1
|
Cui Y, Han D, Bai X, Shi W. Development and applications of enzymatic peptide and protein ligation. J Pept Sci 2025; 31:e3657. [PMID: 39433441 DOI: 10.1002/psc.3657] [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: 08/06/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024]
Abstract
Chemical synthesis of complex peptides and proteins continues to play increasingly important roles in industry and academia, where strategies for covalent ligation of two or more peptide fragments to produce longer peptides and proteins in convergent manners have become critical. In recent decades, efficient and site-selective ligation strategies mediated by exploiting the biocatalytic capacity of nature's diverse toolkit (i.e., enzymes) have been widely recognized as a powerful extension of existing chemical strategies. In this review, we present a chronological overview of the development of proteases, transpeptidases, transglutaminases, and ubiquitin ligases. We survey the different properties between the ligation reactions of various enzymes, including the selectivity and efficiency of the reaction, the ligation "scar" left in the product, the type of amide bond formed (natural or isopeptide), the synthetic availability of the reactants, and whether the enzymes are orthogonal to another. This review also describes how the inherent specificity of these enzymes can be exploited for peptide and protein ligation.
Collapse
Affiliation(s)
- Yan Cui
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Dongyang Han
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Xuerong Bai
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Weiwei Shi
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| |
Collapse
|
2
|
O’Day DH. The Search for a Universal Treatment for Defined and Mixed Pathology Neurodegenerative Diseases. Int J Mol Sci 2024; 25:13424. [PMID: 39769187 PMCID: PMC11678063 DOI: 10.3390/ijms252413424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
The predominant neurodegenerative diseases, Alzheimer's disease, Parkinson's disease, dementia with Lewy Bodies, Huntington's disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are rarely pure diseases but, instead, show a diversity of mixed pathologies. At some level, all of them share a combination of one or more different toxic biomarker proteins: amyloid beta (Aβ), phosphorylated Tau (pTau), alpha-synuclein (αSyn), mutant huntingtin (mHtt), fused in sarcoma, superoxide dismutase 1, and TAR DNA-binding protein 43. These toxic proteins share some common attributes, making them potentially universal and simultaneous targets for therapeutic intervention. First, they all form toxic aggregates prior to taking on their final forms as contributors to plaques, neurofibrillary tangles, Lewy bodies, and other protein deposits. Second, the primary enzyme that directs their aggregation is transglutaminase 2 (TGM2), a brain-localized enzyme involved in neurodegeneration. Third, TGM2 binds to calmodulin, a regulatory event that can increase the activity of this enzyme threefold. Fourth, the most common mixed pathology toxic biomarkers (Aβ, pTau, αSyn, nHtt) also bind calmodulin, which can affect their ability to aggregate. This review examines the potential therapeutic routes opened up by this knowledge. The end goal reveals multiple opportunities that are immediately available for universal therapeutic treatment of the most devastating neurodegenerative diseases facing humankind.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
3
|
Jiang Y, Zhu C, Ma X, Fan D. Smart hydrogel-based trends in future tendon injury repair: A review. Int J Biol Macromol 2024; 282:137092. [PMID: 39489238 DOI: 10.1016/j.ijbiomac.2024.137092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Despite advances in tissue engineering for tendon repair, rapid functional repair is still challenging due to its specificity and is prone to complications such as postoperative infections and tendon adhesions. Smart responsive hydrogels provide new ideas for tendon therapy with their flexibly designed three-dimensional cross-linked polymer networks that respond to specific stimuli. In recent years, a variety of smart-responsive hydrogels have been developed for the treatment of tendon disorders, showing great research promise and ability to address complex challenges. This article provides a comprehensive review of recent advances in the field of smart-responsive hydrogels for the treatment of tendon disorders, with a special focus on their response properties to different physical, chemical and biological stimuli. The multiple functional properties of these innovative materials are discussed in depth, including excellent biocompatibility and biodegradability, excellent mechanical properties, biomimetic structural design, convenient injectability, and unique self-healing capabilities. These properties enable the smart-responsive hydrogels to demonstrate significant advantages in solving difficult problems in the treatment of tendon disorders, such as precise drug delivery, tendon adhesion prevention and postoperative infection control. In addition, the article looks at the future prospects of smart-responsive hydrogels and analyses the challenges they may face in achieving widespread application.
Collapse
Affiliation(s)
- Yingxue Jiang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Xiaoxuan Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China.
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
4
|
Chowdhury MAH, Sarkar F, Reem CSA, Rahman SM, Mahamud AGMSU, Rahman MA, Md Ashrafudoulla. Enzyme applications in baking: From dough development to shelf-life extension. Int J Biol Macromol 2024; 282:137020. [PMID: 39489247 DOI: 10.1016/j.ijbiomac.2024.137020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/10/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
Enzymes play a vital role in baking, providing significant benefits from dough development to extending shelf life, which enhances product quality and consistency. Acting as biological catalysts, enzymes such as proteases and amylases break down proteins and starches, modifying dough rheology and improving fermentation. Lipases and oxidases further refine dough texture through emulsification and oxidation, while lipases also produce fatty acid derivatives during fermentation, contributing to the flavor and aroma of baked goods. Xylanases and cellulases optimize dough handling by altering fiber structure, and amylases help maintain moisture and texture, extending the shelf life of baked products. Ensuring regulatory compliance is essential when incorporating enzymes into baking processes, as bakers must address enzyme stability and determine appropriate dosages for reliable outcomes. Ongoing research is exploring innovative enzyme applications, including customized enzyme blends that target specific product qualities, offering new possibilities for product differentiation and innovation. In summary, enzyme-driven advancements present bakers with opportunities to improve product quality, shelf life, and consistency, while meeting industry regulations. This review emphasizes the critical impact enzymes have on dough properties and finished product characteristics, highlighting their role in driving future innovations within the baking industry.
Collapse
Affiliation(s)
- Md Anamul Hasan Chowdhury
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong, -si, Gyeonggi-Do 17546, Republic of Korea
| | - Feroj Sarkar
- Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Chowdhury Sanat Anjum Reem
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong, -si, Gyeonggi-Do 17546, Republic of Korea
| | - Sk Mustafizur Rahman
- Department of Nutrition and Food Engineering, Daffodil International University, Birulia 1216, Bangladesh
| | - A G M Sofi Uddin Mahamud
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong, -si, Gyeonggi-Do 17546, Republic of Korea
| | - Md Ashikur Rahman
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong, -si, Gyeonggi-Do 17546, Republic of Korea
| | | |
Collapse
|
5
|
Bérot A, Maniti O, El Alaoui S, Granjon T, El Alaoui M. Generation of Anti-Epidermal Growth Factor Receptor-2 (HER2) Immunoliposomes Using Microbial Transglutaminase (mTG)-Mediated Site-Specific Conjugated Antibodies. ACS Pharmacol Transl Sci 2024; 7:3034-3044. [PMID: 39416960 PMCID: PMC11475288 DOI: 10.1021/acsptsci.4c00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
Nanocarriers have found their interests in many fields including drug delivery and labeling of cells with the aim to target and eradicate tumor cells. One of the approaches to specifically address nanocarriers, such as liposomes, to their target is to attach antibodies of interest to their surface. To date, the development of immunoliposomes has been widely explored but has mainly involved chemical and unspecific reactions that could impair antibody stability, integrity, and orientation, thus reducing optimized immunoliposomes generation. In this study, we report the use of the patented COVISOLINK technology and the strain-promoted alkyne-azide cycloaddition (SPAAC) to generate immunoliposomes that target HER2 positive breast cancer with Trastuzumab as the antibody to be coupled. The efficacy of our two-step functionalization strategy and the successful specific coupling of the antibodies were validated by high-performance liquid chromatography-size exclusion chromatography (HPLC-SEC), which allowed a precise quantification of antibodies conjugated to liposomes and confirmed by cryo-TEM and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analyses. We also demonstrate by flow cytometry and epifluorescence microscopy that the produced anti-HER2 immunoliposomes were able to interact specifically with their target cells (SK-BR-3) while remaining negative with cells that express HER2 at a low level (MDA-MB-231). Hence, for the first time, our COVISOLINK strategy using microbial transglutaminase (mTG) enables the preparation and production of well-characterized immunoliposomes that could be used in different applications, including therapies.
Collapse
Affiliation(s)
- Anna Bérot
- Covalab, 1B Rue Jacques Monod, 69500 Bron, France
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires
ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | - Ofelia Maniti
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires
ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | | | - Thierry Granjon
- Institut
de Chimie et Biochimie Moléculaires et Supramoléculaires
ICBMS UMR 5246, Univ Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | | |
Collapse
|
6
|
Ivashchenko SD, Shulga DA, Ivashchenko VD, Zinovev EV, Vlasov AV. In silico studies of the open form of human tissue transglutaminase. Sci Rep 2024; 14:15981. [PMID: 38987418 PMCID: PMC11236986 DOI: 10.1038/s41598-024-66348-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Human tissue transglutaminase (tTG) is an intriguing multifunctional enzyme involved in various diseases, including celiac disease and neurological disorders. Although a number of tTG inhibitors have been developed, the molecular determinants governing ligand binding remain incomplete due to the lack of high-resolution structural data in the vicinity of its active site. In this study, we obtained the complete high-resolution model of tTG by in silico methods based on available PDB structures. We discovered significant differences in the active site architecture between our and known tTG models, revealing an additional loop which affects the ligand binding affinity. We assembled a library of new potential tTG inhibitors based on the obtained complete model of the enzyme. Our library substantially expands the spectrum of possible drug candidates targeting tTG and encompasses twelve molecular scaffolds, eleven of which are novel and exhibit higher binding affinity then already known ones, according to our in silico studies. The results of this study open new directions for structure-based drug design of tTG inhibitors, offering the complete protein model and suggesting a wide range of new compounds for further experimental validation.
