1
|
Wang J, Yang S, Wang X, Zhang L, Zhao Y. Comparative efficacy of natural seed coats in regulating protein aggregation in pre-roasted pine kernels and enhancing associated techno-functionality. Food Chem 2025; 479:143766. [PMID: 40101380 DOI: 10.1016/j.foodchem.2025.143766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
To investigate the regulatory effect of pine nut seed coats on protein techno-functionality during pre-roasting, proteins from kernels subjected to various treatments, including de-shelling, de-skinning, and roasting with or without seed coat, were compared in terms of gelation behavior and interfacial properties. Results indicated that roasting without the seed coat caused disordered unfolding of proteins and the formation of heterogeneous, blocky protein aggregates. In contrast, skin-coating facilitated polyphenol binding with proteins by 2.5-fold, promoting ordered aggregation. Solubility, emulsification activity, emulsion stability, and foaming capacity increased by 34.52 %, 210.46 %, 59.51 %, and 55.54 %, respectively, while the gel network formed uniformly. Shell-coating promoted the formation of heat-stable aggregates, characterized by strong hydrogen bonds, disulfide bonds, and α-helical conformation. The seed coat was found to mediate roasting-induced modifications in protein spatial conformations and aggregate morphological transformations. This study proposes a novel strategy for modulating the functionality of nut proteins.
Collapse
Affiliation(s)
- Jiarong Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, People's Republic of China
| | - Shuang Yang
- College of Life Science, Northeast Forestry University, Harbin 150040, People's Republic of China
| | - Xuemei Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, People's Republic of China
| | - Ligang Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yuhong Zhao
- College of Life Science, Northeast Forestry University, Harbin 150040, People's Republic of China.
| |
Collapse
|
2
|
Elmaci DN, Hopping G, Hoffmann W, Muttenthaler M, Stein M. The structural integrity of human TFF1 under reducing conditions. Redox Biol 2025; 81:103534. [PMID: 39978303 PMCID: PMC11889601 DOI: 10.1016/j.redox.2025.103534] [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: 01/07/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
The trefoil factor family (TFF) comprises three secretory peptides (TFF1, TFF2, TFF3) that regulate diverse physiological processes to maintain gastrointestinal mucosal integrity and homeostasis. The TFF domain is stabilized by six conserved cysteine residues forming three intramolecular disulfide bonds. In this work, we investigated human TFF1 domain stability against increasing concentrations of the reducing agent tris(2-carboxyethyl)phosphine (TCEP). Experiments revealed high resistance of the disulfide bonds within the TFF1 domain to reduction compared to two reference peptides with similar three-disulfide frameworks, namely the bovine pancreatic trypsin inhibitor (BPTI) and the peptide drug linaclotide. Full reduction of TFF1 was only achieved with a large excess of TCEP (150-fold), and no partially reduced intermediates were observed, supporting a compact TFF1 domain. This TFF1 domain stability was supported by extensive all-atom molecular dynamics simulations for a total of 24 μs of all possible combinatorial states of disulfide bond reduction. Despite minor structural and conformational changes observed upon reduction, the domain substantially retained its overall compactness and solvent exposure when only one or two disulfide bonds were removed. The reduced cysteine residues did not undergo large structural rearrangements and remained buried. The loss of covalent disulfide bonds upon reduction was counterbalanced through persistent non-covalent interactions. These molecular simulations explained why TFF1 could not be partially reduced and alkylated during the experiments despite titrating different TCEP concentrations in the presence of alkylating agents. Our findings provide the first insights into the remarkable stability of the human TFF domain under reducing conditions, supporting its functional resilience upon expression and secretion throughout the human body.
Collapse
Affiliation(s)
- Dilsah Nur Elmaci
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany
| | - Gene Hopping
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia
| | - Werner Hoffmann
- Institute for Molecular Biology and Medicinal Chemistry, Medical Faculty Otto von Guericke University, 39120 Magdeburg, Germany
| | - Markus Muttenthaler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane 4072, Australia; Institute of Biological Chemistry, University of Vienna, 1090 Vienna, Austria.
| | - Matthias Stein
- Molecular Simulations and Design Group, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Germany.
| |
Collapse
|
3
|
Willis LF, Kapur N, Radford SE, Brockwell DJ. Biophysical Analysis of Therapeutic Antibodies in the Early Development Pipeline. Biologics 2024; 18:413-432. [PMID: 39723199 PMCID: PMC11669289 DOI: 10.2147/btt.s486345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
The successful progression of therapeutic antibodies and other biologics from the laboratory to the clinic depends on their possession of "drug-like" biophysical properties. The techniques and the resultant biophysical and biochemical parameters used to characterize their ease of manufacture can be broadly defined as developability. Focusing on antibodies, this review firstly discusses established and emerging biophysical techniques used to probe the early-stage developability of biologics, aimed towards those new to the field. Secondly, we describe the inter-relationships and redundancies amongst developability assays and how in silico methods aid the efficient deployment of developability to bring a new generation of cost-effective therapeutic proteins from bench to bedside more quickly and sustainably.
Collapse
Affiliation(s)
- Leon F Willis
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Nikil Kapur
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Sheena E Radford
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - David J Brockwell
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
4
|
Zhou K, Xia Y. High-Coverage Disulfide Mapping Enabled by Programmable Disulfide-Ene Reaction Integrated onto a Bottom-Up Protein Analysis Workflow. Anal Chem 2024; 96:17396-17404. [PMID: 39425647 DOI: 10.1021/acs.analchem.4c04257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Mapping disulfide linkages is crucial for characterizing pharmaceutical proteins during drug development and quality control. Traditional bottom-up protein analysis workflows often suffer from incomplete mapping for tryptic peptides consisting of multiple disulfide bonds. Although the employment of a partial reduction of disulfide bonds can improve disulfide mapping, it becomes a bottleneck of analysis because individual tuning is often needed. Herein, we have developed an online disulfide-ene reaction system in which the composition of the reaction solvent can be programmed to achieve optimal partial reduction of tryptic disulfide peptides after liquid chromatography separation. By coupling this system onto a bottom-up protein analysis workflow, high coverage for sequencing (71-83%) and disulfide mapping (84-100%) was achieved for standard proteins consisting of 4-19 disulfide bonds. The analytical capability was further demonstrated by mapping 13 scrambled disulfide bonds in lysozyme and achieving compositional analysis of IgG isotypes (κ and λ) and subclasses (IgG1-IgG4) from human plasma.
Collapse
Affiliation(s)
- Keting Zhou
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 10084, China
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 10084, China
| |
Collapse
|
5
|
Ma YH, Wang C, Yang J, Li B, Han X, Lu X. Disulfide-Driven Charge and Hydrophobicity Rearrangement of Remodeled Membrane Proteins toward Amyloid-Type Aggregation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:16145-16150. [PMID: 39054779 DOI: 10.1021/acs.langmuir.4c01201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
As a common pathological hallmark, protein aggregation into amyloids is a highly complicated phenomenon, attracting extensive research interest for elucidating its structural details and formation mechanisms. Membrane deposition and disulfide-driven protein misfolding play critical roles in amyloid-type aggregation, yet the underlying molecular process remains unclear. Here, we employed sum frequency generation (SFG) vibrational spectroscopy to comprehensively investigate the remodeling process of lysozyme, as the model protein, into amyloid-type aggregates at the cell membrane interface. It was discovered that disulfide reduction concurrently induced the transition of membrane-bound lysozyme from predominantly α-helical to antiparallel β-sheet structures, under a mode switch of membrane interaction from electrostatic to hydrophobic, and subsequent oligomeric aggregation. These findings shed light on the systematic understanding of dynamic molecular mechanisms underlying membrane-interactive amyloid oligomer formation.
Collapse
Affiliation(s)
- Yong-Hao Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Chu Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Jingjing Yang
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bolin Li
- Anhui Key Laboratory of Condensed Matter Physics at Extreme Conditions, High Magnetic Field Laboratory, HFIPS, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiaofeng Han
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiaolin Lu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
6
|
Melo EP, El-Guendouz S, Correia C, Teodoro F, Lopes C, Martel PJ. A Conformational-Dependent Interdomain Redox Relay at the Core of Protein Disulfide Isomerase Activity. Antioxid Redox Signal 2024; 41:181-200. [PMID: 38497737 DOI: 10.1089/ars.2023.0288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Aims: Protein disulfide isomerases (PDIs) are a family of chaperones resident in the endoplasmic reticulum (ER). In addition to holdase function, some members catalyze disulfide bond formation and isomerization, a crucial step for native folding and prevention of aggregation of misfolded proteins. PDIs are characterized by an arrangement of thioredoxin-like domains, with the canonical protein disulfide isomerase A1 (PDIA1) organized as four thioredoxin-like domains forming a horseshoe with two active sites, a and a', at the extremities. We aimed to clarify important aspects underlying the catalytic cycle of PDIA1 in the context of the full pathways of oxidative protein folding operating in the ER. Results: Using two fluorescent redox sensors, redox green fluorescent protein 2 (roGFP2) and HyPer (circularly permutated yellow fluorescent protein containing the regulatory domain of the H2O2-sensing protein OxyR), either unfolded or native, as client substrates, we identified the N-terminal a active site of PDIA1 as the main oxidant of thiols. From there, electrons can flow to the C-terminal a' active site, with the redox-dependent conformational flexibility of PDIA1 allowing the formation of an interdomain disulfide bond. The a' active site then acts as a crossing point to redirect electrons to ER downstream oxidases or back to client proteins to reduce scrambled disulfide bonds. Innovation and Conclusions: The two active sites of PDIA1 work cooperatively as an interdomain redox relay mechanism that explains PDIA1 oxidative activity to form native disulfides and PDIA1 reductase activity to resolve scrambled disulfides. This mechanism suggests a new rationale for shutting down oxidative protein folding under ER redox imbalance. Whether it applies to physiological substrates in cells remains to be shown.
