validated cleavage efficiency of thrombin-like protease on fusion proteins in vivo. • Study advances bioengineering tools for diatom-based biotechnological applications.
Collapse
Affiliation(s)
- Anis Messaabi
- Department of Chemistry, Biochemistry and Physics, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
| | - Natacha Merindol
- Department of Chemistry, Biochemistry and Physics, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
| | - Lea Bohnenblust
- Department of Chemistry, Biochemistry and Physics, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
| | - Elisa Fantino
- Department of Chemistry, Biochemistry and Physics, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
- Plant Biology Research Group, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
| | - Fatma Meddeb-Mouelhi
- Department of Chemistry, Biochemistry and Physics, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
- Plant Biology Research Group, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada
| | - Isabel Desgagné-Penix
- Department of Chemistry, Biochemistry and Physics, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada.
- Plant Biology Research Group, Université du Québec À Trois-Rivières, Trois-Rivières, QC, Canada.
| |
Collapse
|
18
|
Breimann S, Kamp F, Steiner H, Frishman D. AAontology: An Ontology of Amino Acid Scales for Interpretable Machine Learning. J Mol Biol 2024; 436:168717. [PMID: 39053689 DOI: 10.1016/j.jmb.2024.168717] [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: 06/03/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Amino acid scales are crucial for protein prediction tasks, many of them being curated in the AAindex database. Despite various clustering attempts to organize them and to better understand their relationships, these approaches lack the fine-grained classification necessary for satisfactory interpretability in many protein prediction problems. To address this issue, we developed AAontology-a two-level classification for 586 amino acid scales (mainly from AAindex) together with an in-depth analysis of their relations-using bag-of-word-based classification, clustering, and manual refinement over multiple iterations. AAontology organizes physicochemical scales into 8 categories and 67 subcategories, enhancing the interpretability of scale-based machine learning methods in protein bioinformatics. Thereby it enables researchers to gain a deeper biological insight. We anticipate that AAontology will be a building block to link amino acid properties with protein function and dysfunctions as well as aid informed decision-making in mutation analysis or protein drug design.
Collapse
Affiliation(s)
- Stephan Breimann
- Department of Bioinformatics, School of Life Sciences, Technical University of Munich, Freising, Germany; Ludwig-Maximilians-University Munich, Biomedical Center, Division of Metabolic Biochemistry, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Frits Kamp
- Ludwig-Maximilians-University Munich, Biomedical Center, Division of Metabolic Biochemistry, Munich, Germany
| | - Harald Steiner
- Ludwig-Maximilians-University Munich, Biomedical Center, Division of Metabolic Biochemistry, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Dmitrij Frishman
- Department of Bioinformatics, School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
19
|
Mendes G, Faulk B, Kaparthi B, Irion AR, Fong BL, Bayless K, Bondos SE. Genetic Functionalization of Protein-Based Biomaterials via Protein Fusions. Biomacromolecules 2024; 25:4639-4662. [PMID: 39074364 PMCID: PMC11323028 DOI: 10.1021/acs.biomac.4c00188] [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: 02/09/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024]
Abstract
Proteins implement many useful functions, including binding ligands with unparalleled affinity and specificity, catalyzing stereospecific chemical reactions, and directing cell behavior. Incorporating proteins into materials has the potential to imbue devices with these desirable traits. This review highlights recent advances in creating active materials by genetically fusing a self-assembling protein to a functional protein. These fusion proteins form materials while retaining the function of interest. Key advantages of this approach include elimination of a separate functionalization step during materials synthesis, uniform and dense coverage of the material by the functional protein, and stabilization of the functional protein. This review focuses on macroscale materials and discusses (i) multiple strategies for successful protein fusion design, (ii) successes and limitations of the protein fusion approach, (iii) engineering solutions to bypass any limitations, (iv) applications of protein fusion materials, including tissue engineering, drug delivery, enzyme immobilization, electronics, and biosensing, and (v) opportunities to further develop this useful technique.
Collapse
Affiliation(s)
- Gabriela
Geraldo Mendes
- Department
of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health, Bryan, Texas 77807-3260, United States
- Fralin
Biomedical Research Institute, Virginia
Tech University, Roanoke, Virginia 24016, United States
| | - Britt Faulk
- Department
of Medical Physiology, College of Medicine, Texas A&M Health, Bryan, Texas 77807, United States
- Department
of Biochemistry and Biophysics, Texas A&M
University, College Station, Texas 77843, United States
| | - Bhavika Kaparthi
- Department
of Medical Physiology, College of Medicine, Texas A&M Health, Bryan, Texas 77807, United States
| | - Andrew R. Irion
- Department
of Medical Physiology, College of Medicine, Texas A&M Health, Bryan, Texas 77807, United States
| | - Brandon Look Fong
- Department
of Medical Physiology, College of Medicine, Texas A&M Health, Bryan, Texas 77807, United States
| | - Kayla Bayless
- Department
of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health, Bryan, Texas 77807-3260, United States
- Department
of Medical Physiology, College of Medicine, Texas A&M Health, Bryan, Texas 77807, United States
| | - Sarah E. Bondos
- Department
of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health, Bryan, Texas 77807-3260, United States
- Department
of Medical Physiology, College of Medicine, Texas A&M Health, Bryan, Texas 77807, United States
- Department
of BioSciences, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
20
|
Biswas R, Swetha RG, Basu S, Roy A, Ramaiah S, Anbarasu A. Designing multi-epitope vaccine against human cytomegalovirus integrating pan-genome and reverse vaccinology pipelines. Biologicals 2024; 87:101782. [PMID: 39003966 DOI: 10.1016/j.biologicals.2024.101782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/13/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024] Open
Abstract
Human cytomegalovirus (HCMV) is accountable for high morbidity in neonates and immunosuppressed individuals. Due to the high genetic variability of HCMV, current prophylactic measures are insufficient. In this study, we employed a pan-genome and reverse vaccinology approach to screen the target for efficient vaccine candidates. Four proteins, envelope glycoprotein M, UL41A, US23, and US28, were shortlisted based on cellular localization, high solubility, antigenicity, and immunogenicity. A total of 29 B-cell and 44 T-cell highly immunogenic and antigenic epitopes with high global population coverage were finalized using immunoinformatics tools and algorithms. Further, the epitopes that were overlapping among the finalized B-cell and T-cell epitopes were linked with suitable linkers to form various combinations of multi-epitopic vaccine constructs. Among 16 vaccine constructs, Vc12 was selected based on physicochemical and structural properties. The docking and molecular simulations of VC12 were performed, which showed its high binding affinity (-23.35 kcal/mol) towards TLR4 due to intermolecular hydrogen bonds, salt bridges, and hydrophobic interactions, and there were only minimal fluctuations. Furthermore, Vc12 eliciting a good response was checked for its expression in Escherichia coli through in silico cloning and codon optimization, suggesting it to be a potent vaccine candidate.
Collapse
Affiliation(s)
- Rhitam Biswas
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Rayapadi G Swetha
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biosciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Soumya Basu
- Department of Biotechnology, NIST University, Berhampur, 761008, Odisha, India
| | - Aditi Roy
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biosciences, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India; Department of Biotechnology, SBST, VIT, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
21
|
Jiang Q, Ma Z, Min F, Ding X, Liang Y, Wang J, Liu L, Li N, Sun Y, Zhong Q, Yao G, Ma X. Screening of Bovine Coronavirus Multiepitope Vaccine Candidates: An Immunoinformatics Approach. Transbound Emerg Dis 2024; 2024:5986893. [PMID: 40303060 PMCID: PMC12016961 DOI: 10.1155/2024/5986893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/29/2024] [Accepted: 06/22/2024] [Indexed: 05/02/2025]
Abstract
Bovine coronavirus (BCoV) is a causative agent of enteric and respiratory disease in cattle. BCoV has been reported to cause a variety of animal diseases and is closely related to human coronaviruses; moreover, it has attracted extensive attention from both cattle farmers and researchers. With the rise of BCoV, a vaccine that is prophylactic and immunotherapeutic has to be utilized for a preemptive and adroit therapeutic approach. The aim of this study was to develop a novel multiepitope-based BCoV vaccine that can induce an immune response using a silicon reverse vaccinology approach. In this study, an immunoinformatics approach was employed to identify potential vaccine targets against BCoV, and four candidate antigen proteins were selected to predict B-cell and T-cell epitopes. To identify dominant epitopes, we employed a variety of bioinformatics techniques, including antigenicity prediction, immunogenicity assessment, allergenicity analysis, conservative analysis, and toxicity assessment. Finally, six multiepitope vaccines were developed using dominant epitopes, suitable adjuvants, Pan HLADR-binding epitope (PADRE), and linkers. Then based on the antigenicity score, solubility analysis, allergenicity evaluation, physicochemical property assessment, and tertiary structure verification score, construct 6 was selected as the best candidate vaccine; it was named CY. Molecular modeling and structural validation ensured the high-quality 3D structure of construct CY. The immunogenicity and complex stability of the vaccine were evaluated by molecular docking and kinetic simulation. In silicon clones, the BCoV vaccine had high levels of gene expression in the insect expression system. These results may contribute to the development of experimental BCoV vaccines with higher potency and safety.
