1
|
Li H, Li D, Wang X, Zeng Z, Pahlavan S, Zhang W, Wang X, Wang K. Progress in Biomaterials-Enhanced Vascularization by Modulating Physical Properties. ACS Biomater Sci Eng 2025; 11:33-54. [PMID: 39615049 DOI: 10.1021/acsbiomaterials.4c01106] [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: 01/06/2025]
Abstract
Sufficient vascular system and adequate blood perfusion is crucial for ensuring nutrient and oxygen supply within biomaterials. Actively exploring the optimal physical properties of biomaterials in various application scenarios has provided clues for enhancing vascularization within materials, leading to improved outcomes in tissue engineering and clinical translation. Here we focus on reviewing the physical properties of biomaterials, including pore structure, surface topography, and stiffness, and their effects on promoting vascularization. This angiogenic capability has the potential to provide better standardized research models and personalized treatment strategies for bone regeneration, wound healing, islet transplantation and cardiac repair.
Collapse
Affiliation(s)
- Hao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Dayan Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Ziyuan Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing 102200, China
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing 100191, China
| |
Collapse
|
2
|
Pan X, Nie J, Lei J, Wang P, Zheng K, Wei Q, Liu X. Integrin Subtypes and Lamellipodia Mediate Spatial Sensing of RGD Ligands during Cell Adhesion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:24882-24891. [PMID: 39546750 DOI: 10.1021/acs.langmuir.4c02796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Understanding how the spatial distribution of adhesive ligands regulates cell behavior is crucial for designing biomaterials. This study investigates how precisely controlled ligand spacing affects cell spreading and integrin subtype engagement. Using engineered polyacrylamide hydrogels with gold nanoparticle arrays, we explored the impact of RGD ligand spacings (30 and 150 nm) on human mesenchymal stromal cells. Cells exhibited distinct morphological behaviors: smaller spacings promoted larger spreading areas, while larger spacings resulted in elongated shapes with reduced spreading. Mechanistically, we found that the α5β1 integrin, not the αvβ3 integrin, played a central role in mediating these responses, alongside lamellipodia formation. Our findings provide critical insights into the spatial sensing of ligands, highlighting the influence of ligand spacing on cellular mechanotransduction and integrin-specific responses. This work advances the understanding of cell-material interactions and offers potential strategies for designing biomaterials to guide cell behavior in tissue engineering.
Collapse
Affiliation(s)
- Xiaokai Pan
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Juan Nie
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Jiacheng Lei
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Peng Wang
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Kaikai Zheng
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Qiang Wei
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| |
Collapse
|
3
|
Chee HL, M Y, Kim J, Koo JW, Luo P, Ramli MFH, Young JL, Wang F. Mechanical and Dimensional Stability of Gelatin-Based Hydrogels Through 3D Printing-Facilitated Confined Space Assembly. ACS APPLIED MATERIALS & INTERFACES 2024; 16:61105-61114. [PMID: 39460703 DOI: 10.1021/acsami.4c15025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Hydrogels have emerged as promising biomaterials for tissue regeneration; yet, their inherent swelling can cause deformation and reduced mechanical properties, posing challenges for practical applications in biomedical engineering. Traditional methods to reduce hydrogel swelling often involve complex synthesis procedures with limited flexibility. Inspired by nature's efficient designs, we present here the approach to improve hydrogel performance using 3D printing-assisted microstructure engineering. By utilizing polymerization-induced phase separation of hydrogel from copolymerization of gelatin methacrylate and hydroxyethyl methacrylate (poly(GelMA-co-HEMA)) in the confined space during vat photopolymerization (VPP) 3D printing, we replicate the cuttlebone-like microstructure of hydrogels with enhanced mechanical properties and swelling resistance. We demonstrate here a 4-fold increase in elastic modulus compared to bulk polymerization of poly(GelMA-co-HEMA), together with improved mechanical and dimensional stability. This method offers promising opportunities for practical biomedical and tissue engineering applications, overcoming previous limitations in the design and performance.
Collapse
Affiliation(s)
- Heng Li Chee
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Singapore
| | - Yashaaswini M
- School of Materials Science and Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Jaedeok Kim
- Mechanobiology Institute (MBI), National University of Singapore (NUS), 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Jing Wen Koo
- Mechanobiology Institute (MBI), National University of Singapore (NUS), 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Ping Luo
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Singapore
| | - M Faris H Ramli
- Mechanobiology Institute (MBI), National University of Singapore (NUS), 5A Engineering Drive 1, Singapore 117411, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore (NUS), 4 Engineering Drive 3, Singapore 117583, Singapore
| | - Jennifer L Young
- Mechanobiology Institute (MBI), National University of Singapore (NUS), 5A Engineering Drive 1, Singapore 117411, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore (NUS), 4 Engineering Drive 3, Singapore 117583, Singapore
| | - FuKe Wang
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Singapore
| |
Collapse
|
4
|
Moghaddam AS, Dunne K, Breyer W, Wu Y, Pashuck ET. Hydrogels with Independently Controlled Adhesion Ligand Mobility and Viscoelasticity Increase Cell Adhesion and Spreading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614501. [PMID: 39386463 PMCID: PMC11463488 DOI: 10.1101/2024.09.23.614501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
A primary objective in designing hydrogels for cell culture is recreating the cell-matrix interactions found within human tissues. Identifying the most important biomaterial features for these interactions is challenging because it is difficult to independently adjust variables such as matrix stiffness, stress relaxation, the mobility of adhesion ligands and the ability of these ligands to support cellular forces. In this work we designed a hydrogel platform consisting of interpenetrating polymer networks of covalently crosslinked poly(ethylene glycol) (PEG) and self-assembled peptide amphiphiles (PA). We can tailor the storage modulus of the hydrogel by altering the concentration and composition of each network, and we can tune the stress relaxation half-life through the non-covalent bonding in the PA network. Ligand mobility can be adjusted independently of the matrix mechanical properties by attaching the RGD cell adhesion ligand to either the covalent PEG network, the dynamic PA network, or both networks at once. Interestingly, our findings show that endothelial cell adhesion formation and spreading is maximized in soft, viscoelastic gels in which RGD adhesion ligands are present on both the covalent PEG and non-covalent PA networks. The dynamic nature of cell adhesion domains, coupled with their ability to exert substantial forces on the matrix, suggests that having different presentations of RGD ligands which are either mobile or are capable of withstanding significant forces are needed mimic different aspects of complex cell-matrix adhesions. By demonstrating how different presentations of RGD ligands affect cell behavior independently of viscoelastic properties, these results contribute to the rational design of hydrogels that facilitate desired cell-matrix interactions, with the potential of improving in vitro models and regenerative therapies.
Collapse
Affiliation(s)
| | - Katelyn Dunne
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| | - Wendy Breyer
- Department of Chemistry, Lehigh University, Bethlehem PA, USA, 18015
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| | - E Thomas Pashuck
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| |
Collapse
|
5
|
Liguori A, Zhao J, Di Gesù R, De Marco R, Gualandi C, Calonghi N, Pollicino A, Gentilucci L, Focarete ML. Peptide direct growth on poly(acrylic acid)/poly(vinyl alcohol) electrospun fibers coated with branched poly(ethylenimine): A solid-phase approach for scaffolds biofunctionalization. Colloids Surf B Biointerfaces 2024; 241:114052. [PMID: 38917667 DOI: 10.1016/j.colsurfb.2024.114052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024]
Abstract
Due to their resemblance to the fibrillar structure of the extracellular matrix, electrospun nanofibrous meshes are currently used as porous and mechanically stable scaffolds for cell culture. In this study, we propose an innovative methodology for growing peptide sequences directly onto the surface of electrospun nanofibers. To achieve this, electrospun fibers were produced from a poly(acrylic acid)/poly(vinyl alcohol) blend that was thermally crosslinked and subjected to a covalent coating of branched poly(ethylenimine). The exposed amino functionalities on the fiber surface were then used for the direct solid-phase synthesis of the RGD peptide sequence. In contrast to established strategies, mainly involving the grafting of pre-synthesized peptides onto the polymer chains before electrospinning or onto the nanofibers surface, this method allows for the concurrent synthesis and anchoring of peptides to the substrate, with potential applications in combinatorial chemistry. The incorporation of this integrin-binding motive significantly enhanced the nanofibers' ability to capture human cervical carcinoma (HeLa) cells, selected as a proof of concept to assess the functionalities of the developed material.
Collapse
Affiliation(s)
- Anna Liguori
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy
| | - Junwei Zhao
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy
| | - Roberto Di Gesù
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy; Ri.MED Foundation, Bandiera st. 11, Palermo 90133, Italy
| | - Rossella De Marco
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy
| | - Chiara Gualandi
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy; Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials Technology, CIRI-MAM, University of Bologna, Viale Risorgimento, 2, Bologna 40136, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna 40126, Italy
| | - Antonino Pollicino
- Department of Civil Engineering and Architecture, University of Catania, via S. Sofia 64, Catania 95125, Italy
| | - Luca Gentilucci
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy; Health Sciences & Technologies (HST) CIRI, University of Bologna, Via Tolara di Sopra 41/E, Ozzano Emilia Bologna 40064, Italy.
| | - Maria Letizia Focarete
- Department of Chemistry "G. Ciamician" and INSTM UdR of Bologna, University of Bologna, via Selmi 2, Bologna 40126, Italy; Health Sciences & Technologies (HST) CIRI, University of Bologna, Via Tolara di Sopra 41/E, Ozzano Emilia Bologna 40064, Italy.
| |
Collapse
|
6
|
Olov N, Nour S, Harris AR, Li D, Cook M, Williams RJ, Cheeseman S, Nisbet DR. Using Nanoscale Passports To Understand and Unlock Ion Channels as Gatekeepers of the Cell. ACS NANO 2024; 18:22709-22733. [PMID: 39136685 DOI: 10.1021/acsnano.4c05654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Natural ion channels are proteins embedded in the cell membrane that control many aspects of cell and human physiology by acting as gatekeepers, regulating the flow of ions in and out of cells. Advances in nanotechnology have influenced the methods for studying ion channels in vitro, as well as ways to unlock the delivery of therapeutics by modulating them in vivo. This review provides an overview of nanotechnology-enabled approaches for ion channel research with a focus on the synthesis and applications of synthetic ion channels. Further, the uses of nanotechnology for therapeutic applications are critically analyzed. Finally, we provide an outlook on the opportunities and challenges at the intersection of nanotechnology and ion channels. This work highlights the key role of nanoscale interactions in the operation and modulation of ion channels, which may prompt insights into nanotechnology-enabled mechanisms to study and exploit these systems in the near future.