Collapse
Affiliation(s)
- Eduardo P Melo
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | | | - Cátia Correia
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | - Fernando Teodoro
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | - Carlos Lopes
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | | |
Collapse
|
7
|
Li T, Li Z, Chen F, Zhu L, Tang H, Wang D, Tang Z. Impact of BSA and Au 3+ concentration on the formation and fluorescence properties of Au nanoclusters. RSC Adv 2024; 14:19284-19293. [PMID: 38887651 PMCID: PMC11181134 DOI: 10.1039/d4ra01140f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
Bovine serum albumin-stabilized Au nanoclusters (BSA-Au NCs) have emerged as promising contenders for imaging agents and highly sensitive fluorescence sensors due to their biocompatibility and strong photoluminescence. Optimizing the synthesis conditions of BSA-Au NCs is crucial for enhancing fluorescence imaging and other nanocluster applications. In this study, for the first time, we systematically investigated the effects of BSA concentration and Au3+ on both particle size and optical characteristics of BSA-Au NCs. When the two components achieved a suitable concentration ratio, it was beneficial to form BSA-Au NCs with a high quantum yield (QY = 74.30%) and good fluorescence stability. In contrast, an inappropriate concentration ratio would lead to the formation of gold nanoparticles (Au NPs), and their internal filtration effect (IFE) would attenuate the fluorescence emission of BSA-Au NCs. The BSA-Au NCs were then employed as efficient fluorescence sensors for detecting Hg2+. Furthermore, the growth mechanism of BSA-Au NCs was elucidated by monitoring fluorescence changes during different incubation times. The BSA-Au NCs with a high quantum yield introduce a novel synthetic concept for sensitive fluorescent probes and expanding versatile applications of BSA-Au NCs in catalysis, chemical sensing and biomedicine.
Collapse
Affiliation(s)
- Tao Li
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University Chongqing China
| | - Zhuo Li
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University Chongqing China
| | - Fengjiao Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University Chongqing China
| | - Liying Zhu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University Guiyang China
| | - Hua Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University Chongqing China
| | - Dan Wang
- Post-Doctoral Research Center, The People's Hospital of Rongchang District Chongqing China
| | - Zhenrong Tang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University Chongqing China
| |
Collapse
|
8
|
Liu X, Guo Z, Wang J, Shen W, Jia Z, Jia S, Li L, Wang J, Wang L, Li J, Sun Y, Chen Y, Zhang M, Bai J, Wang L, Li X. Thiolation-Based Protein-Protein Hydrogels for Improved Wound Healing. Adv Healthc Mater 2024; 13:e2303824. [PMID: 38303578 DOI: 10.1002/adhm.202303824] [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: 11/02/2023] [Revised: 01/28/2024] [Indexed: 02/03/2024]
Abstract
The limitations of protein-based hydrogels, including their insufficient mechanical properties and restricted biological functions, arise from the highly specific functions of proteins as natural building blocks. A potential solution to overcome these shortcomings is the development of protein-protein hydrogels, which integrate structural and functional proteins. In this study, a protein-protein hydrogel formed by crosslinking bovine serum albumin (BSA) and a genetically engineered intrinsically disordered collagen-like protein (CLP) through Ag─S bonding is introduced. The approach involves thiolating lysine residues of BSA and crosslinking CLP with Ag+ ions, utilizing thiolation of BSA and the free-cysteines of CLP. The resulting protein-protein hydrogels exhibit exceptional properties, including notable plasticity, inherent self-healing capabilities, and gel-sol transition in response to redox conditions. In comparison to standalone BSA hydrogels, these protein-protein hydrogels demonstrate enhanced cellular viability, and improved cellular migration. In vivo experiments provide conclusive evidence of accelerated wound healing, observed not only in murine models with streptozotocin (Step)-induced diabetes but also in zebrafish models subjected to UV-burn injuries. Detailed mechanistic insights, combined with assessments of proinflammatory cytokines and the expression of epidermal differentiation-related proteins, robustly validate the protein-protein hydrogel's effectiveness in promoting wound repair.
Collapse
Affiliation(s)
- Xing Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Zhao Guo
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jie Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Wenting Shen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhenzhen Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, 250014, P. R. China
| | - Shuang Jia
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Limiao Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jieqi Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Liping Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jiaqi Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Yinan Sun
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Yufang Chen
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Min Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Jia Bai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Liyao Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| | - Xinyu Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Institute of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, P .R. China
| |
Collapse
|
9
|
Swarupa S, Thareja P. Techniques, applications and prospects of polysaccharide and protein based biopolymer coatings: A review. Int J Biol Macromol 2024; 266:131104. [PMID: 38522703 DOI: 10.1016/j.ijbiomac.2024.131104] [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: 07/12/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
The growing relevance of sustainable materials has recently led to the exploration of naturally derived biopolymeric hydrogels as coating materials due to their biodegradability, biocompatibility, ease of fabrication and modification. Although many review articles exist on biopolymeric coatings, they mainly focus on a specific polysaccharide, protein biopolymer, or a particular application- biomedical engineering or food preservation. The current review first summarizes the commonly used polysaccharide and protein-based biopolymers like chitosan, alginate, carrageenan, pectin, cellulose, starch, pullulan, agarose and silk fibroin, gelatin, respectively, with a systematic description of the techniques widely used for physical coating on substrates. Then, broad applications of these biopolymeric coatings on various substrates in biomedical engineering- 3D scaffolds, biomedical implants, and nanoparticles are described in detail. It also entails the application of biopolymeric coatings for food preservation in the form of food packaging and edible coatings. A brief discussion on the newly discovered interest in exploring biopolymers for anticorrosive coating applications is also included. Finally, concluding remarks on the role of biopolymer microstructures in forming homogeneous coatings, prospective alternatives to the currently used biopolymers as coating material and the advent of computer-aided technologies to expedite experimental findings are presented.
Collapse
Affiliation(s)
- Sanchari Swarupa
- Biological Sciences and Engineering, IIT Gandhinagar, Palaj, Gujarat 382355, India
| | - Prachi Thareja
- Chemical Engineering, Dr. Kiran C. Patel Centre for Sustainable Development, IIT Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
10
|
Shao Y, Du G, Luo B, Liu T, Zhao J, Zhang S, Wang J, Chi M, Cai C, Liu Y, Meng X, Liu Z, Wang S, Nie S. A Tough Monolithic-Integrated Triboelectric Bioplastic Enabled by Dynamic Covalent Chemistry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311993. [PMID: 38183330 DOI: 10.1002/adma.202311993] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Electronic waste is a growing threat to the global environment and human health, raising particular concerns. Triboelectric devices synthesized from sustainable and degradable materials are a promising electronic alternative, but the mechanical mismatch at the interface between the polymer substrate and the electrodes remains unresolved in practical applications. This study uses the sulfhydryl silanization reaction and the chemical selectivity and site specificity of the thiol-disulfide exchange reaction in dynamic covalent chemistry to prepare a tough monolithic-integrated triboelectric bioplastic. The stress is dissipated by covalent bond adaptation to the interface interaction, which makes the polymer dielectric layer to the conductive layer have a good interface adhesion effect (220.55 kPa). The interfacial interlocking of the polymer substrate with the conductive layer gives the triboelectric bioplastic excellent tensile strength (87.4 MPa) and fracture toughness (33.3 MJ m-3). Even when subjected to a tension force of 10 000 times its weight, it still maintains a stable triboelectric output with no visible cracks. This study provides new insights into the design of reliable and environmentally friendly self-powered devices, which is significant for the development of flexible wearable electronics.
Collapse
Affiliation(s)
- Yuzheng Shao
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Guoli Du
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Bin Luo
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Tao Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Jiamin Zhao
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Song Zhang
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Jinlong Wang
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Mingchao Chi
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Chenchen Cai
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Yanhua Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Xiangjiang Meng
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Zhaomeng Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Shuangfei Wang
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| | - Shuangxi Nie
- School of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, P. R. China
| |
Collapse
|
11
|
Ng YK, Konermann L. Mechanism of Protein Aggregation Inhibition by Arginine: Blockage of Anionic Side Chains Favors Unproductive Encounter Complexes. J Am Chem Soc 2024; 146:8394-8406. [PMID: 38477601 DOI: 10.1021/jacs.3c14180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Aggregation refers to the assembly of proteins into nonphysiological higher order structures. While amyloid has been studied extensively, much less is known about amorphous aggregation, a process that interferes with protein expression and storage. Free arginine (Arg+) is a widely used aggregation inhibitor, but its mechanism remains elusive. Focusing on myoglobin (Mb), we recently applied atomistic molecular dynamics (MD) simulations for gaining detailed insights into amorphous aggregation (Ng J. Phys. Chem. B 2021, 125, 13099). Building on that approach, the current work for the first time demonstrates that MD simulations can directly elucidate aggregation inhibition mechanisms. Comparative simulations with and without Arg+ reproduced the experimental finding that Arg+ significantly decreased the Mb aggregation propensity. Our data reveal that, without Arg+, protein-protein encounter complexes readily form salt bridges and hydrophobic contacts, culminating in firmly linked dimeric aggregation nuclei. Arg+ promotes the dissociation of encounter complexes. These "unproductive" encounter complexes are favored because Arg+ binding to D- and E- lowers the tendency of these anionic residues to form interprotein salt bridges. Side chain blockage is mediated largely by the guanidinium group of Arg+, which binds carboxylates through H-bond-reinforced ionic contacts. Our MD data revealed Arg+ self-association into a dynamic quasi-infinite network, but we found no evidence that this self-association is important for protein aggregation inhibition. Instead, aggregation inhibition by Arg+ is similar to that mediated by free guanidinium ions. The computational strategy used here should be suitable for the rational design of aggregation inhibitors with enhanced potency.