Collapse
Affiliation(s)
- Qian Jiang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Zhigang Ma
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Fang Min
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Xiaojun Ding
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Yumeng Liang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Lu Liu
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Na Li
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Yawei Sun
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Qi Zhong
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi 830011, China
| | - Gang Yao
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| | - Xuelian Ma
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animal (XJ-KLNDSCHA), Xinjiang Agricultural University, Urumqi 830052, China
| |
Collapse
|
22
|
Roy A, Swetha RG, Basu S, Biswas R, Ramaiah S, Anbarasu A. Integrating pan-genome and reverse vaccinology to design multi-epitope vaccine against Herpes simplex virus type-1. 3 Biotech 2024; 14:176. [PMID: 38855144 PMCID: PMC11153438 DOI: 10.1007/s13205-024-04022-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024] Open
Abstract
Herpes simplex virus type-1 (HSV-1), the etiological agent of sporadic encephalitis and recurring oral (sometimes genital) infections in humans, affects millions each year. The evolving viral genome reduces susceptibility to existing antivirals and, thus, necessitates new therapeutic strategies. Immunoinformatics strategies have shown promise in designing novel vaccine candidates in the absence of a clinically licensed vaccine to prevent HSV-1. However, to encourage clinical translation, the HSV-1 pan-genome was integrated with the reverse-vaccinology pipeline for rigorous screening of universal vaccine candidates. Viral targets were screened from 104 available complete genomes. Among 364 proteins, envelope glycoprotein D being an outer membrane protein with a high antigenicity score (> 0.4) and solubility (> 0.6) was selected for epitope screening. A total of 17 T-cell and 4 B-cell epitopes with highly antigenic, immunogenic, non-toxic properties and high global population coverage were identified. Furthermore, 8 vaccine constructs were designed using different combinations of epitopes and suitable linkers. VC-8 was identified as the most potential vaccine candidate regarding chemical and structural stability. Molecular docking revealed high interactive affinity (low binding energy: - 56.25 kcal/mol) of VC-8 with the target elicited by firm intermolecular H-bonds, salt-bridges, and hydrophobic interactions, which was validated with simulations. Compatibility of the vaccine candidate to be expressed in pET-29(a) + plasmid was established by in silico cloning studies. Immune simulations confirmed the potential of VC-8 to trigger robust B-cell, T-cell, cytokine, and antibody-mediated responses, thereby suggesting a promising candidate for the future of HSV-1 prevention. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04022-6.
Collapse
Affiliation(s)
- Aditi Roy
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014 India
- Department of Biotechnology, SBST, VIT, Vellore, Tamil Nadu 632014 India
| | - Rayapadi G. Swetha
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014 India
- Department of Biosciences, SBST, VIT, Vellore, Tamil Nadu 632014 India
| | - Soumya Basu
- Department of Biotechnology, NIST University, Berhampur, Odisha 761008 India
| | - Rhitam Biswas
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014 India
- Department of Biotechnology, SBST, VIT, Vellore, Tamil Nadu 632014 India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014 India
- Department of Biosciences, SBST, VIT, Vellore, Tamil Nadu 632014 India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014 India
- Department of Biotechnology, SBST, VIT, Vellore, Tamil Nadu 632014 India
| |
Collapse
|
23
|
Park JH, Kwon S, Choi SY, Kim B, Oh J. Optimizing the Amino Acid Sequence Enhances the Productivity and Bioefficacy of the RBP-Albumin Fusion Protein. Bioengineering (Basel) 2024; 11:617. [PMID: 38927853 PMCID: PMC11200973 DOI: 10.3390/bioengineering11060617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
The significant growth of the global protein drug market, including fusion proteins, emphasizes the crucial role of optimizing amino acid sequences to enhance the productivity and bioefficacy. Among these fusion proteins, RBP-IIIA-IB, comprising retinol-binding protein in conjunction with the albumin domains, IIIA and IB, has displayed efficacy in alleviating liver fibrosis by inhibiting the activation of hepatic stellate cells (HSCs). This study aimed to address the issue of the low productivity in RBP-IIIA-IB. To induce structural changes, the linking sequence, EVDD, between domain IIIA and IB in RBP-IIIA-IB was modified to DGPG, AAAA, and GGPA. Among these, RBP-IIIA-AAAA-IB demonstrated an increase in yield (>4-fold) and a heightened inhibition of HSC activation. Furthermore, we identified amino acid residues that could form disulfide bonds when substituted with cysteine. Through the mutation of N453S-V480S in RBP-IIIA-AAAA-IB, the productivity further increased by over 9-fold, accompanied by an increase in anti-fibrotic activity. Overall, there was a more than 30-fold increase in the fusion protein's yield. These findings demonstrate the effectiveness of modifying linker sequences and introducing extra disulfide bonds to improve both the production yield and biological efficacy of fusion proteins.
Collapse
Affiliation(s)
- Ji Hoon Park
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; (J.H.P.); (S.-Y.C.)
| | - Sohyun Kwon
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - So-Young Choi
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; (J.H.P.); (S.-Y.C.)
| | - Bongcheol Kim
- Senelix Co. Ltd., 25, Beobwon-ro 11-gil, Songpa-gu, Seoul 05836, Republic of Korea;
| | - Junseo Oh
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| |
Collapse
|
24
|
Gavade A, Nagraj AK, Patel R, Pais R, Dhanure P, Scheele J, Seiz W, Patil J. Understanding the Specific Implications of Amino Acids in the Antibody Development. Protein J 2024; 43:405-424. [PMID: 38724751 DOI: 10.1007/s10930-024-10201-4] [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] [Accepted: 04/21/2024] [Indexed: 06/01/2024]
Abstract
As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.
Collapse
Affiliation(s)
- Akshata Gavade
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Anil Kumar Nagraj
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Riya Patel
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Roylan Pais
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | - Pratiksha Dhanure
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India
| | | | | | - Jaspal Patil
- Innoplexus Consulting Services Pvt Ltd, 7Th Floor, Midas Tower, Hinjawadi, Pune, Maharashtra, 411057, India.
| |
Collapse
|
25
|
van Boven MA, Mestroni M, Zwijnenburg PJG, Verhage M, Cornelisse LN. A de novo missense mutation in synaptotagmin-1 associated with neurodevelopmental disorder desynchronizes neurotransmitter release. Mol Psychiatry 2024; 29:1798-1809. [PMID: 38321119 PMCID: PMC11371641 DOI: 10.1038/s41380-024-02444-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
Synaptotagmin-1 (Syt1) is a presynaptic calcium sensor with two calcium binding domains, C2A and C2B, that triggers action potential-induced synchronous neurotransmitter release, while suppressing asynchronous and spontaneous release. We identified a de novo missense mutation (P401L) in the C2B domain in a patient with developmental delay and autistic symptoms. Expressing the orthologous mouse mutant (P400L) in cultured Syt1 null mutant neurons revealed a reduction in dendrite outgrowth with a proportional reduction in synapses. This was not observed in single Syt1PL-rescued neurons that received normal synaptic input when cultured in a control network. Patch-clamp recordings showed that spontaneous miniature release events per synapse were increased more than 500% in Syt1PL-rescued neurons, even beyond the increased rates in Syt1 KO neurons. Furthermore, action potential-induced asynchronous release was increased more than 100%, while synchronous release was unaffected. A similar shift to more asynchronous release was observed during train stimulations. These cellular phenotypes were also observed when Syt1PL was overexpressed in wild type neurons. Our findings show that Syt1PL desynchronizes neurotransmission by increasing the readily releasable pool for asynchronous release and reducing the suppression of spontaneous and asynchronous release. Neurons respond to this by shortening their dendrites, possibly to counteract the increased synaptic input. Syt1PL acts in a dominant-negative manner supporting a causative role for the mutation in the heterozygous patient. We propose that the substitution of a rigid proline to a more flexible leucine at the bottom of the C2B domain impairs clamping of release by interfering with Syt1's primary interface with the SNARE complex. This is a novel cellular phenotype, distinct from what was previously found for other SYT1 disease variants, and points to a role for spontaneous and asynchronous release in SYT1-associated neurodevelopmental disorder.