Collapse
Affiliation(s)
- S D Ivashchenko
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
- Laboratory of Microbiology, BIOTECH University, Moscow, Russia, 125080
| | - D A Shulga
- Department of Chemistry, Moscow State University, Moscow, Russia, 119991
| | - V D Ivashchenko
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
| | - E V Zinovev
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
| | - A V Vlasov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701.
- Laboratory of Microbiology, BIOTECH University, Moscow, Russia, 125080.
- Joint Institute for Nuclear Research, Dubna, Russia, 141980.
| |
Collapse
|
7
|
Wu S, Gai T, Chen J, Chen X, Chen W. Smart responsive in situ hydrogel systems applied in bone tissue engineering. Front Bioeng Biotechnol 2024; 12:1389733. [PMID: 38863497 PMCID: PMC11165218 DOI: 10.3389/fbioe.2024.1389733] [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: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 06/13/2024] Open
Abstract
The repair of irregular bone tissue suffers severe clinical problems due to the scarcity of an appropriate therapeutic carrier that can match dynamic and complex bone damage. Fortunately, stimuli-responsive in situ hydrogel systems that are triggered by a special microenvironment could be an ideal method of regenerating bone tissue because of the injectability, in situ gelatin, and spatiotemporally tunable drug release. Herein, we introduce the two main stimulus-response approaches, exogenous and endogenous, to forming in situ hydrogels in bone tissue engineering. First, we summarize specific and distinct responses to an extensive range of external stimuli (e.g., ultraviolet, near-infrared, ultrasound, etc.) to form in situ hydrogels created from biocompatible materials modified by various functional groups or hybrid functional nanoparticles. Furthermore, "smart" hydrogels, which respond to endogenous physiological or environmental stimuli (e.g., temperature, pH, enzyme, etc.), can achieve in situ gelation by one injection in vivo without additional intervention. Moreover, the mild chemistry response-mediated in situ hydrogel systems also offer fascinating prospects in bone tissue engineering, such as a Diels-Alder, Michael addition, thiol-Michael addition, and Schiff reactions, etc. The recent developments and challenges of various smart in situ hydrogels and their application to drug administration and bone tissue engineering are discussed in this review. It is anticipated that advanced strategies and innovative ideas of in situ hydrogels will be exploited in the clinical field and increase the quality of life for patients with bone damage.
Collapse
Affiliation(s)
- Shunli Wu
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Hangzhou Singclean Medical Products Co., Ltd, Hangzhou, China
| | - Tingting Gai
- School of Medicine, Shanghai University, Shanghai, China
| | - Jie Chen
- Jiaxing Vocational Technical College, Department of Student Affairs, Jiaxing, China
| | - Xiguang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, China
- Laoshan Laboratory, Qingdao, China
| | - Weikai Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Munaweera TIK, Damnjanović J, Camagna M, Nezu M, Jia B, Hitomi K, Nemoto N, Nakano H. Substrate profiling of human transglutaminase 1 using cDNA display and next-generation sequencing. Biosci Biotechnol Biochem 2024; 88:620-629. [PMID: 38479783 DOI: 10.1093/bbb/zbae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/04/2024] [Indexed: 05/23/2024]
Abstract
Human transglutaminase 1 (TG1) modulates skin development, while its involvement in diseases remains poorly understood, necessitating comprehensive exploration of its substrate interactions. To study the substrate profile of TG1, an in vitro selection system based on cDNA display technology was used to screen two peptide libraries with mutations at varying distance from the reactive glutamine. Next-generation sequencing and bioinformatics analysis of the selected DNA pools revealed a detailed TG1 substrate profile, indicating preferred and non-preferred amino acid sequences. The peptide sequence, AEQHKLPSKWPF, was identified showing high reactivity and specificity to TG1. The position weight matrix calculated from the per amino acid enrichment factors was employed to search human proteins using an in-house algorithm, revealing six known TG1 substrate proteins with high scores, alongside a list of candidate substrates currently under investigation. Our findings are expected to assist in future medical diagnoses and development of treatments for skin disorders.
Collapse
Affiliation(s)
- T I K Munaweera
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Jasmina Damnjanović
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Maurizio Camagna
- Laboratory of Plant Pathology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Moeri Nezu
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Beixi Jia
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kiyotaka Hitomi
- Laboratory of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Naoto Nemoto
- Laboratory of Evolutionary Molecular Engineering, Graduate School of Science and Engineering, Saitama University , Saitama, Japan
| | - Hideo Nakano
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
9
|
Buccarelli M, Castellani G, Fiorentino V, Pizzimenti C, Beninati S, Ricci-Vitiani L, Scattoni ML, Mischiati C, Facchiano F, Tabolacci C. Biological Implications and Functional Significance of Transglutaminase Type 2 in Nervous System Tumors. Cells 2024; 13:667. [PMID: 38667282 PMCID: PMC11048792 DOI: 10.3390/cells13080667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Transglutaminase type 2 (TG2) is the most ubiquitously expressed member of the transglutaminase family. TG2 catalyzes the transamidation reaction leading to several protein post-translational modifications and it is also implicated in signal transduction thanks to its GTP binding/hydrolyzing activity. In the nervous system, TG2 regulates multiple physiological processes, such as development, neuronal cell death and differentiation, and synaptic plasticity. Given its different enzymatic activities, aberrant expression or activity of TG2 can contribute to tumorigenesis, including in peripheral and central nervous system tumors. Indeed, TG2 dysregulation has been reported in meningiomas, medulloblastomas, neuroblastomas, glioblastomas, and other adult-type diffuse gliomas. The aim of this review is to provide an overview of the biological and functional relevance of TG2 in the pathogenesis of nervous system tumors, highlighting its involvement in survival, tumor inflammation, differentiation, and in the resistance to standard therapies.
Collapse
Affiliation(s)
- Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy;
| | - Simone Beninati
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Carlo Mischiati
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.); (F.F.)
| | - Claudio Tabolacci
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
10
|
Rosseto M, Rigueto CVT, Gomes KS, Krein DDC, Loss RA, Dettmer A, Richards NSPDS. Whey filtration: a review of products, application, and pretreatment with transglutaminase enzyme. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3185-3196. [PMID: 38151774 DOI: 10.1002/jsfa.13248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/28/2023] [Accepted: 12/27/2023] [Indexed: 12/29/2023]
Abstract
In the cheese industry, whey, which is rich in lactose and proteins, is underutilized, causing adverse environmental impacts. The fractionation of its components, typically carried out through filtration membranes, faces operational challenges such as membrane fouling, significant protein loss during the process, and extended operating times. These challenges require attention and specific methods for optimization and to increase efficiency. A promising strategy to enhance industry efficiency and sustainability is the use of enzymatic pre-treatment with the enzyme transglutaminase (TGase). This enzyme plays a crucial role in protein modification, catalyzing covalent cross-links between lysine and glutamine residues, increasing the molecular weight of proteins, facilitating their retention on membranes, and contributing to the improvement of the quality of the final products. The aim of this study is to review the application of the enzyme TGase as a pretreatment in whey protein filtration. The scope involves assessing the enzyme's impact on whey protein properties and its relationship with process performance. It also aims to identify both the optimization of operational parameters and the enhancement of product characteristics. This study demonstrates that the application of TGase leads to improved performance in protein concentration, lactose permeation, and permeate flux rate during the filtration process. It also has the capacity to enhance protein solubility, viscosity, thermal stability, and protein gelation in whey. In this context, it is relevant for enhancing the characteristics of whey, thereby contributing to the production of higher quality final products in the food industry. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Marieli Rosseto
- Rural Science Center, Postgraduate Program in Food Science and Technology (PPGCTA), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Cesar Vinicius Toniciolli Rigueto
- Rural Science Center, Postgraduate Program in Food Science and Technology (PPGCTA), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Karolynne Sousa Gomes
- Graduate Program in Food Engineering and Science, Federal University of Rio Grande, Rio Grande, Brazil
| | | | - Raquel Aparecida Loss
- Food Engineering Department, Faculty of Architecture and Engineering (FAE), Mato Grosso State University (UNEMAT), Barra do Bugres, Brazil
| | - Aline Dettmer
- Postgraduate Program in Food Science and Technology (PPGCTA), Institute of Technology (ITec), University of Passo Fundo (UPF), Passo Fundo, Brazil
| | | |
Collapse
|
11
|
Liu J, Mouradian MM. Pathogenetic Contributions and Therapeutic Implications of Transglutaminase 2 in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2364. [PMID: 38397040 PMCID: PMC10888553 DOI: 10.3390/ijms25042364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases encompass a heterogeneous group of disorders that afflict millions of people worldwide. Characteristic protein aggregates are histopathological hallmark features of these disorders, including Amyloid β (Aβ)-containing plaques and tau-containing neurofibrillary tangles in Alzheimer's disease, α-Synuclein (α-Syn)-containing Lewy bodies and Lewy neurites in Parkinson's disease and dementia with Lewy bodies, and mutant huntingtin (mHTT) in nuclear inclusions in Huntington's disease. These various aggregates are found in specific brain regions that are impacted by neurodegeneration and associated with clinical manifestations. Transglutaminase (TG2) (also known as tissue transglutaminase) is the most ubiquitously expressed member of the transglutaminase family with protein crosslinking activity. To date, Aβ, tau, α-Syn, and mHTT have been determined to be substrates of TG2, leading to their aggregation and implicating the involvement of TG2 in several pathophysiological events in neurodegenerative disorders. In this review, we summarize the biochemistry and physiologic functions of TG2 and describe recent advances in the pathogenetic role of TG2 in these diseases. We also review TG2 inhibitors tested in clinical trials and discuss recent TG2-targeting approaches, which offer new perspectives for the design of future highly potent and selective drugs with improved brain delivery as a disease-modifying treatment for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
12
|
Wu J, Chu T, Hao J, Lin L. SpSrtA-Catalyzed Isopeptide Ligation on Lysine Residues. Microorganisms 2024; 12:179. [PMID: 38258005 PMCID: PMC10818881 DOI: 10.3390/microorganisms12010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Sortase-mediated ligation (SML) is widely used for protein bioconjugation. However, the sortase used in this strategy typically recognizes only the N-terminal oligoglycine, which is absent in most natural proteins. To broaden the spectrum of substrates compatible with SML, we focus on a novel sortase, sortase A from Streptococcus pneumoniae (SpSrtA), known for its expanded substrate specificity (N-terminal glycine, alanine, and serine). We present the first evidence showing that the reported SpSrtA mutant (SpSrtA*) can modify lysine residues in itself and other proteins. The modification sites of SpSrtA* were identified through LC-MS/MS analysis. Moreover, we discovered an optimal lysine-containing peptide tag by fusing it onto sfGFP, resulting in a labeling efficiency of 57%. Inspired by this, we applied the method to modify proteins on microorganism surfaces up to 13.5-fold. To enhance labeling efficiency, we fused the SpSrtA* onto a surface protein and achieved a 2.64-fold improvement. We further developed a high-throughput yeast display screening method for the directed evolution of SpSrtA*, achieving a 10-fold improvement in the labeling efficiency of this surface protein. Our study provides a novel strategy for modifying the lysine residues that will be a powerful addition to the protein bioconjugation toolbox.