Collapse
Affiliation(s)
- Yuen Ki Ng
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Lars Konermann
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| |
Collapse
|
12
|
Senft MD, Maier R, Hiremath A, Zhang F, Schreiber F. Effective interactions and phase behavior of protein solutions in the presence of hexamine cobalt(III) chloride. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:119. [PMID: 38051398 PMCID: PMC10698144 DOI: 10.1140/epje/s10189-023-00376-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023]
Abstract
It is well established that deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) exhibit a reentrant condensation (RC) phase behavior in the presence of the trivalent hexamine cobalt(III) cations (Hac) which can be important for their packing and folding. A similar behavior can be observed for negatively charged globular proteins in the presence of trivalent metal cations, such as Y3+ or La3+. This phase behavior is mainly driven by charge inversion upon an increasing salt concentration for a fixed protein concentration (cp). However, as Hac exhibits structural differences compared to other multivalent metal cations, with six ammonia ligands (NH3) covalently bonded to the central cobalt atom, it is not clear that Hac can induce a similar phase behavior for proteins. In this work, we systematically investigate whether negatively charged globular proteins β-lactoglobulin (BLG), bovine serum albumin (BSA), human serum albumin (HSA) and ovalbumin (OVA) feature Hac-induced RC. Effective protein-protein interactions were investigated by small-angle X-ray scattering. The reduced second virial coefficient (B2/B2HS) was obtained as a function of salt concentration. The virial coefficient analysis performed confirms the reentrant interaction (RI) behavior for BLG without actually inducing RC, given the insufficient strengths of the interactions for the latter to occur. In contrast, the strength of attraction for BSA, HSA and OVA are too weak to show RC. Model free analysis of the inverse intensity [Formula: see text] also supports this finding. Looking at different q-range by employing static (SLS) and dynamic light scattering experiments, the presence of RI behavior can be confirmed. The results are further discussed in view of metal cation binding sites in nucleic acids (DNA and RNA), where Hac induced RC phase behavior.
Collapse
Affiliation(s)
- Maximilian D Senft
- Institut für Angewandte Physik, Universität Tübingen, Auf der Morgenstelle 10, 72076, Tübingen, Germany.
| | - Ralph Maier
- Institut für Angewandte Physik, Universität Tübingen, Auf der Morgenstelle 10, 72076, Tübingen, Germany
| | - Anusha Hiremath
- Institut für Angewandte Physik, Universität Tübingen, Auf der Morgenstelle 10, 72076, Tübingen, Germany
| | - Fajun Zhang
- Institut für Angewandte Physik, Universität Tübingen, Auf der Morgenstelle 10, 72076, Tübingen, Germany.
| | - Frank Schreiber
- Institut für Angewandte Physik, Universität Tübingen, Auf der Morgenstelle 10, 72076, Tübingen, Germany
| |
Collapse
|
13
|
Li J, Sonje J, Suryanarayanan R. Role of Poloxamer 188 in Preventing Ice-Surface-Induced Protein Destabilization during Freeze-Thawing. Mol Pharm 2023; 20:4587-4596. [PMID: 37535010 DOI: 10.1021/acs.molpharmaceut.3c00312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
The phase behavior of poloxamer 188 (P188) in aqueous solutions, characterized by differential scanning calorimetry (DSC) and synchrotron X-ray diffractometry, revealed solute crystallization during both freezing and thawing. Sucrose and trehalose inhibited P188 crystallization during freeze-thawing (FT). While trehalose inhibited P188 crystallization only during cooling, sucrose completely suppressed P188 crystallization during both cooling and heating. Lactate dehydrogenase (LDH) served as a model protein to evaluate the stabilizing effect of P188. The ability of P188, over a concentration range of 0.003-0.800% w/v, to prevent LDH (10 μg/mL) destabilization was evaluated. After five FT cycles, the aggregation behavior (by dynamic light scattering) and activity recovery were evaluated. While LDH alone was sensitive to interfacial stress, P188 at concentrations of ≥0.100% w/v stabilized the protein. However, as the surfactant concentration decreased, protein aggregation after FT increased. The addition of sugar (1.0% w/v; sucrose or trehalose) improved the stabilizing function of P188 at lower concentrations (≤0.010% w/v), possibly due to the inhibition of surfactant crystallization. Based on a comparison with the stabilization effect of polysorbate (both 20 and 80), it was evident that P188 could be a promising alternative surfactant in frozen protein formulations. However, when the surfactant concentration is low, the potential for P188 crystallization and the consequent compromise in its functionality warrant careful consideration.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jayesh Sonje
- Pfizer Biotherapeutics, Pfizer Inc., Andover, Massachusetts 01810, United States
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
14
|
Zhang Y, Liu Y, Liu Y, Zuo P, Miao S, Hu B, Kang Y, Liu W, Yang Q, Ren H, Yang P. α-Helix-Mediated Protein Adhesion. J Am Chem Soc 2023; 145:17125-17135. [PMID: 37505921 DOI: 10.1021/jacs.3c03581] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Proteins have been adopted by natural living organisms to create robust bioadhesive materials, such as biofilms and amyloid plaques formed in microbes and barnacles. In these cases, β-sheet stacking is recognized as a key feature that is closely related to the interfacial adhesion of proteins. Herein, we challenge this well-known recognition by proposing an α-helix-mediated interfacial adhesion model for proteins. By using bovine serum albumin (BSA) as a model protein, it was discovered that the reduction of disulfide bonds in BSA results in random coils from unfolded BSA dragging α-helices to gather at the solid/liquid interface (SLI). The hydrophobic residues in the α-helix then expose and break through the hydration layer of the SLI, followed by the random deposition of hydrophilic and hydrophobic residues to achieve interfacial adhesion. As a result, the first assembled layer is enriched in the α-helix secondary structure, which is then strengthened by intermolecular disulfide bonds and further initiates stepwise layering protein assembly. In this process, β-sheet stacking is transformed from the α-helix in a gradually evolving manner. This finding thus indicates a valuable clue that β-sheet-featuring amyloid may form after the interfacial adhesion of proteins. Furthermore, the finding of the α-helix-mediated interfacial adhesion model of proteins affords a unique strategy to prepare protein nanofilms with a well-defined layer number, presenting robust and modulable adhesion on various substrates and exhibiting good resistance to acid, alkali, organic solvent, ultrasonic, and adhesive tape peeling.
Collapse
Affiliation(s)
- Yingying Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Yongchun Liu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Yonggang Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Ping Zuo
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Shuting Miao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Bowen Hu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Yu Kang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Wei Liu
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Qingmin Yang
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Hao Ren
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
15
|
Mavi PS, Singh S, Kumar A. Media component bovine serum albumin facilitates the formation of mycobacterial biofilms in response to reductive stress. BMC Microbiol 2023; 23:111. [PMID: 37081437 PMCID: PMC10116703 DOI: 10.1186/s12866-023-02853-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Mycobacterium tuberculosis (Mtb) forms physiologically relevant biofilms harboring drug-tolerant bacteria. This observation has brought the study of mycobacterial biofilms to the forefront of tuberculosis research. We established earlier that dithiothreitol (DTT) mediated reductive stress induces cellulose-rich biofilm formation in Mtb cultures. The molecular events associated with the DTT-induced biofilm formation are not known. Furthermore, there are only limited tools for monitoring the presence of cellulose in biofilms. RESULTS To decipher the molecular events associated with DTT-induced biofilm formation, we used Mtb and non-pathogenic, fast-growing Mycobacterium smegmatis (Msm). We observed that DTT induces biofilm formation in Msm cultures. We explored whether media components facilitate biofilm formation in mycobacteria upon exposure to DTT. We observed that media component bovine serum albumin promotes mycobacterial biofilm formation in response to DTT. Furthermore, we analyzed the composition of extracellular polymeric substances of Msm biofilms. We found that, like Mtb biofilms, Msm biofilms are also rich in polysaccharides and proteins. We also developed a novel protein-based molecular probe for imaging cellulose by utilizing the cellulose-binding domain of cellulase CenA from Cellulomonas fimi and fusing it to fluorescent reporter mCherry. Characterization of this new probe revealed that it has a high affinity for cellulose and could be used for visualizing cellulose biosynthesis during the development of Agrobacterium biofilms. Furthermore, we have demonstrated that biological macromolecule cellulose is present in the extracellular polymeric substances of Msm biofilms using this novel probe. CONCLUSIONS This study indicates that DTT-mediated reduction of media component BSA leads to the formation of nucleating foci. These nucleating foci are critical for subsequent attachment of bacterial cells and induction of EPS production. Furthermore, this new tool, IMT-CBD-mC, could be used for monitoring cellulose incorporation in plant cells, understanding cellulose biosynthesis dynamics during biofilm formation, etc.