Collapse
Affiliation(s)
- Maaike A van Boven
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Marta Mestroni
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | | | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) Amsterdam, 1081 HV, Amsterdam, The Netherlands
- Department of Functional Genomics and Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam UMC-Location VUmc, 1081 HV, Amsterdam, The Netherlands
| | - L Niels Cornelisse
- Department of Functional Genomics and Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam UMC-Location VUmc, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Pike A, Pietryski C, Deighan P, Kuehner J, Lau D, Seshan A, March PE. A simple, robust, broadly applicable insertion mutagenesis method to create random fluorescent protein: target protein fusions. G3 (BETHESDA, MD.) 2024; 14:jkae036. [PMID: 38366837 PMCID: PMC11075570 DOI: 10.1093/g3journal/jkae036] [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: 10/18/2023] [Revised: 10/18/2023] [Accepted: 02/05/2024] [Indexed: 02/18/2024]
Abstract
A simple, broadly applicable method was developed using an in vitro transposition reaction followed by transformation into Escherichia coli and screening plates for fluorescent colonies. The transposition reaction catalyzes the random insertion of a fluorescent protein open reading frame into a target gene on a plasmid. The transposition reaction is employed directly in an E. coli transformation with no further procedures. Plating at high colony density yields fluorescent colonies. Plasmids purified from fluorescent colonies contain random, in-frame fusion proteins into the target gene. The plate screen also results in expressed, stable proteins. A large library of chimeric proteins was produced, which was useful for downstream research. The effect of using different fluorescent proteins was investigated as well as the dependence of the linker sequence between the target and fluorescent protein open reading frames. The utility and simplicity of the method were demonstrated by the fact that it has been employed in an undergraduate biology laboratory class without failure over dozens of class sections. This suggests that the method will be useful in high-impact research at small liberal arts colleges with limited resources. However, in-frame fusion proteins were obtained from 8 different targets suggesting that the method is broadly applicable in any research setting.
Collapse
Affiliation(s)
- Andrew Pike
- Department of Biology, Oberlin College and Conservatory, 173 W. Lorain St, Oberlin, OH 44074, USA
| | - Cassandra Pietryski
- Department of Biology, Emmanuel College, 400 The Fenway, Boston, MA 02115, USA
| | - Padraig Deighan
- Department of Biology, Emmanuel College, 400 The Fenway, Boston, MA 02115, USA
| | - Jason Kuehner
- Department of Biology, Emmanuel College, 400 The Fenway, Boston, MA 02115, USA
| | - Derek Lau
- Department of Biology, Emmanuel College, 400 The Fenway, Boston, MA 02115, USA
| | - Anupama Seshan
- Department of Biology, Emmanuel College, 400 The Fenway, Boston, MA 02115, USA
| | - Paul E March
- Department of Biology, Emmanuel College, 400 The Fenway, Boston, MA 02115, USA
| |
Collapse
|
27
|
Urbániková Ľ, Janeček Š. Trehalose synthases from the subfamily GH13_16 involved in α-glucan biosynthesis - a focus on their maltokinase domain. Int J Biol Macromol 2024; 268:131680. [PMID: 38641282 DOI: 10.1016/j.ijbiomac.2024.131680] [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: 02/21/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
The subfamily GH13_16 trehalose synthase (TreS) converts maltose to trehalose and vice versa. Typically, it consists of three domains, but it may contain a C-terminal extension exhibiting clear sequence features of a maltokinase (MaK). The present in silico study was focused on collection of naturally fused TreS-MaKs and their subsequent detailed bioinformatics analysis. Hence a set of total 3354 unique sequences was compared consisting of 1900 single TreSs, 1426 fused TreS-MaKs and 28 single MaKs. Fused TreS-MaKs were divided into five groups, namely with a standard MaK, with mutations in the maltose-binding site, of the catalytic nucleophile, of the general acid/base and of both catalytic residues. Sequence logos bearing the best conserved sequence regions were prepared for both TreSs and MaKs in an effort to find unique sequence features. In addition, linkers connecting the TreS and MaK parts in the fused enzymes were analysed. This analysis revealed that MaKs in fused enzymes have an extended N-terminal regions compared to single MaKs. Finally, the evolutionary relationships were demonstrated by phylogenetic trees of TreS parts from single TreSs and fused TreS-MaKs from the same organism as well as of single TreSs existing in multiple isoforms in the same organism.
Collapse
Affiliation(s)
- Ľubica Urbániková
- Laboratory of Protein Evolution, Institute of Molecular Biology, Slovak Academy of Sciences, SK-84551 Bratislava, Slovakia
| | - Štefan Janeček
- Laboratory of Protein Evolution, Institute of Molecular Biology, Slovak Academy of Sciences, SK-84551 Bratislava, Slovakia; Institute of Biology and Biotechnology, Faculty of Natural Sciences, University of Ss. Cyril and Methodius in Trnava, SK-91701 Trnava, Slovakia.
| |
Collapse
|
28
|
Waghmare S, Guptasarma P. 'Nunchuck' proteins: Short flexible linkers resist proteolysis by facilitating motions in flanking domains to inhibit the approach of proteases. Biochem Biophys Res Commun 2024; 706:149730. [PMID: 38461648 DOI: 10.1016/j.bbrc.2024.149730] [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: 09/06/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024]
Abstract
Peptides linking well-folded and non-interacting domains in fusion proteins can undergo proteolytic degradation. This leads to physical separation of the domains that were originally sought to be joined. In order to identify characteristics that determine linker degradation propensity, we selected a pair of thermostable, proteolytically-resistant domains, and joined them using five different linkers. We then assessed linker degradation propensities through size-exclusion chromatography, and denaturing and non-denaturing electrophoresis. The domains used were Coh2, an all-beta cohesin from C. thermocellum CipA, and BSX, a beta/alpha barrel xylanase from Bacillus sp. NG-27, while the linkers used were Rigid (3 repeats of N-EAAAK-C), Flexible (two repeats of N-SGGGG-C), Nat-full (42 residues of a Coh2-adjacent linker from CipA), Nat-half (a 21 residues-long derivative of Nat-full) and Nat-quarter (a 9 residues-long derivative of Nat-full). Both with proteolysis effected by proteases present in the environment, and with an exogenously-added protease (Subtilisin A), we found that Flexible underwent little or no degradation, whereas linkers of comparable length like Nat-quarter or Rigid underwent extensive degradation, as did longer linkers like Nat-Half and Nat-Full. Our analyses disfavor the likelihood of the sequence of Flexible being naturally resistant to proteolysis, and instead favor the explanation that the flexibility of Flexible facilitates movements of Coh2 relative to BSX which then serve to sterically prevent the approach of proteases. Thus, the construct incorporating Flexible appears to behave like a 'nunchuck' in which rods/spheres flanking a chain collide with approaching swords that are capable of severing the chain, to prevent severance.
Collapse
Affiliation(s)
- Snehal Waghmare
- Centre for Protein Science, Design and Engineering (CPSDE) and Hyperthermophile Enzyme Hydrolase Research Centre (HEHRC), Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Knowledge City, Sector-81, SAS Nagar, Punjab, 140306, India
| | - Purnananda Guptasarma
- Centre for Protein Science, Design and Engineering (CPSDE) and Hyperthermophile Enzyme Hydrolase Research Centre (HEHRC), Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Knowledge City, Sector-81, SAS Nagar, Punjab, 140306, India.
| |
Collapse
|
29
|
Zhang Y, Mariz FC, Sehr P, Spagnoli G, Koenig KM, Çelikyürekli S, Kreuziger T, Zhao X, Bolchi A, Ottonello S, Müller M. Inter-epitope spacer variation within polytopic L2-based human papillomavirus antigens affects immunogenicity. NPJ Vaccines 2024; 9:44. [PMID: 38402256 PMCID: PMC10894200 DOI: 10.1038/s41541-024-00832-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/05/2024] [Indexed: 02/26/2024] Open
Abstract
The human papillomavirus minor capsid protein L2 is being extensively explored in pre-clinical studies as an attractive vaccine antigen capable of inducing broad-spectrum prophylactic antibody responses. Recently, we have developed two HPV vaccine antigens - PANHPVAX and CUT-PANHPVAX- both based on heptameric nanoparticle antigens displaying polytopes of the L2 major cross-neutralizing epitopes of eight mucosal and twelve cutaneous HPV types, respectively. Prompted by the variable neutralizing antibody responses against some of the HPV types targeted by the antigens observed in previous studies, here we investigated the influence on immunogenicity of six distinct glycine-proline spacers inserted upstream to a specific L2 epitope. We show that spacer variants differentially influence antigen immunogenicity in a mouse model, with the antigen constructs M8merV6 and C12merV6 displaying a superior ability in the induction of neutralizing antibodies as determined by pseudovirus-based neutralization assays (PBNAs). L2-peptide enzyme-linked immunosorbent assay (ELISA) assessments determined the total anti-L2 antibody level for each antigen variant, showing for the majority of sera a correlation with their repective neutralizing antibody level. Surface Plasmon Resonance revealed that L2 epitope-specific, neutralizing monoclonal antibodies (mAbs) display distinct avidities to different antigen spacer variants. Furthermore, mAb affinity toward individual spacer variants was well correlated with their neutralizing antibody induction capacity, indicating that the mAb affinity assay predicts L2-based antigen immunogenicity. These observations provide insights on the development and optimization of L2-based HPV vaccines.