Collapse
Affiliation(s)
- Nafiseh Olov
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - Shirin Nour
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Polymer Science Group, Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexander R Harris
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - Dan Li
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark Cook
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Department of Medicine, St Vincent's Hospital, Melbourne, Fitzroy, VIC 3065, Australia
| | - Richard J Williams
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Centre for Sustainable Bioproducts, Deakin University, Waurn Ponds, VIC 3217, Australia
- IMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC 3217, Australia
| | - Samuel Cheeseman
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| | - David R Nisbet
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- The Graeme Clark Institute, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC 3010, Melbourne, Australia
| |
Collapse
|
7
|
Luo W, Zhang H, Wan R, Cai Y, Liu Y, Wu Y, Yang Y, Chen J, Zhang D, Luo Z, Shang X. Biomaterials-Based Technologies in Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2024; 13:e2304196. [PMID: 38712598 DOI: 10.1002/adhm.202304196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
For many clinically prevalent severe injuries, the inherent regenerative capacity of skeletal muscle remains inadequate. Skeletal muscle tissue engineering (SMTE) seeks to meet this clinical demand. With continuous progress in biomedicine and related technologies including micro/nanotechnology and 3D printing, numerous studies have uncovered various intrinsic mechanisms regulating skeletal muscle regeneration and developed tailored biomaterial systems based on these understandings. Here, the skeletal muscle structure and regeneration process are discussed and the diverse biomaterial systems derived from various technologies are explored in detail. Biomaterials serve not merely as local niches for cell growth, but also as scaffolds endowed with structural or physicochemical properties that provide tissue regenerative cues such as topographical, electrical, and mechanical signals. They can also act as delivery systems for stem cells and bioactive molecules that have been shown as key participants in endogenous repair cascades. To achieve bench-to-bedside translation, the typical effect enabled by biomaterial systems and the potential underlying molecular mechanisms are also summarized. Insights into the roles of biomaterials in SMTE from cellular and molecular perspectives are provided. Finally, perspectives on the advancement of SMTE are provided, for which gene therapy, exosomes, and hybrid biomaterials may hold promise to make important contributions.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Hanli Zhang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Renwen Wan
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxi Cai
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yang Wu
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yimeng Yang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiani Chen
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, Hong Kong
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiliang Shang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| |
Collapse
|
8
|
Özcolak B, Erenay B, Odabaş S, Jandt KD, Garipcan B. Effects of bone surface topography and chemistry on macrophage polarization. Sci Rep 2024; 14:12721. [PMID: 38830871 PMCID: PMC11148019 DOI: 10.1038/s41598-024-62484-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Surface structure plays a crucial role in determining cell behavior on biomaterials, influencing cell adhesion, proliferation, differentiation, as well as immune cells and macrophage polarization. While grooves and ridges stimulate M2 polarization and pits and bumps promote M1 polarization, these structures do not accurately mimic the real bone surface. Consequently, the impact of mimicking bone surface topography on macrophage polarization remains unknown. Understanding the synergistic sequential roles of M1 and M2 macrophages in osteoimmunomodulation is crucial for effective bone tissue engineering. Thus, exploring the impact of bone surface microstructure mimicking biomaterials on macrophage polarization is critical. In this study, we aimed to sequentially activate M1 and M2 macrophages using Poly-L-Lactic acid (PLA) membranes with bone surface topographical features mimicked through the soft lithography technique. To mimic the bone surface topography, a bovine femur was used as a model surface, and the membranes were further modified with collagen type-I and hydroxyapatite to mimic the bone surface microenvironment. To determine the effect of these biomaterials on macrophage polarization, we conducted experimental analysis that contained estimating cytokine release profiles and characterizing cell morphology. Our results demonstrated the potential of the hydroxyapatite-deposited bone surface-mimicked PLA membranes to trigger sequential and synergistic M1 and M2 macrophage polarizations, suggesting their ability to achieve osteoimmunomodulatory macrophage polarization for bone tissue engineering applications. Although further experimental studies are required to completely investigate the osteoimmunomodulatory effects of these biomaterials, our results provide valuable insights into the potential advantages of biomaterials that mimic the complex microenvironment of bone surfaces.
Collapse
Affiliation(s)
- Birgün Özcolak
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Berkay Erenay
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
| | - Sedat Odabaş
- Biomaterials and Tissue Engineering Laboratory (bteLAB), Department of Chemistry, Faculty of Science, Ankara University, 06560, Ankara, Turkey
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, 06560, Ankara, Turkey
| | - Klaus D Jandt
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
| | - Bora Garipcan
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey.
| |
Collapse
|
9
|
Jia B, Huang H, Dong Z, Ren X, Lu Y, Wang W, Zhou S, Zhao X, Guo B. Degradable biomedical elastomers: paving the future of tissue repair and regenerative medicine. Chem Soc Rev 2024; 53:4086-4153. [PMID: 38465517 DOI: 10.1039/d3cs00923h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Degradable biomedical elastomers (DBE), characterized by controlled biodegradability, excellent biocompatibility, tailored elasticity, and favorable network design and processability, have become indispensable in tissue repair. This review critically examines the recent advances of biodegradable elastomers for tissue repair, focusing mainly on degradation mechanisms and evaluation, synthesis and crosslinking methods, microstructure design, processing techniques, and tissue repair applications. The review explores the material composition and cross-linking methods of elastomers used in tissue repair, addressing chemistry-related challenges and structural design considerations. In addition, this review focuses on the processing methods of two- and three-dimensional structures of elastomers, and systematically discusses the contribution of processing methods such as solvent casting, electrostatic spinning, and three-/four-dimensional printing of DBE. Furthermore, we describe recent advances in tissue repair using DBE, and include advances achieved in regenerating different tissues, including nerves, tendons, muscle, cardiac, and bone, highlighting their efficacy and versatility. The review concludes by discussing the current challenges in material selection, biodegradation, bioactivation, and manufacturing in tissue repair, and suggests future research directions. This concise yet comprehensive analysis aims to provide valuable insights and technical guidance for advances in DBE for tissue engineering.
Collapse
Affiliation(s)
- Ben Jia
- School of Civil Aviation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Heyuan Huang
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Zhicheng Dong
- School of Civil Aviation, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaoyang Ren
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Yanyan Lu
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Wenzhi Wang
- School of Aeronautics, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Shaowen Zhou
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Baolin Guo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
10
|
Mayer K, Ruhoff A, Chan NJ, Waterhouse A, O'Connor AJ, Scheibel T, Heath DE. REDV-Functionalized Recombinant Spider Silk for Next-Generation Coronary Artery Stent Coatings: Hemocompatible, Drug-Eluting, and Endothelial Cell-Specific Materials. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38470984 DOI: 10.1021/acsami.3c17861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Coronary artery stents are life-saving devices, and millions of these devices are implanted annually to treat coronary heart disease. The current gold standard in treatment is drug-eluting stents, which are coated with a biodegradable polymer layer that elutes antiproliferative drugs to prevent restenosis due to neointimal hyperplasia. Stenting is commonly paired with systemic antiplatelet therapy to prevent stent thrombosis. Despite their clinical success, current stents have significant limitations including inducing local inflammation that drives hyperplasia; a lack of hemocompatibility that promotes thrombosis, increasing need for antiplatelet therapy; and limited endothelialization, which is a critical step in the healing process. In this research, we designed a novel material for use as a next-generation coating for drug-eluting stents that addresses the limitations described above. Specifically, we developed a recombinant spider silk material that is functionalized with an REDV cell-adhesive ligand, a peptide motif that promotes specific adhesion of endothelial cells in the cardiovascular environment. We illustrated that this REDV-modified spider silk variant [eADF4(C16)-REDV] is an endothelial-cell-specific material that can promote the formation of a near-confluent endothelium. We additionally performed hemocompatibility assays using human whole blood and demonstrated that spider silk materials exhibit excellent hemocompatibility under both static and flow conditions. Furthermore, we showed that the material displayed slow enzyme-mediated degradation. Finally, we illustrated the ability to load and release the clinically relevant drug everolimus from recombinant spider silk coatings in a quantity and at a rate similar to that of commercial devices. These results support the use of REDV-functionalized recombinant spider silk as a coating for drug-eluting stents.
Collapse
Affiliation(s)
- Kai Mayer
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
- Chair for Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof. Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
| | - Alexander Ruhoff
- Heart Research Institute, The University of Sydney, Newtown, NSW 2042, Australia
| | - Nicholas J Chan
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Thomas Scheibel
- Chair for Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof. Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Materialzentrum (BayMat), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayrisches Polymerinstitut (BPI), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Daniel E Heath
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
11
|
Van Ryk D, Vimonpatranon S, Hiatt J, Ganesan S, Chen N, McMurry J, Garba S, Min S, Goes LR, Girard A, Yolitz J, Licavoli I, Wei D, Huang D, Soares MA, Martinelli E, Cicala C, Arthos J. The V2 domain of HIV gp120 mimics an interaction between CD4 and integrin ⍺4β7. PLoS Pathog 2023; 19:e1011860. [PMID: 38064524 PMCID: PMC10732398 DOI: 10.1371/journal.ppat.1011860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/20/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023] Open
Abstract
The CD4 receptor, by stabilizing TCR-MHC II interactions, plays a central role in adaptive immunity. It also serves as the HIV docking receptor. The HIV gp120 envelope protein binds directly to CD4. This interaction is a prerequisite for viral entry. gp120 also binds to ⍺4β7, an integrin that is expressed on a subset of memory CD4+ T cells. HIV tropisms for CD4+ T cells and gut tissues are central features of HIV pathogenesis. We report that CD4 binds directly to ⍺4β7 in a dynamic way, consistent with a cis regulatory interaction. The molecular details of this interaction are related to the way in which gp120 interacts with both receptors. Like MAdCAM-1 and VCAM-1, two recognized ligands of ⍺4β7, the binding interface on CD4 includes 2 sites (1° and accessory), distributed across its two N-terminal IgSF domains (D1 and D2). The 1° site includes a sequence in the G β-strand of CD4 D2, KIDIV, that binds directly to ⍺4β7. This pentapeptide sequence occurs infrequently in eukaryotic proteins. However, a closely related and conserved sequence, KLDIV, appears in the V2 domain of gp120. KLDIV mediates gp120-⍺4β7 binding. The accessory ⍺4β7 binding site on CD4 includes Phe43. The Phe43 aromatic ring protrudes outward from one edge of a loop connecting the C'C" strands of CD4 D1. Phe43 is a principal contact for HIV gp120. It interacts with conserved residues in the recessed CD4 binding pocket. Substitution of Phe43 abrogates CD4 binding to both gp120 and ⍺4β7. As such, the interactions of gp120 with both CD4 and ⍺4β7 reflect elements of their interactions with each other. These findings indicate that gp120 specificities for CD4 and ⍺4β7 are interrelated and suggest that selective pressures which produced a CD4 tropic virus that replicates in gut tissues are linked to a dynamic interaction between these two receptors.