Collapse
Affiliation(s)
- Parminder Singh Mavi
- Institute of Microbial Technology, Council of Scientific and Industrial Research, Room No 508, Sector 39 A, Chandigarh, India, 160036
| | - Shweta Singh
- Institute of Microbial Technology, Council of Scientific and Industrial Research, Room No 508, Sector 39 A, Chandigarh, India, 160036
| | - Ashwani Kumar
- Institute of Microbial Technology, Council of Scientific and Industrial Research, Room No 508, Sector 39 A, Chandigarh, India, 160036.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India, 201002.
| |
Collapse
|
16
|
Malik A, Khan JM, Alhomida AS, Ola MS, Alokail MS, Khan MS, Alenad AM, Altwaijry N, Alafaleq NO, Odeibat H. Agitation does not induce fibrillation in reduced hen egg-white lysozyme at physiological temperature and pH. J Mol Recognit 2023; 36:e3009. [PMID: 36841950 DOI: 10.1002/jmr.3009] [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: 10/10/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Several proteins and peptides tend to form an amyloid fibril, causing a range of unrelated diseases, from neurodegenerative to certain types of cancer. In the native state, these proteins are folded and soluble. However, these proteins acquired β-sheet amyloid fibril due to unfolding and aggregation. The conversion mechanism from well-folded soluble into amorphous or amyloid fibril is not well understood yet. Here, we induced unfolding and aggregation of hen egg-white lysozyme (HEWL) by reducing agent dithiothreitol and applied mechanical sheering force by constant shaking (1000 rpm) on the thermostat for 7 days. Our turbidity results showed that reduced HEWL rapidly formed aggregates, and a plateau was attained in nearly 5 h of incubation in both shaking and non-shaking conditions. The turbidity was lower in the shaking condition than in the non-shaking condition. The thioflavin T binding and transmission electron micrographs showed that reduced HEWL formed amorphous aggregates in both conditions. Far-UV circular dichroism results showed that reduced HEWL lost nearly all alpha-helical structure, and β-sheet secondary structure was not formed in both conditions. All the spectroscopic and microscopic results showed that reduced HEWL formed amorphous aggregates under both conditions.
Collapse
Affiliation(s)
- Ajamaluddin Malik
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Javed Masood Khan
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah S Alhomida
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Shamsul Ola
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Majed S Alokail
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Amal M Alenad
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nojood Altwaijry
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nouf Omar Alafaleq
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hamza Odeibat
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Wang Y, Chen X, Xu X, Du M, Wu C. Reducing disulfide bonds as a robust strategy to facilitate the self-assembly of cod protein fabricating potential active ingredients-nanocarrier. Colloids Surf B Biointerfaces 2023; 222:113080. [PMID: 36542952 DOI: 10.1016/j.colsurfb.2022.113080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/24/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
In this study, a novel method was developed to encapsulate hydrophobic compounds by self-assembly of cod protein (CP) triggered by breaking disulfide bonds. Curcumin (Cur), a representative lipid-soluble polyphenol, was selected as a model to evaluate the potential of CP nanoparticles as novel and accessible nanocarriers. Results showed that the protein structure gradually unfolded with increasing dithiothreitol (DTT) concentration, indicating that S-S cleavage was conducive to forming a looser structure. The resultant unfolded CP exposed more hydrophobic sites, facilitating its interaction with hydrophobic compounds. The encapsulation efficiency (EE) of formed CP-Cur nanoparticles was relatively high, reaching 99.09%, 98.8%, and 89.77% when the mass ratios of CP to Cur were 20:1, 10:1, and 5:1 (w/v), respectively. The hydrophobic interaction, weak van der Waals, and hydrogen bond were the forces contributing to the formation of CP-Cur nanoparticles, whereas the hydrophobic interaction played a crucial role. The CP-Cur complex exhibited increased stability and a homogeneous-stable structural phase. Thus, this research not only proposed a novel and simple encapsulation method of hydrophobic bioactive compounds but also provided a theoretical reference for the application of reductants in food or pharmacy system.
Collapse
Affiliation(s)
- Yuying Wang
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China; College of Food Science, Jilin University, Changchun 130015, China
| | - Xufei Chen
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China
| | - Xianbing Xu
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China
| | - Ming Du
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China
| | - Chao Wu
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
18
|
Effects of Molecular Crowding and Betaine on HSPB5 Interactions, with Target Proteins Differing in the Quaternary Structure and Aggregation Mechanism. Int J Mol Sci 2022; 23:ijms232315392. [PMID: 36499725 PMCID: PMC9737104 DOI: 10.3390/ijms232315392] [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: 11/17/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
The aggregation of intracellular proteins may be enhanced under stress. The expression of heat-shock proteins (HSPs) and the accumulation of osmolytes are among the cellular protective mechanisms in these conditions. In addition, one should remember that the cell environment is highly crowded. The antiaggregation activity of HSPB5 and the effect on it of either a crowding agent (polyethylene glycol (PEG)) or an osmolyte (betaine), or their mixture, were tested on the aggregation of two target proteins that differ in the order of aggregation with respect to the protein: thermal aggregation of glutamate dehydrogenase and DTT-induced aggregation of lysozyme. The kinetic analysis of the dynamic light-scattering data indicates that crowding can decrease the chaperone-like activity of HSPB5. Nonetheless, the analytical ultracentrifugation shows the protective effect of HSPB5, which retains protein aggregates in a soluble state. Overall, various additives may either improve or impair the antiaggregation activity of HSPB5 against different protein targets. The mixed crowding arising from the presence of PEG and 1 M betaine demonstrates an extraordinary effect on the oligomeric state of protein aggregates. The shift in the equilibrium of HSPB5 dynamic ensembles allows for the regulation of its antiaggregation activity. Crowding can modulate HSPB5 activity by affecting protein-protein interactions.
Collapse
|
19
|
Wang Y, Chen X, Xu X, Du M, Zhu B, Wu C. Disulfide bond-breaking induced structural unfolding and assembly of soy protein acting as a nanovehicle for curcumin. INNOV FOOD SCI EMERG 2022. [DOI: 10.1016/j.ifset.2022.103188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Identification of peptides with antioxidant, anti-lipoxygenase, anti-xanthine oxidase and anti-tyrosinase activities from velvet antler blood. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
21
|
Cao H, Lu Q, Wei H, Zhang S. Phosphorylcholine zwitterionic shell-detachable mixed micelles for enhanced cancerous cellular uptakes and increased DOX release. J Mater Chem B 2022; 10:5624-5632. [PMID: 35815797 DOI: 10.1039/d2tb01061e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To further enhance the cancerous cellular uptakes and increase the drug release of the drug loaded micelles, herein, we fabricated a series of mixed micelles with different mass ratios using two amphiphilic copolymers P(DMAEMA-co-MaPCL) and PCL-SS-PMPC. The mixed micelles showed a prolonged circulation time due to the zwitterionic shells in a physiological environment (pH 7.4). In addition, because of the protonation of tertiary amine groups in PDMAEMA and the breakage of the disulfide bond in PMPC-SS-PCL in a tumor microenvironment, the mixed micelles aggregated, which led to enhanced cancerous cellular penetration and increased DOX release. Moreover, cytotoxicity assay showed that the mixed micelles had good biocompatibility to L929, HeLa and MCF-7 cells, even at a concentration of up to 1 mg mL-1. Furthermore, enhanced antitumour activity and cellular uptake of HeLa and MCF-7 cells were detected after loading with DOX, which was determined by confocal laser scanning microscopy (CLSM) and flow cytometry (FC), especially for the DOX@MIX 3 micelles (20% mass ratio of the P(DMAEMA-co-MaPCL)). Therefore, the mixed strategy provides a simple and efficient ways to promote anticancer drug delivery.
Collapse
Affiliation(s)
- Haimei Cao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, P. R. China.
| | - Qian Lu
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, P. R. China.
| | - Henan Wei
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, P. R. China.
| | - Shiping Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, P. R. China.
| |
Collapse
|
22
|
Kleinberg A, Joseph R, Mao Y, Li N. Ultrasensitive disulfide scrambling analysis of mAbs by LC-MS with post-column reduction and glycine signal enhancement. Anal Biochem 2022; 653:114773. [PMID: 35688259 DOI: 10.1016/j.ab.2022.114773] [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: 01/12/2022] [Revised: 05/20/2022] [Accepted: 05/31/2022] [Indexed: 11/15/2022]
Abstract
Explicitly confirming the complete disulfide bond linkage pattern of a monoclonal antibody (mAb) presents a challenge in the biopharmaceutical industry. Although proper native disulfide connections are in high abundance for analytical purposes within a peptide mapping digest under non-reducing conditions, disulfide scrambling can also exist but be difficult to detect, let alone characterize, particularly at low levels. Here, we developed an ultrasensitive high-confidence method for identifying explicit disulfide connectivity in mAbs. By applying a post-column addition of tris (2-carboxyethyl)phosphine hydrochloride (TCEP) to the liquid chromatography (LC) eluent of a non-reduced mAb digest, partial reduction of disulfide peptides is achieved after the initial peptide separation, allowing both the parent disulfide and its reduced daughter peptides to co-elute for simultaneous mass spectrometry (MS) detection. Combining this concept with the recently discovered ability of glycine to enhance MS signal when added to the LC eluent, we demonstrate a method for detecting, characterizing and quantifying low-abundance disulfide scrambling in mAbs.