Collapse
Affiliation(s)
- Yueru Zhang
- German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Filipe Colaco Mariz
- German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Peter Sehr
- EMBL-DKFZ Chemical Biology Core Facility, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Gloria Spagnoli
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Karl Moritz Koenig
- German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Simay Çelikyürekli
- German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Tim Kreuziger
- Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Xueer Zhao
- German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany
| | - Angelo Bolchi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Simone Ottonello
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Martin Müller
- German Cancer Research Center, Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| |
Collapse
|
30
|
Park JM, Kim JH, Kim G, Sim HJ, Ahn SM, Choi KS, Kwon HJ. Rapid Antibacterial Activity Assessment of Chimeric Lysins. Int J Mol Sci 2024; 25:2430. [PMID: 38397110 PMCID: PMC10888538 DOI: 10.3390/ijms25042430] [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: 01/23/2024] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024] Open
Abstract
Various chimeric lysins have been developed as efficacious antibiotics against multidrug-resistant bacteria, but direct comparisons of their antibacterial activities have been difficult due to the preparation of multiple recombinant chimeric lysins. Previously, we reported an Escherichia coli cell-free expression method to better screen chimeric lysins against Staphylococcus aureus, but we still needed to increase the amounts of expressed proteins enough to be able to detect them non-isotopically for quantity comparisons. In this study, we improved the previous cell-free expression system by adding a previously reported artificial T7 terminator and reversing the different nucleotides between the T7 promoter and start codon to those of the T7 phage. The new method increased the expressed amount of chimeric lysins enough for us to detect them using Western blotting. Therefore, the qualitative comparison of activity between different chimeric lysins has become possible via the adjustment of the number of variables between samples without protein purification. We applied this method to select more active chimeric lysins derived from our previously reported chimeric lysin (ALS2). Finally, we compared the antibacterial activities of our selected chimeric lysins with reported chimeric lysins (ClyC and ClyO) and lysostaphin and determined the rank orders of antibacterial activities on different Staphylococcus aureus strains in our experimental conditions.
Collapse
Affiliation(s)
- Jin-Mi Park
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- GeNiner Inc., Seoul 08826, Republic of Korea
| | - Jun-Hyun Kim
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- GeNiner Inc., Seoul 08826, Republic of Korea
| | - Gun Kim
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- Laboratory of Pharmacology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hun-Ju Sim
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- Laboratory of Pharmacology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Sun-Min Ahn
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
| | - Kang-Seuk Choi
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyuk-Joon Kwon
- Laboratory of Poultry Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; (J.-M.P.); (J.-H.K.); (S.-M.A.)
- College of Veterinary Medicine and BK21 for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea; (G.K.); (H.-J.S.)
- GeNiner Inc., Seoul 08826, Republic of Korea
| |
Collapse
|
31
|
Qin Q, Jiang X, Huo L, Qian J, Yu H, Zhu H, Du W, Cao Y, Zhang X, Huang Q. Computational design and engineering of self-assembling multivalent microproteins with therapeutic potential against SARS-CoV-2. J Nanobiotechnology 2024; 22:58. [PMID: 38341574 PMCID: PMC10858482 DOI: 10.1186/s12951-024-02329-3] [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: 10/10/2023] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Multivalent drugs targeting homo-oligomeric viral surface proteins, such as the SARS-CoV-2 trimeric spike (S) protein, have the potential to elicit more potent and broad-spectrum therapeutic responses than monovalent drugs by synergistically engaging multiple binding sites on viral targets. However, rational design and engineering of nanoscale multivalent protein drugs are still lacking. Here, we developed a computational approach to engineer self-assembling trivalent microproteins that simultaneously bind to the three receptor binding domains (RBDs) of the S protein. This approach involves four steps: structure-guided linker design, molecular simulation evaluation of self-assembly, experimental validation of self-assembly state, and functional testing. Using this approach, we first designed trivalent constructs of the microprotein miniACE2 (MP) with different trimerization scaffolds and linkers, and found that one of the constructs (MP-5ff) showed high trimerization efficiency, good conformational homogeneity, and strong antiviral neutralizing activity. With its trimerization unit (5ff), we then engineered a trivalent nanobody (Tr67) that exhibited potent and broad neutralizing activity against the dominant Omicron variants, including XBB.1 and XBB.1.5. Cryo-EM complex structure confirmed that Tr67 stably binds to all three RBDs of the Omicron S protein in a synergistic form, locking them in the "3-RBD-up" conformation that could block human receptor (ACE2) binding and potentially facilitate immune clearance. Therefore, our approach provides an effective strategy for engineering potent protein drugs against SARS-CoV-2 and other deadly coronaviruses.
Collapse
Affiliation(s)
- Qin Qin
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xinyi Jiang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Liyun Huo
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jiaqiang Qian
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | | | - Haixia Zhu
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Wenhao Du
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yuhui Cao
- ACROBiosystems Inc, Beijing, 100176, China
| | - Xing Zhang
- ACROBiosystems Inc, Beijing, 100176, China
| | - Qiang Huang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
32
|
Garg A, González-Foutel NS, Gielnik MB, Kjaergaard M. Design of functional intrinsically disordered proteins. Protein Eng Des Sel 2024; 37:gzae004. [PMID: 38431892 DOI: 10.1093/protein/gzae004] [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: 10/02/2023] [Revised: 12/22/2023] [Indexed: 03/05/2024] Open
Abstract
Many proteins do not fold into a fixed three-dimensional structure, but rather function in a highly disordered state. These intrinsically disordered proteins pose a unique challenge to protein engineering and design: How can proteins be designed de novo if not by tailoring their structure? Here, we will review the nascent field of design of intrinsically disordered proteins with focus on applications in biotechnology and medicine. The design goals should not necessarily be the same as for de novo design of folded proteins as disordered proteins have unique functional strengths and limitations. We focus on functions where intrinsically disordered proteins are uniquely suited including disordered linkers, desiccation chaperones, sensors of the chemical environment, delivery of pharmaceuticals, and constituents of biomolecular condensates. Design of functional intrinsically disordered proteins relies on a combination of computational tools and heuristics gleaned from sequence-function studies. There are few cases where intrinsically disordered proteins have made it into industrial applications. However, we argue that disordered proteins can perform many roles currently performed by organic polymers, and that these proteins might be more designable due to their modularity.
Collapse
Affiliation(s)
- Ankush Garg
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | | | - Maciej B Gielnik
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
33
|
Debroy B, Chowdhury S, Pal K. Designing a novel and combinatorial multi-antigenic epitope-based vaccine "MarVax" against Marburg virus-a reverse vaccinology and immunoinformatics approach. J Genet Eng Biotechnol 2023; 21:143. [PMID: 38012426 PMCID: PMC10681968 DOI: 10.1186/s43141-023-00575-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023]
Abstract
CONTEXT Marburg virus (MARV) is a member of the Filoviridae family and causes Marburg virus disease (MVD) among humans and primates. With fatality rates going up to 88%, there is currently no commercialized cure or vaccine to combat the infection. The National Institute of Allergy and Infectious Diseases (NIAID) classified MARV as priority pathogen A, which presages the need for a vaccine candidate which can provide stable, long-term adaptive immunity. The surface glycoprotein (GP) and fusion protein (FP) mediate the adherence, fusion, and entry of the virus into the host cell via the TIM-I receptor. Being important antigenic determinants, studies reveal that GP and FP are prone to evolutionary mutations, underscoring the requirement of a vaccine construct capable of eliciting a robust and sustained immune response. In this computational study, a reverse vaccinology approach was employed to design a combinatorial vaccine from conserved and antigenic epitopes of essential viral proteins of MARV, namely GP, VP24, VP30, VP35, and VP40 along with an endogenous protein large polymerase (L). METHODS Epitopes for T-cell and B-cell were predicted using TepiTool and ElliPro, respectively. The surface-exposed TLRs like TLR2, TLR4, and TLR5 were used to screen high-binding affinity epitopes using the protein-peptide docking platform MdockPeP. The best binding epitopes were selected and assembled with linkers to design a recombinant multi-epitope vaccine construct which was then modeled in Robetta. The in silico biophysical and biochemical analyses of the recombinant vaccine were performed. The docking and MD simulation of the vaccine using WebGro and CABS-Flex against TLRs support the stable binding of vaccine candidates. A virtual immune simulation to check the immediate and long-term immunogenicity was carried out using the C-ImmSim server. RESULTS The biochemical characteristics and docking studies with MD simulation establish the recombinant protein vaccine construct MarVax as a stable, antigenic, and potent vaccine molecule. Immune simulation studies reveal 1-year passive immunity which needs to be validated by in vivo studies.