Collapse
Affiliation(s)
- Donald Van Ryk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Sinmanus Vimonpatranon
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences–United States Component, Bangkok, Thailand
| | - Joe Hiatt
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Sundar Ganesan
- Biological Imaging Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Nathalie Chen
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Jordan McMurry
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Saadiq Garba
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Susie Min
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Livia R. Goes
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
- Oncovirology Program, Instituto Nacional de Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre Girard
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Jason Yolitz
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Isabella Licavoli
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Danlan Wei
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Dawei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Marcelo A. Soares
- Oncovirology Program, Instituto Nacional de Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Genetics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| |
Collapse
|
12
|
Majkowska A, Inostroza-Brito KE, Gonzalez M, Redondo-Gómez C, Rice A, Rodriguez-Cabello JC, Del Rio Hernandez AE, Mata A. Peptide-Protein Coassemblies into Hierarchical and Bioactive Tubular Membranes. Biomacromolecules 2023; 24:4419-4429. [PMID: 36696687 PMCID: PMC10565817 DOI: 10.1021/acs.biomac.2c01095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/16/2022] [Indexed: 01/26/2023]
Abstract
Multicomponent self-assembly offers opportunities for the design of complex and functional biomaterials with tunable properties. Here, we demonstrate how minor modifications in the molecular structures of peptide amphiphiles (PAs) and elastin-like recombinamers (ELs) can be used to generate coassembling tubular membranes with distinct structures, properties, and bioactivity. First, by introducing minor modifications in the charge density of PA molecules (PAK2, PAK3, PAK4), different diffusion-reaction processes can be triggered, resulting in distinct membrane microstructures. Second, by combining different types of these PAs prior to their coassembly with ELs, further modifications can be achieved, tuning the structures and properties of the tubular membranes. Finally, by introducing the cell adhesive peptide RGDS in either the PA or EL molecules, it is possible to harness the different diffusion-reaction processes to generate tubular membranes with distinct bioactivities. The study demonstrates the possibility to trigger and achieve minor but crucial differences in coassembling processes and tune material structure and bioactivity. The study demonstrates the possibility to use minor, yet crucial, differences in coassembling processes to tune material structure and bioactivity.
Collapse
Affiliation(s)
- Anna Majkowska
- William
Harvey Research Institute, Queen Mary University
of London, London EC1M 6BQ, U.K.
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Karla E. Inostroza-Brito
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Mariel Gonzalez
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Carlos Redondo-Gómez
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Alistair Rice
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
| | | | | | - Alvaro Mata
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
- School
of
Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K.
- Biodiscovery
Institute, University of Nottingham, Nottingham NG7 2RD, U.K.
- Department
of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, U.K.
| |
Collapse
|
13
|
Falcone N, Ermis M, Tamay DG, Mecwan M, Monirizad M, Mathes TG, Jucaud V, Choroomi A, de Barros NR, Zhu Y, Vrana NE, Kraatz HB, Kim HJ, Khademhosseini A. Peptide Hydrogels as Immunomaterials and Their Use in Cancer Immunotherapy Delivery. Adv Healthc Mater 2023; 12:e2301096. [PMID: 37256647 PMCID: PMC10615713 DOI: 10.1002/adhm.202301096] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Indexed: 06/01/2023]
Abstract
Peptide-based hydrogel biomaterials have emerged as an excellent strategy for immune system modulation. Peptide-based hydrogels are supramolecular materials that self-assemble into various nanostructures through various interactive forces (i.e., hydrogen bonding and hydrophobic interactions) and respond to microenvironmental stimuli (i.e., pH, temperature). While they have been reported in numerous biomedical applications, they have recently been deemed promising candidates to improve the efficacy of cancer immunotherapies and treatments. Immunotherapies seek to harness the body's immune system to preemptively protect against and treat various diseases, such as cancer. However, their low efficacy rates result in limited patient responses to treatment. Here, the immunomaterial's potential to improve these efficacy rates by either functioning as immune stimulators through direct immune system interactions and/or delivering a range of immune agents is highlighted. The chemical and physical properties of these peptide-based materials that lead to immuno modulation and how one may design a system to achieve desired immune responses in a controllable manner are discussed. Works in the literature that reports peptide hydrogels as adjuvant systems and for the delivery of immunotherapies are highlighted. Finally, the future trends and possible developments based on peptide hydrogels for cancer immunotherapy applications are discussed.
Collapse
Affiliation(s)
- Natashya Falcone
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
| | - Dilara Goksu Tamay
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, 06800, Turkey
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Tess Grett Mathes
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Nihal Engin Vrana
- SPARTHA Medical, CRBS 1 Rue Eugene Boeckel, Strasbourg, 67000, France
| | - Heinz-Bernhard Kraatz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON, M1C 1A4, Canada
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| |
Collapse
|
14
|
Almeida BR, Barros BCSC, Barros DTL, Orikaza CM, Suzuki E. Paracoccidioides brasiliensis Induces α3 Integrin Lysosomal Degradation in Lung Epithelial Cells. J Fungi (Basel) 2023; 9:912. [PMID: 37755020 PMCID: PMC10532483 DOI: 10.3390/jof9090912] [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: 08/14/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Studies on the pathogen-host interaction are crucial for the understanding of the mechanisms involved in the establishment, maintenance, and spread of infection. In recent years, our research group has observed that the P. brasiliensis species interact with integrin family receptors and increase the expression of α3 integrin in lung epithelial cells within 5 h of infection. Interestingly, α3 integrin levels were reduced by approximately 99% after 24 h of infection with P. brasiliensis compared to non-infected cells. In this work, we show that, during infection with this fungus, α3 integrin is increased in the late endosomes of A549 lung epithelial cells. We also observed that the inhibitor of the lysosomal activity bafilomycin A1 was able to inhibit the decrease in α3 integrin levels. In addition, the silencing of the charged multivesicular body protein 3 (CHMP3) inhibited the reduction in α3 integrin levels induced by P. brasiliensis in A549 cells. Thus, together, these results indicate that this fungus induces the degradation of α3 integrin in A549 lung epithelial cells by hijacking the host cell endolysosomal pathway.
Collapse
Affiliation(s)
| | | | | | | | - Erika Suzuki
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, Ed. Antonio C. M. Paiva, São Paulo 04023-062, SP, Brazil; (B.R.A.)
| |
Collapse
|
15
|
Zhang X, Karagöz Z, Swapnasrita S, Habibovic P, Carlier A, van Rijt S. Development of Mesoporous Silica Nanoparticle-Based Films with Tunable Arginine-Glycine-Aspartate Peptide Global Density and Clustering Levels to Study Stem Cell Adhesion and Differentiation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38171-38184. [PMID: 37527490 PMCID: PMC10436245 DOI: 10.1021/acsami.3c04249] [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: 03/24/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023]
Abstract
Stem cell adhesion is mediated via the binding of integrin receptors to adhesion motifs present in the extracellular matrix (ECM). The spatial organization of adhesion ligands plays an important role in stem cell integrin-mediated adhesion. In this study, we developed a series of biointerfaces using arginine-glycine-aspartate (RGD)-functionalized mesoporous silica nanoparticles (MSN-RGD) to study the effect of RGD adhesion ligand global density (ligand coverage over the surface), spacing, and RGD clustering levels on stem cell adhesion and differentiation. To prepare the biointerface, MSNs were chemically functionalized with RGD peptides via an antifouling poly(ethylene glycol) (PEG) linker. The RGD surface functionalization ratio could be controlled to create MSNs with high and low RGD ligand clustering levels. MSN films with varying RGD global densities could be created by blending different ratios of MSN-RGD and non-RGD-functionalized MSNs together. A computational simulation study was performed to analyze nanoparticle distribution and RGD spacing on the resulting surfaces to determine experimental conditions. Enhanced cell adhesion and spreading were observed when RGD global density increased from 1.06 to 5.32 nmol cm-2 using highly clustered RGD-MSN-based films. Higher RGD ligand clustering levels led to larger cell spreading and increased formation of focal adhesions. Moreover, a higher RGD ligand clustering level promoted the expression of alkaline phosphatase in hMSCs. Overall, these findings indicate that both RGD global density and clustering levels are crucial variables in regulating stem cell behaviors. This study provides important information about ligand-integrin interactions, which could be implemented into biomaterial design to achieve optimal performance of adhesive functional peptides.
Collapse
Affiliation(s)
- Xingzhen Zhang
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Zeynep Karagöz
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sangita Swapnasrita
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Pamela Habibovic
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Aurélie Carlier
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sabine van Rijt
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
16
|
Lv J, Wu Y, Cao Z, Liu X, Sun Y, Zhang P, Zhang X, Tang K, Cheng M, Yao Q, Zhu Y. Enhanced Cartilage and Subchondral Bone Repair Using Carbon Nanotube-Doped Peptide Hydrogel-Polycaprolactone Composite Scaffolds. Pharmaceutics 2023; 15:2145. [PMID: 37631359 PMCID: PMC10458387 DOI: 10.3390/pharmaceutics15082145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
A carbon nanotube-doped octapeptide self-assembled hydrogel (FEK/C) and a hydrogel-based polycaprolactone PCL composite scaffold (FEK/C3-S) were developed for cartilage and subchondral bone repair. The composite scaffold demonstrated modulated microstructure, mechanical properties, and conductivity by adjusting CNT concentration. In vitro evaluations showed enhanced cell proliferation, adhesion, and migration of articular cartilage cells, osteoblasts, and bone marrow mesenchymal stem cells. The composite scaffold exhibited good biocompatibility, low haemolysis rate, and high protein absorption capacity. It also promoted osteogenesis and chondrogenesis, with increased mineralization, alkaline phosphatase (ALP) activity, and glycosaminoglycan (GAG) secretion. The composite scaffold facilitated accelerated cartilage and subchondral bone regeneration in a rabbit knee joint defect model. Histological analysis revealed improved cartilage tissue formation and increased subchondral bone density. Notably, the FEK/C3-S composite scaffold exhibited the most significant cartilage and subchondral bone formation. The FEK/C3-S composite scaffold holds great promise for cartilage and subchondral bone repair. It offers enhanced mechanical support, conductivity, and bioactivity, leading to improved tissue regeneration. These findings contribute to the advancement of regenerative strategies for challenging musculoskeletal tissue defects.
Collapse
Affiliation(s)
- Jiayi Lv
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xu Liu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xin Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Kexin Tang
- College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Min Cheng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
17
|
Ge B, Wei M, Bao B, Pan Z, Elango J, Wu W. The Role of Integrin Receptor's α and β Subunits of Mouse Mesenchymal Stem Cells on the Interaction of Marine-Derived Blacktip Reef Shark ( Carcharhinus melanopterus) Skin Collagen. Int J Mol Sci 2023; 24:ijms24119110. [PMID: 37298062 DOI: 10.3390/ijms24119110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 06/12/2023] Open
Abstract
Marine collagen (MC) has recently attracted more attention in tissue engineering as a biomaterial substitute due to its significant role in cellular signaling mechanisms, especially in mesenchymal stem cells (MSCs). However, the actual signaling mechanism of MC in MSC growth, which is highly influenced by their molecular pattern, is poorly understood. Hence, we investigated the integrin receptors (α1β1, α2β1, α10β1, and α11β1) binding mechanism and proliferation of MCs (blacktip reef shark collagen (BSC) and blue shark collagen (SC)) compared to bovine collagen (BC) on MSCs behavior through functionalized collagen molecule probing for the first time. The results showed that BSC and SC had higher proliferation rates and accelerated scratch wound healing by increasing migratory rates of MSCs. Cell adhesion and spreading results demonstrated that MC had a better capacity to anchor MSCs and maintain cell morphology than controls. Living cell observations showed that BSC was gradually assembled by cells into the ECM network within 24 h. Interestingly, qRT-PCR and ELISA revealed that the proliferative effect of MC was triggered by interacting with specific integrin receptors such as α2β1, α10β1, and α11β1 of MSCs. Accordingly, BSC accelerated MSCs' growth, adhesion, shape, and spreading by interacting with specific integrin subunits (α2 and β1) and thereby triggering further signaling cascade mechanisms.