Collapse
Affiliation(s)
- Jiajia Wu
- Department of Chemistry, Shanghai University, Shanghai 200444, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Tianyu Chu
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jian Hao
- Department of Chemistry, Shanghai University, Shanghai 200444, China
| | - Liang Lin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
13
|
Lu S, Xiong W, Yao Y, Zhang J, Wang L. Investigating the physicochemical properties and air-water interface adsorption behavior of transglutaminase-crosslinking rapeseed protein isolate. Food Res Int 2023; 174:113505. [PMID: 37986500 DOI: 10.1016/j.foodres.2023.113505] [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: 07/28/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 11/22/2023]
Abstract
Improving the technical functionality to adapt to the application of complex food systems is an important challenge for the development of plant protein ingredients. Herein, the correlation between the physicochemical properties and interfacial adsorption behavior of rapeseed protein isolate (RPI) at the air-water interface after transglutaminase (TG) treatment was investigated. The results of cross-linking degree, Fourier transform infrared spectroscopy (FTIR) and sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) showed that the TG enzyme was able to catalyse cross-linking between lysine and glutamine residues of RPI. The foaming capacity of RPI was enhanced from 120 % to 150 % after TG cross-linking 5 h, whereas the average size (210-219 nm) of the RPI determined by dynamic light scattering did not change significantly. Besides, the hydrophobicity tended to increase overall under the enzyme treatment, while the surface electrostatic potential decreased. The former indicates the unfolding of the protein and reduces the kinetic barriers to protein adsorption at the air-water interface, with a consequent increase in disulfide bonding and surface pressure. Furthermore, as the enzyme treatment time increased, a significant increase in protein content of foam by 33.86 %. These findings provide novel insight into the foaming mechanism of TG cross-linking RPI.
Collapse
Affiliation(s)
- Shanshan Lu
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu, China
| | - Wenfei Xiong
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu, China
| | - Yijun Yao
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu, China
| | - Jing Zhang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu, China
| | - Lifeng Wang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
14
|
Soltani F, Kaartinen MT. Transglutaminases in fibrosis-overview and recent advances. Am J Physiol Cell Physiol 2023; 325:C885-C894. [PMID: 37642242 DOI: 10.1152/ajpcell.00322.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Transglutaminases (TGs) are a family of protein cross-linking enzymes that are capable of stiffening and insolubilizing proteins and creating protein networks, and thereby altering biological functions of proteins. Their role in fibrosis progression has been widely investigated with a focus on kidney, lung, liver, and heart where activity is triggered by various stimuli including hypoxia, inflammation, and hyperglycemia. TG2 has been considered one of the key enzymes in the pathogenesis of fibrosis mainly through transforming growth factor beta (TGF-beta) signaling and matrix cross-linking mechanisms. Although TG2 has been most widely studied in this context, the involvement of other TGs, TG1 and Factor XIII-A (FXIII-A), is beginning to emerge. This mini-review highlights the major steps taken in the TG and fibrosis research and summarizes the most recent advances and contributions of TG2, TG1, and FXIII-A to the progression of fibrosis in various animal models. Also, their mechanisms of action as well as therapeutic prospects are discussed.
Collapse
Affiliation(s)
- Fatemeh Soltani
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Mari T Kaartinen
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Faculty of Dental Medicine and Oral Health Sciences (Biomedical Sciences), McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Amirthalingam S, Rajendran AK, Moon YG, Hwang NS. Stimuli-responsive dynamic hydrogels: design, properties and tissue engineering applications. MATERIALS HORIZONS 2023; 10:3325-3350. [PMID: 37387121 DOI: 10.1039/d3mh00399j] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
The field of tissue engineering and regenerative medicine has been evolving at a rapid pace with numerous novel and interesting biomaterials being reported. Hydrogels have come a long way in this regard and have been proven to be an excellent choice for tissue regeneration. This could be due to their innate properties such as water retention, and ability to carry and deliver a multitude of therapeutic and regenerative elements to aid in better outcomes. Over the past few decades, hydrogels have been developed into an active and attractive system that can respond to various stimuli, thereby presenting a wider control over the delivery of the therapeutic agents to the intended site in a spatiotemporal manner. Researchers have developed hydrogels that respond dynamically to a multitude of external as well as internal stimuli such as mechanics, thermal energy, light, electric field, ultrasonics, tissue pH, and enzyme levels, to name a few. This review gives a brief overview of the recent developments in such hydrogel systems which respond dynamically to various stimuli, some of the interesting fabrication strategies, and their application in cardiac, bone, and neural tissue engineering.
Collapse
Affiliation(s)
- Sivashanmugam Amirthalingam
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Arun Kumar Rajendran
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Gi Moon
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
16
|
Cao J, Yuan P, Wu B, Liu Y, Hu C. Advances in the Research and Application of Smart-Responsive Hydrogels in Disease Treatment. Gels 2023; 9:662. [PMID: 37623116 PMCID: PMC10454421 DOI: 10.3390/gels9080662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/12/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Smart-responsive hydrogels have been widely used in various fields, particularly in the biomedical field. Compared with traditional hydrogels, smart-responsive hydrogels not only facilitate the encapsulation and controlled release of drugs, active substances, and even cells but, more importantly, they enable the on-demand and controllable release of drugs and active substances at the disease site, significantly enhancing the efficacy of disease treatment. With the rapid advancement of biomaterials, smart-responsive hydrogels have received widespread attention, and a wide variety of smart-responsive hydrogels have been developed for the treatment of different diseases, thus presenting tremendous research prospects. This review summarizes the latest advancements in various smart-responsive hydrogels used for disease treatment. Additionally, some of the current shortcomings of smart-responsive hydrogels and the strategies to address them are discussed, as well as the future development directions and prospects of smart-responsive hydrogels.
Collapse
Affiliation(s)
- Juan Cao
- School of Fashion and Design Art, Sichuan Normal University, Chengdu 610066, China;
| | - Ping Yuan
- School of Mechanical Engineering, Chengdu University, Chengdu 610106, China;
| | - Bo Wu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China; (B.W.); (Y.L.)
| | - Yeqi Liu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China; (B.W.); (Y.L.)
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
17
|
Li G, Liu S, Chen Y, Zhao J, Xu H, Weng J, Yu F, Xiong A, Udduttula A, Wang D, Liu P, Chen Y, Zeng H. An injectable liposome-anchored teriparatide incorporated gallic acid-grafted gelatin hydrogel for osteoarthritis treatment. Nat Commun 2023; 14:3159. [PMID: 37258510 DOI: 10.1038/s41467-023-38597-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 05/10/2023] [Indexed: 06/02/2023] Open
Abstract
Intra-articular injection of therapeutics is an effective strategy for treating osteoarthritis (OA), but it is hindered by rapid drug diffusion, thereby necessitating high-frequency injections. Hence, the development of a biofunctional hydrogel for improved delivery is required. In this study, we introduce a liposome-anchored teriparatide (PTH (1-34)) incorporated into a gallic acid-grafted gelatin injectable hydrogel (GLP hydrogel). We show that the GLP hydrogel can form in situ and without affecting knee motion after intra-articular injection in mice. We demonstrate controlled, sustained release of PTH (1-34) from the GLP hydrogel. We find that the GLP hydrogel promotes ATDC5 cell proliferation and protects the IL-1β-induced ATDC5 cells from further OA progression by regulating the PI3K/AKT signaling pathway. Further, we show that intra-articular injection of hydrogels into an OA-induced mouse model promotes glycosaminoglycans synthesis and protects the cartilage from degradation, supporting the potential of this biomaterial for OA treatment.
Collapse
Affiliation(s)
- Guoqing Li
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Su Liu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Yixiao Chen
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Jin Zhao
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Huihui Xu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Jian Weng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Fei Yu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Ao Xiong
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Anjaneyulu Udduttula
- Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Deli Wang
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China
| | - Peng Liu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China.
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China.
| | - Yingqi Chen
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China.
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China.
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China.
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, PR China.
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen, Guangdong Province, PR China.
| |
Collapse
|
18
|
Zheng N, Long M, Zhang Z, Du S, Huang X, Osire T, Xia X. Behavior of enzymes under high pressure in food processing: mechanisms, applications, and developments. Crit Rev Food Sci Nutr 2023; 64:9829-9843. [PMID: 37243343 DOI: 10.1080/10408398.2023.2217268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
High pressure processing (HPP) offers the benefits of safety, uniformity, energy-efficient, and low waste, which is widely applied for microbial inactivation and shelf-life extension for foods. Over the past forty years, HPP has been extensively researched in the food industry, enabling the inactivation or activation of different enzymes in future food by altering their molecular structure and active site conformation. Such activation or inactivation of enzymes effectively hinders the spoilage of food and the production of beneficial substances, which is crucial for improving food quality. This paper reviews the mechanism in which high pressure affects the stability and activity of enzymes, concludes the roles of key enzymes in the future food processed using high pressure technologies. Moreover, we discuss the application of modified enzymes based on high pressure, providing insights into the future direction of enzyme evolution under complex food processing conditions (e.g. high temperature, high pressure, high shear, and multiple elements). Finally, we conclude with prospects of high pressure technology and research directions in the future. Although HPP has shown positive effects in improving the future food quality, there is still a pressing need to develop new and effective combined processing methods, upgrade processing modes, and promote sustainable lifestyles.