Collapse
Affiliation(s)
- Andrew Kleinberg
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, United States
| | - Rachel Joseph
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, United States
| | - Yuan Mao
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, United States.
| | - Ning Li
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, United States
| |
Collapse
|
23
|
Kang K, Platten F. Electric-field induced modulation of amorphous protein aggregates: polarization, deformation, and reorientation. Sci Rep 2022; 12:3061. [PMID: 35197521 PMCID: PMC8866516 DOI: 10.1038/s41598-022-06995-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/09/2022] [Indexed: 11/09/2022] Open
Abstract
Proteins in their native state are only marginally stable and tend to aggregate. However, protein misfolding and condensation are often associated with undesired processes, such as pathogenesis, or unwanted properties, such as reduced biological activity, immunogenicity, or uncontrolled materials properties. Therefore, controlling protein aggregation is very important, but still a major challenge in various fields, including medicine, pharmacology, food processing, and materials science. Here, flexible, amorphous, micron-sized protein aggregates composed of lysozyme molecules reduced by dithiothreitol are used as a model system. The preformed amorphous protein aggregates are exposed to a weak alternating current electric field. Their field response is followed in situ by time-resolved polarized optical microscopy, revealing field-induced deformation, reorientation and enhanced polarization as well as the disintegration of large clusters of aggregates. Small-angle dynamic light scattering was applied to probe the collective microscopic dynamics of amorphous aggregate suspensions. Field-enhanced local oscillations of the intensity auto-correlation function are observed and related to two distinguishable elastic moduli. Our results validate the prospects of electric fields for controlling protein aggregation processes.
Collapse
Affiliation(s)
- Kyongok Kang
- Forschungszentrum Jülich, Institute of Biological Information Processing IBI-4, Biomacromolecular Systems and Processes, Jülich, Germany.
| | - Florian Platten
- Forschungszentrum Jülich, Institute of Biological Information Processing IBI-4, Biomacromolecular Systems and Processes, Jülich, Germany.
- Condensed Matter Physics Laboratory, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
24
|
Ng YK, Tajoddin NN, Scrosati PM, Konermann L. Mechanism of Thermal Protein Aggregation: Experiments and Molecular Dynamics Simulations on the High-Temperature Behavior of Myoglobin. J Phys Chem B 2021; 125:13099-13110. [PMID: 34808050 DOI: 10.1021/acs.jpcb.1c07210] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Proteins that encounter unfavorable solvent conditions are prone to aggregation, a phenomenon that remains poorly understood. This work focuses on myoglobin (Mb) as a model protein. Upon heating, Mb produces amorphous aggregates. Thermal unfolding experiments at low concentration (where aggregation is negligible), along with centrifugation assays, imply that Mb aggregation proceeds via globally unfolded conformers. This contrasts studies on other proteins that emphasized the role of partially folded structures as aggregate precursors. Molecular dynamics (MD) simulations were performed to gain insights into the mechanism by which heat-unfolded Mb molecules associate with one another. A prerequisite for these simulations was the development of a method for generating monomeric starting structures. Periodic boundary condition artifacts necessitated the implementation of a partially immobilized water layer lining the walls of the simulation box. Aggregation simulations were performed at 370 K to track the assembly of monomeric Mb into pentameric species. Binding events were preceded by multiple unsuccessful encounters. Even after association, protein-protein contacts remained in flux. Binding was mediated by hydrophobic contacts, along with salt bridges that involved hydrophobically embedded Lys residues. Overall, this work illustrates that atomistic MD simulations are well suited for garnering insights into protein aggregation mechanisms.
Collapse
Affiliation(s)
- Yuen Ki Ng
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Nastaran N Tajoddin
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Pablo M Scrosati
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Lars Konermann
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| |
Collapse
|
25
|
Shiratori T, Goto S, Sakaguchi T, Kasai T, Otsuka Y, Higashi K, Makino K, Takahashi H, Komatsu K. Singular value decomposition analysis of the secondary structure features contributing to the circular dichroism spectra of model proteins. Biochem Biophys Rep 2021; 28:101153. [PMID: 34712848 PMCID: PMC8528683 DOI: 10.1016/j.bbrep.2021.101153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Amyloid fibril formation occurs in restricted environment, such as the interface between intercellular fluids and bio-membranes. Conformational interconversion from α-helix to β-structure does not progress in fluids; however, it can occur after sedimentary aggregation during amyloid fibril formation induced by heat treatment of hen egg white lysozyme (HEWL). Secondary structures of various proteins and denatured proteins titrated with 2,2,2-trifluoroethanol (TFE) were examined using their CD spectra. Gaussian peak/trough and singular value decompositions (SVD) showed that the spectral pattern of the α-helix comprised a sharp trough at wavelength 207 nm and a broad trough at 220 nm. Conversely, we distinguished two patterns for β-sheet-a spread barrel type, corresponding to ConA, and a tightly weaved type, corresponding to the soybean trypsin inhibitor. Herein, we confirmed that the spectral/conformational interconversion of the heat-treated HEWL was not observed in the dissolved fluid.
Collapse
Affiliation(s)
- Tomoki Shiratori
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Satoru Goto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Tomoyo Sakaguchi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Takahiro Kasai
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Yuta Otsuka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kyohei Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kosho Makino
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Hideyo Takahashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kazushi Komatsu
- Department of Mathematics, Faculty of Science, Kochi University, 2-5-1 Akebonocho, Kochi, 780-8520, Japan
| |
Collapse
|
26
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
27
|
Wang X, Bowman J, Tu S, Nykypanchuk D, Kuksenok O, Minko S. Polyethylene Glycol Crowder's Effect on Enzyme Aggregation, Thermal Stability, and Residual Catalytic Activity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:8474-8485. [PMID: 34236863 DOI: 10.1021/acs.langmuir.1c00872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Protein stability and performance in various natural and artificial systems incorporating many other macromolecules for therapeutic, diagnostic, sensor, and biotechnological applications attract increasing interest with the expansion of these technologies. Here we address the catalytic activity of lysozyme protein (LYZ) in the presence of a polyethylene glycol (PEG) crowder in a broad range of concentrations and temperatures in aqueous solutions of two different molecular mass PEG samples (Mw = 3350 and 10000 g/mol). The phase behavior of PEG-protein solutions is examined by using dynamic light scattering (DLS) and small-angle X-ray scattering (SAXS), while the enzyme denaturing is monitored by using an activity assay (AS) and circular dichroism (CD) spectroscopy. Molecular dynamic (MD) simulations are used to illustrate the effect of PEG concentration on protein stability at high temperatures. The results demonstrate that LYZ residual activity after 1 h incubation at 80 °C is improved from 15% up to 55% with the addition of PEG. The improvement is attributed to two underlying mechanisms. (i) Primarily, the stabilizing effect is due to the suppression of the enzyme aggregation because of the stronger PEG-protein interactions caused by the increased hydrophobicity of PEG and lysozyme at elevated temperatures. (ii) The MD simulations showed that the addition of PEG to some degree stabilizes the secondary structures of the enzyme by delaying unfolding at elevated temperatures. The more pronounced effect is observed with an increase in PEG concentration. This trend is consistent with CD and AS experimental results, where the thermal stability is strengthened with increasing of PEG concentration and molecular mass. The results show that the highest stabilizing effect is approached at the critical overlap concentration of PEG.
Collapse
Affiliation(s)
- Xue Wang
- Nanostructured Materials Lab, University of Georgia, Athens, Georgia 30602, United States
| | - Jeremy Bowman
- Nanostructured Materials Lab, University of Georgia, Athens, Georgia 30602, United States
| | - Sidong Tu
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Dmytro Nykypanchuk
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, New York 11973, United States
| | - Olga Kuksenok
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Sergiy Minko
- Nanostructured Materials Lab, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
28
|
Jain K, Salamat-Miller N, Taylor K. Freeze-thaw characterization process to minimize aggregation and enable drug product manufacturing of protein based therapeutics. Sci Rep 2021; 11:11332. [PMID: 34059716 PMCID: PMC8166975 DOI: 10.1038/s41598-021-90772-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 05/04/2021] [Indexed: 11/29/2022] Open
Abstract
Physical instabilities of proteins in the form of protein aggregation continue to be a major challenge in the development of protein drug candidates. Aggregation can occur during different stages of product lifecycle such as freeze–thaw, manufacturing, shipping, and storage, and can potentially delay commercialization of candidates. A lack of clear understanding of the underlying mechanism(s) behind protein aggregation and the potential immunogenic reactions renders the presence of aggregates in biotherapeutic products undesirable. Understanding and minimizing aggregation can potentially reduce immunogenic responses and make protein therapeutics safer. Therefore, it is imperative to identify, understand, and control aggregation during early formulation development and develop reliable and orthogonal analytical methodologies to detect and monitor levels of aggregation. Freezing and thawing are typical steps involved in the manufacturing of drug product and could result in complex physical and chemical changes, which in turn could potentially cause protein aggregation. This study provides a systematic approach in understanding and selecting the ideal freeze–thaw conditions for manufacturing of protein-based therapeutics. It identifies the importance of balancing different excipients with an overall goal of sufficiently reducing or eliminating aggregation and developing a stable and scalable formulation. The results demonstrated that the freeze–thaw damage of mAb-1 in aqueous solutions was significantly reduced by identification of optimal freeze–thaw conditions using first a small-scale model with subsequent at-scale verifications. The work provides a framework for successful transfer of drug product manufacturing process from small-scale to the manufacturing scale production environment especially for molecules that are susceptible to freeze–thaw induced degradations.