Collapse
Affiliation(s)
- Baolin Ge
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Mingjun Wei
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Bin Bao
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zhilin Pan
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Jeevithan Elango
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Católica San Antonio de Murcia, Guadalupe, 30107 Murcia, Spain
| | - Wenhui Wu
- Department of Marine Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
18
|
Xia X, Zhang G, Pica Ciamarra M, Jiao Y, Ni R. The Role of Receptor Uniformity in Multivalent Binding. JACS AU 2023; 3:1385-1391. [PMID: 37234107 PMCID: PMC10207130 DOI: 10.1021/jacsau.3c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/13/2023] [Accepted: 04/11/2023] [Indexed: 05/27/2023]
Abstract
Multivalency is prevalent in various biological systems and applications due to the superselectivity that arises from the cooperativity of multivalent binding. Traditionally, it was thought that weaker individual binding would improve the selectivity in multivalent targeting. Here, using analytical mean field theory and Monte Carlo simulations, we discover that, for receptors that are highly uniformly distributed, the highest selectivity occurs at an intermediate binding energy and can be significantly greater than the weak binding limit. This is caused by an exponential relationship between the bound fraction and receptor concentration, which is influenced by both the strength and combinatorial entropy of binding. Our findings not only provide new guidelines for the rational design of biosensors using multivalent nanoparticles but also introduce a new perspective in understanding biological processes involving multivalency.
Collapse
Affiliation(s)
- Xiuyang Xia
- School
of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Drive, 637459 Singapore
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Ge Zhang
- Department
of Physics, City University of Hong Kong, 518057 Kowloon, Hong Kong China
| | - Massimo Pica Ciamarra
- Division
of Physics and Applied Physics, School of Physical and Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, 637371 Singapore
| | - Yang Jiao
- Materials
Science and Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Ran Ni
- School
of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Drive, 637459 Singapore
| |
Collapse
|
19
|
Kuwar R, Wen X, Zhang N, Sun D. Integrin binding peptides facilitate growth and interconnected vascular-like network formation of rat primary cortical vascular endothelial cells in vitro. Neural Regen Res 2023; 18:1052-1056. [PMID: 36254992 PMCID: PMC9827785 DOI: 10.4103/1673-5374.355760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/28/2022] [Accepted: 07/11/2022] [Indexed: 11/07/2022] Open
Abstract
Neovascularization and angiogenesis in the brain are important physiological processes for normal brain development and repair/regeneration following insults. Integrins are cell surface adhesion receptors mediating important function of cells such as survival, growth and development during tissue organization, differentiation and organogenesis. In this study, we used an integrin-binding array platform to identify the important types of integrins and their binding peptides that facilitate adhesion, growth, development, and vascular-like network formation of rat primary brain microvascular endothelial cells. Brain microvascular endothelial cells were isolated from rat brain on post-natal day 7. Cells were cultured in a custom-designed integrin array system containing short synthetic peptides binding to 16 types of integrins commonly expressed on cells in vertebrates. After 7 days of culture, the brain microvascular endothelial cells were processed for immunostaining with markers for endothelial cells including von Willibrand factor and platelet endothelial cell adhesion molecule. 5-Bromo-2'-dexoyuridine was added to the culture at 48 hours prior to fixation to assess cell proliferation. Among 16 integrins tested, we found that α5β1, αvβ5 and αvβ8 greatly promoted proliferation of endothelial cells in culture. To investigate the effect of integrin-binding peptides in promoting neovascularization and angiogenesis, the binding peptides to the above three types of integrins were immobilized to our custom-designed hydrogel in three-dimensional (3D) culture of brain microvascular endothelial cells with the addition of vascular endothelial growth factor. Following a 7-day 3D culture, the culture was fixed and processed for double labeling of phalloidin with von Willibrand factor or platelet endothelial cell adhesion molecule and assessed under confocal microscopy. In the 3D culture in hydrogels conjugated with the integrin-binding peptide, brain microvascular endothelial cells formed interconnected vascular-like network with clearly discernable lumens, which is reminiscent of brain microvascular network in vivo. With the novel integrin-binding array system, we identified the specific types of integrins on brain microvascular endothelial cells that mediate cell adhesion and growth followed by functionalizing a 3D hydrogel culture system using the binding peptides that specifically bind to the identified integrins, leading to robust growth and lumenized microvascular-like network formation of brain microvascular endothelial cells in 3D culture. This technology can be used for in vitro and in vivo vascularization of transplants or brain lesions to promote brain tissue regeneration following neurological insults.
Collapse
Affiliation(s)
- Ram Kuwar
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Ning Zhang
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Dong Sun
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
20
|
Valencia-Gallardo C, Aguilar-Salvador DI, Khakzad H, Cocom-Chan B, Bou-Nader C, Velours C, Zarrouk Y, Le Clainche C, Malosse C, Lima DB, Quenech'Du N, Mazhar B, Essid S, Fontecave M, Asnacios A, Chamot-Rooke J, Malmström L, Tran Van Nhieu G. Shigella IpaA mediates actin bundling through diffusible vinculin oligomers with activation imprint. Cell Rep 2023; 42:112405. [PMID: 37071535 DOI: 10.1016/j.celrep.2023.112405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023] Open
Abstract
Upon activation, vinculin reinforces cytoskeletal anchorage during cell adhesion. Activating ligands classically disrupt intramolecular interactions between the vinculin head and tail domains that bind to actin filaments. Here, we show that Shigella IpaA triggers major allosteric changes in the head domain, leading to vinculin homo-oligomerization. Through the cooperative binding of its three vinculin-binding sites (VBSs), IpaA induces a striking reorientation of the D1 and D2 head subdomains associated with vinculin oligomerization. IpaA thus acts as a catalyst producing vinculin clusters that bundle actin at a distance from the activation site and trigger the formation of highly stable adhesions resisting the action of actin relaxing drugs. Unlike canonical activation, vinculin homo-oligomers induced by IpaA appear to keep a persistent imprint of the activated state in addition to their bundling activity, accounting for stable cell adhesion independent of force transduction and relevant to bacterial invasion.
Collapse
Affiliation(s)
- Cesar Valencia-Gallardo
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France
| | - Daniel-Isui Aguilar-Salvador
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France
| | - Hamed Khakzad
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France
| | - Benjamin Cocom-Chan
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France; Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198/INSERM U1280, Team "Ca(2+) Signaling and Microbial Infections," CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Charles Bou-Nader
- Laboratoire de Chimie des Processus Biologiques, Collège De France, CNRS UMR8229, 75005 Paris, France
| | - Christophe Velours
- Fundamental Microbiology and Pathogenicity Laboratory, UMR 5234 CNRS-University of Bordeaux, SFR TransBioMed, 33076 Bordeaux, France
| | - Yosra Zarrouk
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198/INSERM U1280, Team "Ca(2+) Signaling and Microbial Infections," CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Christophe Le Clainche
- Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198, Team "Cytoskeletal Dynamics and Motility", CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Christian Malosse
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, F-75015 Paris
| | - Diogo Borges Lima
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, F-75015 Paris
| | - Nicole Quenech'Du
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France
| | - Bilal Mazhar
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France
| | - Sami Essid
- Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France
| | - Marc Fontecave
- Laboratoire de Chimie des Processus Biologiques, Collège De France, CNRS UMR8229, 75005 Paris, France
| | - Atef Asnacios
- Université Paris Cité, CNRS, Laboratoire Matière et Systèmes Complexes, UMR7057, F-75013 Paris, France
| | - Julia Chamot-Rooke
- Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Mass Spectrometry for Biology Unit, F-75015 Paris
| | - Lars Malmström
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Guy Tran Van Nhieu
- Center for Interdisciplinary Research in Biology (CIRB), Team "Ca(2+) Signaling and Microbial Infections," Collège de France, CNRS UMR7241/INSERM U1050, PSL Research University, 75005 Paris, France; Laboratoire de biologie et Pharmacie Appliquée (LBPA), CNRS UMR8113/INSERM U1282, Team "Ca(2+) Signaling and Microbial Infections," Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, 91190 Gif-sur-Yvette, France; Institute for Integrative Biology of the Cell (I2BC), CNRS UMR9198/INSERM U1280, Team "Ca(2+) Signaling and Microbial Infections," CEA, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
21
|
Yang Q, Guo J, Zhang S, Guan F, Yu Y, Feng S, Yao Q, Bao D. Improved biomedical bioactivity of polyvinyl alcohol/polyethylene oxide composite system-based nanofiber membranes via incorporating Antarctic krill protein. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.111888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
22
|
Tvaroška I, Kozmon S, Kóňa J. Molecular Modeling Insights into the Structure and Behavior of Integrins: A Review. Cells 2023; 12:cells12020324. [PMID: 36672259 PMCID: PMC9856412 DOI: 10.3390/cells12020324] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Integrins are heterodimeric glycoproteins crucial to the physiology and pathology of many biological functions. As adhesion molecules, they mediate immune cell trafficking, migration, and immunological synapse formation during inflammation and cancer. The recognition of the vital roles of integrins in various diseases revealed their therapeutic potential. Despite the great effort in the last thirty years, up to now, only seven integrin-based drugs have entered the market. Recent progress in deciphering integrin functions, signaling, and interactions with ligands, along with advancement in rational drug design strategies, provide an opportunity to exploit their therapeutic potential and discover novel agents. This review will discuss the molecular modeling methods used in determining integrins' dynamic properties and in providing information toward understanding their properties and function at the atomic level. Then, we will survey the relevant contributions and the current understanding of integrin structure, activation, the binding of essential ligands, and the role of molecular modeling methods in the rational design of antagonists. We will emphasize the role played by molecular modeling methods in progress in these areas and the designing of integrin antagonists.
Collapse
Affiliation(s)
- Igor Tvaroška
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Correspondence:
| | - Stanislav Kozmon
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Medical Vision o. z., Záhradnícka 4837/55, 821 08 Bratislava, Slovakia
| | - Juraj Kóňa
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravska cesta 9, 845 38 Bratislava, Slovakia
- Medical Vision o. z., Záhradnícka 4837/55, 821 08 Bratislava, Slovakia
| |
Collapse
|
23
|
Cao D, Ding J. Recent advances in regenerative biomaterials. Regen Biomater 2022; 9:rbac098. [PMID: 36518879 PMCID: PMC9745784 DOI: 10.1093/rb/rbac098] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 07/22/2023] Open
Abstract
Nowadays, biomaterials have evolved from the inert supports or functional substitutes to the bioactive materials able to trigger or promote the regenerative potential of tissues. The interdisciplinary progress has broadened the definition of 'biomaterials', and a typical new insight is the concept of tissue induction biomaterials. The term 'regenerative biomaterials' and thus the contents of this article are relevant to yet beyond tissue induction biomaterials. This review summarizes the recent progress of medical materials including metals, ceramics, hydrogels, other polymers and bio-derived materials. As the application aspects are concerned, this article introduces regenerative biomaterials for bone and cartilage regeneration, cardiovascular repair, 3D bioprinting, wound healing and medical cosmetology. Cell-biomaterial interactions are highlighted. Since the global pandemic of coronavirus disease 2019, the review particularly mentions biomaterials for public health emergency. In the last section, perspectives are suggested: (i) creation of new materials is the source of innovation; (ii) modification of existing materials is an effective strategy for performance improvement; (iii) biomaterial degradation and tissue regeneration are required to be harmonious with each other; (iv) host responses can significantly influence the clinical outcomes; (v) the long-term outcomes should be paid more attention to; (vi) the noninvasive approaches for monitoring in vivo dynamic evolution are required to be developed; (vii) public health emergencies call for more research and development of biomaterials; and (viii) clinical translation needs to be pushed forward in a full-chain way. In the future, more new insights are expected to be shed into the brilliant field-regenerative biomaterials.