Collapse
Affiliation(s)
- Nan Zheng
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Mengfei Long
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zehua Zhang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Shuang Du
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xinlei Huang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Tolbert Osire
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, China
| | - Xiaole Xia
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
19
|
Park JW, Lee SH, Kim HW, Park HJ. Application of extrusion-based 3D food printing to regulate marbling patterns of restructured beef steak. Meat Sci 2023; 202:109203. [PMID: 37120978 DOI: 10.1016/j.meatsci.2023.109203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/31/2023] [Accepted: 04/22/2023] [Indexed: 05/02/2023]
Abstract
Consumers prefer marbled meat and are willing to pay a higher price, in addition, to the potential wastage of meat that is considered a lower value. In this study, meat production with varying levels of marbling was investigated using a multifilament printing approach. Different amounts of fat sticks were embedded into lean meat paste ink and used to produce 3D-printed meat that would cater to the diverse range of consumer preferences. The rheological behaviors of the meat and fat paste used in the multifilament were assessed and indicated that the ink would maintain shape stability after deposition. When the multifilament was used for printing, the intramuscular fat area of the cross-sectional surface was proportional to the fat added to the ink. The meat protein formed a three-dimensional gel network and showed a clear contraction pattern after heat treatment. As the fat content increased, the cutting strength of the printed meat after cooking decreased, and the cooking loss increased. All the printed steaks were well-texturized; in particular, the product with 10% fat paste had a high degree of texturization. This study will provide a market for less popular cuts of beef and guidelines for using various grades of meat to generate an improved quality product through a multifilament 3D printing approach.
Collapse
Affiliation(s)
- Jung Whee Park
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Anam-dong, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Su Hyun Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Anam-dong, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hyun Woo Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Anam-dong, Seongbuk-gu, Seoul 02841, Republic of Korea.
| | - Hyun Jin Park
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Anam-dong, Seongbuk-gu, Seoul 02841, Republic of Korea.
| |
Collapse
|
20
|
Dejager L, Jairaj M, Jones K, Johnson T, Dudal S, Dudal Y, Shahgaldian P, Correro R, Qu J, An B, Lucey R, Szarka S, Wheller R, Pruna A, Kettell S, Pitt A, Cutler P. Development and validation of a liquid chromatography-triple quadrupole mass spectrometry method for the determination of isopeptide ε-(γ-glutamyl) lysine in human urine as biomarker for transglutaminase 2 cross-linked proteins. J Chromatogr A 2023; 1699:464002. [PMID: 37126878 DOI: 10.1016/j.chroma.2023.464002] [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: 02/03/2023] [Revised: 04/05/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Determination of the levels of protein cross-linking catalysed by the activity of transglutaminase 2 in various disease states has remained a significant challenge. The ability to quantify the isopeptide ε-(γ-glutamyl) lysine, which can form as a heterogeneous bond within or between proteins has significant analytical and clinical potential as a biomarker in biofluids such as human urine. Increased transglutaminase 2 activity is associated with a number of diseases, such as fibrosis. Previously published methods have been based on classical amino acid analysis, however they require a complex multi-enzyme digestion in order to achieve complete protein digestion, whilst leaving the isopeptide cross link intact. These methods require high levels of enzymes, which contaminate the analysis and alter the dynamics of digestion. The amino acid analysis detection also lacked selectivity, especially where the levels of crosslink are expected to be low relative to the background protein levels. We have systematically addressed these challenges, by optimising the precipitation of the protein in urine, the use of innovative immobilised enzyme technology, which allows for efficient digestion without enzyme contamination and LC-MS/MS detection based on multiple reaction monitoring. This method was validated for its analytical performance characteristics, showing the method has a sensitivity of 0.1 ng/mL of ε-(γ-glutamyl) lysine in human urine with precision of less than 20% CV, and is selective as no interferences were observed that may adversely affect the analysis. As such this approach represents a significant advance in the ability to detect and quantify ε-(γ-glutamyl) lysine.
Collapse
Affiliation(s)
- Lien Dejager
- UCB Pharma, Chemin du Foriest, B-1420 Braine-l'Alleud, Belgium.
| | - Mark Jairaj
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE United Kingdom
| | - Kieran Jones
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE United Kingdom
| | | | - Sherri Dudal
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE United Kingdom
| | - Yves Dudal
- INOFEA AG, Hofackerstrasse 40B, Muttenz CH-4132, Switzerland
| | | | - Rita Correro
- INOFEA AG, Hofackerstrasse 40B, Muttenz CH-4132, Switzerland
| | - Jun Qu
- University of Buffalo, Buffalo, NY 14260, United States
| | - Bo An
- University of Buffalo, Buffalo, NY 14260, United States
| | - Richard Lucey
- DDS (Drug Development Solutions), Newmarket Rd, Fordham, CB7 5WW, United Kingdom
| | - Szabolcs Szarka
- DDS (Drug Development Solutions), Newmarket Rd, Fordham, CB7 5WW, United Kingdom
| | - Robert Wheller
- DDS (Drug Development Solutions), Newmarket Rd, Fordham, CB7 5WW, United Kingdom
| | - Alina Pruna
- DDS (Drug Development Solutions), Newmarket Rd, Fordham, CB7 5WW, United Kingdom
| | - Sarah Kettell
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE United Kingdom
| | - Andrew Pitt
- University of Aston, Birmingham, B4 7ET, United Kingdom
| | - Paul Cutler
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE United Kingdom
| |
Collapse
|
21
|
Abstract
The ability to manipulate the chemical composition of proteins and peptides has been central to the development of improved polypeptide-based therapeutics and has enabled researchers to address fundamental biological questions that would otherwise be out of reach. Protein ligation, in which two or more polypeptides are covalently linked, is a powerful strategy for generating semisynthetic products and for controlling polypeptide topology. However, specialized tools are required to efficiently forge a peptide bond in a chemoselective manner with fast kinetics and high yield. Fortunately, nature has addressed this challenge by evolving enzymatic mechanisms that can join polypeptides using a diverse set of chemical reactions. Here, we summarize how such nature-inspired protein ligation strategies have been repurposed as chemical biology tools that afford enhanced control over polypeptide composition.
Collapse
Affiliation(s)
- Rasmus Pihl
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Qingfei Zheng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA.
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA.
| | - Yael David
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
22
|
Naranjo-Alcazar R, Bendix S, Groth T, Gallego Ferrer G. Research Progress in Enzymatically Cross-Linked Hydrogels as Injectable Systems for Bioprinting and Tissue Engineering. Gels 2023; 9:gels9030230. [PMID: 36975679 PMCID: PMC10048521 DOI: 10.3390/gels9030230] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Hydrogels have been developed for different biomedical applications such as in vitro culture platforms, drug delivery, bioprinting and tissue engineering. Enzymatic cross-linking has many advantages for its ability to form gels in situ while being injected into tissue, which facilitates minimally invasive surgery and adaptation to the shape of the defect. It is a highly biocompatible form of cross-linking, which permits the harmless encapsulation of cytokines and cells in contrast to chemically or photochemically induced cross-linking processes. The enzymatic cross-linking of synthetic and biogenic polymers also opens up their application as bioinks for engineering tissue and tumor models. This review first provides a general overview of the different cross-linking mechanisms, followed by a detailed survey of the enzymatic cross-linking mechanism applied to both natural and synthetic hydrogels. A detailed analysis of their specifications for bioprinting and tissue engineering applications is also included.
Collapse
Affiliation(s)
- Raquel Naranjo-Alcazar
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain
- Correspondence:
| | - Sophie Bendix
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany
| | - Thomas Groth
- Department of Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany
- Interdisciplinary Center of Material Research, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Gloria Gallego Ferrer
- Centre for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine, Carlos III Health Institute (CIBER-BBN, ISCIII), 46022 Valencia, Spain
| |
Collapse
|
23
|
Holz E, Darwish M, Tesar DB, Shatz-Binder W. A Review of Protein- and Peptide-Based Chemical Conjugates: Past, Present, and Future. Pharmaceutics 2023; 15:600. [PMID: 36839922 PMCID: PMC9959917 DOI: 10.3390/pharmaceutics15020600] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Over the past few decades, the complexity of molecular entities being advanced for therapeutic purposes has continued to evolve. A main propellent fueling innovation is the perpetual mandate within the pharmaceutical industry to meet the needs of novel disease areas and/or delivery challenges. As new mechanisms of action are uncovered, and as our understanding of existing mechanisms grows, the properties that are required and/or leveraged to enable therapeutic development continue to expand. One rapidly evolving area of interest is that of chemically enhanced peptide and protein therapeutics. While a variety of conjugate molecules such as antibody-drug conjugates, peptide/protein-PEG conjugates, and protein conjugate vaccines are already well established, others, such as antibody-oligonucleotide conjugates and peptide/protein conjugates using non-PEG polymers, are newer to clinical development. This review will evaluate the current development landscape of protein-based chemical conjugates with special attention to considerations such as modulation of pharmacokinetics, safety/tolerability, and entry into difficult to access targets, as well as bioavailability. Furthermore, for the purpose of this review, the types of molecules discussed are divided into two categories: (1) therapeutics that are enhanced by protein or peptide bioconjugation, and (2) protein and peptide therapeutics that require chemical modifications. Overall, the breadth of novel peptide- or protein-based therapeutics moving through the pipeline each year supports a path forward for the pursuit of even more complex therapeutic strategies.