Collapse
Affiliation(s)
- Bishal Debroy
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India
| | - Sribas Chowdhury
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India
| | - Kuntal Pal
- Cancer Biology Laboratory, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India.
- School of Biosciences and Technology (SBST), Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
34
|
Debatis M, Danz H, Tremblay JM, Gaspie K, Kudej RK, Vigdorovich V, Sather N, Jaskiewicz JJ, Tzipori S, Shoemaker CB. Enteric pharmacokinetics of monomeric and multimeric camelid nanobody single-domain antibodies. PLoS One 2023; 18:e0291937. [PMID: 38011121 PMCID: PMC10681176 DOI: 10.1371/journal.pone.0291937] [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: 06/07/2023] [Accepted: 09/10/2023] [Indexed: 11/29/2023] Open
Abstract
Single-domain antibodies (sdAbs) derived from Camelidae heavy-chain-only antibodies (also called nanobodies or VHHs) have advantages over conventional antibodies in terms of their small size and stability to pH and temperature extremes, their ability to express well in microbial hosts, and to be functionally multimerized for enhanced properties. For these reasons, VHHs are showing promise as enteric disease therapeutics, yet little is known as to their pharmacokinetics (PK) within the digestive tract. To improve understanding of enteric VHH PK, we investigated the functional and structural stability of monomeric and multimeric camelid VHH-agents following in vitro incubation with intestinal extracts (chyme) from rabbits and pigs or fecal extracts from human sources, and in vivo in rabbits. The results showed that unstructured domains such as epitopic tags and flexible spacers composed of different amino acid sequences were rapidly degraded by enteric proteases while the functional core VHHs were much more stable to these treatments. Individual VHHs were widely variable in their functional stability to GI tract proteases. Some VHH-based agents which neutralize enteric Shiga toxin Stx2 displayed a functional stability to chyme incubations comparable to that of Stx2-neutralizing IgG and IgA mAbs, thus indicating that selected nanobodies can approach the functional stability of conventional immunoglobulins. Enteric PK data obtained from in vitro incubation studies were consistent with similar incubations performed in vivo in rabbit surgical gut loops. These findings have broad implications for enteric use of VHH-based agents, particularly VHH fusion proteins.
Collapse
Affiliation(s)
- Michelle Debatis
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Hillary Danz
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Jacqueline M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Kimberly Gaspie
- Division of Animal Resources, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Raymond K. Kudej
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States of America
| | - Noah Sather
- Center for Global Infectious Disease Research, Seattle Children’s Hospital, Seattle, WA, United States of America
| | - Justyna J. Jaskiewicz
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States of America
| |
Collapse
|
35
|
Wu Q, Xu C, Shi W, Li L, Zhang H, Liu T, Fan J, Cui L, Li J. Suitable carrier protein and linker peptide significantly increase the secretory expression of human lysozyme in Aspergillus niger. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1677-1680. [PMID: 37661695 PMCID: PMC10577450 DOI: 10.3724/abbs.2023153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/06/2023] [Indexed: 09/05/2023] Open
Affiliation(s)
- Qi Wu
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Can Xu
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Wei Shi
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Lifang Li
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Hui Zhang
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Tianqi Liu
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Junbo Fan
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Lingmeng Cui
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| | - Jie Li
- />College of Life SciencesNortheast Agricultural UniversityHarbin150030China
| |
Collapse
|
36
|
Wang X, Jiang Y, Liu H, Yuan H, Huang D, Wang T. Research progress of multi-enzyme complexes based on the design of scaffold protein. BIORESOUR BIOPROCESS 2023; 10:72. [PMID: 38647916 PMCID: PMC10992622 DOI: 10.1186/s40643-023-00695-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/04/2023] [Indexed: 04/25/2024] Open
Abstract
Multi-enzyme complexes designed based on scaffold proteins are a current topic in molecular enzyme engineering. They have been gradually applied to increase the production of enzyme cascades, thereby achieving effective biosynthetic pathways. This paper reviews the recent progress in the design strategy and application of multi-enzyme complexes. First, the metabolic channels in the multi-enzyme complex have been introduced, and the construction strategies of the multi-enzyme complex emerging in recent years have been summarized. Then, the discovered enzyme cascades related to scaffold proteins are discussed, emphasizing on the influence of the linker on the fusion enzyme (fusion protein) and its possible mechanism. This review is expected to provide a more theoretical basis for the modification of multi-enzyme complexes and broaden their applications in synthetic biology.
Collapse
Affiliation(s)
- Xiangyi Wang
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
| | - Yi Jiang
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
| | - Hongling Liu
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
| | - Haibo Yuan
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
| | - Di Huang
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China
| | - Tengfei Wang
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China.
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, People's Republic of China.
| |
Collapse
|
37
|
Mathony J, Aschenbrenner S, Becker P, Niopek D. Dissecting the Determinants of Domain Insertion Tolerance and Allostery in Proteins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303496. [PMID: 37562980 PMCID: PMC10558690 DOI: 10.1002/advs.202303496] [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: 05/30/2023] [Revised: 07/21/2023] [Indexed: 08/12/2023]
Abstract
Domain insertion engineering is a promising approach to recombine the functions of evolutionarily unrelated proteins. Insertion of light-switchable receptor domains into a selected effector protein, for instance, can yield allosteric effectors with light-dependent activity. However, the parameters that determine domain insertion tolerance and allostery are poorly understood. Here, an unbiased screen is used to systematically assess the domain insertion permissibility of several evolutionary unrelated proteins. Training machine learning models on the resulting data allow to dissect features informative for domain insertion tolerance and revealed sequence conservation statistics as the strongest indicators of suitable insertion sites. Finally, extending the experimental pipeline toward the identification of switchable hybrids results in opto-chemogenetic derivatives of the transcription factor AraC that function as single-protein Boolean logic gates. The study reveals determinants of domain insertion tolerance and yielded multimodally switchable proteins with unique functional properties.
Collapse
Affiliation(s)
- Jan Mathony
- Center for Synthetic BiologyTechnical University of Darmstadt64287DarmstadtGermany
- Department of BiologyTechnical University of Darmstadt64287DarmstadtGermany
- Institute of Pharmacy and Molecular Biotechnology (IPMB)Faculty of Engineering SciencesHeidelberg University69120HeidelbergGermany
| | - Sabine Aschenbrenner
- Institute of Pharmacy and Molecular Biotechnology (IPMB)Faculty of Engineering SciencesHeidelberg University69120HeidelbergGermany
| | - Philipp Becker
- Center for Synthetic BiologyTechnical University of Darmstadt64287DarmstadtGermany
- Department of BiologyTechnical University of Darmstadt64287DarmstadtGermany
- Department of Biotechnology and BiomedicineTechnical University of DenmarkKongens Lyngby2800Denmark
| | - Dominik Niopek
- Institute of Pharmacy and Molecular Biotechnology (IPMB)Faculty of Engineering SciencesHeidelberg University69120HeidelbergGermany
| |
Collapse
|
38
|
Dirvelyte E, Bujanauskiene D, Jankaityte E, Daugelaviciene N, Kisieliute U, Nagula I, Budvytyte R, Neniskyte U. Genetically encoded phosphatidylserine biosensor for in vitro, ex vivo and in vivo labelling. Cell Mol Biol Lett 2023; 28:59. [PMID: 37501184 PMCID: PMC10373266 DOI: 10.1186/s11658-023-00472-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND The dynamics of phosphatidylserine in the plasma membrane is a tightly regulated feature of eukaryotic cells. Phosphatidylserine (PS) is found preferentially in the inner leaflet of the plasma membrane. Disruption of this asymmetry leads to the exposure of phosphatidylserine on the cell surface and is associated with cell death, synaptic pruning, blood clotting and other cellular processes. Due to the role of phosphatidylserine in widespread cellular functions, an efficient phosphatidylserine probe is needed to study them. Currently, a few different phosphatidylserine labelling tools are available; however, these labels have unfavourable signal-to-noise ratios and are difficult to use in tissues due to limited permeability. Their application in living tissue requires injection procedures that damage the tissue and release damage-associated molecular patterns, which in turn stimulates phosphatidylserine exposure. METHODS For this reason, we developed a novel genetically encoded phosphatidylserine probe based on the C2 domain of the lactadherin (MFG-E8) protein, suitable for labelling exposed phosphatidylserine in various research models. We tested the C2 probe specificity to phosphatidylserine on hybrid bilayer lipid membranes by observing surface plasmon resonance angle shift. Then, we analysed purified fused C2 proteins on different cell culture lines or engineered AAVs encoding C2 probes on tissue cultures after apoptosis induction. For in vivo experiments, neurotropic AAVs were intravenously injected into perinatal mice, and after 2 weeks, brain slices were collected to observe C2-SNAP expression. RESULTS The biophysical analysis revealed the high specificity of the C2 probe for phosphatidylserine. The fused recombinant C2 proteins were suitable for labelling phosphatidylserine on the surface of apoptotic cells in various cell lines. We engineered AAVs and validated them in organotypic brain tissue cultures for non-invasive delivery of the genetically encoded C2 probe and showed that these probes were expressed in the brain in vivo after intravenous AAV delivery to mice. CONCLUSIONS We have demonstrated that the developed genetically encoded PS biosensor can be utilised in a variety of assays as a two-component system of C2 and C2m2 fusion proteins. This system allows for precise quantification and PS visualisation at directly specified threshold levels, enabling the evaluation of PS exposure in both physiological and cell death processes.