Collapse
Affiliation(s)
- Dinglingge Cao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
24
|
Merino-Casallo F, Gomez-Benito MJ, Hervas-Raluy S, Garcia-Aznar JM. Unravelling cell migration: defining movement from the cell surface. Cell Adh Migr 2022; 16:25-64. [PMID: 35499121 PMCID: PMC9067518 DOI: 10.1080/19336918.2022.2055520] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Cell motility is essential for life and development. Unfortunately, cell migration is also linked to several pathological processes, such as cancer metastasis. Cells' ability to migrate relies on many actors. Cells change their migratory strategy based on their phenotype and the properties of the surrounding microenvironment. Cell migration is, therefore, an extremely complex phenomenon. Researchers have investigated cell motility for more than a century. Recent discoveries have uncovered some of the mysteries associated with the mechanisms involved in cell migration, such as intracellular signaling and cell mechanics. These findings involve different players, including transmembrane receptors, adhesive complexes, cytoskeletal components , the nucleus, and the extracellular matrix. This review aims to give a global overview of our current understanding of cell migration.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Silvia Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
25
|
Krasilnikova OA, Baranovskii DS, Yakimova AO, Arguchinskaya N, Kisel A, Sosin D, Sulina Y, Ivanov SA, Shegay PV, Kaprin AD, Klabukov ID. Intraoperative Creation of Tissue-Engineered Grafts with Minimally Manipulated Cells: New Concept of Bone Tissue Engineering In Situ. Bioengineering (Basel) 2022; 9:704. [PMID: 36421105 PMCID: PMC9687730 DOI: 10.3390/bioengineering9110704] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 07/22/2023] Open
Abstract
Transfer of regenerative approaches into clinical practice is limited by strict legal regulation of in vitro expanded cells and risks associated with substantial manipulations. Isolation of cells for the enrichment of bone grafts directly in the Operating Room appears to be a promising solution for the translation of biomedical technologies into clinical practice. These intraoperative approaches could be generally characterized as a joint concept of tissue engineering in situ. Our review covers techniques of intraoperative cell isolation and seeding for the creation of tissue-engineered grafts in situ, that is, directly in the Operating Room. Up-to-date, the clinical use of tissue-engineered grafts created in vitro remains a highly inaccessible option. Fortunately, intraoperative tissue engineering in situ is already available for patients who need advanced treatment modalities.
Collapse
Affiliation(s)
- Olga A. Krasilnikova
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Denis S. Baranovskii
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Anna O. Yakimova
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Nadezhda Arguchinskaya
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Anastas Kisel
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Dmitry Sosin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, Pogodinskaya St. 10 Bld. 1, 119121 Moscow, Russia
| | - Yana Sulina
- Department of Obstetrics and Gynecology, Sechenov University, Bolshaya Pirogovskaya St. 2 Bld. 3, 119435 Moscow, Russia
| | - Sergey A. Ivanov
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Peter V. Shegay
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
| | - Ilya D. Klabukov
- Department of Regenerative Medicine, National Medical Research Radiological Center, Koroleva St. 4, 249036 Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering, National Research Nuclear University MEPhI, Studgorodok 1, 249039 Obninsk, Russia
| |
Collapse
|
26
|
Revete A, Aparicio A, Cisterna BA, Revete J, Luis L, Ibarra E, Segura González EA, Molino J, Reginensi D. Advancements in the Use of Hydrogels for Regenerative Medicine: Properties and Biomedical Applications. Int J Biomater 2022; 2022:3606765. [PMID: 36387956 PMCID: PMC9663251 DOI: 10.1155/2022/3606765] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/29/2022] [Accepted: 10/05/2022] [Indexed: 07/29/2023] Open
Abstract
Due to their particular water absorption capacity, hydrogels are the most widely used scaffolds in biomedical studies to regenerate damaged tissue. Hydrogels can be used in tissue engineering to design scaffolds for three-dimensional cell culture, providing a novel alternative to the traditional two-dimensional cell culture as hydrogels have a three-dimensional biomimetic structure. This material property is crucial in regenerative medicine, especially for the nervous system, since it is a highly complex and delicate structure. Hydrogels can move quickly within the human body without physically disturbing the environment and possess essential biocompatible properties, as well as the ability to form a mimetic scaffold in situ. Therefore, hydrogels are perfect candidates for biomedical applications. Hydrogels represent a potential alternative to regenerating tissue lost after removing a brain tumor and/or brain injuries. This reason presents them as an exciting alternative to highly complex human physiological problems, such as injuries to the central nervous system and neurodegenerative disease.
Collapse
Affiliation(s)
- Andrea Revete
- Biological Engineering, Faculty of Biosciences and Public Health, Universidad Especializada de las Americas (UDELAS), Panama City, Panama
- Biomedical Engineering, Faculty of Health Sciences and Engineering, Universidad Latina de Panama (ULATINA), Panama City, Panama
| | - Andrea Aparicio
- Biological Engineering, Faculty of Biosciences and Public Health, Universidad Especializada de las Americas (UDELAS), Panama City, Panama
| | - Bruno A. Cisterna
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Javier Revete
- Experimentia S.A, Development of Innovative Strategies in Biomedicine and Sustainable Development, Panama, Panama
| | - Luis Luis
- Experimentia S.A, Development of Innovative Strategies in Biomedicine and Sustainable Development, Panama, Panama
| | - Ernesto Ibarra
- Biomedical Engineering, Faculty of Health Sciences and Engineering, Universidad Latina de Panama (ULATINA), Panama City, Panama
| | | | - Jay Molino
- Biological Engineering, Faculty of Biosciences and Public Health, Universidad Especializada de las Americas (UDELAS), Panama City, Panama
| | - Diego Reginensi
- Biological Engineering, Faculty of Biosciences and Public Health, Universidad Especializada de las Americas (UDELAS), Panama City, Panama
- Biomedical Engineering, Faculty of Health Sciences and Engineering, Universidad Latina de Panama (ULATINA), Panama City, Panama
- Integrative Neurobiology, School of Medicine, Universidad de Panama (UP), Panama, Panama
- Center for Biodiversity and Drug Discovery, INDICASAT-AIP, City of Knowledge, Panama, Panama
| |
Collapse
|
27
|
Matrix Regeneration Ability In Situ Induced by a Silk Fibroin Small-Caliber Artificial Blood Vessel In Vivo. Polymers (Basel) 2022; 14:polym14183754. [PMID: 36145899 PMCID: PMC9502482 DOI: 10.3390/polym14183754] [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: 08/14/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
The success of a small-caliber artificial vascular graft in the host in order to obtain functional tissue regeneration and remodeling remains a great challenge in clinical application. In our previous work, a silk-based, small-caliber tubular scaffold (SFTS) showed excellent mechanical properties, long-term patency and rapid endothelialization capabilities. On this basis, the aim of the present study was to evaluate the vascular reconstruction process after implantation to replace the common carotid artery in rabbits. The new tissue on both sides of the SFTSs at 1 month was clearly observed. Inside the SFTSs, the extracellular matrix (ECM) was deposited on the pore wall at 1 month and continued to increase during the follow-up period. The self-assembled collagen fibers and elastic fibers were clearly visible in a circumferential arrangement at 6 months and were similar to autologous blood vessels. The positive expression rate of Lysyl oxidase-1 (LOXL-1) was positively correlated with the formation and maturity of collagen fibers and elastic fibers. In summary, the findings of the tissue regeneration processes indicated that the bionic SFTSs induced in situ angiogenesis in defects.
Collapse
|
28
|
Gao X, Xu J, Yao T, Liu X, Zhang H, Zhan C. Peptide-decorated nanocarriers penetrating the blood-brain barrier for imaging and therapy of brain diseases. Adv Drug Deliv Rev 2022; 187:114362. [PMID: 35654215 DOI: 10.1016/j.addr.2022.114362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Blood-Brain Barrier (BBB) is one of the most important physiological barriers strictly restricting the substance exchange between blood and brain tissues. While the BBB protects the brain from infections and toxins and maintains brain homeostasis, it is also recognized as the main obstacle to the penetration of therapeutics and imaging agents into the brain. Due to high specificity and affinity, peptides are frequently exploited to decorate nanocarriers across the BBB for diagnosis and/or therapy purposes. However, there are still some challenges that restrict their clinical application, such as stability, safety and immunocompatibility. In this review, we summarize the biological and pathophysiological characteristics of the BBB, strategies across the BBB, and recent progress on peptide decorated nanocarriers for brain diseases diagnosis and therapy. The challenges and opportunities for their translation are also discussed.
Collapse
|
29
|
Park HJ, Hong H, Thangam R, Song MG, Kim JE, Jo EH, Jang YJ, Choi WH, Lee MY, Kang H, Lee KB. Static and Dynamic Biomaterial Engineering for Cell Modulation. NANOMATERIALS 2022; 12:nano12081377. [PMID: 35458085 PMCID: PMC9028203 DOI: 10.3390/nano12081377] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/01/2023]
Abstract
In the biological microenvironment, cells are surrounded by an extracellular matrix (ECM), with which they dynamically interact during various biological processes. Specifically, the physical and chemical properties of the ECM work cooperatively to influence the behavior and fate of cells directly and indirectly, which invokes various physiological responses in the body. Hence, efficient strategies to modulate cellular responses for a specific purpose have become important for various scientific fields such as biology, pharmacy, and medicine. Among many approaches, the utilization of biomaterials has been studied the most because they can be meticulously engineered to mimic cellular modulatory behavior. For such careful engineering, studies on physical modulation (e.g., ECM topography, stiffness, and wettability) and chemical manipulation (e.g., composition and soluble and surface biosignals) have been actively conducted. At present, the scope of research is being shifted from static (considering only the initial environment and the effects of each element) to biomimetic dynamic (including the concepts of time and gradient) modulation in both physical and chemical manipulations. This review provides an overall perspective on how the static and dynamic biomaterials are actively engineered to modulate targeted cellular responses while highlighting the importance and advance from static modulation to biomimetic dynamic modulation for biomedical applications.
Collapse
Affiliation(s)
- Hyung-Joon Park
- Department of Interdisciplinary Biomicrosystem Technology, College of Engineering, Korea University, Seoul 02841, Korea;
| | - Hyunsik Hong
- Department of Materials Science and Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (H.H.); (R.T.)
| | - Ramar Thangam
- Department of Materials Science and Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (H.H.); (R.T.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
| | - Min-Gyo Song
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 02841, Korea; (M.-G.S.); (W.-H.C.); (M.-Y.L.)
| | - Ju-Eun Kim
- Department of Biomedical Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (J.-E.K.); (E.-H.J.)
| | - Eun-Hae Jo
- Department of Biomedical Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (J.-E.K.); (E.-H.J.)
| | - Yun-Jeong Jang
- Department of Biomedical Engineering, Armour College of Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA;
| | - Won-Hyoung Choi
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 02841, Korea; (M.-G.S.); (W.-H.C.); (M.-Y.L.)
| | - Min-Young Lee
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 02841, Korea; (M.-G.S.); (W.-H.C.); (M.-Y.L.)
| | - Heemin Kang
- Department of Interdisciplinary Biomicrosystem Technology, College of Engineering, Korea University, Seoul 02841, Korea;
- Department of Materials Science and Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (H.H.); (R.T.)