Collapse
Affiliation(s)
- Emily Holz
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Martine Darwish
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Whitney Shatz-Binder
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
24
|
Peptidic Inhibitors and a Fluorescent Probe for the Selective Inhibition and Labelling of Factor XIIIa Transglutaminase. Molecules 2023; 28:molecules28041634. [PMID: 36838622 PMCID: PMC9960274 DOI: 10.3390/molecules28041634] [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: 12/22/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Factor XIIIa (FXIIIa) is a transglutaminase of major therapeutic interest for the development of anticoagulants due to its essential role in the blood coagulation cascade. While numerous FXIIIa inhibitors have been reported, they failed to reach clinical evaluation due to their lack of metabolic stability and low selectivity over transglutaminase 2 (TG2). Furthermore, the chemical tools available for the study of FXIIIa activity and localization are extremely limited. To combat these shortcomings, we designed, synthesised, and evaluated a library of 21 novel FXIIIa inhibitors. Electrophilic warheads, linker lengths, and hydrophobic units were varied on small molecule and peptidic scaffolds to optimize isozyme selectivity and potency. A previously reported FXIIIa inhibitor was then adapted for the design of a probe bearing a rhodamine B moiety, producing the innovative KM93 as the first known fluorescent probe designed to selectively label active FXIIIa with high efficiency (kinact/KI = 127,300 M-1 min-1) and 6.5-fold selectivity over TG2. The probe KM93 facilitated fluorescent microscopy studies within bone marrow macrophages, labelling FXIIIa with high efficiency and selectivity in cell culture. The structure-activity trends with these novel inhibitors and probes will help in the future study of the activity, inhibition, and localization of FXIIIa.
Collapse
|
25
|
Syeddan SA. Research Methodology and Mechanisms of Action of Current Orthopaedic Implant Coatings. J Long Term Eff Med Implants 2023; 33:51-66. [PMID: 36734927 DOI: 10.1615/jlongtermeffmedimplants.2022040062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Orthopedic implants are crucial interventions that are gaining greater importance in modern medicine to restore function to commonly affected joints. Each implantation carries the risk of implant-associated infection and loosening of the implant due to improper integration with soft tissue. Coating strategies have been developed to aid the growth of bone into the implant (osteointegration) and prevent biofilm formation to avoid infection. In this review, primary articles highlighting recent developments and advancements in orthopedic implant coating will be presented. Additionally, the methodology of the articles will be critiqued based on this research criteria: establishment of function on a theoretical basis, validation of coating function, and potential next steps/improvements based on results. A theoretical basis based on understanding the mechanisms at play of these various coatings allows for systems to be developed to tackle the tasks of osteointegration, subversion of infection, and avoidance of cytotoxicity. The current state of research methodology in coating design focuses too heavily on either osteointegration or the prevention of infection, thus, future development in medical implant coating needs to investigate the creation of a coating that accomplishes both tasks. Additionally, next steps and improvements to systems need to be better highlighted to move forward when problems arise within a system. Research currently showcasing new coatings is performed primarily in vitro and in vivo. More clinical trials need to be performed to highlight long-term sustainability, the structural integrity, and the safety of the implant.
Collapse
|
26
|
Zou L, Geng X, Li Z, Li T. Design of highly active substrates using molecular docking for microbial transglutaminase detection. RSC Adv 2023; 13:5259-5265. [PMID: 36793302 PMCID: PMC9923216 DOI: 10.1039/d2ra06467g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
The transglutaminase (TGase) family catalyzes a transamidation reaction between glutamine (Gln) and lysine (Lys) residues on protein substrates. Highly active substrates are important for cross-linking and modifying proteins of TGase. In the present work, high-activity substrates have been designed based on the principles of enzyme-substrate interaction, using microbial transglutaminase (mTGase) as a research model of the TGase family. Substrates with high activity were screened using a combination of molecular docking and traditional experiments. Twenty-four sets of peptide substrates all produced good catalytic activity with mTGase. FFKKAYAV as the acyl acceptor and VLQRAY as the acyl donor group had the best reaction efficiency with highly sensitive detection of 26 nM mTGase. In addition, the substrate grouping, KAYAV and AFQSAY, detected 130 nM mTGase under physiological conditions (37 °C, pH 7.4), producing 20-fold higher activity than the natural substrate, collagen. The experimental results confirmed the potential for design of high-activity substrates by a combination of molecular docking and traditional experiments under physiological conditions.
Collapse
Affiliation(s)
- Longhao Zou
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, School of Life Sciences, Jilin University Changchun China
| | - Xu Geng
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, School of Life Sciences, Jilin University Changchun China
| | - Zhengqiang Li
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, School of Life Sciences, Jilin University Changchun China
| | - Tao Li
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, School of Life Sciences, Jilin University Changchun China
| |
Collapse
|
27
|
Yu Y, Li F, Li J, Zheng X, Tian H, Mahmut Z, Du Y, Dai Y, Wang L. Lipase-catalyzed hydrazine insertion for the synthesis of N'-alkyl benzohydrazides. Biotechnol Appl Biochem 2023; 70:130-136. [PMID: 35285069 DOI: 10.1002/bab.2335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/22/2022] [Indexed: 11/07/2022]
Abstract
N'-alkyl benzohydrazides are classic organic compounds that have been widely utilized in organic chemistry. In this study, an efficient method was developed for the synthesis of N'-alkyl benzohydrazides by hydrazine insertion catalyzed by lipase. Under the optimal conditions (Morita-Baylis-Hillman ketone [1 mmol], phenylhydrazine [1.3 mmol], N,N-dimethylformamide [2 ml], lipase [20 mg], room temperature, 12 h), satisfactory yields (71-97%) and substrate tolerance were obtained when porcine pancreatic lipase was used as biocatalyst. These findings imply the great potential for the lipase-catalyzed synthesis of N'-alkyl benzohydrazides and extend the utilization of lipase in organic chemistry.
Collapse
Affiliation(s)
- Yue Yu
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Fengxi Li
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Jiapeng Li
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Xin Zheng
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Haochen Tian
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Zulpiya Mahmut
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yanan Du
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yuyin Dai
- Department of Nuclear Medicine, the First Hospital of Jilin University, Changchun, China
| | - Lei Wang
- Key Laboratory of Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
28
|
Enzymatic Crosslinked Hydrogels of Gelatin and Poly (Vinyl Alcohol) Loaded with Probiotic Bacteria as Oral Delivery System. Pharmaceutics 2022; 14:pharmaceutics14122759. [PMID: 36559253 PMCID: PMC9784308 DOI: 10.3390/pharmaceutics14122759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Probiotic bacteria are widely used to prepare pharmaceutical products and functional foods because they promote and sustain health. Nonetheless, probiotic viability is prone to decrease under gastrointestinal conditions. In this investigation, Lactiplantibacillus plantarum spp. CM-CNRG TB98 was entrapped in a gelatin−poly (vinyl alcohol) (Gel−PVA) hydrogel which was prepared by a “green” route using microbial transglutaminase (mTGase), which acts as a crosslinking agent. The hydrogel was fully characterized and its ability to entrap and protect L. plantarum from the lyophilization process and under simulated gastric and intestine conditions was explored. The Gel−PVA hydrogel showed a high probiotic loading efficiency (>90%) and survivability from the lyophilization process (91%) of the total bacteria entrapped. Under gastric conditions, no disintegration of the hydrogel was observed, keeping L. plantarum protected with a survival rate of >94%. While in the intestinal fluid the hydrogel is completely dissolved, helping to release probiotics. A Gel−PVA hydrogel is suitable for a probiotic oral administration system due to its physicochemical properties, lack of cytotoxicity, and the protection it offers L. plantarum under gastric conditions.
Collapse
|
29
|
A comprehensive review on gelatin: Understanding impact of the sources, extraction methods, and modifications on potential packaging applications. Food Packag Shelf Life 2022. [DOI: 10.1016/j.fpsl.2022.100945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
30
|
Inorganic/Biopolymers Hybrid Hydrogels Dual Cross-Linked for Bone Tissue Regeneration. Gels 2022; 8:gels8120762. [PMID: 36547286 PMCID: PMC9777565 DOI: 10.3390/gels8120762] [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: 11/04/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
In tissue engineering, the potential of re-growing new tissue has been considered, however, developments towards such clinical and commercial outcomes have been modest. One of the most important elements here is the selection of a biomaterial that serves as a "scaffold" for the regeneration process. Herein, we designed hydrogels composed of two biocompatible natural polymers, namely gelatin with photopolymerizable functionalities and a pectin derivative amenable to direct protein conjugation. Aiming to design biomimetic hydrogels for bone regeneration, this study proposes double-reinforcement by way of inorganic/biopolymer hybrid filling composed of Si-based compounds and cellulose nanofibers. To attain networks with high flexibility and elastic modulus, a double-crosslinking strategy was envisioned-photochemical and enzyme-mediated conjugation reactions. The dual cross-linked procedure will generate intra- and intermolecular interactions between the protein and polysaccharide and might be a resourceful strategy to develop innovative scaffolding materials.
Collapse
|
31
|
Sood A, Ji SM, Kumar A, Han SS. Enzyme-Triggered Crosslinked Hybrid Hydrogels for Bone Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6383. [PMID: 36143697 PMCID: PMC9506111 DOI: 10.3390/ma15186383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 06/16/2023]
Abstract
The quest to develop state-of-the-art hydrogels for bone tissue engineering has accompanied substantial innovation and significant progression in the field of bioactive hydrogels. Still, there is scope for advancement in this cell-friendly and biocompatible scaffold system. The crosslinking approaches used for hydrogel synthesis plays a decisive role in guiding and regulating the mechanical stability, network framework, macroscopic architect, immunological behaviors, and cellular responses. Until recently, enzyme-based crosslinking strategies were considered as the pinnacle in designing efficient hybrid hydrogel systems. A variety of enzymes have been explored for manufacturing hydrogels while taking the advantage of the biocompatible nature, specificity, ability to produce nontoxic by products and high efficiency of enzymes. The current review focuses on the utility of different enzymes as crosslinking agents for hydrogel formation with their application in bone tissue engineering. The field of enzyme crosslinked hydrogel synthesis is rapidly maturing with a lot of opportunities to be explored in bone tissue engineering. Enzyme-based in situ and externally crosslinked hydrogels for bone regeneration is an attractive field, and with innovation in using engineered enzymes this field will continue to flourish with clinical orientation.