Collapse
Affiliation(s)
- Keethkumar Jain
- Takeda Pharmaceutical Company Limited, 200 Shire Way, Lexington, MA, 02421, USA.
| | | | - Katherine Taylor
- Takeda Pharmaceutical Company Limited, 200 Shire Way, Lexington, MA, 02421, USA
| |
Collapse
|
29
|
Tzeng SR, Tseng YC, Lin CC, Hsu CY, Huang SJ, Kuo YT, Chang CI. Molecular insights into substrate recognition and discrimination by the N-terminal domain of Lon AAA+ protease. eLife 2021; 10:64056. [PMID: 33929321 PMCID: PMC8087443 DOI: 10.7554/elife.64056] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/02/2021] [Indexed: 11/13/2022] Open
Abstract
The Lon AAA+ protease (LonA) is a ubiquitous ATP-dependent proteolytic machine, which selectively degrades damaged proteins or native proteins carrying exposed motifs (degrons). Here we characterize the structural basis for substrate recognition and discrimination by the N-terminal domain (NTD) of LonA. The results reveal that the six NTDs are attached to the hexameric LonA chamber by flexible linkers such that the formers tumble independently of the latter. Further spectral analyses show that the NTD selectively interacts with unfolded proteins, protein aggregates, and degron-tagged proteins by two hydrophobic patches of its N-lobe, but not intrinsically disordered substrate, α-casein. Moreover, the NTD selectively binds to protein substrates when they are thermally induced to adopt unfolded conformations. Collectively, our findings demonstrate that NTDs enable LonA to perform protein quality control to selectively degrade proteins in damaged states and suggest that substrate discrimination and selective degradation by LonA are mediated by multiple NTD interactions. There are many different types of protein which each have different roles in biology. Most proteins are surrounded by water and are folded so that their water-attracting regions are on the outside and more fat-like regions, which repel water, are on the inside. When a protein becomes damaged or is assembled incorrectly, some of the fat-like regions end up on the outside of the protein and become exposed to water. This can prevent the protein from performing its role and harm the cell instead. LonA proteases are responsible for dismantling and recycling these harmful proteins, as well as proteins that have been labelled for destruction. They do this by unfolding the unwanted protein and transporting it into an enclosed chamber made of six LonA molecules. Once inside the chamber, the target protein is broken down into smaller fragments that can be used to build other structures. LonA proteases contain a region called the N-terminal domain, or NTD for short, which is thought to be responsible for identifying which proteins need degrading. Yet it remained unclear how the NTD recognizes and binds to these target proteins. To answer this question, Tzeng et al. studied the detailed structure of a LonA protease that had been purified from bacteria cells. This revealed that the NTD of LonA contains two water-repelling regions which bind to fat-like segments on the surface of proteins that have become unfolded or tagged for destruction. Further experiments showed that the NTD is bound to the main body of LonA via a ‘flexible linker’. This led Tzeng et al. to propose that the NTD sways around loosely at the end of LonA searching for proteins with exposed water-repelling regions. Once an NTD identifies and attaches to a target, the NTDs of the other LonA molecules then bind to the protein and help insert it into the chamber. Proteases are a vital component of all biological systems. Controlling protein destruction and recycling is a key factor in how cells divide and respond to a changing environment. This study provides new insights into how LonA operates in bacteria, which may apply to proteases more widely. This contributes to our knowledge of fundamental biology and may also be relevant in a range of diseases where protein recycling is defective or inefficient.
Collapse
Affiliation(s)
- Shiou-Ru Tzeng
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yin-Chu Tseng
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chu Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chia-Ying Hsu
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shing-Jong Huang
- Instrumentation Center, National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Kuo
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-I Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Sarimov RM, Binhi VN, Matveeva TA, Penkov NV, Gudkov SV. Unfolding and Aggregation of Lysozyme under the Combined Action of Dithiothreitol and Guanidine Hydrochloride: Optical Studies. Int J Mol Sci 2021; 22:2710. [PMID: 33800175 PMCID: PMC7962454 DOI: 10.3390/ijms22052710] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 11/16/2022] Open
Abstract
Using a number of optical techniques (interferometry, dynamic light scattering, and spectroscopy), denaturation of hen egg white lysozyme (HEWL) by treatment with a combination of dithiothreitol (DTT) and guanidine hydrochloride (GdnHCl) has been investigated. The denaturing solutions were selected so that protein denaturation occurred with aggregation (Tris-HCl pH = 8.0, 50 mM, DTT 30 mM) or without aggregation (Tris-HCl pH = 8.0, 50 mM, DTT 30 mM, GdnHCl 6 M) and can be evaluated after 60 min of treatment. It has been found that denatured by solution with 6 M GdnHCl lysozyme completely loses its enzymatic activity after 30 min and the size of the protein molecule increases by 1.5 times, from 3.8 nm to 5.7 nm. Denaturation without of GdnHCl led to aggregation with preserving about 50% of its enzymatic activity. Denaturation of HEWL was examined using interferometry. Previously, it has been shown that protein denaturation that occurs without subsequent aggregation leads to an increase in the refractive index (Δn ~ 4.5 × 10-5). This is most likely due to variations in the HEWL-solvent interface area. By applying modern optical techniques conjointly, it has been possible to obtain information on the nature of time-dependent changes that occur inside a protein and its hydration shell as it undergoes denaturation.
Collapse
Affiliation(s)
- Ruslan M. Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St., 38, 119991 Moscow, Russia; (R.M.S.); (V.N.B.); (T.A.M.)
| | - Vladimir N. Binhi
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St., 38, 119991 Moscow, Russia; (R.M.S.); (V.N.B.); (T.A.M.)
| | - Tatiana A. Matveeva
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St., 38, 119991 Moscow, Russia; (R.M.S.); (V.N.B.); (T.A.M.)
| | - Nikita V. Penkov
- Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS, Institutskaya St., 3, Pushchino, 142290 Moscow, Russia;
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St., 38, 119991 Moscow, Russia; (R.M.S.); (V.N.B.); (T.A.M.)
| |
Collapse
|
31
|
Catalini S, Perinelli DR, Sassi P, Comez L, Palmieri GF, Morresi A, Bonacucina G, Foggi P, Pucciarelli S, Paolantoni M. Amyloid Self-Assembly of Lysozyme in Self-Crowded Conditions: The Formation of a Protein Oligomer Hydrogel. Biomacromolecules 2021; 22:1147-1158. [PMID: 33600168 PMCID: PMC8023603 DOI: 10.1021/acs.biomac.0c01652] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
A method
is designed to quickly form protein hydrogels, based on
the self-assembly of highly concentrated lysozyme solutions in acidic
conditions. Their properties can be easily modulated by selecting
the curing temperature. Molecular insights on the gelation pathway,
derived by in situ FTIR spectroscopy, are related to calorimetric
and rheological results, providing a consistent picture on structure–property
correlations. In these self-crowded samples, the thermal unfolding
induces the rapid formation of amyloid aggregates, leading to temperature-dependent
quasi-stationary levels of antiparallel cross β-sheet links,
attributed to kinetically trapped oligomers. Upon subsequent cooling,
thermoreversible hydrogels develop by the formation of interoligomer
contacts. Through heating/cooling cycles, the starting solutions can
be largely recovered back, due to oligomer-to-monomer dissociation
and refolding. Overall, transparent protein hydrogels can be easily
formed in self-crowding conditions and their properties explained,
considering the formation of interconnected amyloid oligomers. This
type of biomaterial might be relevant in different fields, along with
analogous systems of a fibrillar nature more commonly considered.
Collapse
Affiliation(s)
- Sara Catalini
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Italy
| | | | - Paola Sassi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Lucia Comez
- IOM-CNR c/o Department of Physics and Geology, University of Perugia, 060123 Perugia, Italy
| | | | - Assunta Morresi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | | | - Paolo Foggi
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Italy.,Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy.,National Metrological Research Institute (INRIM), Strada delle Cacce 91, 10135 Torino, Italy
| | - Stefania Pucciarelli
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Marco Paolantoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
32
|
Sakuma C, Tomioka Y, Li C, Shibata T, Nakagawa M, Kurosawa Y, Arakawa T, Akuta T. Analysis of protein denaturation, aggregation and post-translational modification by agarose native gel electrophoresis. Int J Biol Macromol 2021; 172:589-596. [PMID: 33454336 DOI: 10.1016/j.ijbiomac.2021.01.075] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Agarose native gel electrophoresis has been developed to separate proteins and protein complexes in the native state. Here, we applied this technology to analyze proteins that undergo degradation, post-translational modification or chemical/physical changes. Antibodies showed aggregation/association upon acid or heat treatment. Limited reduction of disulfide bonds resulted in non-covalent aggregation of bovine serum albumin and cleavage of only inter-chain linkages of an antibody that had no effects on its overall structure. Native agarose gel analysis showed changes in mobility of human transferrin upon Fe3+ binding. Analysis of a commercial glycated human hemoglobin A1c showed no difference in electrophoretic pattern from un-modified hemoglobin. Native agarose gel showed aggregation of a virus upon acid or heat treatment. We have extracted bands of bovine serum albumin from the agarose native gel for sodium dodecylsulfate gel electrophoresis analysis, showing degradation of aged sample. Lastly, we analyzed phosphorylation of Zap70 kinase by native gel and Western blotting. These applications should expand the utility of this native gel electrophoresis technology.