Collapse
Affiliation(s)
- Eimina Dirvelyte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daina Bujanauskiene
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Evelina Jankaityte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Neringa Daugelaviciene
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ugne Kisieliute
- Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Igor Nagula
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rima Budvytyte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
- Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| |
Collapse
|
39
|
Guo WH, Zhu YJ, Haimiti G, Xie XR, Niu C, Li M, Shi J, Yin ZW, Yu MK, Ding JB, Zhang FB. Bioinformatics-based design of a fusion vaccine with CTLA-4 variable region to combat Brucella. Braz J Med Biol Res 2023; 56:e12938. [PMID: 37493775 PMCID: PMC10361638 DOI: 10.1590/1414-431x2023e12938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/16/2023] [Indexed: 07/27/2023] Open
Abstract
Brucellosis has become a global zoonotic disease, seriously endangering the health of people all over the world. Vaccination is an effective strategy for protection against Brucella infection in livestock in developed countries. However, current vaccines are pathogenic to humans and pregnant animals, which limits their use. Therefore, it is very important to improve the safety and immune protection of Brucella vaccine. In this study, different bioinformatics approaches were carried out to predict the physicochemical properties, T/B epitope, and tertiary structure of Omp2b and Omp31. Then, these two proteins were sequentially linked, and the Cytotoxic T lymphocyte associated antigen-4 (CTLA-4) variable region was fused to the N-terminal of the epitope sequence. In addition, molecular docking was performed to show that the structure of the fusion protein vaccine had strong affinity with B7 (B7-1, B7-2). This study showed that the designed vaccine containing CTLA-4 had high potency against Brucella, which could provide a reference for the future development of efficient brucellosis vaccines.
Collapse
Affiliation(s)
- W H Guo
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Y J Zhu
- Department of Reproductive Assistance, Center for Reproductive Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - G Haimiti
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - X R Xie
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - C Niu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - M Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - J Shi
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Z W Yin
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - M K Yu
- School of Life Science and Technology, Southeast University, Nanjing, China
| | - J B Ding
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - F B Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of Central Asian High Incidence Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
40
|
Bouin A, Zhang C, Lindley ND, Truan G, Lautier T. Exploring linker's sequence diversity to fuse carotene cyclase and hydroxylase for zeaxanthin biosynthesis. Metab Eng Commun 2023; 16:e00222. [PMID: 37168436 PMCID: PMC10165439 DOI: 10.1016/j.mec.2023.e00222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/13/2023] Open
Abstract
Fusion of catalytic domains can accelerate cascade reactions by bringing enzymes in close proximity. However, the design of a protein fusion and the choice of a linker are often challenging and lack of guidance. To determine the impact of linker parameters on fusion proteins, a library of linkers featuring various lengths, secondary structures, extensions and hydrophobicities was designed. Linkers were used to fuse the lycopene cyclase (crtY) and β-carotene hydroxylase (crtZ) from Pantoea ananatis to create fusion proteins to produce zeaxanthin. The fusion efficiency was assessed by comparing the carotenoids content in a carotenoid-producer Escherichia coli strain. It was shown that in addition to the orientation of the enzymes and the size of the linker, the first amino acid of the linker is also a key factor in determining the efficiency of a protein fusion. The wide range of sequence diversity in our linker library enables the fine tuning of protein fusion and this approach can be easily transferred to other enzyme couples.
Collapse
Affiliation(s)
- Aurélie Bouin
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Congqiang Zhang
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Nic D. Lindley
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Gilles Truan
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Thomas Lautier
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
- CNRS@CREATE, 1 Create Way, #08-01 Create Tower, 138602, Singapore
| |
Collapse
|
41
|
Skalidis I, Kyrilis FL, Tüting C, Hamdi F, Träger TK, Belapure J, Hause G, Fratini M, O'Reilly FJ, Heilmann I, Rappsilber J, Kastritis PL. Structural analysis of an endogenous 4-megadalton succinyl-CoA-generating metabolon. Commun Biol 2023; 6:552. [PMID: 37217784 DOI: 10.1038/s42003-023-04885-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
The oxoglutarate dehydrogenase complex (OGDHc) participates in the tricarboxylic acid cycle and, in a multi-step reaction, decarboxylates α-ketoglutarate, transfers succinyl to CoA, and reduces NAD+. Due to its pivotal role in metabolism, OGDHc enzymatic components have been studied in isolation; however, their interactions within the endogenous OGDHc remain elusive. Here, we discern the organization of a thermophilic, eukaryotic, native OGDHc in its active state. By combining biochemical, biophysical, and bioinformatic methods, we resolve its composition, 3D architecture, and molecular function at 3.35 Å resolution. We further report the high-resolution cryo-EM structure of the OGDHc core (E2o), which displays various structural adaptations. These include hydrogen bonding patterns confining interactions of OGDHc participating enzymes (E1o-E2o-E3), electrostatic tunneling that drives inter-subunit communication, and the presence of a flexible subunit (E3BPo), connecting E2o and E3. This multi-scale analysis of a succinyl-CoA-producing native cell extract provides a blueprint for structure-function studies of complex mixtures of medical and biotechnological value.
Collapse
Affiliation(s)
- Ioannis Skalidis
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120, Halle/Saale, Germany
| | - Fotis L Kyrilis
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120, Halle/Saale, Germany
| | - Christian Tüting
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany
| | - Farzad Hamdi
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany
| | - Toni K Träger
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120, Halle/Saale, Germany
| | - Jaydeep Belapure
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany
| | - Gerd Hause
- Biozentrum, Martin Luther University Halle-Wittenberg, Weinbergweg 22, 06120, Halle/Saale, Germany
| | - Marta Fratini
- Department of Plant Biochemistry, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle/Saale, Germany
| | - Francis J O'Reilly
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute (NCI), Frederick, MD, 21702-1201, USA
| | - Ingo Heilmann
- Department of Plant Biochemistry, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle/Saale, Germany
| | - Juri Rappsilber
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355, Berlin, Germany
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3BF, Scotland, United Kingdom
| | - Panagiotis L Kastritis
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120, Halle/Saale, Germany.
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120, Halle/Saale, Germany.
- Biozentrum, Martin Luther University Halle-Wittenberg, Weinbergweg 22, 06120, Halle/Saale, Germany.
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, 11635, Greece.
| |
Collapse
|
42
|
Zheng J, Yang J, Zhang Z, Liang X, Liu S, Pan Y, Wei J, Huang Y, Huang X, Qin Q. An improved oral vaccine with molecular adjuvant β-defensin protects grouper against nervous necrosis virus infection. FISH & SHELLFISH IMMUNOLOGY 2023; 136:108709. [PMID: 36972841 DOI: 10.1016/j.fsi.2023.108709] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 06/18/2023]
Abstract
Nervous necrosis virus (NNV) is one of the most important fish viral pathogens infecting more than 120 fish species worldwide. Due to the mass mortality rates often seen among larvae and juveniles, few effective vaccines against NNV were developed up to now. Here, the protective effect of recombinant coat protein (CP) from red-spotted grouper nervous necrosis virus (RGNNV) fused with grouper β-defensin (DEFB) as an oral vaccine was evaluated using Artemia as a biocarrier delivery system in pearl gentian grouper (Epinephelus lanceolatus♂ × Epinephelus fuscoguttatus♀). Feeding with Artemia encapsulated with E. coli expressing control vector (control group), CP, or CP-DEFB showed no obvious side effects on the growth of groupers. ELISA and antibody neutralization assay showed that CP-DEFB oral vaccination group induced higher anti-RGNNV CP specific antibodies and exhibited higher neutralization potency than the CP and control group. Meanwhile, the expression levels of several immune and inflammatory factors in the spleen and kidney after feeding with CP-DEFB were also significantly increased compared with the CP group. Consistently, after challenge with RGNNV, groupers fed CP-DEFB and CP exhibited 100% and 88.23% relative percentage survival (RPS), respectively. Moreover, the lower transcription levels of viral genes and milder pathological changes in CP-DEFB group were detected compared with the CP and control group. Thus, we proposed that grouper β-defensin functioned as an efficient molecular adjuvant for an improved oral vaccine against nervous necrosis virus infection.