Collapse
Affiliation(s)
- Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
| | - Seong Min Ji
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
- Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
- Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Korea
| |
Collapse
|
32
|
L-Glutamine-, peptidyl- and protein-glutaminases: structural features and applications in the food industry. World J Microbiol Biotechnol 2022; 38:204. [PMID: 36002753 DOI: 10.1007/s11274-022-03391-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022]
Abstract
L-Glutaminases are enzymes that catalyze the cleavage of the gamma-amido bond of L-glutamine residues, producing ammonia and L-glutamate. These enzymes have several applications in food and pharmaceutical industries. However, the L-glutaminases that hydrolyze free L-glutamine (L-glutamine glutaminases, EC 3.5.1.2) have different structures and properties with respect to the L-glutaminases that hydrolyze the same amino acid covalently bound in peptides (peptidyl glutaminases, EC 3.5.1.43) and proteins (protein-glutamine glutaminase, EC 3.5.1.44). In the food industry, L-glutamine glutaminases are applied to enhance the flavor of foods, whereas protein glutaminases are useful to improve the functional properties of proteins. This review will focus on structural backgrounds and differences between these enzymes, the methodology available to measure the activity as well as strengths and limitations. Production methods, applications, and challenges in the food industry will be also discussed. This review will provide useful information to search and identify the suitable L-glutaminase that best fits to the intended application.
Collapse
|
33
|
Wachendörfer M, Schräder P, Buhl EM, Palkowitz AL, Ben Messaoud G, Richtering W, Fischer H. A defined heat pretreatment of gelatin enables control of hydrolytic stability, stiffness, and microstructural architecture of fibrin-gelatin hydrogel blends. Biomater Sci 2022; 10:5552-5565. [PMID: 35969162 DOI: 10.1039/d2bm00214k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fibrin-gelatin hydrogel blends exhibit high potential for tissue engineering in vitro applications. However, the means to tailor these blends in order to control their properties, thus opening up a broad range of new target applications, have been insufficiently explored. We hypothesized that a controlled heat treatment of gelatin prior to blend synthesis enables control of hydrolytic swelling and shrinking, stiffness, and microstructural architecture of fibrin-gelatin based hydrogel blends while providing tremendous long-term stability. We investigated these hydrogel blends' compressive strength, in vitro degradation stability, and microstructure in order to test this hypothesis. In addition, we examined the gel's ability to support endothelial cell proliferation and stretching of encapsulated smooth muscle cells. This research showed that a controlled heat pretreatment of the gelatin component strongly influenced the stiffness, swelling, shrinking, and microstructural architecture of the final blends regardless of identical gelatin mass fractions. All blends offered high long-term hydrolytic stability. In conclusion, the results of this study open the possibility to use this technique in order to tune low-concentrated, open-porous fibrin-based hydrogels, even in long-term tissue engineering in vitro experiments.
Collapse
Affiliation(s)
- Mattis Wachendörfer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Philipp Schräder
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Alena L Palkowitz
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Ghazi Ben Messaoud
- Institute of Physical Chemistry, RWTH Aachen University, Landoltweg 2, 52074 Aachen, Germany.,DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany
| | - Walter Richtering
- Institute of Physical Chemistry, RWTH Aachen University, Landoltweg 2, 52074 Aachen, Germany.,DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| |
Collapse
|
34
|
Suzuki M, Date M, Kashiwagi T, Suzuki E, Yokoyama K. Rational design of a disulfide bridge increases the thermostability of microbial transglutaminase. Appl Microbiol Biotechnol 2022; 106:4553-4562. [PMID: 35729274 DOI: 10.1007/s00253-022-12024-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022]
Abstract
Microbial transglutaminase (MTG) has numerous industrial applications in the food and pharmaceutical sectors. Unfortunately, the thermostability of MTG is too low to tolerate the desired conditions used in many of these commercial processes. In a previous study, we used protein engineering to improve the thermostability of MTG. Specifically, we generated a T7C/E58C mutant of MTG from Streptomyces mobaraensis that displayed enhanced resistance to thermal inactivation. In this study, a rational structure-based approach was adopted to introduce a disulfide bridge to further increase the thermostability of MTG. In all, four new mutants, each containing a novel disulfide bond, were engineered. Of these four mutants, D3C/G283C showed the most promising thermostability with a significantly higher ∆T50 (defined as the temperature of incubation at which 50% of the initial activity remains) of + 9 °C by comparison to wild-type MTG. Indeed, D3C/G283C combined enhanced thermostability with a 2.1-fold increased half-life at 65 °C compared with the wild-type enzyme. By structure-based rational design, we were able to create an MTG variant which might be useful for expanding the scope of application in food. KEY POINTS: • Microbial transglutaminase (MTG) is an enzyme used in many food applications • The applicability of MTG to various industrial processes other than the food sector is being investigated • Improvement of thermostability was confirmed for the disulfide bridge mutant D3C/G283C.
Collapse
Affiliation(s)
- Mototaka Suzuki
- Institute for Innovation, Ajinomoto Co., Inc., 1-1, Suzuki-cho, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Masayo Date
- Institute for Innovation, Ajinomoto Co., Inc., 1-1, Suzuki-cho, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Tatsuki Kashiwagi
- Institute for Innovation, Ajinomoto Co., Inc., 1-1, Suzuki-cho, Kawasaki-shi, Kanagawa, 210-8681, Japan
| | - Eiichiro Suzuki
- Institute for Innovation, Ajinomoto Co., Inc., 1-1, Suzuki-cho, Kawasaki-shi, Kanagawa, 210-8681, Japan.,Kihara Memorial Yokohama Foundation for the Advancement of Life Sciences Yokohama, Bio Industry Center, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Keiichi Yokoyama
- Institute for Innovation, Ajinomoto Co., Inc., 1-1, Suzuki-cho, Kawasaki-shi, Kanagawa, 210-8681, Japan. .,R&B Planning Department, Ajinomoto Co., Inc, Tokyo, 104-8315, Japan.
| |
Collapse
|
35
|
Lai E, Bao B, Zhu Y, Lin H. Transglutaminase-Catalyzed Bottom-Up Synthesis of Polymer Hydrogel. Front Bioeng Biotechnol 2022; 10:824747. [PMID: 35392400 PMCID: PMC8980521 DOI: 10.3389/fbioe.2022.824747] [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: 11/29/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Enzyme catalysis has attracted increasing attention for application in the synthesis of polymer hydrogel due to the eco-friendly process and the devisable catalytic reaction. Moreover, bottom-up approaches combining enzyme catalysts and molecular self-assembly have been explored for synthesizing hydrogel with complex architectures. An enzyme widely distributed in nature, transglutaminase (TGase) has been confirmed to catalyze the formation of isopeptide bonds between proteins, which can effectively improve the gelation of proteins. In this mini-review, TGase-catalyzed synthesis of polymer hydrogels, including fibrin hydrogels, polyethylene glycol hydrogels, soy protein hydrogels, collagen hydrogels, gelatin hydrogels and hyaluronan hydrogels, has been reviewed in detail. The catalytic process and gel formation mechanism by TGase have also been considered. Furthermore, future perspectives and challenges in the preparation of polymer hydrogels by TGase are also highlighted.
Collapse
|
36
|
Gray VP, Amelung CD, Duti IJ, Laudermilch EG, Letteri RA, Lampe KJ. Biomaterials via peptide assembly: Design, characterization, and application in tissue engineering. Acta Biomater 2022; 140:43-75. [PMID: 34710626 PMCID: PMC8829437 DOI: 10.1016/j.actbio.2021.10.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A core challenge in biomaterials, with both fundamental significance and technological relevance, concerns the rational design of bioactive microenvironments. Designed properly, peptides can undergo supramolecular assembly into dynamic, physical hydrogels that mimic the mechanical, topological, and biochemical features of native tissue microenvironments. The relatively facile, inexpensive, and automatable preparation of peptides, coupled with low batch-to-batch variability, motivates the expanded use of assembling peptide hydrogels for biomedical applications. Integral to realizing dynamic peptide assemblies as functional biomaterials for tissue engineering is an understanding of the molecular and macroscopic features that govern assembly, morphology, and biological interactions. In this review, we first discuss the design of assembling peptides, including primary structure (sequence), secondary structure (e.g., α-helix and β-sheets), and molecular interactions that facilitate assembly into multiscale materials with desired properties. Next, we describe characterization tools for elucidating molecular structure and interactions, morphology, bulk properties, and biological functionality. Understanding of these characterization methods enables researchers to access a variety of approaches in this ever-expanding field. Finally, we discuss the biological properties and applications of peptide-based biomaterials for engineering several important tissues. By connecting molecular features and mechanisms of assembling peptides to the material and biological properties, we aim to guide the design and characterization of peptide-based biomaterials for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Engineering peptide-based biomaterials that mimic the topological and mechanical properties of natural extracellular matrices provide excellent opportunities to direct cell behavior for regenerative medicine and tissue engineering. Here we review the molecular-scale features of assembling peptides that result in biomaterials that exhibit a variety of relevant extracellular matrix-mimetic properties and promote beneficial cell-biomaterial interactions. Aiming to inspire and guide researchers approaching this challenge from both the peptide biomaterial design and tissue engineering perspectives, we also present characterization tools for understanding the connection between peptide structure and properties and highlight the use of peptide-based biomaterials in neural, orthopedic, cardiac, muscular, and immune engineering applications.