Collapse
Affiliation(s)
- Chiaki Sakuma
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., 3333-26, Aza-Asayama, Kamitezuna, Takahagi-shi, Ibaraki 318-0004, Japan
| | - Yui Tomioka
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., 3333-26, Aza-Asayama, Kamitezuna, Takahagi-shi, Ibaraki 318-0004, Japan
| | - Cynthia Li
- HTL Biosolutions, Inc., 4010 Adoflo Road, Ste. B, Camrillo, CA 93012, USA
| | - Takashi Shibata
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., 3333-26, Aza-Asayama, Kamitezuna, Takahagi-shi, Ibaraki 318-0004, Japan
| | - Masataka Nakagawa
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., 3333-26, Aza-Asayama, Kamitezuna, Takahagi-shi, Ibaraki 318-0004, Japan
| | - Yasunori Kurosawa
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., 3333-26, Aza-Asayama, Kamitezuna, Takahagi-shi, Ibaraki 318-0004, Japan; Abwiz Bio Inc., 9823 Pacific Heights Blvd, San Diego, CA 92121, USA
| | - Tsutomu Arakawa
- Alliance Protein Laboratories, 13380 Pantera Rd, San Diego, CA 92130, USA.
| | - Teruo Akuta
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., 3333-26, Aza-Asayama, Kamitezuna, Takahagi-shi, Ibaraki 318-0004, Japan.
| |
Collapse
|
33
|
Khanna S, Singh AK, Behera SP, Gupta S. Thermoresponsive BSA hydrogels with phase tunability. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111590. [PMID: 33321635 DOI: 10.1016/j.msec.2020.111590] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/26/2022]
Abstract
Amyloids are fibrillar structures formed due to protein aggregation or misfolding when the molecules undergo a conformational change from α-helix to β-sheet. Although this self-assembly is associated with many neurodegenerative diseases in vivo, the highly ordered amyloidic structures formed in vitro are ideal scaffolds for many bionanotechnological applications. Amyloid fibrillar networks under specific stimuli can also form stable hydrogels. We have used bovine serum albumin (BSA) as a model amyloidogenic protein to obtain thermally-induced hydrogels that display tunable sol-gel-sol transitions spanning over minutes to days. High concentrations of BSA (14-22% w/v) were heated at 65 °C for less than 3 min without any cross-linking agent to yield soft, injectable gels that were non-toxic to mammalian cells. A detailed investigation of temperature, concentration, incubation time and ionic strength on the formation and reversal of these gels was carried out using visual inspection, rheology, electron microscopy, fluorescence spectroscopy, UV-visible spectroscopy and circular dichroism spectroscopy. The optimum gelation temperature (Tg) for phase reversal of BSA gels was found to lie between 60 and 70 °C. An increase in protein concentration led to a reduction in the gelation time and increase in the gel-to-rev sol transition time. Gels heated for longer duration than their minimum gelation time yielded irreversible gels suggesting that low incubation periods were favourable for partial protein denaturation and hydrogel formation. This was supported by time-resolved secondary and tertiary structural ensemble studies. Further, the hydrogel networks demonstrated a zero-order drug release kinetics and the rev sol was found to be cytocompatible with HaCaT skin cell lines. Overall, our approach demonstrates rapid, crosslinker-free thermoresponsive BSA gelation with wide tunability and control on the time and material property, ideal for topical drug delivery applications.
Collapse
Affiliation(s)
- Shruti Khanna
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Ajay Kumar Singh
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Soumya Prakash Behera
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Shalini Gupta
- Dept. of Chemical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
34
|
Liu XY, Chen W, Yu HQ. Probing protein-induced membrane fouling with in-situ attenuated total reflectance fourier transform infrared spectroscopy and multivariate curve resolution-alternating least squares. WATER RESEARCH 2020; 183:116052. [PMID: 32622234 DOI: 10.1016/j.watres.2020.116052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 06/11/2023]
Abstract
Proteins are one of the major contributors to membrane fouling. The interaction between proteins and the polymer membrane at the molecular level is essential for the alleviation/prevention of membrane fouling, but remains unclear. In this work, time-dependent in-situ attenuated total reflectance Fourier transform infrared spectroscopy is applied to investigate the interaction process between two model proteins, bovine serum albumin and lysozyme, and the poly(vinylidene fluoride) (PVDF) membrane. Multivariate curve resolution-alternating least squares is integrated with two-dimensional correlation spectroscopy analysis to resolve the membrane-induced conformational changes of proteins. The multivariate curve resolution-alternating least squares analysis reveals a two-step process in the protein-membrane interaction and provides the kinetics of the conformational transition, which aids the segmentation of the spectral dataset. By applying two-dimensional correlation spectroscopy analysis to different groups of the time-dependent spectra, the sequential order of the secondary structural changes of proteins is determined. The proteins initially undergo unfolding transition to a more open, less structured state, which appears to be triggered by the hydrophobic membrane surface. Afterwards, the proteins become aggregated with the high anti-parallel β-sheet content, aggravating the membrane fouling. The conformational transition process of proteins was also confirmed by the atomic force microscopic images and quartz crystal microbalance measurement. Overall, this work provides an in-depth understanding of the interaction between proteins and the membrane surface, which is helpful for the development of membrane anti-fouling strategies.
Collapse
Affiliation(s)
- Xiao-Yang Liu
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Wei Chen
- School of Metallurgy and Environment, Central South University, Changsha, 410083, China
| | - Han-Qing Yu
- CAS Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
35
|
Huo XZ, Wang X, Yang R, Qu LB, Zeng HJ. Studies on the effect of a Fupenzi glycoprotein on the fibrillation of bovine serum albumin and its antioxidant activity. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 237:118387. [PMID: 32416513 DOI: 10.1016/j.saa.2020.118387] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/04/2020] [Accepted: 04/18/2020] [Indexed: 06/11/2023]
Abstract
In this study, the effect of a glycoprotein obtained from Fupenzi (FPZ) (Rubus chingii Hu.) on the fibrillation of bovine serum album (BSA) was investigated by multi-spectroscopic methods and transmission electron microscopy. Moreover, the cytotoxicity of the glycoprotein and the effect of it on H2O2-induced cell viability were investigated by cell counting kit and β-galactosidase kit, respectively. The experimental results indicated that the glycoprotein showed very low toxicity to NRK-52E cells and could obviously delay cell senescence and improve cell viability. Moreover, the glycoprotein could effectively inhibit the formation of BSA fibrils and destroy the stability of preformed BSA fibrils in a concentration-dependent manner. Generally, antioxidant capacities are thought to be related to the anti-amyloidogenic activity of inhibitors; therefore, to reveal the inhibitory mechanism, the anti-oxidative property of the glycoprotein was examined by DPPH and ABTS assays. The results demonstrated that FPZ glycoprotein had a remarkable antioxidant activity and the IC50 values of DPPH and ABTS were 0.249 mg mL-1 and 0.092 mg mL-1, respectively. This work suggested that the FPZ glycoprotein had the potential to be designed a new therapeutic agent for attenuating aging and preventing the age-related diseases.
Collapse
Affiliation(s)
- Xiu-Zhu Huo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Xia Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ran Yang
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ling-Bo Qu
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou University, Zhengzhou 450001, PR China
| | - Hua-Jin Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
36
|
Okami M, Sunada Y, Hatori K. Lysozyme-induced suppression of enzymatic and motile activities of actin-myosin: Impact of basic proteins. Int J Biol Macromol 2020; 163:1147-1153. [PMID: 32668307 DOI: 10.1016/j.ijbiomac.2020.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 10/23/2022]
Abstract
Electrostatic interactions between actin filaments and myosin molecules, which are ubiquitous proteins in eukaryotes, are crucial for their enzymatic activity and motility. Nonspecific electrostatic interactions between proteins are unavoidable in cells; therefore, it is worth exploring how ambient proteins, such as polyelectrolytes, affect actin-myosin functions. To understand the effect of counterionic proteins on actin-myosin, we examined ATPase activity and sliding velocity via actin-myosin interactions in the presence of the basic model protein hen egg lysozyme. In an in vitro motility assay with ATP, the sliding velocity of actin filaments on heavy meromyosin (HMM) decreased with increasing lysozyme concentrations. Actin filaments were completely stalled at a lysozyme concentration above 0.08 mg/mL. Lysozyme decreased the ATP hydrolysis rate of the actin-HMM complex but not that HMM alone. Co-sedimentation assays revealed that lysozyme enhanced the binding of HMM to actin filaments in the presence of ATP. Additionally, lysozyme could bind to actin and myosin filaments. The inhibitory effect of poly-l-lysine, histone mixture, and lactoferrin on the motility of actin-myosin was higher than that of lysozyme. Thus, nonspecific electrostatic interactions of basic proteins are involved in the bundling of actin filaments and modulation of essential functions of the actomyosin complex.
Collapse
Affiliation(s)
- Masaki Okami
- Department of Bio-Systems Engineering, Graduate School of Science and Engineering, Yamagata University, 4-3-16, Jyonan, Yonezawa, Yamagata 992-8510, Japan
| | - Yuma Sunada
- Department of Bio-Systems Engineering, Graduate School of Science and Engineering, Yamagata University, 4-3-16, Jyonan, Yonezawa, Yamagata 992-8510, Japan
| | - Kuniyuki Hatori
- Department of Bio-Systems Engineering, Graduate School of Science and Engineering, Yamagata University, 4-3-16, Jyonan, Yonezawa, Yamagata 992-8510, Japan.
| |
Collapse
|
37
|
Seaberg J, Flynn N, Cai A, Ramsey JD. Effect of redox‐responsive DTSSP crosslinking on poly(
l
‐lysine)‐grafted‐poly(ethylene glycol) nanoparticles for delivery of proteins. Biotechnol Bioeng 2020; 117:2504-2515. [DOI: 10.1002/bit.27369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/20/2020] [Accepted: 05/01/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Joshua Seaberg
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma
| | - Nicholas Flynn
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma
| | - Amanda Cai
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma
| | - Joshua D. Ramsey
- School of Chemical Engineering Oklahoma State University Stillwater Oklahoma
| |
Collapse
|
38
|
Sakaguchi T, Wada T, Kasai T, Shiratori T, Minami Y, Shimada Y, Otsuka Y, Komatsu K, Goto S. Effects of ionic and reductive atmosphere on the conformational rearrangement in hen egg white lysozyme prior to amyloid formation. Colloids Surf B Biointerfaces 2020; 190:110845. [DOI: 10.1016/j.colsurfb.2020.110845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/08/2020] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
|
39
|
Anti-aggregation effect of Ascorbic Acid and Quercetin on aggregated Bovine Serum Albumin Induced by Dithiothreitol: Comparison of Turbidity and Soluble Protein Fraction Methods. JURNAL KIMIA SAINS DAN APLIKASI 2020. [DOI: 10.14710/jksa.23.4.129-134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Studies on the anti-aggregation of dithiothreitol (DTT) induced - protein is generally determined by the fraction soluble (non-aggregated) protein. While the turbidity method is commonly used in studies of anti-aggregation, in which protein is induced by heat, in this study, both methods are compared in observing the anti-aggregation activity of ascorbic acid and quercetin toward bovine serum albumin induced by DTT. The DTT is a reducing agent for protein disulfide bonds and capable of inducing protein aggregation at physiological pH and temperature. The work was performed by the formation of Bovine Serum Albumin (BSA) aggregates induced by DTT under physiological conditions, which are pH 7.4 and 37°C. The aggregated protein profile was observed using the turbidity method at the end of incubation and measuring the difference of concentration between the fraction of soluble protein before and after incubation. The measurement was carried out using a spectrophotometer UV-Vis. The results indicate that both methods show similar inhibition profiles. The potential inhibition of ascorbic acid (AA) toward BSA protein aggregation induced by DTT increased along with incubation time. While quercetin shows the highest inhibition at 12 hours but decreased at 18 hours, this study reveals that both methods can observe the anti-aggregation activity of ascorbic acid and quercetin.