- Correspondence: (H.K.); (K.-B.L.)
| | - Kyu-Back Lee
- Department of Interdisciplinary Biomicrosystem Technology, College of Engineering, Korea University, Seoul 02841, Korea;
- Department of Biomedical Engineering, College of Health Science, Korea University, Seoul 02841, Korea; (M.-G.S.); (W.-H.C.); (M.-Y.L.)
- Department of Biomedical Engineering, College of Engineering, Korea University, Seoul 02841, Korea; (J.-E.K.); (E.-H.J.)
- Correspondence: (H.K.); (K.-B.L.)
| |
Collapse
|
30
|
Bandzerewicz A, Gadomska-Gajadhur A. Into the Tissues: Extracellular Matrix and Its Artificial Substitutes: Cell Signalling Mechanisms. Cells 2022; 11:914. [PMID: 35269536 PMCID: PMC8909573 DOI: 10.3390/cells11050914] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The existence of orderly structures, such as tissues and organs is made possible by cell adhesion, i.e., the process by which cells attach to neighbouring cells and a supporting substance in the form of the extracellular matrix. The extracellular matrix is a three-dimensional structure composed of collagens, elastin, and various proteoglycans and glycoproteins. It is a storehouse for multiple signalling factors. Cells are informed of their correct connection to the matrix via receptors. Tissue disruption often prevents the natural reconstitution of the matrix. The use of appropriate implants is then required. This review is a compilation of crucial information on the structural and functional features of the extracellular matrix and the complex mechanisms of cell-cell connectivity. The possibilities of regenerating damaged tissues using an artificial matrix substitute are described, detailing the host response to the implant. An important issue is the surface properties of such an implant and the possibilities of their modification.
Collapse
|
31
|
Chi Y, Chen Y, Jiang W, Huang W, Ouyang M, Liu L, Pan Y, Li J, Qu X, Liu H, Liu C, Deng L, Qin X, Xiang Y. Deficiency of Integrin β4 Results in Increased Lung Tissue Stiffness and Responds to Substrate Stiffness via Modulating RhoA Activity. Front Cell Dev Biol 2022; 10:845440. [PMID: 35309934 PMCID: PMC8926985 DOI: 10.3389/fcell.2022.845440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
The interaction between extracellular matrix (ECM) and epithelial cells plays a key role in lung development. Our studies found that mice with conditional integrin β4 (ITGB4) knockout presented lung dysplasia and increased stiffness of lung tissues. In accordance with our previous studies regarding the functions of ITGB4 in bronchial epithelial cells (BECs), we hypothesize that the decreased ITGB4 expression during embryonic stage leads to abnormal ECM remodeling and increased tissue stiffness, thus impairing BECs motility and compromising lung development. In this study, we examined lung tissue stiffness in normal and ITGB4 deficiency mice using Atomic Force Microscopy (AFM), and demonstrated that ITGB4 deficiency resulted in increased lung tissue stiffness. The examination of ECM components collagen, elastin, and lysyl oxidase (LOX) family showed that the expression of type VI collagen, elastin and LOXL4 were significantly elevated in the ITGB4-deficiency mice, compared with those in normal groups. Airway epithelial cell migration and proliferation capacities on normal and stiff substrates were evaluated through video-microscopy and flow cytometry. The morphology of the cytoskeleton was detected by laser confocal microscopy, and RhoA activities were determined by fluorescence resonance energy transfer (FRET) microscopy. The results showed that migration and proliferation of ITGB4 deficiency cells were noticeably inhibited, along decreased cytoskeleton stabilization, and hampered RhoA activity, especially for cells cultured on the stiff substrate. These results suggest that decreased ITGB4 expression results in increased lung tissue stiffness and impairs the adaptation of bronchial epithelial cells to substrate stiffness, which may be related to the occurrence of broncho pulmonary dysplasia.
Collapse
Affiliation(s)
- Yinxiu Chi
- School of Basic Medicine, Central South University, Changsha, China
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, China
- Longdong College, Qingyang, China
| | - Yu Chen
- School of Basic Medicine, Central South University, Changsha, China
| | - Wang Jiang
- School of Basic Medicine, Central South University, Changsha, China
| | - Wenjie Huang
- School of Basic Medicine, Central South University, Changsha, China
- Affiliated Liuzhou Maternity and Child Healthcare Hospital of Guangxi University of Science and Technology, Liuzhou, China
| | - Mingxing Ouyang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, China
| | - Lei Liu
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, China
| | - Yan Pan
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, China
| | - Jingjing Li
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, China
| | - Xiangping Qu
- School of Basic Medicine, Central South University, Changsha, China
| | - Huijun Liu
- School of Basic Medicine, Central South University, Changsha, China
| | - Chi Liu
- School of Basic Medicine, Central South University, Changsha, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, China
- *Correspondence: Linhong Deng, ; Xiaoqun Qin, ; Yang Xiang,
| | - Xiaoqun Qin
- School of Basic Medicine, Central South University, Changsha, China
- *Correspondence: Linhong Deng, ; Xiaoqun Qin, ; Yang Xiang,
| | - Yang Xiang
- School of Basic Medicine, Central South University, Changsha, China
- *Correspondence: Linhong Deng, ; Xiaoqun Qin, ; Yang Xiang,
| |
Collapse
|
32
|
Hybrid Surface Nanostructures Using Chemical Vapor Deposition and Colloidal Self-Assembled Patterns for Human Mesenchymal Stem Cell Culture—A Preliminary Study. COATINGS 2022. [DOI: 10.3390/coatings12030311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Surface coatings are critical in biomaterials and biomedical devices. Chemical vapor deposition (CVD) is a well-known technology for the generation of thin films on a surface. However, the granular structures produced using CVD are rare. Recently, we used PPX-C, an excellent insulating material, for granular structure coating using CVD. Colloidal self-assembly is also a well-established method to generate granular structures named colloidal self-assembled patterns (cSAPs). In this study, we combined these two technologies to generate hierarchical granular structures and tested the biophysical effect of these hybrid surfaces on human bone marrow mesenchymal stem cells (hBMSCs). Two CVD-derived granular structures were made using water or glycerin droplets (i.e., CVD or GlyCVD surfaces). Water drops generate porous particles, while glycerin drops generate core–shell particles on the surface. These particles were dispersed randomly on the surface with sizes ranging from 1 to 20 μm. These CVD surfaces were hydrophobic (WCA ~ 80–110 degrees). On the other hand, a binary colloidal crystal (BCC), one type of cSAPs, composed of 5 μm Si and 400 nm carboxylated polystyrene (PSC) particles, had a close-packed structure and a hydrophilic surface (WCA ~ 45 degrees). The hybrid surfaces (i.e., CVD-BCC and GlyCVD-BCC) were smooth (Ra ~ 1.1–1.5 μm) and hydrophilic (WCA ~ 50 degrees), indicating a large surface coverage of BCC dominating the surface property. The hybrid surfaces were expected to be slightly negatively charged due to naturally charged CVD particles and negatively charged BCC particles. Cell adhesion was reduced on the hybrid surfaces, leading to an aggregated cell morphology, without reducing cell activity, compared to the flat control after 5 days. qPCR analysis showed that gene expression of type II collagen (COL2) was highly expressed on the GlyCVD-BCC without chemical induction after 3 and 14 days compared to the flat control. This proof-of-concept study demonstrates the potential of combining two technologies to make hybrid structures that can modulate stem cell attachment and differentiation.
Collapse
|
33
|
Jeong W, Bu J, Jafari R, Rehak P, Kubiatowicz LJ, Drelich AJ, Owen RH, Nair A, Rawding PA, Poellmann MJ, Hopkins CM, Král P, Hong S. Hierarchically Multivalent Peptide-Nanoparticle Architectures: A Systematic Approach to Engineer Surface Adhesion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103098. [PMID: 34894089 PMCID: PMC8811846 DOI: 10.1002/advs.202103098] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/04/2021] [Indexed: 05/20/2023]
Abstract
The multivalent binding effect has been the subject of extensive studies to modulate adhesion behaviors of various biological and engineered systems. However, precise control over the strong avidity-based binding remains a significant challenge. Here, a set of engineering strategies are developed and tested to systematically enhance the multivalent binding of peptides in a stepwise manner. Poly(amidoamine) (PAMAM) dendrimers are employed to increase local peptide densities on a substrate, resulting in hierarchically multivalent architectures (HMAs) that display multivalent dendrimer-peptide conjugates (DPCs) with various configurations. To control binding behaviors, effects of the three major components of the HMAs are investigated: i) poly(ethylene glycol) (PEG) linkers as spacers between conjugated peptides; ii) multiple peptides on the DPCs; and iii) various surface arrangements of HMAs (i.e., a mixture of DPCs each containing different peptides vs DPCs cofunctionalized with multiple peptides). The optimized HMA configuration enables significantly enhanced target cell binding with high selectivity compared to the control surfaces directly conjugated with peptides. The engineering approaches presented herein can be applied individually or in combination, providing guidelines for the effective utilization of biomolecular multivalent interactions using DPC-based HMAs.
Collapse
Affiliation(s)
- Woo‐jin Jeong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
- Department of Biological Sciences and BioengineeringInha University100 Inha‐ro, Michuhol‐guIncheon22212Republic of Korea
| | - Jiyoon Bu
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Roya Jafari
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Pavel Rehak
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Luke J. Kubiatowicz
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Adam J. Drelich
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Randall H. Owen
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Ashita Nair
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Piper A. Rawding
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Michael J. Poellmann
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Caroline M. Hopkins
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Petr Král
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
- Departments of Physics, Pharmaceutical Sciences and Chemical EngineeringUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
- Department of Biomedical EngineeringThe University of Wisconsin‐Madison1550 Engineering Dr.MadisonWI53705USA
- Yonsei Frontier LabDepartment of PharmacyYonsei University50 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| |
Collapse
|
34
|
Cui T, Wu S, Wei Y, Qin H, Ren J, Qu X. A Topologically Engineered Gold Island for Programmed In Vivo Stem Cell Manipulation. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202113103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Tingting Cui
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology Changchun Institute of Applied Chemistry Chinese Academy of Science Changchun, Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Si Wu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology Changchun Institute of Applied Chemistry Chinese Academy of Science Changchun, Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Yue Wei
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology Changchun Institute of Applied Chemistry Chinese Academy of Science Changchun, Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Hongshuang Qin
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology Changchun Institute of Applied Chemistry Chinese Academy of Science Changchun, Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology Changchun Institute of Applied Chemistry Chinese Academy of Science Changchun, Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resource Utilization and Laboratory of Chemical Biology Changchun Institute of Applied Chemistry Chinese Academy of Science Changchun, Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 China
| |
Collapse
|
35
|
Critical parameters for design and development of multivalent nanoconstructs: recent trends. Drug Deliv Transl Res 2022; 12:2335-2358. [PMID: 35013982 PMCID: PMC8747862 DOI: 10.1007/s13346-021-01103-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
A century ago, the groundbreaking concept of the magic bullet was given by Paul Ehrlich. Since then, this concept has been extensively explored in various forms to date. The concept of multivalency is among such advancements of the magic bullet concept. Biologically, the concept of multivalency plays a critical role in significantly huge numbers of biochemical interactions. This concept is the sole reason behind the higher affinity of biological molecules like viruses to more selectively target the host cell surface receptors. Multivalent nanoconstructs are a promising approach for drug delivery by the active targeting principle. Designing and developing effective and target-specific multivalent drug delivery nanoconstructs, on the other hand, remain a challenge. The underlying reason for this is a lack of understanding of the crucial interactions between ligands and cell surface receptors, as well as the design of nanoconstructs. This review highlights the need for a better theoretical understanding of the multivalent effect of what happens to the receptor-ligand complex after it has been established. Furthermore, the critical parameters for designing and developing robust multivalent systems have been emphasized. We have also discussed current advances in the design and development of multivalent nanoconstructs for drug delivery. We believe that a thorough knowledge of theoretical concepts and experimental methodologies may transform a brilliant idea into clinical translation.