Collapse
Affiliation(s)
- Vincent P Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Connor D Amelung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Israt Jahan Duti
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Emma G Laudermilch
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| |
Collapse
|
37
|
Haas D, Hauptstein N, Dirauf M, Driessen MD, Ruopp M, Schubert US, Lühmann T, Meinel L. Chemo-Enzymatic PEGylation/POxylation of Murine Interleukin-4. Bioconjug Chem 2022; 33:97-104. [PMID: 34967625 DOI: 10.1021/acs.bioconjchem.1c00495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interleukin-4 (IL-4) is a potentially interesting anti-inflammatory therapeutic, which is rapidly excreted. Therefore, serum half-life extension by polymer conjugation is desirable, which may be done by PEGylation. Here, we use PEtOx as an alternative to PEG for bioconjugate engineering. We genetically extended murine IL-4 (mIL-4) with the d-domain of insulin-like growth factor I (IGF-I), a previously identified substrate of transglutaminase (TG) Factor XIIIa (FXIIIa). Thereby, engineered mIL-4 (mIL-4-TG) became an educt for TG catalyzed C-terminal, site-directed conjugation. This was deployed to enzymatically couple an azide group containing peptide sequence to mIL-4, allowing C-terminal bioconjugation of polyethylene glycol or poly(2-ethyl-2-oxazoline). Both bioconjugates had wild-type potency and alternatively polarized macrophages.
Collapse
Affiliation(s)
- Dorothee Haas
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Niklas Hauptstein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Michael Dirauf
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Marc D Driessen
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Matthias Ruopp
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| |
Collapse
|
38
|
Elham Badali, Hosseini M, Mohajer M, Hassanzadeh S, Saghati S, Hilborn J, Khanmohammadi M. Enzymatic Crosslinked Hydrogels for Biomedical Application. POLYMER SCIENCE SERIES A 2021. [DOI: 10.1134/s0965545x22030026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
da S. Pereira A, Souza CPL, Moraes L, Fontes-Sant’Ana GC, Amaral PFF. Polymers as Encapsulating Agents and Delivery Vehicles of Enzymes. Polymers (Basel) 2021; 13:polym13234061. [PMID: 34883565 PMCID: PMC8659040 DOI: 10.3390/polym13234061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 01/15/2023] Open
Abstract
Enzymes are versatile biomolecules with broad applications. Since they are biological molecules, they can be easily destabilized when placed in adverse environmental conditions, such as variations in temperature, pH, or ionic strength. In this sense, the use of protective structures, as polymeric capsules, has been an excellent approach to maintain the catalytic stability of enzymes during their application. Thus, in this review, we report the use of polymeric materials as enzyme encapsulation agents, recent technological developments related to this subject, and characterization methodologies and possible applications of the formed bioactive structures. Our search detected that the most explored methods for enzyme encapsulation are ionotropic gelation, spray drying, freeze-drying, nanoprecipitation, and electrospinning. α-chymotrypsin, lysozyme, and β-galactosidase were the most used enzymes in encapsulations, with chitosan and sodium alginate being the main polymers. Furthermore, most studies reported high encapsulation efficiency, enzyme activity maintenance, and stability improvement at pH, temperature, and storage. Therefore, the information presented here shows a direction for the development of encapsulation systems capable of stabilizing different enzymes and obtaining better performance during application.
Collapse
Affiliation(s)
- Adejanildo da S. Pereira
- Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (A.d.S.P.); (C.P.L.S.); (L.M.)
| | - Camila P. L. Souza
- Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (A.d.S.P.); (C.P.L.S.); (L.M.)
| | - Lidiane Moraes
- Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (A.d.S.P.); (C.P.L.S.); (L.M.)
| | - Gizele C. Fontes-Sant’Ana
- Biochemical Processes Technology Department, Chemistry Institute, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil;
| | - Priscilla F. F. Amaral
- Escola de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (A.d.S.P.); (C.P.L.S.); (L.M.)
- Correspondence: ; Tel.: +55-21-3938-7623
| |
Collapse
|
40
|
Kurt-Celep İ, Nihan Kilinc A, Griffin M, Telci D. Nitrosylation of Tissue Transglutaminase enhances fibroblast migration and regulates MMP activation. Matrix Biol 2021; 105:1-16. [PMID: 34763097 DOI: 10.1016/j.matbio.2021.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022]
Abstract
In wound healing, the TG2 enzyme plays a dual functional role. TG2 has been shown to regulate extracellular matrix (ECM) stabilization by its transamidase activity while increasing cell migration by acting as a cell adhesion molecule. In this process, nitric oxide (NO) plays a particularly important role by nitrosylation of free cysteine residues on TG2, leading to the irreversible inactivation of the catalytic activity. In this study, transfected fibroblasts expressing TG2 under the control of the tetracycline-off promoter were treated with NO donor s-nitroso-n-acetyl penicillamine (SNAP) to analyze the interplay between NO and TG2 in the regulation of cell migration/invasion as well as TGF-β1-dependent MMP activation. Our results demonstrated that inhibition of TG2 cross-linking activity by SNAP promoted the migration and invasion capacity of fibroblasts by hindering TG2-mediated TGF-β1 activation. While the inhibition of TG2 activity by NO downregulated the biosynthesis and activity of MMP-2 and MMP-9, that of MMP-1a and MMP-13 shown to be upregulated in a TGF-β1-dependent manner under the same conditions. In the presence of SNAP, interaction of TG2 with its cell surface binding partners Integrin-β1 and Syndecan-4 was reduced, which was paralleled by an increase in TG2 and PDGF association. These findings suggests that migratory phenotype of fibroblasts can be regulated by the interplay between nitric oxide and TG2 activity.
Collapse
Affiliation(s)
- İnci Kurt-Celep
- Department of Genetics and Bioengineering, Yeditepe University, 26 August Campus, Kayisdagi, Atasehir, Istanbul 34755, Turkey
| | - Ayse Nihan Kilinc
- Department of Genetics and Bioengineering, Yeditepe University, 26 August Campus, Kayisdagi, Atasehir, Istanbul 34755, Turkey; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Martin Griffin
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Dilek Telci
- Department of Genetics and Bioengineering, Yeditepe University, 26 August Campus, Kayisdagi, Atasehir, Istanbul 34755, Turkey.
| |
Collapse
|
41
|
Wang H, Wang Y, Yuan Z, Wang Y, Li X, Song P, Lu F, Liu Y. Insight into the cross-linking preferences and characteristics of the transglutaminase from Bacillus subtilis by in vitro RNA display. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.112152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
42
|
Fatima SW, Imtiyaz K, Alam Rizvi MM, Khare SK. Microbial transglutaminase nanoflowers as an alternative nanomedicine for breast cancer theranostics. RSC Adv 2021; 11:34613-34630. [PMID: 35494746 PMCID: PMC9042677 DOI: 10.1039/d1ra04513j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the most common malignancy among women. With the aim of decreasing the toxicity of conventional breast cancer treatments, an alternative that could provide appropriate and effective drug utilization was envisioned. Thus, we contemplated and compared the in vitro effects of microbial transglutaminase nanoflowers (MTGase NFs) on breast cancer cells (MCF-7). Transglutaminase is an important regulatory enzyme acting as a site-specific cross-linker for proteins. With the versatility of MTGase facilitating the nanoflower formation by acting as molecular glue, it was demonstrated to have anti-cancer properties. The rational drug design based on a transglutaminase enzyme-assisted approach led to the uniform shape of petals in these nanoflowers, which had the capacity to act directly as an anti-cancer drug. Herein, we report the anti-cancer characteristics portrayed by enzymatic MTGase NFs, which are biocompatible in nature. This study demonstrated the prognostic and therapeutic significance of MTGase NFs as a nano-drug in breast cancer treatment. The results on MCF-7 cells showed a significantly improved in vitro therapeutic efficacy. MTGase NFs were able to exhibit inhibitory effects on cell viability (IC50-8.23 μg ml−1) within 24 h of dosage. To further substantiate its superior anti-proliferative role, the clonogenic potential was measured to be 62.8%, along with migratory inhibition of cells (3.76-fold change). Drastic perturbations were induced (4.61-fold increase in G0/G1 phase arrest), pointed towards apoptotic induction with a 58.9% effect. These results validated the role of MTGase NFs possessing a cytotoxic nature in mitigating breast cancer. Thus, MTGase bestows distinct functionality towards therapeutic nano-modality, i.e., nanoflowers, which shows promise in cancer treatment. Development of a novel therapeutic nano-modality in the form of enzymatic transglutaminase nanoflowers; endowed with anti-cancerous action against breast cancers.![]()
Collapse
Affiliation(s)
- Syeda Warisul Fatima
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi New Delhi-110016 India +91-112659 6533
| | - Khalid Imtiyaz
- Genome Biology Laboratory, Department of Biosciences, Jamia Millia Islamia New Delhi-110025 India
| | - Mohammad M Alam Rizvi
- Genome Biology Laboratory, Department of Biosciences, Jamia Millia Islamia New Delhi-110025 India
| | - Sunil K Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi New Delhi-110016 India +91-112659 6533
| |
Collapse
|
43
|
Fabrication of Soft Tissue Scaffold-Mimicked Microelectrode Arrays Using Enzyme-Mediated Transfer Printing. MICROMACHINES 2021; 12:mi12091057. [PMID: 34577700 PMCID: PMC8472004 DOI: 10.3390/mi12091057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022]
Abstract
Hydrogels are the ideal materials in the development of implanted bioactive neural interfaces because of the nerve tissue-mimicked physical and biological properties that can enhance neural interfacing compatibility. However, the integration of hydrogels and rigid/dehydrated electronic microstructure is challenging due to the non-reliable interfacial bonding, whereas hydrogels are not compatible with most conditions required for the micromachined fabrication process. Herein, we propose a new enzyme-mediated transfer printing process to design an adhesive biological hydrogel neural interface. The donor substrate was fabricated via photo-crosslinking of gelatin methacryloyl (GelMA) containing various conductive nanoparticles (NPs), including Ag nanowires (NWs), Pt NWs, and PEDOT:PSS, to form a stretchable conductive bioelectrode, called NP-doped GelMA. On the other hand, a receiver substrate composed of microbial transglutaminase-incorporated gelatin (mTG-Gln) enabled simultaneous temporally controlled gelation and covalent bond-enhanced adhesion to achieve one-step transfer printing of the prefabricated NP-doped GelMA features. The integrated hydrogel microelectrode arrays (MEA) were adhesive, and mechanically/structurally bio-compliant with stable conductivity. The devices were structurally stable in moisture to support the growth of neuronal cells. Despite that the introduction of AgNW and PEDOT:PSS NPs in the hydrogels needed further study to avoid cell toxicity, the PtNW-doped GelMA exhibited a comparable live cell density. This Gln-based MEA is expected to be the next-generation bioactive neural interface.