Collapse
|
40
|
Varela AE, England KA, Cavagnero S. Kinetic trapping in protein folding. Protein Eng Des Sel 2020; 32:103-108. [PMID: 31390019 DOI: 10.1093/protein/gzz018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
The founding principles of protein folding introduced by Christian Anfinsen, together with the numerous mechanistic investigations that followed, assume that protein folding is a thermodynamically controlled process. On the other hand, this review underscores the fact that thermodynamic control is far from being the norm in protein folding, as long as one considers an extended chemical-potential landscape encompassing aggregates, in addition to native, unfolded and intermediate states. Here, we highlight the key role of kinetic trapping of the protein native state relative to unfolded, intermediate and, most importantly, aggregated states. We propose that kinetic trapping serves an important role in biology by protecting the bioactive states of a large number of proteins from deleterious aggregation. In the event that undesired aggregates were somehow formed, specialized intracellular disaggregation machines have evolved to convert any aberrant populations back to the native state, thus restoring a fully bioactive and aggregation-protected protein cohort.
Collapse
Affiliation(s)
- Angela E Varela
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kevin A England
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Silvia Cavagnero
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
41
|
Al Adem K, Lukman S, Kim TY, Lee S. Inhibition of lysozyme aggregation and cellular toxicity by organic acids at acidic and physiological pH conditions. Int J Biol Macromol 2020; 149:921-930. [DOI: 10.1016/j.ijbiomac.2020.01.267] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/16/2022]
|
42
|
Nathiya D, Gurunathan K, Wilson J. Size controllable, pH triggered reduction of bovine serum albumin and its adsorption behavior with SnO2/SnS2 quantum dots for biosensing application. Talanta 2020; 210:120671. [DOI: 10.1016/j.talanta.2019.120671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 12/06/2019] [Accepted: 12/22/2019] [Indexed: 01/17/2023]
|
43
|
Li G, Delafield DG, Li L. Improved structural elucidation of peptide isomers and their receptors using advanced ion mobility-mass spectrometry. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2019.05.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
44
|
Xu Y, Liu Y, Hu X, Qin R, Su H, Li J, Yang P. The Synthesis of a 2D Ultra‐Large Protein Supramolecular Nanofilm by Chemoselective Thiol–Disulfide Exchange and its Emergent Functions. Angew Chem Int Ed Engl 2020; 59:2850-2859. [DOI: 10.1002/anie.201912848] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/29/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Yan Xu
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Yongchun Liu
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Xinyi Hu
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Rongrong Qin
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Hao Su
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Juling Li
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| |
Collapse
|
45
|
Xu Y, Liu Y, Hu X, Qin R, Su H, Li J, Yang P. The Synthesis of a 2D Ultra‐Large Protein Supramolecular Nanofilm by Chemoselective Thiol–Disulfide Exchange and its Emergent Functions. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201912848] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yan Xu
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Yongchun Liu
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Xinyi Hu
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Rongrong Qin
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Hao Su
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Juling Li
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid ChemistryMinistry of EducationSchool of Chemistry and Chemical EngineeringShaanxi Normal University Xi'an 710119 China
| |
Collapse
|
46
|
Liu X, Xu J, Xie X, Ma Z, Zheng T, Wu L, Li B, Li W. Heteropoly acid-driven assembly of glutathione into redox-responsive underwater adhesive. Chem Commun (Camb) 2020; 56:11034-11037. [DOI: 10.1039/d0cc03746j] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A glutathione-based underwater adhesive with dynamic attachment/detachment behaviour was achieved via the reversible formation and breakage of disulfide bonds of glutathione.
Collapse
Affiliation(s)
- Xiaohuan Liu
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Jing Xu
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Xiaoming Xie
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Zhiyuan Ma
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Tingting Zheng
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Lixin Wu
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Bao Li
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Wen Li
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| |
Collapse
|
47
|
Kalhor HR, Jabbary M. Investigating Reliable Conditions for HEWL as an Amyloid Model in Computational Studies and Drug Interactions. J Chem Inf Model 2019; 59:5218-5229. [DOI: 10.1021/acs.jcim.9b00819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Hamid R. Kalhor
- Biochemistry Research Laboratory, Department of Chemistry, Sharif University of Technology, PO Box: 11365-11155 Tehran, Iran
| | - Mohammadparsa Jabbary
- Biochemistry Research Laboratory, Department of Chemistry, Sharif University of Technology, PO Box: 11365-11155 Tehran, Iran
| |
Collapse
|
48
|
Streamlining the polishing step development process via physicochemical characterization of monoclonal antibody aggregates. J Chromatogr A 2019; 1598:101-112. [DOI: 10.1016/j.chroma.2019.03.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/08/2019] [Accepted: 03/21/2019] [Indexed: 01/07/2023]
|
49
|
Cao Y, Mezzenga R. Food protein amyloid fibrils: Origin, structure, formation, characterization, applications and health implications. Adv Colloid Interface Sci 2019; 269:334-356. [PMID: 31128463 DOI: 10.1016/j.cis.2019.05.002] [Citation(s) in RCA: 303] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/27/2023]
Abstract
Amyloid fibrils have traditionally been considered only as pathological aggregates in human neurodegenerative diseases, but it is increasingly becoming clear that the propensity to form amyloid fibrils is a generic property for all proteins, including food proteins. Differently from the pathological amyloid fibrils, those derived from food proteins can be used as advanced materials in biomedicine, tissue engineering, environmental science, nanotechnology, material science as well as in food science, owing to a combination of highly desirable feature such as extreme aspect ratios, outstanding stiffness and a broad availability of functional groups on their surfaces. In food science, protein fibrillization is progressively recognized as an appealing strategy to broaden and improve food protein functionality. This review article discusses the various classes of reported food protein amyloid fibrils and their formation conditions. It furthermore considers amyloid fibrils in a broad context, from their structural characterization to their forming mechanisms and ensued physical properties, emphasizing their applications in food-related fields. Finally, the biological fate and the potential toxicity mechanisms of food amyloid fibrils are discussed, and an experimental protocol for their health safety validation is proposed in the concluding part of the review.
Collapse
Affiliation(s)
- Yiping Cao
- Food and Soft Materials, Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland
| | - Raffaele Mezzenga
- Food and Soft Materials, Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 9, Zurich 8092, Switzerland.
| |
Collapse
|
50
|
Saotome T, Yamazaki T, Kuroda Y. Misfolding of a Single Disulfide Bonded Globular Protein into a Low-Solubility Species Conformationally and Biophysically Distinct from the Native One. Biomolecules 2019; 9:biom9060250. [PMID: 31242697 PMCID: PMC6627273 DOI: 10.3390/biom9060250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 11/16/2022] Open
Abstract
In practice and despite Anfinsen’s dogma, the refolding of recombinant multiple SS-bonded proteins is famously difficult because misfolded species with non-native SS-bonds appear upon the oxidization of their cysteine residues. On the other hand, single SS-bond proteins are thought to be simple to refold because their cysteines have only one SS-bond partner. Here, we report that dengue 4 envelope protein domain 3 (DEN4 ED3), a single SS-bonded protein can be irreversibly trapped into a misfolded species through the formation of its sole intramolecular SS-bond. The misfolded species had a much lower solubility than the native one at pHs higher than about 7, and circular dichroism measurements clearly indicated that its secondary structure content was different from the native species. Furthermore, the peaks in the Heteronuclear Single Quantum Correlation spectroscopy (HSQC) spectrum of DEN4 ED3 from the supernatant fraction were sharp and well dispersed, reflecting the beta-sheeted native structure, whereas the spectrum of the precipitated fraction showed broad signals clustered near its center suggesting no or little structure and a strong tendency to aggregate. The two species had distinct biophysical properties and could interconvert into each other only by cleaving and reforming the SS-bond, strongly suggesting that they are topologically different. This phenomenon can potentially happen with any single SS-bonded protein, and our observation emphasizes the need for assessing the conformation and biophysical properties of bacterially produced therapeutic proteins in addition to their chemical purities.
Collapse
Affiliation(s)
- Tomonori Saotome
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology (TUAT), Tokyo 184-8588, Japan
| | - Toshio Yamazaki
- NMR Facility, Division of Structural and Synthetic Biology, Center for Life Science Technologies, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology (TUAT), Tokyo 184-8588, Japan.
| |
Collapse
|