Collapse
Affiliation(s)
- Jiaying Zheng
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Jiahui Yang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Zemiao Zhang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Xia Liang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Shijia Liu
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Ying Pan
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Jingguang Wei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Youhua Huang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China
| | - Xiaohong Huang
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China; Fishery Institute of South China Agricultural University, Nansha, Guangzhou, China.
| |
Collapse
|
43
|
Pang Y, Liu B. TransDFL: Identification of Disordered Flexible Linkers in Proteins by Transfer Learning. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:359-369. [PMID: 36272675 PMCID: PMC10626177 DOI: 10.1016/j.gpb.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 09/21/2022] [Accepted: 10/14/2022] [Indexed: 11/27/2022]
Abstract
Disordered flexible linkers (DFLs) are the functional disordered regions in proteins, which are the sub-regions of intrinsically disordered regions (IDRs) and play important roles in connecting domains and maintaining inter-domain interactions. Trained with the limited available DFLs, the existing DFL predictors based on the machine learning techniques tend to predict the ordered residues as DFLs, leading to a high falsepositive rate (FPR) and low prediction accuracy. Previous studies have shown that DFLs are extremely flexible disordered regions, which are usually predicted as disordered residues with high confidence [P(D) > 0.9] by an IDR predictor. Therefore, transferring an IDR predictor to an accurate DFL predictor is of great significance for understanding the functions of IDRs. In this study, we proposed a new predictor called TransDFL for identifying DFLs by transferring the RFPR-IDP predictor for IDR identification to the DFL prediction. The RFPR-IDP was pre-trained with IDR sequences to learn the general features between IDRs and DFLs, which is helpful to reduce the false positives in the ordered regions. RFPR-IDP was fine-tuned with the DFL sequences to capture the specific features of DFLs so as to be transferred into the TransDFL. Experimental results of two application scenarios (prediction of DFLs only in IDRs or prediction of DFLs in entire proteins) showed that TransDFL consistently outperformed other existing DFL predictors with higher accuracy. The corresponding web server of TransDFL can be freely accessed at http://bliulab.net/TransDFL/.
Collapse
Affiliation(s)
- Yihe Pang
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Bin Liu
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China; Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
44
|
Larijani A, Kia-Karimi A, Roostaei D. Design of a multi-epitopic vaccine against Epstein-Barr virus via computer-based methods. Front Immunol 2023; 14:1115345. [PMID: 36999015 PMCID: PMC10043181 DOI: 10.3389/fimmu.2023.1115345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
BackgroundScientific findings have shown that Epstein-Barr virus (EBV) plays a key role in the development of some tumor diseases. Therefore, this study intends to take a practical step in controlling the pathogenicity of this virus by designing an effective vaccine based on the virus Capsid Envelope and Epstein–Barr nuclear immunogen (EBNA) Proteins Epitopes. Currently, there are no effective drugs or vaccines to treat or prevent EBV infection. So, we applied a computer-based strategy to design an epitope vaccineResultsWe designed a powerful multi-epitope peptide vaccine against EBV using in silico analysis. The vaccine is made up of 844 amino acids derived from three different types of proteins (Envelope, Capsid, EBNA) found in two different viral strains. responses. These epitopes have a high immunogenic capacity and are not likely to cause allergies. To enhance the vaccine immunogenicity, we used rOv-ASP-1, a recombinant Onchocerca volvulus activation associated protein-1, as an adjuvant and linked it to the vaccine’s N and C terminus. The physicochemical and immunological properties of the vaccine structure were evaluated. The proposed vaccine was stable, with a stability index of 33.57 and a pI of 10.10, according to bioinformatic predictions. Docking analysis revealed that the vaccine protein binds correctly with immunological receptors.ConclusionOur results demonstrated that the multi-epitope vaccine might be potentially immunogenic and induce humoral and cellular immune responses against EBV. This vaccine can interact appropriately with immunological receptors Also, it has a high-quality structure and suitable characteristics such as high stability.
Collapse
Affiliation(s)
- Amirhossein Larijani
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Kia-Karimi
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Davoud Roostaei
- Department of Pharmacology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- *Correspondence: Davoud Roostaei,
| |
Collapse
|
45
|
Kowalczyk A, Nisiewicz MK, Bamburowicz-Klimkowska M, Kasprzak A, Ruzycka-Ayoush M, Koszytkowska-Stawińska M, Nowicka AM. Effective voltammetric tool for simultaneous detection of MMP-1, MMP-2, and MMP-9; important non-small cell lung cancer biomarkers. Biosens Bioelectron 2023; 229:115212. [PMID: 36958204 DOI: 10.1016/j.bios.2023.115212] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
Simultaneous detection of multiple biomarkers can allow to reduce the costs of medical diagnostics, and thus improve the accuracy and effectiveness of disease diagnosis and prognosis. Here, for the first time, we present a low-cost, simple, and rapid method for simultaneous detection of three matrix metalloproteinases (MMP-1, MMP-2, and MMP-9) that play important roles in the progression of lung cancer. The sensor matrix was constructed using a G2 polyamidoamine dendrimer (PAMAM) containing amino, carboxyl, and sulfhydryl groups. The recognition process was based on specific enzymatic cleavage of the Gly-Ile peptide bond by MMP-1, Gly-Leu bond by MMP-2, and Gly-Met bond by MMP-9, and monitoring was done by square wave voltammetry. The activity of metalloproteinases was detected based on the change of current signals of redox receptors (dipeptides labeled with electroactive compounds) covalently anchored onto the electrode surface. The conditions of the biosensor construction, including the concentration of receptors on the sensor surface and the time of interaction of the receptor with the analyte, were carefully optimized. Under optimal conditions, the linear response of the developed method ranged from 1.0⋅10-8 to 1.0 mg⋅L-1, and the limit of detection for MMP-1, MMP-2, and MMP-9 was 0.35, 0.62, and 1.10 fg⋅mL-1, respectively. The constructed biosensor enabled us to efficiently profile the levels of active forms of MMP-1, MMP-2, and MMP-9 in tissue samples (plasma and lung and tumor extracts). Thus, the developed biosensor can aid in the early detection and diagnosis of lung cancer.
Collapse
Affiliation(s)
- Agata Kowalczyk
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland
| | - Monika K Nisiewicz
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland; Faculty of Chemistry, Warsaw University of Technology, Noakowskiego Str. 3, PL 00-664, Warsaw, Poland
| | - Magdalena Bamburowicz-Klimkowska
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland; Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, PL 02-097, Warsaw, Poland
| | - Artur Kasprzak
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego Str. 3, PL 00-664, Warsaw, Poland
| | - Monika Ruzycka-Ayoush
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland; Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, PL 02-097, Warsaw, Poland
| | | | - Anna M Nowicka
- Faculty of Chemistry, University of Warsaw, Pasteura Str. 1, PL 02-093, Warsaw, Poland.
| |
Collapse
|
46
|
Liu J, Maxwell M, Cuddihy T, Crawford T, Bassetti M, Hyde C, Peigneur S, Tytgat J, Undheim EAB, Mobli M. ScrepYard: An online resource for disulfide-stabilized tandem repeat peptides. Protein Sci 2023; 32:e4566. [PMID: 36644825 PMCID: PMC9885460 DOI: 10.1002/pro.4566] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023]
Abstract
Receptor avidity through multivalency is a highly sought-after property of ligands. While readily available in nature in the form of bivalent antibodies, this property remains challenging to engineer in synthetic molecules. The discovery of several bivalent venom peptides containing two homologous and independently folded domains (in a tandem repeat arrangement) has provided a unique opportunity to better understand the underpinning design of multivalency in multimeric biomolecules, as well as how naturally occurring multivalent ligands can be identified. In previous work, we classified these molecules as a larger class termed secreted cysteine-rich repeat-proteins (SCREPs). Here, we present an online resource; ScrepYard, designed to assist researchers in identification of SCREP sequences of interest and to aid in characterizing this emerging class of biomolecules. Analysis of sequences within the ScrepYard reveals that two-domain tandem repeats constitute the most abundant SCREP domain architecture, while the interdomain "linker" regions connecting the functional domains are found to be abundant in amino acids with short or polar sidechains and contain an unusually high abundance of proline residues. Finally, we demonstrate the utility of ScrepYard as a virtual screening tool for discovery of putatively multivalent peptides, by using it as a resource to identify a previously uncharacterized serine protease inhibitor and confirm its predicted activity using an enzyme assay.