Collapse
|
44
|
Tatsukawa H, Hitomi K. Role of Transglutaminase 2 in Cell Death, Survival, and Fibrosis. Cells 2021; 10:cells10071842. [PMID: 34360011 PMCID: PMC8307792 DOI: 10.3390/cells10071842] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022] Open
Abstract
Transglutaminase 2 (TG2) is a ubiquitously expressed enzyme catalyzing the crosslinking between Gln and Lys residues and involved in various pathophysiological events. Besides this crosslinking activity, TG2 functions as a deamidase, GTPase, isopeptidase, adapter/scaffold, protein disulfide isomerase, and kinase. It also plays a role in the regulation of hypusination and serotonylation. Through these activities, TG2 is involved in cell growth, differentiation, cell death, inflammation, tissue repair, and fibrosis. Depending on the cell type and stimulus, TG2 changes its subcellular localization and biological activity, leading to cell death or survival. In normal unstressed cells, intracellular TG2 exhibits a GTP-bound closed conformation, exerting prosurvival functions. However, upon cell stimulation with Ca2+ or other factors, TG2 adopts a Ca2+-bound open conformation, demonstrating a transamidase activity involved in cell death or survival. These functional discrepancies of TG2 open form might be caused by its multifunctional nature, the existence of splicing variants, the cell type and stimulus, and the genetic backgrounds and variations of the mouse models used. TG2 is also involved in the phagocytosis of dead cells by macrophages and in fibrosis during tissue repair. Here, we summarize and discuss the multifunctional and controversial roles of TG2, focusing on cell death/survival and fibrosis.
Collapse
|
45
|
Bolzati C, Spolaore B. Enzymatic Methods for the Site-Specific Radiolabeling of Targeting Proteins. Molecules 2021; 26:3492. [PMID: 34201280 PMCID: PMC8229434 DOI: 10.3390/molecules26123492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/19/2022] Open
Abstract
Site-specific conjugation of proteins is currently required to produce homogenous derivatives for medicine applications. Proteins derivatized at specific positions of the polypeptide chain can actually show higher stability, superior pharmacokinetics, and activity in vivo, as compared with conjugates modified at heterogeneous sites. Moreover, they can be better characterized regarding the composition of the derivatization sites as well as the conformational and activity properties. To this aim, several site-specific derivatization approaches have been developed. Among these, enzymes are powerful tools that efficiently allow the generation of homogenous protein-drug conjugates under physiological conditions, thus preserving their native structure and activity. This review will summarize the progress made over the last decade on the use of enzymatic-based methodologies for the production of site-specific labeled immunoconjugates of interest for nuclear medicine. Enzymes used in this field, including microbial transglutaminase, sortase, galactosyltransferase, and lipoic acid ligase, will be overviewed and their recent applications in the radiopharmaceutical field will be described. Since nuclear medicine can benefit greatly from the production of homogenous derivatives, we hope that this review will aid the use of enzymes for the development of better radio-conjugates for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Cristina Bolzati
- Institute of Condensed Matter Chemistry and Technologies for Energy ICMATE-CNR, Corso Stati Uniti, 4, I-35127 Padova, Italy
| | - Barbara Spolaore
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, I-35131 Padova, Italy
- CRIBI Biotechnology Center, University of Padua, Viale G. Colombo, 3, I-35131 Padova, Italy
| |
Collapse
|
46
|
Song W, Ko J, Choi YH, Hwang NS. Recent advancements in enzyme-mediated crosslinkable hydrogels: In vivo-mimicking strategies. APL Bioeng 2021; 5:021502. [PMID: 33834154 PMCID: PMC8018798 DOI: 10.1063/5.0037793] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
Enzymes play a central role in fundamental biological processes and have been traditionally used to trigger various processes. In recent years, enzymes have been used to tune biomaterial responses and modify the chemical structures at desired sites. These chemical modifications have allowed the fabrication of various hydrogels for tissue engineering and therapeutic applications. This review provides a comprehensive overview of recent advancements in the use of enzymes for hydrogel fabrication. Strategies to enhance the enzyme function and improve biocompatibility are described. In addition, we describe future opportunities and challenges for the production of enzyme-mediated crosslinkable hydrogels.
Collapse
Affiliation(s)
- Wonmoon Song
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Junghyeon Ko
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Hwan Choi
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Nathaniel S. Hwang
- Author to whom correspondence should be addressed:. Tel.: 82-2-880-1635. Fax: 82-2-880-7295
| |
Collapse
|
47
|
Pagar AD, Patil MD, Flood DT, Yoo TH, Dawson PE, Yun H. Recent Advances in Biocatalysis with Chemical Modification and Expanded Amino Acid Alphabet. Chem Rev 2021; 121:6173-6245. [PMID: 33886302 DOI: 10.1021/acs.chemrev.0c01201] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The two main strategies for enzyme engineering, directed evolution and rational design, have found widespread applications in improving the intrinsic activities of proteins. Although numerous advances have been achieved using these ground-breaking methods, the limited chemical diversity of the biopolymers, restricted to the 20 canonical amino acids, hampers creation of novel enzymes that Nature has never made thus far. To address this, much research has been devoted to expanding the protein sequence space via chemical modifications and/or incorporation of noncanonical amino acids (ncAAs). This review provides a balanced discussion and critical evaluation of the applications, recent advances, and technical breakthroughs in biocatalysis for three approaches: (i) chemical modification of cAAs, (ii) incorporation of ncAAs, and (iii) chemical modification of incorporated ncAAs. Furthermore, the applications of these approaches and the result on the functional properties and mechanistic study of the enzymes are extensively reviewed. We also discuss the design of artificial enzymes and directed evolution strategies for enzymes with ncAAs incorporated. Finally, we discuss the current challenges and future perspectives for biocatalysis using the expanded amino acid alphabet.
Collapse
Affiliation(s)
- Amol D Pagar
- Department of Systems Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Mahesh D Patil
- Department of Systems Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Dillon T Flood
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Tae Hyeon Yoo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon 16499, Korea
| | - Philip E Dawson
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Hyungdon Yun
- Department of Systems Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| |
Collapse
|
48
|
Strategies to Use Nanofiber Scaffolds as Enzyme-Based Biocatalysts in Tissue Engineering Applications. Catalysts 2021. [DOI: 10.3390/catal11050536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nanofibers are considered versatile materials with remarkable potential in tissue engineering and regeneration. In addition to their extracellular matrix-mimicking properties, nanofibers can be functionalized with specific moieties (e.g., antimicrobial nanoparticles, ceramics, bioactive proteins, etc.) to improve their overall performance. A novel approach in this regard is the use of enzymes immobilized onto nanofibers to impart biocatalytic activity. These nanofibers are capable of carrying out the catalysis of various biological processes that are essential in the healing process of tissue. In this review, we emphasize the use of biocatalytic nanofibers in various tissue regeneration applications. Biocatalytic nanofibers can be used for wound edge or scar matrix digestion, which reduces the hindrance for cell migration and proliferation, hence displaying applications in fast tissue repair, e.g., spinal cord injury. These nanofibers have potential applications in bone regeneration, mediating osteogenic differentiation, biomineralization, and matrix formation through direct enzyme activity. Moreover, enzymes can be used to undertake efficient crosslinking and fabrication of nanofibers with better physicochemical properties and tissue regeneration potential.
Collapse
|
49
|
Hirono-Hara Y, Yui M, Hara KY. Production of transglutaminase in glutathione-producing recombinant Saccharomyces cerevisiae. AMB Express 2021; 11:13. [PMID: 33415535 PMCID: PMC7790930 DOI: 10.1186/s13568-020-01176-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/30/2020] [Indexed: 11/18/2022] Open
Abstract
Transglutaminase (TG) catalyzes the formation of cross-links between proteins. TG from Streptoverticillium mobaraense (SmTG) is used widely in food, cosmetic, biomaterial and medical industries. SmTG is occasionally supplied as a mixture with the activator peptide glutathione. Currently, glutathione is industrially produced using a budding yeast, Saccharomyces cerevisiae, because of its intracellular high content of glutathione. In this study, active SmTG was produced together with glutathione in S. cerevisiae. SmTG extracted from S. cerevisiae expressing SmTG showed cross-linking activity when BSA and sodium caseinate were substrates. The cross-linking activity of SmTG increased proportionally as the concentration of added glutathione increased. Furthermore, SmTG was prepared by extracting SmTG from an engineered S. cerevisiae whose glutathione synthetic pathway was enhanced. The SmTG solution showed higher activity when compared with a SmTG solution prepared from a S. cerevisiae strain without enhanced glutathione production. This result indicates that a high content of intracellular glutathione further enhances active SmTG production in S. cerevisiae. S. cerevisiae co-producing SmTG and a higher content of glutathione has the potential to supply a ready-to-use industrial active TG solution.![]()
Collapse
|
50
|
Rodríguez-Alba E, Dionisio N, Pérez-Calixto M, Huerta L, García-Uriostegui L, Hautefeuille M, Vázquez-Victorio G, Burillo G. Surface modification of polyethylenterephthalate film with primary amines using gamma radiation and aminolysis reaction for cell adhesion studies. Radiat Phys Chem Oxf Engl 1993 2020. [DOI: 10.1016/j.radphyschem.2020.109070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|