Collapse
|
36
|
Ladeira B, Custodio C, Mano J. Core-Shell Microcapsules: Biofabrication and Potential Applications in Tissue Engineering and Regenerative Medicine. Biomater Sci 2022; 10:2122-2153. [DOI: 10.1039/d1bm01974k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The construction of biomaterial scaffolds that accurately recreate the architecture of living tissues in vitro is a major challenge in the field of tissue engineering and regenerative medicine. Core-shell microcapsules...
Collapse
|
37
|
Hu X, Xia Z, Cai K. Recent advances of 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B 2022; 10:1486-1507. [DOI: 10.1039/d1tb02537f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly recognized as resources for disease treatments and regenerative medicine. Meanwhile, the unique chemical and physical properties of hydrogels provide innate advantages to achieve...
Collapse
|
38
|
Qu X, Cui T, Wu S, Wei Y, Qin H, Ren J. A Topologically Engineered Gold Island for Programmed In Vivo Stem Cell Manipulation. Angew Chem Int Ed Engl 2021; 61:e202113103. [PMID: 34939267 DOI: 10.1002/anie.202113103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Indexed: 11/06/2022]
Abstract
E ven a well-designed system can only control stem cell adhesion, release, and differentiation, while other cell manipulations such as in situ labeling and retention in target tissues, are difficult to achieve in the same system. Herein, native ligand cluster-mimicking islands, composed of topologically engineered ligand, anchoring point AuNP, nuclease mimetics Ce IV complexes and magnetic core Fe 3 O 4 , are designed to facilitate comprehensive cell manipulations in a programmable manner. Three islands with different amounts of AuNPs are constructed, which means tunable interligand spacing within a cluster. These nanostructures are chemically coupled to a substrate using DNA tethers. Under tissue-penetrative magnetic field, this integrated system promotes stem cell adhesion, proliferation, mechanosensing, differentiation, detachment, in situ effective magnetic labeling and retention both in vitro and in vivo , offering fascinating opportunities for biomimetic matrix in regenerative medicine.
Collapse
Affiliation(s)
- Xiaogang Qu
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Laboratory of Chemical Biology, Division of Biological Inorganic Chemistry, 5625 Renmin Street, 130022, Changchun, CHINA
| | - Tingting Cui
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences: Chang Chun Institute of Applied Chemistry Chinese Academy of Sciences, State Key Laboratory of Rare Earth Resource Utilization, CHINA
| | - Si Wu
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences: Chang Chun Institute of Applied Chemistry Chinese Academy of Sciences, State Key Laboratory of Rare Earth Resource Utilization, CHINA
| | - Yue Wei
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences: Chang Chun Institute of Applied Chemistry Chinese Academy of Sciences, State Key Laboratory of Rare Earth Resource Utilization, CHINA
| | - Hongshuang Qin
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences: Chang Chun Institute of Applied Chemistry Chinese Academy of Sciences, State Key Laboratory of Rare Earth Resource Utilization, CHINA
| | - Jinsong Ren
- Changchun Institute of Applied Chemistry Chinese Academy of Sciences: Chang Chun Institute of Applied Chemistry Chinese Academy of Sciences, State Key Laboratory of Rare Earth Resource Utilization, Remnin Street #5625, 130022, Changchun, CHINA
| |
Collapse
|
39
|
Siepi M, Oliva R, Masino A, Gaglione R, Arciello A, Russo R, Di Maro A, Zanfardino A, Varcamonti M, Petraccone L, Del Vecchio P, Merola M, Pizzo E, Notomista E, Cafaro V. Environment-Sensitive Fluorescent Labelling of Peptides by Luciferin Analogues. Int J Mol Sci 2021; 22:ijms222413312. [PMID: 34948103 PMCID: PMC8706149 DOI: 10.3390/ijms222413312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Environment-sensitive fluorophores are very valuable tools in the study of molecular and cellular processes. When used to label proteins and peptides, they allow for the monitoring of even small variations in the local microenvironment, thus acting as reporters of conformational variations and binding events. Luciferin and aminoluciferin, well known substrates of firefly luciferase, are environment-sensitive fluorophores with unusual and still-unexploited properties. Both fluorophores show strong solvatochromism. Moreover, luciferin fluorescence is influenced by pH and water abundance. These features allow to detect local variations of pH, solvent polarity and local water concentration, even when they occur simultaneously, by analyzing excitation and emission spectra. Here, we describe the characterization of (amino)luciferin-labeled derivatives of four bioactive peptides: the antimicrobial peptides GKY20 and ApoBL, the antitumor peptide p53pAnt and the integrin-binding peptide RGD. The two probes allowed for the study of the interaction of the peptides with model membranes, SDS micelles, lipopolysaccharide micelles and Escherichia coli cells. Kd values and binding stoichiometries for lipopolysaccharide were also determined. Aminoluciferin also proved to be very well-suited to confocal laser scanning microscopy. Overall, the characterization of the labeled peptides demonstrates that luciferin and aminoluciferin are previously neglected environment-sensitive labels with widespread potential applications in the study of proteins and peptides.
Collapse
Affiliation(s)
- Marialuisa Siepi
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| | - Rosario Oliva
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; (R.O.); (R.G.); (A.A.); (L.P.); (P.D.V.)
| | - Antonio Masino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| | - Rosa Gaglione
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; (R.O.); (R.G.); (A.A.); (L.P.); (P.D.V.)
| | - Angela Arciello
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; (R.O.); (R.G.); (A.A.); (L.P.); (P.D.V.)
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (R.R.); (A.D.M.)
| | - Antimo Di Maro
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (R.R.); (A.D.M.)
| | - Anna Zanfardino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| | - Mario Varcamonti
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| | - Luigi Petraccone
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; (R.O.); (R.G.); (A.A.); (L.P.); (P.D.V.)
| | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; (R.O.); (R.G.); (A.A.); (L.P.); (P.D.V.)
| | - Marcello Merola
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| | - Elio Pizzo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| | - Eugenio Notomista
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
- Correspondence:
| | - Valeria Cafaro
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (M.S.); (A.M.); (A.Z.); (M.V.); (M.M.); (E.P.); (V.C.)
| |
Collapse
|
40
|
Hu X, Zhang Y. Developing biomaterials to mediate the spatial distribution of integrins. BIOPHYSICS REVIEWS 2021; 2:041302. [PMID: 38504718 PMCID: PMC10903404 DOI: 10.1063/5.0055746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 10/21/2021] [Indexed: 03/21/2024]
Abstract
Innovation in material design to regulate cell behavior and function is one of the primary tasks in materials science. Integrins, a family of cell surface-adhesion receptors that mechanically connect the extracellular matrix (ECM) to the intracellular cytoskeleton, have long served as primary targets for the design of biomaterials because their activity is not only critical to a wide range of cell and tissue functions but also subject to very tight and complex regulations from the outside environment. To review the recent progress of material innovations targeting the spatial distribution of integrins, we first introduce the interaction mechanisms between cells and the ECM by highlighting integrin-based cell adhesions, describing how integrins respond to environmental stimuli, including variations in ligand presentation, mechanical cues, and topographical variations. Then, we overview the current development of soft materials in guiding cell behaviors and functions via spatial regulation of integrins. Finally, we discuss the current limitations of these technologies and the advances that may be achieved in the future. Undoubtedly, synthetic soft materials that mediate the spatial distribution of integrins play an important role in biomaterial innovations for advancing biomedical applications and addressing fundamental biological questions.
Collapse
Affiliation(s)
- Xunwu Hu
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Ye Zhang
- Bioinspired Soft Matter Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| |
Collapse
|
41
|
Mu R, Zhang Y, Yan L, Liao Z, Yang Y, Su H, Dong L, Wang C. A "Bridge-Building" Glycan Scaffold Mimicking Microbial Invasion for In Situ Endothelialization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103490. [PMID: 34476850 DOI: 10.1002/adma.202103490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/21/2021] [Indexed: 06/13/2023]
Abstract
The globally high prevalence of peripheral artery diseases poses a pressing need for biomaterials grafts to rebuild vasculature. When implanted, they should promote endothelial cells (ECs) adhesion both profoundly and selectively-but the latter expectation remains unfulfilled. Here, this work is inspired by fungi that invade blood vessels via the "bridge" of galectins that, secreted by ECs, can simultaneously bind carbohydrates on fungal surface and integrin receptors on ECs. A glucomannan decanoate (GMDE) substrate mimicking fungal carbohydrates that highly and preferentially supports ECs adhesion while rejecting several other cell types is designed. Electrospun GMDE scaffolds efficiently sequester endogenous galectin-1-which bridges ECs to the scaffolds as it functions in fungal invasions-and promote blood perfusion in a murine limb ischemic model. Meanwhile, the application of GMDE requires no exogenous pro-angiogenic agents and causes no organ toxicity or adverse inflammation in mice, highlighting its high safety of potential translation. This glycan material, uniquely mimicking a microbial action and harnessing a secreted protein as a "bridge," represents an effective, safe, and different strategy for ischemic vascular therapy.