Collapse
Affiliation(s)
- Junyu Liu
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Michael Maxwell
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Thom Cuddihy
- Queensland Cyber Infrastructure Foundation Ltd.The University of QueenslandSt. LuciaQueenslandAustralia,Centre for Clinical ResearchThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Theo Crawford
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| | - Madeline Bassetti
- Queensland Cyber Infrastructure Foundation Ltd.The University of QueenslandSt. LuciaQueenslandAustralia
| | - Cameron Hyde
- Queensland Cyber Infrastructure Foundation Ltd.The University of QueenslandSt. LuciaQueenslandAustralia,University of the Sunshine CoastMaroochydoreQueenslandAustralia
| | - Steve Peigneur
- Toxicology and PharmacologyUniversity of Leuven (KU Leuven)LeuvenBelgium
| | - Jan Tytgat
- Toxicology and PharmacologyUniversity of Leuven (KU Leuven)LeuvenBelgium
| | - Eivind A. B. Undheim
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia,Centre for Ecological and Evolutionary Synthesis, Department of BiosciencesUniversity of OsloOsloNorway
| | - Mehdi Mobli
- Centre for Advanced ImagingThe University of QueenslandSt. LuciaQueenslandAustralia
| |
Collapse
|
47
|
Velazquez MB, Busi MV, Gomez-Casati DF, Nag-Dasgupta C, Barchiesi J. Molecular insight into cellulose degradation by the phototrophic green alga Scenedesmus. Proteins 2023; 91:750-770. [PMID: 36607613 DOI: 10.1002/prot.26464] [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: 08/11/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Lignocellulose is the most abundant natural biopolymer on earth and a potential raw material for the production of fuels and chemicals. However, only some organisms such as bacteria and fungi produce enzymes that metabolize this polymer. In this work we have demonstrated the presence of cellulolytic activity in the supernatant of Scenedesmus quadricauda cultures and we identified the presence of extracellular cellulases in the genome of five Scenedesmus species. Scenedesmus is a green alga which grows in both freshwater and saltwater regions as well as in soils, showing highly flexible metabolic properties. Sequence comparison of the different identified cellulases with hydrolytic enzymes from other organisms using multisequence alignments and phylogenetic trees showed that these proteins belong to the families of glycosyl hydrolases 1, 5, 9, and 10. In addition, most of the Scenedesmus cellulases showed greater sequence similarity with those from invertebrates, fungi, bacteria, and other microalgae than with the plant homologs. Furthermore, the data obtained from the three dimensional structure showed that both, their global structure and the main amino acid residues involved in catalysis and substrate binding are well conserved. Based on our results, we propose that different species of Scenedesmus could act as biocatalysts for the hydrolysis of cellulosic biomass produced from sunlight.
Collapse
Affiliation(s)
- María B Velazquez
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - María V Busi
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Diego F Gomez-Casati
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | | | - Julieta Barchiesi
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
48
|
Conselheiro JA, Barone GT, Miyagi SAT, de Souza Silva SO, Agostinho WC, Aguiar J, Brandão PE. Evolution of Rabies Virus Isolates: Virulence Signatures and Effects of Modulation by Neutralizing Antibodies. Pathogens 2022; 11:pathogens11121556. [PMID: 36558890 PMCID: PMC9782306 DOI: 10.3390/pathogens11121556] [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: 11/22/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Lyssavirus rabies (RABV) is an RNA virus and, therefore, is subject to mutations due to low RNA polymerase replication fidelity, forming a population structure known as a viral quasispecies, which is the core of RNA viruses' adaptive strategy. Under new microenvironmental conditions, the fittest populations are selected, and the study of this process on the molecular level can help determine molecular signatures related to virulence. Our aim was to survey gene signatures on nucleoprotein and glycoprotein genes that might be involved in virulence modulation during the in vitro evolution of RABV lineages after serial passages in a neuronal cell system with or without the presence of neutralizing antibodies based on replicative fitness, in vivo neurotropism and protein structure and dynamics. The experiments revealed that amino acids at positions 186 and 188 of the glycoprotein are virulence factors of Lyssavirus rabies, and site 186 specifically might allow the attachment to heparan as a secondary cell receptor, while polymorphism at position 333 might allow the selection of escape mutants under suboptimal neutralizing antibodies titers.
Collapse
Affiliation(s)
- Juliana Amorim Conselheiro
- Laboratory of Diagnostics of Zoonosis and Vector-borne Diseases (LabZoo), Zoonosis Surveillance Division, Health Surveillance Coordination, Municipal Health Department, São Paulo 02031-020, SP, Brazil
- Correspondence:
| | - Gisely Toledo Barone
- Laboratory of Diagnostics of Zoonosis and Vector-borne Diseases (LabZoo), Zoonosis Surveillance Division, Health Surveillance Coordination, Municipal Health Department, São Paulo 02031-020, SP, Brazil
| | - Sueli Akemi Taniwaki Miyagi
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Sheila Oliveira de Souza Silva
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Washington Carlos Agostinho
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Joana Aguiar
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Paulo Eduardo Brandão
- Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine, University of São Paulo, São Paulo 05508-270, SP, Brazil
| |
Collapse
|
49
|
Huang TC, Fischer WB. Predicting the Assembly of the Transmembrane Domains of Viral Channel Forming Proteins and Peptide Drug Screening Using a Docking Approach. Biomolecules 2022; 12:biom12121844. [PMID: 36551274 PMCID: PMC9775931 DOI: 10.3390/biom12121844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
A de novo assembly algorithm is provided to propose the assembly of bitopic transmembrane domains (TMDs) of membrane proteins. The algorithm is probed using, in particular, viral channel forming proteins (VCPs) such as M2 of influenza A virus, E protein of severe acute respiratory syndrome corona virus (SARS-CoV), 6K of Chikungunya virus (CHIKV), SH of human respiratory syncytial virus (hRSV), and Vpu of human immunodeficiency virus type 2 (HIV-2). The generation of the structures is based on screening a 7-dimensional space. Assembly of the TMDs can be achieved either by simultaneously docking the individual TMDs or via a sequential docking. Scoring based on estimated binding energies (EBEs) of the oligomeric structures is obtained by the tilt to decipher the handedness of the bundles. The bundles match especially well for all-atom models of M2 referring to an experimentally reported tetrameric bundle. Docking of helical poly-peptides to experimental structures of M2 and E protein identifies improving EBEs for positively charged (K,R,H) and aromatic amino acids (F,Y,W). Data are improved when using polypeptides for which the coordinates of the amino acids are adapted to the Cα coordinates of the respective experimentally derived structures of the TMDs of the target proteins.
Collapse
|
50
|
Vij S, Thakur R, Rishi P. Reverse engineering approach: a step towards a new era of vaccinology with special reference to Salmonella. Expert Rev Vaccines 2022; 21:1763-1785. [PMID: 36408592 DOI: 10.1080/14760584.2022.2148661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Salmonella is responsible for causing enteric fever, septicemia, and gastroenteritis in humans. Due to high disease burden and emergence of multi- and extensively drug-resistant Salmonella strains, it is becoming difficult to treat the infection with existing battery of antibiotics as we are not able to discover newer antibiotics at the same pace at which the pathogens are acquiring resistance. Though vaccines against Salmonella are available commercially, they have limited efficacy. Advancements in genome sequencing technologies and immunoinformatics approaches have solved the problem significantly by giving rise to a new era of vaccine designing, i.e. 'Reverse engineering.' Reverse engineering/vaccinology has expedited the vaccine identification process. Using this approach, multiple potential proteins/epitopes can be identified and constructed as a single entity to tackle enteric fever. AREAS COVERED This review provides details of reverse engineering approach and discusses various protein and epitope-based vaccine candidates identified using this approach against typhoidal Salmonella. EXPERT OPINION Reverse engineering approach holds great promise for developing strategies to tackle the pathogen(s) by overcoming the limitations posed by existing vaccines. Progressive advancements in the arena of reverse vaccinology, structural biology, and systems biology combined with an improved understanding of host-pathogen interactions are essential components to design new-generation vaccines.
Collapse
Affiliation(s)
- Shania Vij
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Reena Thakur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|