Collapse
Affiliation(s)
- Ruoyu Mu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Yuhan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Zhencheng Liao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Yushun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
42
|
Doron G, Temenoff JS. Culture Substrates for Improved Manufacture of Mesenchymal Stromal Cell Therapies. Adv Healthc Mater 2021; 10:e2100016. [PMID: 33930252 DOI: 10.1002/adhm.202100016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Recent developments in mesenchymal stromal cell (MSC) therapies have increased the demand for tools to improve their manufacture, including the selection of optimal culture substrate materials. While many clinical manufacturers use planar tissue culture plastic (TCP) surfaces for MSC production, others have begun exploring the use of alternative culture substrates that present a variety of spatial, mechanical, and biochemical cues that influence cell expansion and resulting cell quality. In this review, the effects of culture and material properties distinct from traditional planar TCP surfaces on MSC proliferation, surface marker expression, and commonly used indications for therapeutic potency are examined. The different properties summarized include the use of alternative culture formats such as cellular aggregates or 3D scaffolds, as well as the effects of culture substrate stiffness and presentation of specific adhesive ligands and topographical cues. Specific substrate properties can be related to greater cell expansion and improvement in specific therapeutic functionalities, demonstrating the utility of culture materials in further improving the clinical-scale manufacture of highly secretory MSC products.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University 313 Ferst Drive Atlanta GA 30332 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology Atlanta GA 30332 USA
| | - Johnna S. Temenoff
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University 313 Ferst Drive Atlanta GA 30332 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology Atlanta GA 30332 USA
| |
Collapse
|
43
|
Jeong SH, Kim M, Kim TY, Choi H, Hahn SK. Biomimetic Supramolecular Drug Delivery Hydrogels for Accelerated Skin Tissue Regeneration. ACS Biomater Sci Eng 2021; 7:4581-4590. [PMID: 34254791 DOI: 10.1021/acsbiomaterials.1c00705] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Skin tissue is regenerated by the combinational function of skin cells, extracellular matrix (ECM), and bioactive molecules. As an artificial ECM, supramolecular hydrogels exhibited outstanding capability to mimic the physical properties of ECM. However, the lack of biochemical function in supramolecular hydrogels has limited further tissue engineering applications. Here, we developed self-assembling supramolecular drug delivery hydrogels to mimic the skin tissue regeneration process. The supramolecular hydrogels were prepared to encapsulate fibroblasts by the host-guest interaction of cyclodextrin-modified gelatin (GE-CD) and adamantane-modified hyaluronate (Ad-HA) in conjugation with human growth hormone (hGH) for accelerated skin tissue regeneration. In vitro, GE-CD/Ad-HA-hGH hydrogels showed highly facilitated cell growth by the controlled hGH delivery. After a subcutaneous injection into the back of mice, IVIS imaging of bioengineered fibroblasts to express red fluorescence protein (RFP) revealed prolonged cell survival and proliferation in the supramolecular hydrogels for more than 21 days. We could also observe the improved skin tissue regeneration by the facilitated fibroblast proliferation with angiogenesis. Taken together, we could confirm the feasibility of biomimetic supramolecular drug delivery GE-CD/Ad-HA-hGH hydrogels for various tissue engineering applications.
Collapse
Affiliation(s)
- Sang Hoon Jeong
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk 790-784, Korea
| | - Mungu Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk 790-784, Korea.,PHI Biomed Co., 175 Yeoksam-ro, Gangnam-gu, Seoul 06247, South Korea
| | - Tae Yeon Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk 790-784, Korea
| | - Hyunsik Choi
- PHI Biomed Co., 175 Yeoksam-ro, Gangnam-gu, Seoul 06247, South Korea
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk 790-784, Korea.,PHI Biomed Co., 175 Yeoksam-ro, Gangnam-gu, Seoul 06247, South Korea
| |
Collapse
|
44
|
Chen X, Yang X, Yuan P, Jin R, Bao L, Qiu X, Liu S, Liu T, Gooding JJ, Chen W, Liu G, Bai Y, Liu S, Jin Y. Modular immune-homeostatic microparticles promote immune tolerance in mouse autoimmune models. Sci Transl Med 2021; 13:13/584/eaaw9668. [PMID: 33692135 DOI: 10.1126/scitranslmed.aaw9668] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/14/2020] [Accepted: 01/26/2021] [Indexed: 12/29/2022]
Abstract
The therapeutic goal for autoimmune diseases is disease antigen-specific immune tolerance without nonspecific immune suppression. However, it is a challenge to induce antigen-specific immune tolerance in a dysregulated immune system. In this study, we developed immune-homeostatic microparticles (IHMs) that treat multiple mouse models of autoimmunity via induction of apoptosis in activated T cells and reestablishment of regulatory T cells. Specifically, in an experimental model of colitis, IHMs rapidly released monocyte chemotactic protein-1 after intravenous administration, which recruited activated T cells and then induced their apoptosis by conjugated Fas ligand on the IHM surface. This triggered professional macrophages to ingest apoptotic T cells and produce high quantities of transforming growth factor-β, which drove regulatory T cell differentiation. Furthermore, the modular design of IHMs allowed IHMs to be engineered with the autoantigen peptides that can reduce disease in an experimental autoimmune encephalomyelitis mouse model and a nonobese diabetic mouse model. This was accomplished by sustained release of the autoantigens after induction of T cell apoptosis and transforming growth factor-β production by macrophages, which promoted to establish an immune tolerant environment. Thus, IHMs may be an efficient therapeutic strategy for autoimmune diseases through induction of apoptosis and reestablishment of tolerant immune responses.
Collapse
Affiliation(s)
- Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China.
| | - Xiaoshan Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Pingyun Yuan
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Ronghua Jin
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Lili Bao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xinyu Qiu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Siying Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Tao Liu
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - John Justin Gooding
- School of Chemistry, Australian Centre for NanoMedicine and ARC Australian Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney 2052, Australia
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guozhen Liu
- Graduate School of Biomedical Engineering, ARC Centre of Excellence for Nanoscale BioPhotonics and Australian Centre for NanoMedicine, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
| | - Yongkang Bai
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
45
|
Lopes SV, Collins MN, Reis RL, Oliveira JM, Silva-Correia J. Vascularization Approaches in Tissue Engineering: Recent Developments on Evaluation Tests and Modulation. ACS APPLIED BIO MATERIALS 2021; 4:2941-2956. [DOI: 10.1021/acsabm.1c00051] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Soraia V. Lopes
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maurice N. Collins
- Bernal Institute, School of Engineering, University of Limerick, Limerick V94 T9PX, Ireland
| | - Rui L. Reis
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B’s Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães 4805-017, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
46
|
Tronolone JJ, Jain A. Engineering new microvascular networks on-chip: ingredients, assembly, and best practices. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007199. [PMID: 33994903 PMCID: PMC8114943 DOI: 10.1002/adfm.202007199] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 05/23/2023]
Abstract
Tissue engineered grafts show great potential as regenerative implants for diseased or injured tissues within the human body. However, these grafts suffer from poor nutrient perfusion and waste transport, thus decreasing their viability post-transplantation. Graft vascularization is therefore a major area of focus within tissue engineering because biologically relevant conduits for nutrient and oxygen perfusion can improve viability post-implantation. Many researchers utilize microphysiological systems as testing platforms for potential grafts due to an ability to integrate vascular networks as well as biological characteristics such as fluid perfusion, 3D architecture, compartmentalization of tissue-specific materials, and biophysical and biochemical cues. While many methods of vascularizing these systems exist, microvascular self-assembly has great potential for bench-to-clinic translation as it relies on naturally occurring physiological events. In this review, we highlight the past decade of literature and critically discuss the most important and tunable components yielding a self-assembled vascular network on chip: endothelial cell source, tissue-specific supporting cells, biomaterial scaffolds, biochemical cues, and biophysical forces. This article discusses the bioengineered systems of angiogenesis, vasculogenesis, and lymphangiogenesis, and includes a brief overview of multicellular systems. We conclude with future avenues of research to guide the next generation of vascularized microfluidic models and future tissue engineered grafts.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Abhishek Jain
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77808, USA
| |
Collapse
|
47
|
Jia Q, Xu B, Zhang Y, Ali A, Liao X. CCN Family Proteins in Cancer: Insight Into Their Structures and Coordination Role in Tumor Microenvironment. Front Genet 2021; 12:649387. [PMID: 33833779 PMCID: PMC8021874 DOI: 10.3389/fgene.2021.649387] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
The crosstalk between tumor cells and the tumor microenvironment (TME), triggers a variety of critical signaling pathways and promotes the malignant progression of cancer. The success rate of cancer therapy through targeting single molecule of this crosstalk may be extremely low, whereas co-targeting multiple components could be complicated design and likely to have more side effects. The six members of cellular communication network (CCN) family proteins are scaffolding proteins that may govern the TME, and several studies have shown targeted therapy of CCN family proteins may be effective for the treatment of cancer. CCN protein family shares similar structures, and they mutually reinforce and neutralize each other to serve various roles that are tightly regulated in a spatiotemporal manner by the TME. Here, we review the current knowledge on the structures and roles of CCN proteins in different types of cancer. We also analyze CCN mRNA expression, and reasons for its diverse relationship to prognosis in different cancers. In this review, we conclude that the discrepant functions of CCN proteins in different types of cancer are attributed to diverse TME and CCN truncated isoforms, and speculate that targeting CCN proteins to rebalance the TME could be a potent anti-cancer strategy.
Collapse
Affiliation(s)
- Qingan Jia
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Binghui Xu
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Yaoyao Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Arshad Ali
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xia Liao
- Department of Nutrition, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
48
|
Dynamics and Physics of Integrin Activation in Tumor Cells by Nano-Sized Extracellular Ligands and Electromagnetic Fields. Methods Mol Biol 2021; 2217:197-233. [PMID: 33215383 DOI: 10.1007/978-1-0716-0962-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrins are stress-sensing proteins expressed on the surface of cells. They regulate bidirectional signal transduction during cell-cell or cell-extracellular matrix (ECM) contacts. Integrins link the ECM with the cytoplasm through interaction with their ligands. Biophysically, such interactions can be understood as changes in stress fields at specific integrin stress-sensing domains, such as the MIDAS and ADMIDAS domains. Stress changes between ligands and cytoskeletal structures are involved in cancer cell growth by altering signal transduction pathways dependent on integrin activation. In this chapter, previous results regarding integrin activation and tumor cell growth using nanoparticles (NPs) of different materials, sizes and shapes are placed within a framework of polarized NPs in the ECM by external electromagnetic fields, in which the synergic action between polarized NPs and electromagnetic fields activates the integrins. Small size NPs activate integrins via the polar component of the dipole force between NPs and integrin sensing stress sites, while large size NPs exercise a similar action via the radial component. A quantum electrodynamic model also accounts for ECM overstressing by electromagnetic mode trapping between coherent symmetric and antisymmetric quantum states.
Collapse
|
49
|
Karagöz Z, Rijns L, Dankers PY, van Griensven M, Carlier A. Towards understanding the messengers of extracellular space: Computational models of outside-in integrin reaction networks. Comput Struct Biotechnol J 2020; 19:303-314. [PMID: 33425258 PMCID: PMC7779863 DOI: 10.1016/j.csbj.2020.12.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The interactions between cells and their extracellular matrix (ECM) are critically important for homeostatic control of cell growth, proliferation, differentiation and apoptosis. Transmembrane integrin molecules facilitate the communication between ECM and the cell. Since the characterization of integrins in the late 1980s, there has been great advancement in understanding the function of integrins at different subcellular levels. However, the versatility in molecular pathways integrins are involved in, the high diversity in their interaction partners both outside and inside the cell as well as on the cell membrane and the short lifetime of events happening at the cell-ECM interface make it difficult to elucidate all the details regarding integrin function experimentally. To overcome the experimental challenges and advance the understanding of integrin biology, computational modeling tools have been used extensively. In this review, we summarize the computational models of integrin signaling while we explain the function of integrins at three main subcellular levels (outside the cell, cell membrane, cytosol). We also discuss how these computational modeling efforts can be helpful in other disciplines such as biomaterial design. As such, this review is a didactic modeling summary for biomaterial researchers interested in complementing their experimental work with computational tools or for seasoned computational scientists that would like to advance current in silico integrin models.
Collapse
Affiliation(s)
- Zeynep Karagöz
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Laura Rijns
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, the Netherlands
| | - Patricia Y.W. Dankers
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, the Netherlands
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
50
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|